[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
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 (2,602)

Search Parameters:
Keywords = model-based drug development

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
14 pages, 3899 KiB  
Article
Development and Application of an In Vitro Drug Screening Assay for Schistosoma mansoni Schistosomula Using YOLOv5
by María Alejandra Villamizar-Monsalve, Javier Sánchez-Montejo, Julio López-Abán, Belén Vicente, Miguel Marín, Noelia Fernández-Ceballos, Rafael Peláez and Antonio Muro
Biomedicines 2024, 12(12), 2894; https://doi.org/10.3390/biomedicines12122894 - 19 Dec 2024
Viewed by 103
Abstract
Background: Schistosomiasis impacts over 230 million people globally, with 251.4 million needing treatment. The disease causes intestinal and urinary symptoms, such as hepatic fibrosis, hepatomegaly, splenomegaly, and bladder calcifications. While praziquantel (PZQ) is the primary treatment, its effectiveness against juvenile stages (schistosomula) is [...] Read more.
Background: Schistosomiasis impacts over 230 million people globally, with 251.4 million needing treatment. The disease causes intestinal and urinary symptoms, such as hepatic fibrosis, hepatomegaly, splenomegaly, and bladder calcifications. While praziquantel (PZQ) is the primary treatment, its effectiveness against juvenile stages (schistosomula) is limited, highlighting the need for new therapeutic agents, repurposed drugs, or reformulated compounds. Existing microscopy methods for assessing schistosomula viability are labor-intensive, subjective, and time-consuming. Methods: An artificial intelligence (AI)-assisted culture system using YOLOv5 was developed to evaluate compounds against Schistosoma mansoni schistosomula. The AI model, based on object detection, was trained on 4390 images distinguishing between healthy and damaged schistosomula. The system was externally validated against human counters, and a small-scale assay was performed to demonstrate its potential for larger-scale assays in the future. Results: The AI model exhibited high accuracy, achieving a mean average precision (mAP) of 0.966 (96.6%) and effectively differentiating between healthy and damaged schistosomula. External validation demonstrated significantly improved accuracy and counting time compared to human counters. A small-scale assay was conducted to validate the system, identifying 28 potential compounds with schistosomicidal activity against schistosomula in vitro and providing their preliminary LC50 values. Conclusions: This AI-powered method significantly improves accuracy and time efficiency compared to traditional microscopy. It enables the evaluation of compounds for potential schistosomiasis drugs without the need for dyes or specialized equipment, facilitating more efficient drug assessment. Full article
Show Figures

Figure 1

Figure 1
<p>Morphology of healthy and damaged schistosomula in culture. (<b>A</b>) Healthy schistosomula with well-defined shape, refringent appearance, and structured internal body. (<b>B<sub>1</sub></b>–<b>B<sub>3</sub></b>) Predominantly damaged schistosomula displaying an irregular shape, granularity, and dark color, indicating tegmental and internal damage.</p>
Full article ">Figure 2
<p>(<b>A</b>) Methods flowchart. Symbols: oval (start or end), rectangle (process), parallelogram (input/output), diamond (decision). (<b>B</b>) AxiWorm minikit. (<b>C</b>) RoboFlow software class annotation: damaged (red), healthy (green) (<b>D</b>) YOLOv5 model’s output predictions (0: damage, 1: healthy).</p>
Full article ">Figure 3
<p>Performance evaluation metrics for schistosomula detection models. (<b>A</b>) Comparative performance evaluation of eight models. (<b>B</b>) mAP 0.5 score for Model 8. (<b>C</b>) Precision metric for Model 8. (<b>D</b>) Recall metric for Model 8.</p>
Full article ">Figure 4
<p>Detection of schistosomula using YOLOv5 showing different morphologies under different conditions after 72 h treatment: damaged (red), healthy (green): (<b>A</b>) Healthy schistosomula. (<b>B</b>) Schistosomula with high granularity. (<b>C</b>) Mixed healthy and damaged schistosomula. (<b>D</b>) Arrows indicating untransformed cercaria and tails are excluded. (<b>E</b>) Schistosomula with high contrast. (<b>F</b>) Predominantly damaged schistosomula with evident deformation.</p>
Full article ">Figure 5
<p>Bland–Altman model evaluation (YOLOv5 vs. manual counting) at different confidence values. (<b>A</b>) Total count difference at 0.4 confidence threshold. (<b>B</b>) Viability percentage difference at 0.4 confidence threshold. (<b>C</b>) YOLOv5 prediction at 0.4 confidence threshold. Damaged schistosomula are inside red boxes. (<b>D</b>) Total count difference at 0.8 confidence threshold. (<b>E</b>) Viability percentage difference at 0.8 confidence threshold. (<b>F</b>) YOLOv5 prediction at 0.8 confidence threshold. Damaged schistosomula are inside red boxes All differences were calculated = (YOLOv5 counting − human counting). Means are shown in black dot lines and standard deviations shown in red dot lines.</p>
Full article ">Figure 6
<p>Differences between YOLOv5 and manual counting by counters who were not involved in the model training. (<b>A</b>) Absolute manual count difference (between three counts). (<b>B</b>) Difference in counts: Bland–Altman plot, means are in black dot line and standard deviations are in red dot lines.</p>
Full article ">Figure 7
<p>Percentage of inhibition of the anti-tubulin compound library at 72 h. A &gt; 60% inhibition threshold corresponds to moderate activity, while a 100% threshold corresponds to positive activity, marked in a dot line.</p>
Full article ">Figure 8
<p>Ten preliminary LC<sub>50</sub> values of active compounds identified through AI-assisted screening assay.</p>
Full article ">
15 pages, 7714 KiB  
Article
Gemcitabine-Loaded Microbeads for Transarterial Chemoembolization of Rabbit Renal Tumor Monitored by 18F-FDG Positron Emission Tomography/X-Ray Computed Tomography Imaging
by Xiaoli Zhang, Tingting Li, Jindong Tong, Meihong Zhou, Zi Wang, Xingdang Liu, Wei Lu, Jingjing Lou and Qingtong Yi
Pharmaceutics 2024, 16(12), 1609; https://doi.org/10.3390/pharmaceutics16121609 - 17 Dec 2024
Viewed by 489
Abstract
Background/Objectives: The purpose of this study was to develop the gemcitabine-loaded drug-eluting beads (G-DEBs) for transarterial chemoembolization (TACE) in rabbit renal tumors and to evaluate their antitumor effect using 2-deoxy-2-[(18)F]fluoro-D-glucose positron emission tomography/X-ray computed tomography (18F-FDG PET/CT). Methods: DEBs were prepared [...] Read more.
Background/Objectives: The purpose of this study was to develop the gemcitabine-loaded drug-eluting beads (G-DEBs) for transarterial chemoembolization (TACE) in rabbit renal tumors and to evaluate their antitumor effect using 2-deoxy-2-[(18)F]fluoro-D-glucose positron emission tomography/X-ray computed tomography (18F-FDG PET/CT). Methods: DEBs were prepared by polyvinyl alcohol-based macromer crosslinked with N-acryl tyrosine and N,N′-methylenebis(acrylamide). Gemcitabine was loaded through ion change to obtain G-DEBs. Their particle size and drug release profile were characterized. VX2 tumors were implanted in the right kidney of rabbits to establish the renal tumor model. The tumor-bearing rabbits received pre-scan by 18F-FDG PET/CT, followed by targeted transarterial injection of G-DEBs under digital subtraction angiography (DSA) guidance. The rabbits received another 18F-FDG PET/CT scan 10 or 14 days after the treatment. The therapeutic effect was further validated by histopathological analysis of the dissected tumors. Results: The average particle size of the microspheres was 58.06 ± 0.50 µm, and the polydisperse index was 0.26 ± 0.002. The maximum loading rate of G-DEBs was 18.09 ± 0.35%, with almost 100% encapsulation efficiency. Within 24 h, GEM was eluted from G-DEBs rapidly and completely, and more than 20% was released in different media. DSA illustrated that G-DEBs were delivered to rabbit renal tumors. Compared with the untreated control group with increased tumor volume and intense 18F -FDG uptake, the G-DEBs group showed significant reductions in tumor volume and maximum standard uptake value (SUVmax) 10 or 14 days after the treatment. Histopathological analysis confirmed that the proliferating area of tumor cells was significantly reduced in the G-DEBs group. Conclusions: Our results demonstrated that G-DEBs are effective in TACE treatment of rabbit VX2 renal tumors, and 18F-FDG PET/CT provides a non-invasive imaging modality to monitor the antitumor effects of TACE in renal tumors. Full article
(This article belongs to the Section Drug Targeting and Design)
Show Figures

Figure 1

Figure 1
<p>Characterization of DEBs. Size distribution (<b>A</b>) and scanning electron micrograph (SEM) (<b>B</b>) of DEBs. Size distribution (<b>C</b>) and SEM (<b>D</b>) of G-DEBs. (<b>E</b>) SEM of the cross-section of G-DEBs. Bar, 10 μm.</p>
Full article ">Figure 2
<p>Drug loading and release profiles of DEBs. (<b>A</b>) Gemcitabine (GEM) loaded by G-DEBs over time at different mass ratios of DEBs to GEM. Data are presented as mean ± SD (<span class="html-italic">n</span> = 3). (<b>B</b>) GEM encapsulation (green) or loading (red) efficiency of G-DEBs over different mass ratios. Data are presented as mean ± SD (<span class="html-italic">n</span> = 3). (<b>C</b>) Percentage of the cumulatively eluted GEM from G-DEBs over time in pH 7.4 PBS. Data are presented as mean ± SD (<span class="html-italic">n</span> = 3). (<b>D</b>) The cumulative drug release of G-DEBs in pH 6.5 and pH 7.4 PBS with and without 10% FBS. Data are presented as mean ± SD (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 3
<p>The viability of VX2 tumor cells incubated with GEM and G-DEBs at different concentrations for 24 h (<b>A</b>) and 72 h (<b>B</b>). Data are presented as mean ± SD (<span class="html-italic">n</span> = 3). ** <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 between the compared groups. Two-way ANOVA with Tukey’s post hoc test was used for statistical significance.</p>
Full article ">Figure 4
<p>Embolization of rabbit renal tumor with G-DEBs under the guidance of DSA. (<b>A</b>,<b>B</b>) CT image (<b>A</b>) and the corresponding <sup>18</sup>F-FDG PET/CT image (<b>B</b>) of rabbit bearing orthotopic VX2 renal tumor. White arrow, two pieces of micro-guide wires adjacent to the VX2 tumor tissue implanted. Green arrow, the VX2 tumor with positive signals of <sup>18</sup>F-FDG. Blue arrow, renal pelvis. (<b>C</b>,<b>D</b>) DSA imaging of VX2 renal tumor before (<b>C</b>) and after (<b>D</b>) intraarterial infusion of G-DEBs. White dotted circles, tumor. (<b>E</b>,<b>F</b>) Microscopic images of tumor (<b>E</b>) and adjacent kidney tissue (<b>F</b>) stained with H&amp;E one day after the embolization. Black arrows, G-DEBs. Bar, 20 µm.</p>
Full article ">Figure 5
<p>Antitumor effect following G-DEB embolization monitored by <sup>18</sup>F-FDG PET/CT imaging. (<b>A</b>) <sup>18</sup>F-FDG PET/CT images of rabbits bearing VX2 renal tumor before (Pre) and 10 or 14 days after transarterial infusion of DEBs (TAE) and G-DEBs (TACE) and without treatment (Control) (<span class="html-italic">n</span> = 3). Green arrows, tumor. (<b>B</b>) SUV<sub>max</sub> values of <sup>18</sup>F-FDG in tumors before (0) and 10 or 14 days after different treatments in (<b>A</b>). --, no <sup>18</sup>F FDG PET/CT imaging acquisition. (<b>C</b>) Photographs of the resected tumors of each group 14 days following the treatment in (<b>A</b>). Bar, 1 cm. Green dotted circles, tumor.</p>
Full article ">Figure 6
<p>Histological analysis of antitumor effect following G-DEB embolization. (<b>A</b>–<b>C</b>) Micrographs of H&amp;E staining, TUNEL, and Ki67 immunofluorescence of the renal tumors after transarterial infusion of (<b>A</b>) DEBs and (<b>B</b>) G-DEBs (TACE) and (<b>C</b>) without treatment. In H&amp;E images, the tumor regions are separated from the normal kidney tissues by green dotted lines. T, tumor. NT, necrotic tumor. The box area of tumors in the images is enlarged and presented on the right. In immunofluorescence images, the tumor regions are separated from the normal kidney tissues by white dotted circles.</p>
Full article ">Figure 7
<p>Micrographs of the major organs of rabbits with H&amp;E staining. (<b>A</b>) Representative micrographs of H&amp;E staining of the major organs 14 days after transarterial infusion of DEBs (TAE) and G-DEBs (TACE) and without treatment (Control). (<b>B</b>) Microscopic images of the lung tissue stained with H&amp;E after the embolization of DEB or G-DEBs.</p>
Full article ">
37 pages, 4688 KiB  
Review
Cell-Based Glioma Models for Anticancer Drug Screening: From Conventional Adherent Cell Cultures to Tumor-Specific Three-Dimensional Constructs
by Daria Lanskikh, Olga Kuziakova, Ivan Baklanov, Alina Penkova, Veronica Doroshenko, Ivan Buriak, Valeriia Zhmenia and Vadim Kumeiko
Cells 2024, 13(24), 2085; https://doi.org/10.3390/cells13242085 - 17 Dec 2024
Viewed by 344
Abstract
Gliomas are a group of primary brain tumors characterized by their aggressive nature and resistance to treatment. Infiltration of surrounding normal tissues limits surgical approaches, wide inter- and intratumor heterogeneity hinders the development of universal therapeutics, and the presence of the blood–brain barrier [...] Read more.
Gliomas are a group of primary brain tumors characterized by their aggressive nature and resistance to treatment. Infiltration of surrounding normal tissues limits surgical approaches, wide inter- and intratumor heterogeneity hinders the development of universal therapeutics, and the presence of the blood–brain barrier reduces the efficiency of their delivery. As a result, patients diagnosed with gliomas often face a poor prognosis and low survival rates. The spectrum of anti-glioma drugs used in clinical practice is quite narrow. Alkylating agents are often used as first-line therapy, but their effectiveness varies depending on the molecular subtypes of gliomas. This highlights the need for new, more effective therapeutic approaches. Standard drug-screening methods involve the use of two-dimensional cell cultures. However, these models cannot fully replicate the conditions present in real tumors, making it difficult to extrapolate the results to humans. We describe the advantages and disadvantages of existing glioma cell-based models designed to improve the situation and build future prospects to make drug discovery comprehensive and more effective for each patient according to personalized therapy paradigms. Full article
Show Figures

Figure 1

Figure 1
<p>Key molecular pathways, onco-associated molecules, and the microenvironment involved in gliomagenesis as prospective targets for glioma therapy that should be modeled in vitro.</p>
Full article ">Figure 2
<p>Application of the patient-derived glioma cell-based models.</p>
Full article ">Figure 3
<p>Approaches employed to create genetically modified glioma cell-based models and their subsequent applications. The left section shows the cell types most frequently utilized for modification, while the right section illustrates the various genetic and epigenetic modifications.</p>
Full article ">Figure 4
<p>Approaches for creating 3D cell-based glioma models. The left side of the figure shows the main cell sources for scaffold-free and scaffold-based models, while the right side shows the scaffold options for scaffold-based models.</p>
Full article ">
14 pages, 567 KiB  
Article
Defining “High-In” Saturated Fat, Sugar, and Sodium to Help Inform Front-of-Pack Labeling Efforts for Packaged Foods and Beverages in the United States
by Elizabeth K. Dunford, Donna R. Miles, Bridget A. Hollingsworth, Samantha Heller, Barry M. Popkin, Shu Wen Ng and Lindsey Smith Taillie
Nutrients 2024, 16(24), 4345; https://doi.org/10.3390/nu16244345 - 17 Dec 2024
Viewed by 386
Abstract
Background: To help consumers make healthier choices, the US Food and Drug Administration (FDA) has been charged with developing a front-of-package label (FOPL) to appear on US packaged foods and beverages. One option being explored is the use of “high-in” FOPLs for [...] Read more.
Background: To help consumers make healthier choices, the US Food and Drug Administration (FDA) has been charged with developing a front-of-package label (FOPL) to appear on US packaged foods and beverages. One option being explored is the use of “high-in” FOPLs for added sugar, sodium, and saturated fat using a threshold of ≥20% of the recommended daily value (%DV) per portion/serving size to define “high-in”. While research has addressed what FOPL designs are most effective at visually communicating “high-in”, less attention has been paid to the nutrient profile model (NPM) used to decide which products should receive these labels. In addition, several established regional NPMs already exist that identify products that are high in nutrients of concern, but it is unclear how these compare to the FDA’s %DV approach. Methods: We used a dataset of 51,809 US products from Mintel’s Global New Products Database to examine how the FDA’s current definition of “high-in” compares to three established regional NPMs: the Canadian NPM, the Pan American Health Organization (PAHO) NPM, and Chile’s NPM. Results: Overall agreement between the four NPMs was 51% for foods and 72% for beverages, with highest agreement in categories such as sweetened sodas (87%), and lowest agreement in categories such as bread (14%) and salty snacks (29%). The Canadian NPM showed the highest agreement to the FDA “high-in” criteria while the Chilean and PAHO models had lower agreement. For many food categories, the FDA’s definition of “high-in” would require the fewest products to carry a “high-in” label. This issue was particularly pronounced in categories that tend to be served in small portions (e.g., salty snacks, bars), but disappeared or reversed for categories that are served in larger portions (e.g., frozen and non-frozen main dishes). Conclusions: The NPM chosen has important policy implications for an FOPL system’s ability to identify unhealthy foods and incentivize companies to reformulate products. Based on these results, the FDA should consider using a stronger NPM similar to those used elsewhere in the Americas region when deciding the final thresholds for “high-in” for US packaged foods and beverages. Full article
(This article belongs to the Section Carbohydrates)
Show Figures

Figure 1

Figure 1
<p>Percent of US food products that would be considered ‘high in’ added sugar, saturated fat, and/or sodium among four nutrient profile models (NPMs) based on % Daily Value (DV), Chile, or the Pan American Health Organization (PAHO) using data from Mintel USA (2019–2023; <span class="html-italic">n</span> = 47,503 food products). “Any ‘high in’” refers to products that meet criteria for one or more nutrients (i.e., added sugar, saturated fat, sodium); “1 ‘high in’”, “2 ‘high in’”, or “3 ‘high in’” refers to the number of nutrients a product meets criteria for high in added sugar, saturated fat, and/or sodium.</p>
Full article ">Figure 2
<p>Percent of US beverage products that would be considered ‘high in’ added sugar, saturated fat, and/or sodium among four nutrient profile models (NPMs) based on % Daily Value (DV), Chile, or the Pan American Health Organization (PAHO) using data from Mintel USA (2019–2023; <span class="html-italic">n</span> = 4306 beverage products). “Any ‘high in’” refers to products that meet criteria for one or more nutrients (i.e., added sugar, saturated fat, sodium); “1 ‘high in’”, “2 ‘high in’”, or “3 ‘high in’” refers to the number of nutrients a product meets criteria for high in added sugar, saturated fat, and/or sodium.</p>
Full article ">
14 pages, 2279 KiB  
Article
Evaluation of the Drug–Drug Interaction Potential of Cannabidiol Against UGT2B7-Mediated Morphine Metabolism Using Physiologically Based Pharmacokinetic Modeling
by Shelby Coates, Keti Bardhi, Bhagwat Prasad and Philip Lazarus
Pharmaceutics 2024, 16(12), 1599; https://doi.org/10.3390/pharmaceutics16121599 - 16 Dec 2024
Viewed by 330
Abstract
Background: Morphine is a commonly prescribed opioid analgesic used to treat chronic pain. Morphine undergoes glucuronidation by UDP-glucuronosyltransferase (UGT) 2B7 to form morphine-3-glucuronide and morphine-6-glucuronide. Morphine is the gold standard for chronic pain management and has a narrow therapeutic index. Reports have shown [...] Read more.
Background: Morphine is a commonly prescribed opioid analgesic used to treat chronic pain. Morphine undergoes glucuronidation by UDP-glucuronosyltransferase (UGT) 2B7 to form morphine-3-glucuronide and morphine-6-glucuronide. Morphine is the gold standard for chronic pain management and has a narrow therapeutic index. Reports have shown that chronic pain patients have increasingly used other supplements to treat their chronic pain, including cannabidiol (CBD). Up to 50% of chronic pain patients report that they co-use cannabis with their prescribed opioid for pain management, including morphine. Previous work has shown that cannabidiol is a potent inhibitor of UGT2B7, including morphine-mediated metabolism. Co-use of morphine and CBD may result in unwanted drug–drug interactions (DDIs). Methods: Using available physiochemical and clinical parameters, morphine and CBD physiologically based pharmacokinetic (PBPK) models were developed and validated in both healthy and cirrhotic populations. Models for the two populations were then combined to predict the severity and clinical relevance of the potential DDIs during coadministration of both morphine and CBD in both healthy and hepatic-impaired virtual populations. Results: The predictive DDI model suggests that a ~5% increase in morphine exposure is to be expected in healthy populations. A similar increase in exposure of morphine is predicted in severe hepatic-impaired populations with an increase of ~10. Conclusions: While these predicted increases in morphine exposure are below the Food and Drug Administration’s cutoff (1.25-fold increase), morphine has a narrow therapeutic index and a 5–10% increase in exposure may be clinically relevant. Future clinical studies are needed to fully characterize the clinical relevance of morphine-related DDIs. Full article
Show Figures

Figure 1

Figure 1
<p>PBPK model of predicted and observed morphine plasma pharmacokinetic profiles after (<b>A</b>) IV (Lotsch et al., 2002) [<a href="#B41-pharmaceutics-16-01599" class="html-bibr">41</a>], (<b>B</b>) IR oral (Hoskin et al., 1989) [<a href="#B42-pharmaceutics-16-01599" class="html-bibr">42</a>], and (<b>C</b>) CR (Kotb et al., 2005) [<a href="#B43-pharmaceutics-16-01599" class="html-bibr">43</a>] oral administration in healthy adults and (<b>D</b>) IV (Hasselstrom et al., 1990) [<a href="#B44-pharmaceutics-16-01599" class="html-bibr">44</a>], (<b>E</b>) IR oral (Hasselstrom et al., 1990) [<a href="#B44-pharmaceutics-16-01599" class="html-bibr">44</a>], and (<b>F</b>) CR oral (Kotb et al., 2005) [<a href="#B43-pharmaceutics-16-01599" class="html-bibr">43</a>] administration in adults with cirrhosis. Data points are the observed values, and the green line represents the model’s predicted morphine plasma concentration–time profile. Gray lines indicate the 5th–95th percentile of the virtual population.</p>
Full article ">Figure 2
<p>PBPK model of predicted and observed CBD plasma pharmacokinetic profiles after (<b>A</b>) oral administration in healthy adults (Taylor et al., 2018) [<a href="#B45-pharmaceutics-16-01599" class="html-bibr">45</a>] and (<b>B</b>) oral administration in adults with cirrhosis (Taylor et al., 2019) [<a href="#B26-pharmaceutics-16-01599" class="html-bibr">26</a>]. Data points are the observed values, and the green line represents the model-predicted morphine plasma concentration–time profiles. Gray lines indicate the 5th–95th percentile of the virtual population.</p>
Full article ">Figure 3
<p>PBPK model of predicted and observed CBD plasma pharmacokinetic profiles after oral administration of 1500 mg twice daily for 7 days in healthy adults plotted on a (<b>A</b>) non-log, and (<b>B</b>) log scale. Data points are the observed values, and green the line represents the model-predicted CBD plasma concentration–time profile. Gray lines indicate the 5th–95th percentile of the virtual population.</p>
Full article ">Figure 4
<p>Brain tissue concentrations of morphine after oral administration of (<b>A</b>) oral solution (11.7 mg) in healthy adults, (<b>B</b>) oral solution (11.7 mg) in adults with severe hepatic impairment, (<b>C</b>) immediate-release tablet (15.2 mg) in healthy adults, (<b>D</b>) immediate-release tablet (15.2 mg) in adults with severe hepatic impairment, (<b>E</b>) controlled-release tablet (22.6 mg) in healthy adults, and (<b>F</b>) controlled-release tablet (22.6 mg) in adults with severe hepatic impairment. Shown are the mean brain morphine concentrations with (black dashed line or without (green line) cannabidiol over time.</p>
Full article ">Figure 5
<p>Morphine intrinsic clearance (CL<sub>int</sub>) after CBD administration (1500 mg twice daily for 7 days) with 15.2 mg of immediate-release morphine in (<b>A</b>) healthy and (<b>B</b>) hepatically impaired adults. The red and black lines represent morphine CL<sub>int</sub> with and without the presence of CBD, respectively. There was a 7.6, 21.8, and 4.8% decrease in hepatic, small intestine, and renal CL<sub>int</sub>, respectively, in healthy adults, and a 17.2, 19.6, and 21.4% decrease in hepatic, small intestine, and renal CL<sub>int</sub>, respectively, in adults with cirrhosis.</p>
Full article ">
27 pages, 9670 KiB  
Article
Application of Microsponge Drug Platform to Enhance Methotrexate Administration in Rheumatoid Arthritis Therapy
by Noemi Fiaschini, Patrizia Nadia Hanieh, Daniela Ariaudo, Rita Cimino, Carlo Abbate, Elena Romano, Francesca Cavalieri, Mariano Venanzi, Valeria Palumbo, Manuel Scimeca, Roberta Bernardini, Maurizio Mattei, Alberto Migliore and Antonio Rinaldi
Pharmaceutics 2024, 16(12), 1593; https://doi.org/10.3390/pharmaceutics16121593 - 13 Dec 2024
Viewed by 418
Abstract
Background/Objectives: This study aimed to develop a novel nanotechnological slow-release drug delivery platform based on hyaluronic acid Microsponge (MSP) for the subcutaneous administration of methotrexate (MTX) in the treatment of rheumatoid arthritis (RA). RA is a chronic autoimmune disease characterized by joint inflammation [...] Read more.
Background/Objectives: This study aimed to develop a novel nanotechnological slow-release drug delivery platform based on hyaluronic acid Microsponge (MSP) for the subcutaneous administration of methotrexate (MTX) in the treatment of rheumatoid arthritis (RA). RA is a chronic autoimmune disease characterized by joint inflammation and damage, while MTX is a common disease-modifying antirheumatic drug (DMARD), the conventional use of which is limited by adverse effects and the lack of release control. Methods: MSP were synthesized as freeze-dried powder to increase their stability and allow for a facile reconstitution prior to administration and precise MTX dosing. Results: A highly stable and rounded-shaped micrometric MSP, characterized by an open porosity inner structure, achieved both a high MTX loading efficiency and a slow release of MTX after injection. Our drug release assays indeed demonstrated a characteristic drug release profile consisting of a very limited burst release in the first few hours, followed by a slow release of MTX sustained for over a month. By means of a preclinical rat model of RA, the administration of MTX-loaded MSP proved to nearly double the therapeutic efficacy compared to sole MTX, according to a steep reduction in arthritic score compared to control groups. The preclinical study was replicated twice to confirm this improvement in performance and the safety profile of the MSP. Conclusions: This study suggests that the MSP drug delivery platform holds significant potential for clinical use in improving RA therapy by enabling the sustained slow release of MTX, thereby enhancing therapeutic outcomes and minimizing side effects associated with conventional burst-release drug administration. Full article
Show Figures

Figure 1

Figure 1
<p>Experimental design of safety study.</p>
Full article ">Figure 2
<p>Experimental design of efficacy study.</p>
Full article ">Figure 3
<p>SEM images of the MSP before (Panel (<b>A</b>)) and after (Panel (<b>B</b>)) the freeze-dry step; MSP loaded with MTX after centrifugation (Panel (<b>C</b>)) and after freeze-drying (Panel (<b>D</b>)); scale bar: 1 μm; morphology of the MTX-MSP population by SEM (Panel (<b>E</b>); scale bar: 20 μm) and by confocal analysis (Panel (<b>F</b>), 3D rendering with isosurfaces where green color indicates MTX, and red color indicates MSP; scale bar: 5 μm)). MTX spectra of MTX-MSP by UV-Vis after centrifugation and lyophilization processes (Panel (<b>G</b>)). Picture of the MSP loaded with MTX, centrifuged and lyophilized, after reconstitution with a PBS buffer (Panel (<b>H</b>)).</p>
Full article ">Figure 4
<p>Entrapment Efficiency (EE%) and Loading Efficiency (LE%) of MTX-MSP over time. Results are presented as the average ± standard deviation (SD) (n = 3).</p>
Full article ">Figure 5
<p>Methotrexate release profile. Panel (<b>A</b>): comparison of the MTX release percentage among free MTX, lyophilized MTX-MPS, and centrifuged MTX-MPS over a period of one month. Panel (<b>B</b>): enlargement of the section depicting the release profile within the first 2 h for the three formulations. The results are expressed as mean ± standard deviation and were performed in triplicate. **** <span class="html-italic">p</span> &lt; 0.0001 compared with Free MTX.</p>
Full article ">Figure 6
<p>Body weight measurement at three key experimental time points: T0 (the day before MSP administration), T1 (the fourth experimental day), and T2 (the eighth experimental day, preceding the sacrifice). Data are presented as mean ± SD.</p>
Full article ">Figure 7
<p>Histopathological analysis for safety study. Histological analysis of knee joint sections by H&amp;E staining: liver; spleen; lung; kidney. 20× magnification: Scale bar = 50 μm.</p>
Full article ">Figure 8
<p>Heat map illustrating Architectural Damage and Inflammation Scores for each tested organ. In the heat map, orange indicates lower scores, while blue represents higher scores.</p>
Full article ">Figure 9
<p>(<b>a</b>) Body weight measurement; (<b>b</b>) ELISA assay against Ab α-Collagen II. Data are presented as mean ± SD. * <span class="html-italic">p</span> &lt; 0.05 compared with negative control.</p>
Full article ">Figure 10
<p>Index of spleen and thymus at sacrifice for all groups. Data presented as mean ± SEM.</p>
Full article ">Figure 11
<p>(<b>a</b>) Histological results of the knee joints stained with hematoxylin and eosin. BM: bone marrow; BV: blood vessel; CA: cartridge; GP: growth plate; Me: meniscus; SB: subchondral bone; Sy: synovia; SyC: synovial cavity; Te: tendon. (<b>b</b>) Rheumatoid Arthritis scores of rats in different groups for both analyzed knees; (<b>c</b>) Rheumatoid Arthritis scores of rats in different groups for right analyzed knees; (<b>d</b>) Rheumatoid Arthritis scores of rats in different groups for left analyzed knees. Data are presented as mean ± SD. * <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 compared with positive control.</p>
Full article ">Figure 12
<p>RA histological scores for the right knee, left knee, and pooled knees under various conditions. Scores range from 0 to 8, with darker shades indicating higher scores. Data are presented as mean ± SD.</p>
Full article ">Figure 13
<p>The levels of IL-1β cytokine from serum determined by ELISA assay at the end of the experiment. Data are presented as mean ± SD.</p>
Full article ">Figure 14
<p>(<b>a</b>) Histological results of the knee joints stained with H&amp;E; BM: bone marrox; CA: cartridge; F: fibrin; GP: growth plate; Me: meniscus; SB: subchondral bone; Sy: synovia; SyC: synovial cavity; (<b>b</b>) Rheumatoid Arthritis scoring of rats in different groups for analyzed right knees. Data are presented as mean ± SD. ** <span class="html-italic">p</span> &lt; 0.01; **** <span class="html-italic">p</span> &lt; 0.0001 compared with positive control.</p>
Full article ">
20 pages, 2172 KiB  
Article
Metabolic Fluxes Using Deep Learning Based on Enzyme Variations: Application to Glycolysis in Entamoeba histolytica
by Freddy Oulia, Philippe Charton, Ophélie Lo-Thong-Viramoutou, Carlos G. Acevedo-Rocha, Wei Liu, Du Huynh, Cédric Damour, Jingbo Wang and Frederic Cadet
Int. J. Mol. Sci. 2024, 25(24), 13390; https://doi.org/10.3390/ijms252413390 - 13 Dec 2024
Viewed by 360
Abstract
Metabolic pathway modeling, essential for understanding organism metabolism, is pivotal in predicting genetic mutation effects, drug design, and biofuel development. Enhancing these modeling techniques is crucial for achieving greater prediction accuracy and reliability. However, the limited experimental data or the complexity of the [...] Read more.
Metabolic pathway modeling, essential for understanding organism metabolism, is pivotal in predicting genetic mutation effects, drug design, and biofuel development. Enhancing these modeling techniques is crucial for achieving greater prediction accuracy and reliability. However, the limited experimental data or the complexity of the pathway makes it challenging for researchers to predict phenotypes. Deep learning (DL) is known to perform better than other Machine Learning (ML) approaches if the right conditions are met (i.e., a large database and good choice of parameters). Here, we use a knowledge-based model to massively generate synthetic data and extend a small initial dataset of experimental values. The main objective is to assess if DL can perform at least as well as other ML approaches in flux prediction, using 68,950 instances. Two processing methods are used to generate DL models: cross-validation and repeated holdout evaluation. DL models predict the metabolic fluxes with high precision and slightly outperform the best-known ML approach (the Cubist model) with a lower RMSE (≤0.01) in both cases. They also outperform the PLS model (RMSE ≥ 30). This study is the first to use DL to predict the overall flux of a metabolic pathway only from variations of enzyme concentrations. Full article
(This article belongs to the Section Molecular Informatics)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The glycolytic metabolic pathway of <span class="html-italic">Entamoeba histolytica</span> drives the conversion of glucose into energy and essential biomolecules. 3-phosphoglycerate mutase (PGAM), enolase (ENO), and pyruvate phosphate dikinase (PPDK) are the three enzymes involved in the metabolic pathway. Red arrows indicate retroinhibition of PGAM and ENO enzymes by 3PG and PPi.</p>
Full article ">Figure 2
<p>Evolution of the RMSE on the training and validation data over the training epochs. The RMSE values shown in this figure are calculated using the normalized output. The number of epochs was set to 3000, and the RMSE values stop at epoch 894 since the RMSE has not decreased for 100 epochs. After this interruption, the model parameters at epoch 794 were reloaded.</p>
Full article ">Figure 3
<p>Heat map representing the Pearson’s correlation coefficient on the five test sets (numbered from 0 to 4) during repeated cross-validation for RMSE. The heat map of Pearson’s correlation coefficient for R<sup>2</sup> is in <a href="#app1-ijms-25-13390" class="html-app">Supplementary Figure S8</a> and MAE in <a href="#app1-ijms-25-13390" class="html-app">Figure S9</a>. The correlation of the RMSE results between the different test sets is very strong (always higher than 0.8): The models have a good generalization capacity.</p>
Full article ">Figure 4
<p>Heat map with the Pearson’s correlation coefficient on the RMSE results obtained on the test sets with the repeated holdout evaluation procedure. Pearson’s correlation coefficient results for R<sup>2</sup> are available in <a href="#app1-ijms-25-13390" class="html-app">Supplementary Figure S21</a>, and Pearson’s rank correlation results for MAE are available in <a href="#app1-ijms-25-13390" class="html-app">Supplementary Figure S22</a>. The correlation of the RMSE results on the different test sets is strong: there is a good generalization capacity of the models that have been trained.</p>
Full article ">Figure 5
<p>Overlay of the RMSE boxplots obtained during the two procedures (repeated cross-validation and repeated holdout evaluation) in order to better observe the differences in performance, if any. For each set, results are represented through a density wave with a boxplot below it, and the diamonds represent outlier results. Find in green the performance of cross-validation models and in orange repeated holdout evaluation models. The average performance on each set is close between each approach (difference less than 0.01), but the repeated hold-out evaluation approach has more dispersed results. Similar plots for R<sup>2</sup> are available in <a href="#app1-ijms-25-13390" class="html-app">Supplementary Figure S29</a> and for MAE in <a href="#app1-ijms-25-13390" class="html-app">Supplementary Figure S30</a>.</p>
Full article ">Figure 6
<p>Comparison between expected and predicted output flux on the test set (the five test sets are concatenated into one). Find in the bottom right rectangle the difference between the expected and predicted output flux on the experimental data.</p>
Full article ">Figure 7
<p>Distribution of the different variables in the Glycolysis database. Find in this figure, the number of occurrences of concentration values for the features (PGAM, ENO, and PPDK) and flux values for the target (Jpred). The variables PGAM, ENO, and PPDK lie in a uniform distribution. The variable J<sub>pred</sub>, corresponding to the output of the model, seems to follow a gamma distribution.</p>
Full article ">Figure 8
<p>Holdout evaluation workflow for a deep learning model. First, the initial dataset is separated into two datasets: a training set and a test set. Given the size of our dataset, a random separation of 80/20 generally allows us to have both two sub-datasets respecting the distribution of the initial dataset and a test set with a satisfactory size. Then, the training set is split in two (80/20) to obtain a validation set in addition to the training set. The remaining training set will be used to train the model, and the validation set will be used to check if the model is overfitting. Once the training is completed, the model is evaluated on the test set.</p>
Full article ">Figure 9
<p>Workflow of the repeated cross-validation approach. First, the initial dataset is separated into 2 data subsets: one set to train the models and the other to evaluate them. The test dataset is separated into 5 test datasets in order to test the generalization capabilities of the models. In the large yellow box, the training data is separated into several folds. One fold will act as a validation set and the others as a training set when training a model. To complete a repetition, each fold will act as a validation set one time. This process will be repeated m times. All the generated models are evaluated on the test sets in order to collect all the performances of each model.</p>
Full article ">
19 pages, 4683 KiB  
Article
Multifractal Analysis and Experimental Evaluation of MCM-48 Mesoporous Silica as a Drug Delivery System for Metformin Hydrochloride
by Mousa Sha’at, Maria Ignat, Liviu Sacarescu, Adrian Florin Spac, Alexandra Barsan (Bujor), Vlad Ghizdovat, Emanuel Nazaretian, Catalin Dumitras, Maricel Agop, Cristina Marcela Rusu and Lacramioara Ochiuz
Biomedicines 2024, 12(12), 2838; https://doi.org/10.3390/biomedicines12122838 - 13 Dec 2024
Viewed by 361
Abstract
Background: This study explored the potential of MCM-48 mesoporous silica matrices as a drug delivery system for metformin hydrochloride, aimed at improving the therapeutic management of type 2 diabetes mellitus. The objectives included the synthesis and characterization of MCM-48, assessment of its [...] Read more.
Background: This study explored the potential of MCM-48 mesoporous silica matrices as a drug delivery system for metformin hydrochloride, aimed at improving the therapeutic management of type 2 diabetes mellitus. The objectives included the synthesis and characterization of MCM-48, assessment of its drug loading capacity, analysis of drug release profiles under simulated physiological conditions, and the development of a multifractal dynamics-based theoretical framework to model and interpret the release kinetics. Methods: MCM-48 was synthesized using a sol–gel method and characterized by SEM-EDX, TEM, and nitrogen adsorption techniques. Drug loading was performed via adsorption at pH 12 using metformin hydrochloride solutions of 1 mg/mL (P-1) and 3 mg/mL (P-2). In vitro dissolution studies were conducted to evaluate the release profiles in simulated gastric and intestinal fluids. A multifractal dynamics model was developed to interpret the release kinetics. Results: SEM-EDX confirmed the uniform distribution of silicon and oxygen, while TEM images revealed a highly ordered cubic mesoporous structure. Nitrogen adsorption analyses showed a high specific surface area of 1325.96 m²/g for unloaded MCM-48, which decreased with drug loading, confirming efficient incorporation of metformin hydrochloride. The loading capacities were 59.788 mg/g (P-1) and 160.978 mg/g (P-2), with efficiencies of 99.65% and 89.43%, respectively. In vitro dissolution studies showed a biphasic release profile: an initial rapid release in gastric conditions followed by sustained release in intestinal fluids, achieving cumulative releases of 92.63% (P-1) and 82.64% (P-2) after 14 hours. The multifractal dynamics-based theoretical release curves closely matched the experimental data. Conclusions: MCM-48 mesoporous silica effectively enhanced metformin delivery, offering a controlled release profile well-suited for type 2 diabetes management. The multifractal theoretical framework provided valuable insights into drug release dynamics, contributing to the advancement of innovative drug delivery systems. Full article
(This article belongs to the Special Issue Nano-Based Drug Delivery and Drug Discovery)
Show Figures

Figure 1

Figure 1
<p>Elemental composition (EDX spectra) of MCM-48 sample unloaded (<b>a</b>); MCM-48 sample P-1 (<b>b</b>); and MCM-48 sample P-2 (<b>c</b>).</p>
Full article ">Figure 2
<p>SEM images: MCM-48 sample unloaded (<b>a</b>); MCM-48 sample P-1 (<b>b</b>); and MCM-48 sample P-2 (<b>c</b>).</p>
Full article ">Figure 3
<p>TEM images of MCM-48 sample unloaded; MCM-48 sample P-1 and MCM-48 sample P-2: (<b>a</b>) 1 µm, (<b>b</b>) 200 nm, and (<b>c</b>) 50 nm.</p>
Full article ">Figure 3 Cont.
<p>TEM images of MCM-48 sample unloaded; MCM-48 sample P-1 and MCM-48 sample P-2: (<b>a</b>) 1 µm, (<b>b</b>) 200 nm, and (<b>c</b>) 50 nm.</p>
Full article ">Figure 4
<p>Nitrogen adsorption isotherm of MCM-48 sample unloaded (<b>a</b>); MCM-48 sample P-1 (<b>b</b>); and MCM-48 sample P-2 (<b>c</b>).</p>
Full article ">Figure 5
<p>In vitro dissolution release of metformin from mesoporous silica.</p>
Full article ">Figure 6
<p>Release rate dependences: (<b>a</b>) 3D plot in non-dimensional coordinates; (<b>b</b>) 2D plot in non-dimensional coordinates; (<b>c</b>) <math display="inline"><semantics> <mrow> <mi>ϕ</mi> <mo>≡</mo> <mi>ρ</mi> <mfenced separators="|"> <mrow> <mi>x</mi> <mo>,</mo> <mn>2</mn> </mrow> </mfenced> </mrow> </semantics></math>; (<b>d</b>) <math display="inline"><semantics> <mrow> <mi>ϕ</mi> <mo>≡</mo> <mi>ρ</mi> <mfenced separators="|"> <mrow> <mn>2</mn> <mo>,</mo> <mi>y</mi> </mrow> </mfenced> </mrow> </semantics></math>.</p>
Full article ">Figure 6 Cont.
<p>Release rate dependences: (<b>a</b>) 3D plot in non-dimensional coordinates; (<b>b</b>) 2D plot in non-dimensional coordinates; (<b>c</b>) <math display="inline"><semantics> <mrow> <mi>ϕ</mi> <mo>≡</mo> <mi>ρ</mi> <mfenced separators="|"> <mrow> <mi>x</mi> <mo>,</mo> <mn>2</mn> </mrow> </mfenced> </mrow> </semantics></math>; (<b>d</b>) <math display="inline"><semantics> <mrow> <mi>ϕ</mi> <mo>≡</mo> <mi>ρ</mi> <mfenced separators="|"> <mrow> <mn>2</mn> <mo>,</mo> <mi>y</mi> </mrow> </mfenced> </mrow> </semantics></math>.</p>
Full article ">Figure 7
<p>Drug release kinetics for various fractality degrees expressed as different resolution scales: 1, 1.5, and 2 (in coordinates <math display="inline"><semantics> <mrow> <mi>ϕ</mi> <mo>≡</mo> <mi>θ</mi> <mo>,</mo> <mo> </mo> <mi>y</mi> <mo>≡</mo> <mi>τ</mi> </mrow> </semantics></math>). The dot circle indicates where the resolution scale changes.</p>
Full article ">
25 pages, 2670 KiB  
Article
The Development of an Age-Appropriate Fixed Dose Combination for Tuberculosis Using Physiologically-Based Pharmacokinetic Modeling (PBBM) and Risk Assessment
by Xavier J. H. Pepin, Juliana Johansson Soares Medeiros, Livia Deris Prado and Sandra Suarez Sharp
Pharmaceutics 2024, 16(12), 1587; https://doi.org/10.3390/pharmaceutics16121587 - 12 Dec 2024
Viewed by 535
Abstract
Background/Objectives: The combination of isoniazid (INH) and rifampicin (RIF) is indicated for the treatment maintenance phase of tuberculosis (TB) in adults and children. In Brazil, there is no current reference listed drug for this indication in children. Farmanguinhos has undertaken the development of [...] Read more.
Background/Objectives: The combination of isoniazid (INH) and rifampicin (RIF) is indicated for the treatment maintenance phase of tuberculosis (TB) in adults and children. In Brazil, there is no current reference listed drug for this indication in children. Farmanguinhos has undertaken the development of an age-appropriate dispersible tablet to be taken with water for all age groups from birth to adolescence. The primary objective of this work was to develop and validate a physiologically-based biopharmaceutics model (PBBM) in GastroPlusTM, to link the product’s in vitro performance to the observed pharmacokinetic (PK) data in adults and children. Methods: The PBBM was developed based on measured or predicted physico-chemical and biopharmaceutical properties of INH and RIF. The metabolic clearance was specified mechanistically in the gut and liver for both parent drugs and acetyl-isoniazid. The model incorporated formulation related measurements such as dosage form disintegration and dissolution as inputs and was validated using extensive literature as well as in house clinical data. Results: The model was used to predict the exposure in children across the targeted dosing regimen for each age group using the new age-appropriate formulation. Probabilistic models of efficacy and safety versus exposure, combined with real world data on children, were utilized to assess drug efficacy and safety in the target populations. Conclusions: The model predictions (systemic exposure) along with clinical data from the literature linking systemic exposure to clinical outcomes confirmed that the proposed dispersible pediatric tablet and dosing regimen are anticipated to be as safe and as effective as adult formulations at similar doses. Full article
Show Figures

Figure 1

Figure 1
<p>Overview of the modeling strategy.</p>
Full article ">Figure 2
<p>Structure of INH (<b>left</b>), Ac-INH (<b>middle</b>), and RIF (<b>right</b>).</p>
Full article ">Figure 3
<p>Prediction of INH AUC (<b>A</b>), RIF AUC (<b>B</b>), INH C<sub>max</sub> (<b>C</b>), and RIF C<sub>max</sub> and plasma concentrations (<b>D</b>) across all the validation clinical datasets for the adult and pediatric studies.</p>
Full article ">Figure 4
<p>C<sub>max</sub> predicted for populations of pediatric subjects for INH (<b>upper panel</b>) and RIF (<b>lower panel</b>) by age group according to the dosing schedule of <a href="#pharmaceutics-16-01587-t001" class="html-table">Table 1</a>. The horizontal line shows the minimum threshold for efficacy according to Kiser et al. [<a href="#B57-pharmaceutics-16-01587" class="html-bibr">57</a>] for INH (<b>upper panel</b>) and Pasipanodya et al. [<a href="#B68-pharmaceutics-16-01587" class="html-bibr">68</a>] for RIF (<b>lower panel</b>).</p>
Full article ">Figure 5
<p>AUC predictions for pediatric populations of INH SA (<b>A</b>), INH RA (<b>B</b>), INH IA (<b>C</b>), and RIF (<b>D</b>) according to the schedule of <a href="#pharmaceutics-16-01587-t001" class="html-table">Table 1</a>. The horizontal lines show the minimum AUC for efficacy and maximum adult AUC for INH DILI according to Zheng et al. [<a href="#B69-pharmaceutics-16-01587" class="html-bibr">69</a>]. The horizontal green line for panel (<b>D</b>) shows the threshold for efficacy according to [<a href="#B68-pharmaceutics-16-01587" class="html-bibr">68</a>].</p>
Full article ">Figure 6
<p>PK profile prediction for 600 mg RIF administered in the fasted state (<b>A</b>), fed state (<b>B</b>), and following ARAs (<b>C</b>). The PK data are reported by Peloquin et al. [<a href="#B80-pharmaceutics-16-01587" class="html-bibr">80</a>]. Panel (<b>D</b>) shows the log degradation half-life for RIF in the fasted state, fed state, and following ARA administration.</p>
Full article ">Figure 7
<p>Evolution of percent abnormal liver markers in children treated prophylactically or with active pulmonary TB as a function of INH dose from Donald [<a href="#B78-pharmaceutics-16-01587" class="html-bibr">78</a>], compared to predictions resulting from this work using the average Brazil genotype reported in [<a href="#B86-pharmaceutics-16-01587" class="html-bibr">86</a>] and risk exposure thresholds reported by Zheng et al. [<a href="#B69-pharmaceutics-16-01587" class="html-bibr">69</a>].</p>
Full article ">
14 pages, 5880 KiB  
Article
Functional Mechanical Behavior and Biocompatible Characteristics of Graphene-Coated Cardiovascular Stents
by Łukasz Wasyluk, Dariusz Hreniak, Vitalii Boiko, Beata Sobieszczańska, Emanuela Bologna, Massimiliano Zingales, Robert Pasławski, Jacek Arkowski, Przemysław Sareło and Magdalena Wawrzyńska
Int. J. Mol. Sci. 2024, 25(24), 13345; https://doi.org/10.3390/ijms252413345 - 12 Dec 2024
Viewed by 381
Abstract
Percutaneous Coronary Intervention (PCI) is a treatment method that involves reopening narrowed arteries with a balloon catheter that delivers a cylindrical, mesh-shaped implant device to the site of the stenosis. Currently, by applying a coating to a bare metal stent (BMS) surface to [...] Read more.
Percutaneous Coronary Intervention (PCI) is a treatment method that involves reopening narrowed arteries with a balloon catheter that delivers a cylindrical, mesh-shaped implant device to the site of the stenosis. Currently, by applying a coating to a bare metal stent (BMS) surface to improve biocompatibility, the main risks after PCI, such as restenosis and thrombosis, are reduced while maintaining the basic requirements for the mechanical behavior of the stent itself. In this work, for the first time, the development and optimization process of the spatial structure of the Co-Cr stent (L-605) with a graphene-based coating using cold-wall chemical vapor deposition (CW-CVD) to ensure uniform coverage of the implant was attempted. The CW-CVD process allows the coating of 3D structures, minimizing thermal stress on the surrounding equipment and allowing the deposition of coatings on temperature-sensitive materials. It produces uniform and high-purity films with control over the thickness and composition. The reduced heating of the chamber walls minimizes unwanted reactions, leading to fewer impurities in the final coating. The graphene layers obtained using Raman spectroscopy at different parameters of the CW-CVD process were verified, their properties were investigated, and the functional mechanical behavior of the studied graphene-covered stent was confirmed. In vitro, graphene-coated stents promoted rapid endothelial cell repopulation, an advantage over gold-standard drug-eluting stents delaying re-endothelialization. Also, full-range biocompatibility studies on potential allergic, irritation, toxicological, and pyrogenic reactions of new material in vivo on small animal models demonstrated excellent biocompatibility of the graphene-coated stents. Full article
(This article belongs to the Special Issue Biofunctional Coatings for Medical Applications)
Show Figures

Figure 1

Figure 1
<p>Raman spectra (λ<sub>ex</sub>—514 nm) of the cardiovascular stents before (black line) and after CW-CVD (red, green, and blue line) with different deposition temperatures (700 °C, 900 °C, and 1100 °C, respectively).</p>
Full article ">Figure 2
<p>SEM images of a cardiovascular stent (<b>a</b>) before and (<b>b</b>) after CW-CVD. (<b>c</b>) The stent fracture after crimping. (<b>d</b>–<b>f</b>) Images of critical areas for properly crimped and expanded stent. The scale bar is presented in the appropriate image.</p>
Full article ">Figure 3
<p>(<b>a</b>) The radial force measurement device. (<b>b</b>) The values of the obtained radial forces of graphene-coated stent (GC-stent) and uncoated stent (BM-stent). The <span class="html-italic">p</span>-value according to the non-parametric Mann–Whitney U test.</p>
Full article ">Figure 4
<p>(<b>a</b>) The coronary stents used in the test and the BOSE 9400 MAPS system. (<b>b</b>) The macro-photography of the bare metal stent. (<b>c</b>) The macro-photography of the graphene-coated stent. (<b>d</b>) Time evolution of the stent diameter for the reference and graphene-coated stents and (<b>e</b>) pressure-diameter elastic behavior of the stent in the cyclic load-unload test for the reference and graphene-coated stents. The response to cyclic loading confirms that graphene-coated stents are just as safe as uncoated stents, which have been used clinically for many years. The mechanical properties of graphene-coated stents are similar to those of other coatings. The main advantage of graphene coating is increased biocompatibility.</p>
Full article ">Figure 5
<p>(<b>a</b>) HUVEC cell proliferation on bare metal (BM) stents and graphene-coated (GC) bare metal stents after 72 h quantified in the WST-1 assay. * <span class="html-italic">p</span> &lt; 0.001. (<b>b</b>) The proliferation of HUVEC cells on bare metal (BM) stent and graphene-coated bare metal (GC) stent after 72 h. Cells were visualized by staining the cell’s actin cytoskeleton with phalloidin-FITC and the cell’s nuclei with DAPI. Magnification 400×.</p>
Full article ">Figure 6
<p>Photographic documentation of the allergy and skin irritation tests performed, where (<b>a</b>) the method of applying the tested graphene-coated samples on the shaved skin of a guinea pig during the GPMT test is presented. The tested implant was placed on the skin of a rabbit similarly during the Rabbit Skin Primary Irritation Test. (<b>b</b>) The site after applying the graphene-coated stent and after a 14-day break and re-applying of the stent. The site was assessed using the Magnusson and Kligman scale in the GPMT test. (<b>c</b>) The site after 72 h where the graphene-coated stent was applied and subjected to erythema and edema assessments on a scale of 0 to 4 in the Rabbit Skin Primary Irritation Test.</p>
Full article ">Figure 7
<p>Photographic documentation (<b>a</b>–<b>c</b>) of individual stages of intraperitoneal insertion of the tested graphene-coated stents. (<b>d</b>) The autopsy did not show any symptoms of reaction to the tested material. The implanted material samples were loose in the peritoneal cavity and could be easily removed.</p>
Full article ">Figure 8
<p>The histopathological microscope images show no organ changes following the introduction of graphene-coated stents: chronic response study. No changes were observed in (<b>a</b>) lungs, (<b>b</b>) heart, (<b>c</b>) kidneys, and (<b>d</b>) liver. The results do not differ from typical images characteristic of healthy organs. Below, histopathological images of the skin after (<b>e</b>) 24 h, (<b>f</b>) 48 h, and (<b>g</b>) 72 h, respectively, are shown in the skin irritation tests. The tests were performed on the White New Zealand rabbit. The scale shown in the images indicates 400 μm.</p>
Full article ">
20 pages, 3540 KiB  
Review
Forward Computational Modeling of Respiratory Airflow
by Emmanuel A. Akor, Bing Han, Mingchao Cai, Ching-Long Lin and David W. Kaczka
Appl. Sci. 2024, 14(24), 11591; https://doi.org/10.3390/app142411591 - 12 Dec 2024
Viewed by 351
Abstract
The simulation of gas flow in the bronchial tree using computational fluid dynamics (CFD) has become a useful tool for the analysis of gas flow mechanics, structural deformation, ventilation, and particle deposition for drug delivery during spontaneous and assisted breathing. CFD allows for [...] Read more.
The simulation of gas flow in the bronchial tree using computational fluid dynamics (CFD) has become a useful tool for the analysis of gas flow mechanics, structural deformation, ventilation, and particle deposition for drug delivery during spontaneous and assisted breathing. CFD allows for new hypotheses to be tested in silico, and detailed results generated without performing expensive experimental procedures that could be potentially harmful to patients. Such computational techniques are also useful for analyzing structure–function relationships in healthy and diseased lungs, assessing regional ventilation at various time points over the course of clinical treatment, or elucidating the changes in airflow patterns over the life span. CFD has also allowed for the development and use of image-based (i.e., patient-specific) models of three-dimensional (3D) airway trees with realistic boundary conditions to achieve more meaningful and personalized data that may be useful for planning effective treatment protocols. This focused review will present a summary of the techniques used in generating realistic 3D airway tree models, the limitations of such models, and the methodologies used for CFD airflow simulation. We will discuss mathematical and image-based geometric models, as well as the various boundary conditions that may be imposed on these geometric models. The results from simulations utilizing mathematical and image-based geometric models of the airway tree will also be discussed in terms of similarities to actual gas flow in the human lung. Full article
(This article belongs to the Special Issue Applications of Fluid Mechanics in Biomedical Engineering)
Show Figures

Figure 1

Figure 1
<p>Projected images showing the three dimensional structural and spatial model of the airway tree based on mathematical algorithm developed by Kitaoka et al. [<a href="#B27-applsci-14-11591" class="html-bibr">27</a>]. There are about 30,000 terminal branches and 54,611 total branches in this model. The airway branches distal to given segmental bronchi are shown by the same color. (<b>A</b>) anterior view (<b>B</b>) lateral view. This work is made available under the terms of the Creative Commons Attribution CC BY 4.0 (<a href="https://creativecommons.org/licenses/by/4.0/" target="_blank">https://creativecommons.org/licenses/by/4.0/</a>).</p>
Full article ">Figure 2
<p>A two-dimensional branching network of the lung airway tree showing the Monte Carlo method for any distributive system. The dots represent random points in 2D space that determine how the network branches. The thick lines within the enclosed lung surface represent the airway branches for the first two generations, and the thin lines are the dividing lines that are generated from the calculation of the center of mass of the points in any given region.</p>
Full article ">Figure 3
<p>3D Spatial, 1D Structural Model generated using the Volume Filling Method. The generated airways are shown in different colors for each lobe: green for left upper lobe (LUL), magenta for left lower lobe (LLL), blue for right upper lobe (RUL), red for right middle lobe (RML) and orange for right lower lobe (RLL). Modified from reference [<a href="#B44-applsci-14-11591" class="html-bibr">44</a>], with permission.</p>
Full article ">Figure 4
<p>Workflow for attaining subject-specific boundary flow conditions at the distal airway tree [<a href="#B55-applsci-14-11591" class="html-bibr">55</a>]. Regional ventilation is obtained from the registration of two 3D images taken at different time points during inhalation, and matched to the structural 1D airway tree using the volume filling branching method. For both regional ventilation and the ventilation map, cool colors correspond to regions of high ventilation, while warm colors correspond to regions of low ventilation. For the volume-filling method, the five different colors correspond to distinct anatomic lobes. This work is made available under the terms of the Creative Commons Attribution CC BY 4.0 (<a href="https://creativecommons.org/licenses/by/4.0/" target="_blank">https://creativecommons.org/licenses/by/4.0/</a>).</p>
Full article ">Figure 5
<p>Velocity vectors (pink) and pressure drop distributions in a three-dimensional model with: (<b>a</b>) patient-specific; (<b>b</b>) uniform velocity; and (<b>c</b>) uniform pressure boundary conditions at the distal airway outlets. The patient-specific boundary condition yielded a greater pressure drop in the left lower lobe and the right lower lobe compared with the uniform velocity condition, the uniform velocity boundary condition yielded a greater pressure drop in the right middle lobe than the others, and the uniform pressure boundary condition yielded a uniform pressure drop across all five lobes. From reference [<a href="#B2-applsci-14-11591" class="html-bibr">2</a>], with permission.</p>
Full article ">Figure 6
<p>Flow velocity contours measured experimentally by PIV (<b>a</b>) and computed from a CFD model (<b>b1</b>,<b>b2</b>), using boundary conditions from the in vitro study. (<b>a</b>) The in-plane velocity was evaluated in the coronal plane at the center of the geometry (z = 0). Velocities at the outlets (labeled numerically) and segments (labeled alphabetically) are shown. (<b>b1</b>) Velocity magnitudes in coronal section were derived from the CFD model using a healthy boundary condition. Flow streamlines are displayed with arrows highlighting areas of flow separation and recirculation. (<b>b2</b>) Axial velocity magnitudes and directions (vectors) are shown across branches a–i. Color bar indicates the velocity spectrum, with red corresponding to maximum velocity and blue corresponding to minimum velocity. From reference [<a href="#B75-applsci-14-11591" class="html-bibr">75</a>], with permission.</p>
Full article ">
29 pages, 8061 KiB  
Article
System Proposal for Supervision of Critical Adverse Processes in Patients with Implanted Ventricular Assist Devices
by José R. C. Sousa Sobrinho, Marcosiris A. O. Pessoa, Fabrício Junqueira, Paulo E. Miyagi and Diolino J. Santos Filho
Appl. Sci. 2024, 14(24), 11551; https://doi.org/10.3390/app142411551 - 11 Dec 2024
Viewed by 355
Abstract
Ventricular assist devices (VADs) are designed to provide sufficient blood flow to patients with severe heart failure. Once implanted, the patient becomes dependent on the VAD, making it essential to prevent situations that could harm the patient while receiving circulatory support. VADs are [...] Read more.
Ventricular assist devices (VADs) are designed to provide sufficient blood flow to patients with severe heart failure. Once implanted, the patient becomes dependent on the VAD, making it essential to prevent situations that could harm the patient while receiving circulatory support. VADs are classified as critical systems (CS), and adverse events (AEs) can lead to serious consequences, including hospitalization or even death. At present, patient care is provided through in-person consultations, with incidents reported via medical device reports (MDRs) to the Food and Drug Administration (FDA). However, there is no real-time monitoring of AEs or oversight of these events. In response to this gap, a system for supervising critical adverse processes in patients with implanted VADs (SCVAD) is proposed, based on horizontally and vertically integrated architecture. This system aims to address the complexity of AEs by considering multiple domains of operation: the device, the patient, and the medical team, as well as the interactions between these entities. In this context, the formalism of Petri nets (PN) is used to develop models that represent adverse processes based on the actions recommended by the medical team. These models allow for the mapping of events with the potential to cause harm to the patient. Therefore, the medical team will be able to monitor adverse processes, as the models in interpreted PN can be isomorphically transcribed into computable algorithms that can be processed on compatible devices, enabling the tracking of complications caused by adverse processes. Full article
(This article belongs to the Section Biomedical Engineering)
Show Figures

Figure 1

Figure 1
<p>Essential elements of a PFS graph. The square brackets represent the ‘activity’ element, the circle represents the ‘distributor’ element, and the arrow represents the ‘arc’ element.</p>
Full article ">Figure 2
<p>Conversion of the model from PFS to PN through successive top-down refinement. In (<b>A</b>), an activity in PFS; in (<b>B</b>), another possible representation of the same activity element; in (<b>C</b>), a combined PFS/PN representation (the activity is represented by a discrete place between two discrete transitions); and in (<b>D</b>), the corresponding PN is represented.</p>
Full article ">Figure 3
<p>PFS model of the exemplified process.</p>
Full article ">Figure 4
<p>Detailed process model in PN.</p>
Full article ">Figure 5
<p>Introduction of resource control elements for sharing.</p>
Full article ">Figure 6
<p>Representation of data flow to the external environment.</p>
Full article ">Figure 7
<p>Event-oriented structure model of the SCVAD for digitizing patient care processes.</p>
Full article ">Figure 8
<p>The model for structuring SCVAD processes is represented. Note: The red numbering highlights areas susceptible to AEs (5, 6, and 7), while the black numbering identifies areas unrelated to AEs (1, 2, 3, and 4).</p>
Full article ">Figure 9
<p>PFS and detailing of the digital model for adverse event diagnosis (DMAD).</p>
Full article ">Figure 10
<p>PFS and detailing of the diagnostic module for thrombosis in the device (AE1).</p>
Full article ">Figure 11
<p>PFS and detailing of the adverse activity ‘pre-pump thrombosis AE1.1’.</p>
Full article ">Figure 12
<p>PN corresponding to the adverse activity ‘pre-pump thrombosis AE1.1’.</p>
Full article ">Figure 13
<p>PFS and detailing of the adverse activity ‘intra-pump thrombosis AE1.2’.</p>
Full article ">Figure 14
<p>PN corresponding to the adverse activity ‘intra-pump thrombosis AE1.2’.</p>
Full article ">Figure 15
<p>PFS and detailing of the adverse activity ‘post-pump thrombosis AE1.3’.</p>
Full article ">Figure 16
<p>PN corresponding to the adverse activity ‘post-pump thrombosis AE1.3’.</p>
Full article ">Figure 17
<p>PFS and detailing of the activity ‘final diagnosis of pump thrombosis (AE1)’.</p>
Full article ">Figure 18
<p>PN corresponds to the ‘final diagnosis of pump thrombosis (AE1)’ activity.</p>
Full article ">Figure 19
<p>PFS and detailing of the activity ‘time counting’.</p>
Full article ">Figure 20
<p>PFS and detailing of the activity ‘end of counting time’.</p>
Full article ">Figure 21
<p>Corresponding PN for the ‘time counting’ activity.</p>
Full article ">Figure 22
<p>PFS and detailing of the digital model for adverse process mitigation (DMAPM).</p>
Full article ">Figure 23
<p>PFS and detailing of the thrombosis mitigation module for the device (AE1).</p>
Full article ">Figure 24
<p>PFS and detailing of the activity ‘pre-pump thrombosis mitigation AE1.1’.</p>
Full article ">Figure 25
<p>PN corresponding to the activity ‘pre-pump thrombosis mitigation AE1.1’.</p>
Full article ">Figure 26
<p>PFS and detailing of the activity ‘intra-pump thrombosis mitigation AE1.2’.</p>
Full article ">Figure 27
<p>PN corresponding to the activity ‘intra-pump thrombosis mitigation AE1.2’.</p>
Full article ">Figure 28
<p>PFS and detailing of the activity ‘post-pump thrombosis mitigation AE1.3’.</p>
Full article ">Figure 29
<p>PN corresponding to the activity ‘post-pump thrombosis mitigation AE1.3’.</p>
Full article ">Figure 30
<p>PFS of the activity ‘end of pump thrombosis mitigation (AE1)’.</p>
Full article ">Figure 31
<p>PN corresponding to the ‘end of pump thrombosis mitigation (AE1)’ activity.</p>
Full article ">
21 pages, 8687 KiB  
Article
Development and Characterization of Dual-Loaded Niosomal Ion-Sensitive In Situ Gel for Ocular Delivery
by Viliana Gugleva, Rositsa Mihaylova, Katya Kamenova, Dimitrina Zheleva-Dimitrova, Denitsa Stefanova, Virginia Tzankova, Maya Margaritova Zaharieva, Hristo Najdenski, Aleksander Forys, Barbara Trzebicka, Petar D. Petrov and Denitsa Momekova
Gels 2024, 10(12), 816; https://doi.org/10.3390/gels10120816 - 11 Dec 2024
Viewed by 477
Abstract
The study investigates the development and characterization of dual-loaded niosomes incorporated into ion-sensitive in situ gel as a potential drug delivery platform for ophthalmic application. Cannabidiol (CBD) and epigallocatechin-3-gallate (EGCG) simultaneously loaded niosomes were prepared via the thin film hydration (TFH) method followed [...] Read more.
The study investigates the development and characterization of dual-loaded niosomes incorporated into ion-sensitive in situ gel as a potential drug delivery platform for ophthalmic application. Cannabidiol (CBD) and epigallocatechin-3-gallate (EGCG) simultaneously loaded niosomes were prepared via the thin film hydration (TFH) method followed by pulsatile sonication and were subjected to comprehensive physicochemical evaluation. The optimal composition was included in a gellan gum-based in situ gel, and the antimicrobial activity, in vitro toxicity in a suitable corneal epithelial model (HaCaT cell line), and antioxidant potential of the hybrid system were further assessed. Dual-loaded niosomes based on Span 60, Tween 60, and cholesterol (3.5:3.5:3 mol/mol) were characterized by appropriate size (250 nm), high entrapment efficiency values for both compounds (85% for CBD and 50% for EGCG) and sustained release profiles. The developed hybrid in situ gel exhibited suitable rheological characteristics to enhance the residence time on the ocular surface. The conducted microbiological studies reveal superior inhibition of methicillin-resistant Staphylococcus aureus (MRSA) adhesion by means of the niosomal in situ gel compared to the blank gel and untreated control. Regarding the antioxidant potential, the dual loading of CBD and EGCG in niosomes enhances their protective properties, and the inclusion of niosomes in gel form preserves these effects. The obtained outcomes indicate the developed niosomal in situ gel as a promising drug delivery platform in ophthalmology. Full article
(This article belongs to the Special Issue Composite Hydrogels for Biomedical Applications)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Size distributions of empty and drug-loaded niosomes.</p>
Full article ">Figure 2
<p>Cryo-TEM images of (<b>a</b>) empty niosomes (N6); (<b>b</b>) CBD-loaded niosomes (N5).</p>
Full article ">Figure 3
<p>Viscosity as a function of the shear rate of G6, G6N, and G6N:CBD:EGCG formulations at 25 °C.</p>
Full article ">Figure 4
<p>Variation in elastic (G′) and loss (G″) moduli as a function of shear stress (τ) of G6, G6N, and G6N:CBD:EGCG formulations. All measurements were carried out at 35 °C.</p>
Full article ">Figure 5
<p>In vitro release profile of CBD and EGCG from optimal niosomal formulation (N3) and its hybrid in situ gelling system (G6N:CBD:EGCG). N3:CBD, G6N:CBD denote cannabidiol release from niosomal suspension and hybrid niosomal gel, respectively, whereas N3:EGCG and G6N:EGCG represent the release profiles of epigallocatechin-3-gallate from niosomes and its hybrid niosomal gel formulation. Each value is presented as mean ± SD (n = 3).</p>
Full article ">Figure 6
<p>Quantitative evaluation of MRSA biofilm formation after exposure to blank (G6N) or hybrid niosomal drug-loaded gel G6N:CBD:EGCG (1/0.5 mg/mL). Legend: Co—untreated control; Dilution 1:8 = 0.125/0.06125 mg/mL; Dilution 1:16 = 0.0625/0.03125 mg/mL; Dilution 1:32 = 0.03125/0.0156 mg/mL. Each value is presented as mean ± SD (n = 4).</p>
Full article ">Figure 7
<p>Cytotoxicity on HaCaT cells of: (<b>A</b>) empty niosomes (N6); (<b>B</b>) free cannabidiol (CBD); (<b>C</b>) free epigallocatechin (EGCG); (<b>D</b>) combination of free cannabidiol and free epigallocatechin (CBD + EGCG); (<b>E</b>) dual-loaded CBD and EGCG vesicles (N:CBD:GCG, formulation) niosomes and (<b>F</b>) niosomal in situ gel based on double-loaded niosomes (G6N:CBD:EGCG), measured by MTT assay. All groups were compared statistically vs. untreated controls by one-way ANOVA with Dunnet’s post hoc test. The results are expressed as means ± SD of triplicate assays (n = 8). *** <span class="html-italic">p</span> &lt; 0.001 vs. control (CTRL, untreated control cells).</p>
Full article ">Figure 8
<p>Protective effects of (<b>A</b>) empty niosomes; (<b>B</b>) free CBD; (<b>C</b>) free EGCG; (<b>D</b>) combination of free CBD and free EGCG (CBD + EGCG); (<b>E</b>) dual-loaded CBD and EGCG (N:CBD:EGCG) niosomes and (<b>F</b>) niosomal in situ gel based on double-loaded niosomes (G6N:CBD:EGCG) in a H<sub>2</sub>O<sub>2</sub>-induced damage model in human keratinocyte HaCaT cell line. The results are expressed as means ± SD of triplicate assays (n = 8). ANOVA with Dunnett’s post-test. * <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 vs. H<sub>2</sub>O<sub>2</sub> CTRL (untreated control cells); H<sub>2</sub>O<sub>2</sub> cells treated with H<sub>2</sub>O<sub>2</sub> (200 µM).</p>
Full article ">
19 pages, 1645 KiB  
Review
Bridging the Gap: Endothelial Dysfunction and the Role of iPSC-Derived Endothelial Cells in Disease Modeling
by Chiara Sgromo, Alessia Cucci, Giorgia Venturin, Antonia Follenzi and Cristina Olgasi
Int. J. Mol. Sci. 2024, 25(24), 13275; https://doi.org/10.3390/ijms252413275 - 11 Dec 2024
Viewed by 406
Abstract
Endothelial cells (ECs) are crucial for vascular health, regulating blood flow, nutrient exchange, and modulating immune responses and inflammation. The impairment of these processes causes the endothelial dysfunction (ED) characterized by oxidative stress, inflammation, vascular permeability, and extracellular matrix remodeling. While primary ECs [...] Read more.
Endothelial cells (ECs) are crucial for vascular health, regulating blood flow, nutrient exchange, and modulating immune responses and inflammation. The impairment of these processes causes the endothelial dysfunction (ED) characterized by oxidative stress, inflammation, vascular permeability, and extracellular matrix remodeling. While primary ECs have been widely used to study ED in vitro, their limitations—such as short lifespan and donor variability—pose challenges. In this context, induced iECs derived from induced pluripotent stem cells offer an innovative solution, providing an unlimited source of ECs to explore disease-specific features of ED. Recent advancements in 3D models and microfluidic systems have enhanced the physiological relevance of iEC-based models by better mimicking the vascular microenvironment. These innovations bridge the gap between understanding ED mechanisms and drug developing and screening to prevent or treat ED. This review highlights the current state of iEC technology as a model to study ED in vascular and non-vascular disorders, including diabetes, cardiovascular, and neurodegenerative diseases. Full article
(This article belongs to the Special Issue hiPSC-Based Disease Models as Replacements of Animal Models)
Show Figures

Figure 1

Figure 1
<p>Mechanisms of endothelial dysfunction. (<b>A</b>) Oxidative stress: An imbalance between endothelium-derived constricting factors (EDCFs) and relaxing factors (EDFRs) results in reduced nitric oxide (NO) levels and increased reactive oxygen species (ROS). This activates NF-κB, promoting the expression of pro-inflammatory mediators such as IL-1β, IL-6, TNF-α, VCAM-1, and ICAM-1. (<b>B</b>) Low or oscillatory shear stress: Disruption of glycocalyx integrity activates the transcriptional regulators AP-1, YAP/TAZ/TEAD, and HIF-1α. This promotes the release of inflammatory mediators such as VEGF, histamine, thrombin, IL-1B, and TNF-α, driving inflammation and increasing endothelial permeability. (<b>C</b>) ECM and cytoskeletal remodeling: Alterations in ECM and cytoskeletal dynamics, mediated by GRK2 and vinculin, exacerbate vascular permeability. These structural changes further amplify the endothelial barrier dysfunction. Together, these processes culminate in inflammation, increased permeability, and ultimately, ED. ECM: extracellular matrix; eNOS: endothelial nitric oxide synthase; NO: nitric oxide; AP-1: activating protein 1; YAP: Yes1-associated transcriptional regulator; TAZ: Tafazzin; TEAD: TEA domain transcription factor 1; HIF-1α: hypoxia-inducible factor 1 subunit alpha; VEGF: vascular endothelial growth factor; IL-1B: interleukin beta1; TNF-α: tumor necrosis factor alpha; GRK2: G protein-coupled receptor kinase 2; and NF-Kb: nuclear factor kappa-light-chain-enhancer of activated B cells.</p>
Full article ">Figure 2
<p>Differentiation of iPSCs into endothelial cell subtypes. Differentiation protocols for deriving arterial, venous, and lymphatic ECs from iPSCs. Mesoderm induction (yellow panel): iPSC-derived EBs are cultured in EC medium and treated sequentially with BMP4 (days 2–4) and a combination of BMP4 and FGF2 (days 4–6) to induce mesoderm specification. Arterial ECs (red panel): EBs, after mesoderm specification, are plated on gelatin-coated plates and treated with EC serum-free medium supplemented with 8Br-cAMP and a high level of VEGF-A (50 ng/mL). Activation of VEGF and Notch signaling pathways promotes arterial differentiation, marked by EphB2 expression, by day 14. Venous ECs (blue panel): After mesoderm specification, EBs are cultured with FGF and VEGF (days 6–8), followed by low VEGF (10 ng/mL) from day 10 onward. Modulation of COUP-TFII expression drives venous differentiation, as indicated by COUP-TFII and EphB4 expression. Lymphatic ECs (green panel): After mesoderm specification, EBs are cultured in suspension, co-cultured with OP9 stromal cells, or cultured in feeder-free conditions. The addition of BMP-4, VEGF-A, VEGF-C, angiopoietin-1 (Ang-1), and EGF activates the ERK/Sox18/Prox1 pathways, driving lymphatic differentiation. Key markers, including Prox1, Lyve1, VEGFR3, and Podoplanin, are upregulated by day 7, with mature lymphatic cells obtained by day 30. BMP4: bone morphogenetic protein 4; FGF2: fibroblast growth factor 2; cAMP: 8-bromoadenosine 3′,5′-cyclic adenosine monophosphate; VEGF: vascular endothelial growth factor; COUP-TFII: Chicken ovalbumin upstream promoter-transcription factor II; EphB2: Ephrin type-B receptor 2; EphB4: Ephrin receptor B4; Lyve1: lymphatic vessel endothelial hyaluronan receptor 1; Prox1: Prospero homeobox 1; VEGFR3: vascular endothelial growth factor receptor 3; ERK: extracellular signal-regulated kinase; and Sox18: SRY-Box transcription factor 18.</p>
Full article ">Figure 3
<p>Modeling the blood–brain barrier (BBB) with iPSC-derived brain microvascular endothelial cells (iBMECs). Induced pluripotent stem cells (iPSCs) are differentiated into brain microvascular endothelial cells (iBMECs) and combined with key components of the neurovascular unit (NVU), including neurons, astrocytes, microglia, and pericytes. These cellular elements are assembled in several BBB models, including transwell systems, organ-on-chip platforms, spheroids, hydrogel-based cultures, and vascularized organoids, to mimic the structure and function of the human BBB. Each model offers unique advantages for studying the interactions within the NVU and assessing BBB integrity, permeability, and response to external factors.</p>
Full article ">
17 pages, 3363 KiB  
Article
Pharmacodynamic Evaluation of Phage Therapy in Ameliorating ETEC-Induced Diarrhea in Mice Models
by Yangjing Xiong, Lu Xia, Yumin Zhang, Guoqing Zhao, Shidan Zhang, Jingjiao Ma, Yuqiang Cheng, Hengan Wang, Jianhe Sun, Yaxian Yan and Zhaofei Wang
Microorganisms 2024, 12(12), 2532; https://doi.org/10.3390/microorganisms12122532 - 8 Dec 2024
Viewed by 740
Abstract
Enterotoxigenic Escherichia coli (ETEC) is a major pathogen causing diarrhea in humans and animals, with increasing antimicrobial resistance posing a growing challenge in recent years. Lytic bacteriophages (phages) offer a targeted and environmentally sustainable approach to combating bacterial infections, particularly in eliminating drug-resistant [...] Read more.
Enterotoxigenic Escherichia coli (ETEC) is a major pathogen causing diarrhea in humans and animals, with increasing antimicrobial resistance posing a growing challenge in recent years. Lytic bacteriophages (phages) offer a targeted and environmentally sustainable approach to combating bacterial infections, particularly in eliminating drug-resistant strains. In this study, ETEC strains were utilized as indicators, and a stable, high-efficiency phage, designated vB_EcoM_JE01 (JE01), was isolated from pig farm manure. The genome of JE01 was a dsDNA molecule, measuring 168.9 kb, and a transmission electron microscope revealed its characteristic T4-like Myoviridae morphology. JE01 effectively lysed multi-drug-resistant ETEC isolates. Stability assays demonstrated that JE01 retained its activity across a temperature range of 20 °C to 50 °C and a pH range of 3–11, showing resilience to ultraviolet radiation and chloroform exposure. Furthermore, JE01 effectively suppressed ETEC adhesion to porcine intestinal epithelial cells (IPEC-J2), mitigating the inflammatory response triggered by ETEC. To investigate the in vivo antibacterial efficacy of phage JE01 preparations, a diarrhea model was established using germ-free mice infected with a drug-resistant ETEC strain. The findings indicated that 12 h post-ETEC inoculation, intragastric administration of phage JE01 significantly reduced mortality, alleviated gastrointestinal lesions, decreased ETEC colonization in the jejunum, and suppressed the expression of the cytokines IL-6 and IL-8. These results demonstrate a therapeutic benefit of JE01 in treating ETEC-induced diarrhea in mice. Additionally, a fluorescent phage incorporating red fluorescent protein (RFP) was engineered, and the pharmacokinetics of phage therapy were preliminarily assessed through intestinal fluorescence imaging in mice. The results showed that the phage localized to ETEC in the jejunum rapidly, within 45 min. Moreover, the pharmacokinetics of the phage were markedly slowed in the presence of its bacterial target in the gut, suggesting sustained bacteriolytic activity in the ETEC-infected intestine. In conclusion, this study establishes a foundation for the development of phage-based therapies against ETEC. Full article
(This article belongs to the Special Issue Advances in Microbial Synthetic Biology)
Show Figures

Figure 1

Figure 1
<p>Analysis of the phage JE01′s antibacterial activity. (<b>A</b>) Optimal multiplicity of infection (MOI) of JE01. JE01 was used to infect strain U74 at different MOIs, indicating the most suitable concentration for lysing bacteria. (<b>B</b>) The in vitro bactericidal effect of the phage JE01. Strain U74 suspensions were mixed with the phage (MOI = 1), and OD600 was measured for 360 min. (<b>C</b>) The one-step growth curve of the phage JE01 in ETEC strains U74. The burst size was estimated at 120 PFU per infected cell, which was the ratio of the final count of liberated phage particles to the initial count of infected bacterial cells. (<b>D</b>) Transmission electron microscopy of a negatively stained JE01 phage. For (<b>A</b>,<b>B</b>), experiments were conducted in triplicate, and the results are shown as the mean ± SEM. For (<b>C</b>), experiments were conducted in quadruplicate, and the results are shown as the mean ± SEM. For (<b>A</b>), statistical significance was determined using one-way ANOVA (Dunnett’s multiple comparisons test) to compare the phage titers between MOI 1 and other MOIs. *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 2
<p>The genomic features of the phage JE01 were determined by making a Proksee map (<a href="https://proksee.ca" target="_blank">https://proksee.ca</a>, accessed on 1 November 2023). The functional modules in the phage genomes are shown in different colors.</p>
Full article ">Figure 3
<p>Biological properties of the phage JE01. (<b>A</b>) Thermal stability assay: phages were incubated for 1 h at various temperatures ranging from 20 °C to 80 °C at 5 °C intervals. (<b>B</b>) pH stability assay: phages were incubated for 1 h at pH values ranging from 2 to 12. (<b>C</b>) UV radiation stability assay: phages were exposed to UV light (30 w, 30 cm) for 15, 30, 45, 60, and 75 min. (<b>D</b>) Chloroform sensitivity assay: phages were treated with various chloroform concentrations (0%, 1%, 2%, 5%, 10%, and 20%) at 37 °C for 30 min. The surviving phage particles were quantified by double-layer tests. Experiments were conducted in triplicate, and the results are shown as the mean ± SEM.</p>
Full article ">Figure 4
<p>JE01 alleviated ETEC-induced IPEC-J2 cell damage. (<b>A</b>) JE01 could alleviate the CFU burden of ETEC adhering to IPEC-J2 cells. (<b>B</b>,<b>C</b>) JE01 inhibited ETEC-induced production of inflammatory cytokines in IPEC-J2 cells. The levels of the inflammatory cytokines IL-6 and IL-8 were determined. For (<b>A</b>–<b>C</b>), experiments were conducted in triplicate, and the results are shown as the mean ± SEM. Statistical significance was determined using an unpaired <span class="html-italic">t</span> test (<b>A</b>) and one-way ANOVA (Tukey’s multiple comparisons test) for multiple comparisons (<b>B</b>,<b>C</b>). * <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, ns = not significant.</p>
Full article ">Figure 5
<p>The protective effect of the phage JE01 on an ETEC-induced mouse diarrhea model. Mice that developed diarrhea due to ETEC U74 challenge were dosed by oral gavage with the JE01 phage or GEN, and the survival rate (<b>A</b>), diarrhea scores (<b>B</b>), and bacterial load of jejunum samples (<b>C</b>) were recorded at each of the selected time points. The transcriptional levels of IL-6, IL-8, and TNF-α (<b>D</b>) in the jejunum tissues were measured using qRT-PCR. For (<b>A</b>), Blue (PBS Control) and black line (PBS GEN) coincided with Blue dotted line (PBS JE01), which of them showed 100% survival. statistical significance was determined using log-rank Mantel–Cox tests for multiple comparisons (<span class="html-italic">n</span> = 15/group). For (<b>B</b>), data are shown as five murine biological replicates’ mean ± SEM. For (<b>C</b>), data are shown as three murine biological replicates’ mean ±SEM at each of the selected time points. For (<b>D</b>–<b>F</b>), data are shown as three murine biological replicates’ mean ± SEM at 72 h after infection. Statistical significance was determined using one-way ANOVA with Tukey’s multiple comparisons test. * <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; 0.0001, ns = not significant.</p>
Full article ">Figure 6
<p>JE01 reduced ETEC-induced intestinal lesions in mice. Mice were infected with ETEC and treated with PBS, JE01, or GEN. Uninfected mice were treated with JE01, GEN, or PBS (PBS Control) and served as controls. Jejunum tissues were collected and processed for H&amp;E staining and microscopic examination. The red arrows indicate villous damage. Magnification: 100×; scale bars: 200 nm.</p>
Full article ">Figure 7
<p>Fluorescence of RFP-displaying phages in the murine jejunum tissues after phage treatment. ETEC-infected jejunum tissues were dissected from mice (N = 3/time points) 15, 30, 45, 90, and 150 min after oral gavage of Phage<sup>RFP</sup> (Group C). Uninfected mice treated with PBS (Group A) or Phage<sup>RFP</sup> (Group B) served as controls; Group A served as the background control to adjust for relative fluorescence values across groups, and regions of interest (ROIs) were selected to assess the average fluorescence intensity per unit area. Avg Radiant Efficiency = <math display="inline"><semantics> <mrow> <mstyle scriptlevel="0" displaystyle="true"> <mfrac> <mrow> <mrow> <mi mathvariant="normal">p</mi> <mo>/</mo> <mi>sec</mi> <mo>/</mo> </mrow> <msup> <mrow> <mi mathvariant="normal">c</mi> <mi mathvariant="normal">m</mi> </mrow> <mrow> <mn>2</mn> </mrow> </msup> <mo>/</mo> <mi mathvariant="normal">s</mi> <mi mathvariant="normal">r</mi> </mrow> <mrow> <mi mathvariant="sans-serif">μ</mi> <mrow> <mi mathvariant="normal">w</mi> <mo>/</mo> </mrow> <msup> <mrow> <mi mathvariant="normal">c</mi> <mi mathvariant="normal">m</mi> </mrow> <mrow> <mn>2</mn> </mrow> </msup> </mrow> </mfrac> </mstyle> </mrow> </semantics></math>. Data are shown as three murine biological replicates’ mean ± SEM at every time points. Statistical significance was determined using one-way ANOVA with Tukey’s multiple comparisons test. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, ns = not significant.</p>
Full article ">
Back to TopTop