[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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (3,499)

Search Parameters:
Keywords = ECMS

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
22 pages, 2905 KiB  
Review
Physical Exercise: A Promising Treatment Against Organ Fibrosis
by Xiaojie Ma, Bing Liu, Ziming Jiang, Zhijian Rao and Lifang Zheng
Int. J. Mol. Sci. 2025, 26(1), 343; https://doi.org/10.3390/ijms26010343 - 2 Jan 2025
Abstract
Fibrosis represents a terminal pathological manifestation encountered in numerous chronic diseases. The process involves the persistent infiltration of inflammatory cells, the transdifferentiation of fibroblasts into myofibroblasts, and the excessive deposition of extracellular matrix (ECM) within damaged tissues, all of which are characteristic features [...] Read more.
Fibrosis represents a terminal pathological manifestation encountered in numerous chronic diseases. The process involves the persistent infiltration of inflammatory cells, the transdifferentiation of fibroblasts into myofibroblasts, and the excessive deposition of extracellular matrix (ECM) within damaged tissues, all of which are characteristic features of organ fibrosis. Extensive documentation exists on fibrosis occurrence in vital organs such as the liver, heart, lungs, kidneys, and skeletal muscles, elucidating its underlying pathological mechanisms. Regular exercise is known to confer health benefits through its anti-inflammatory, antioxidant, and anti-aging effects. Notably, exercise exerts anti-fibrotic effects by modulating multiple pathways, including transforming growth factor-β1/small mother decapentaplegic protein (TGF-β1/Samd), Wnt/β-catenin, nuclear factor kappa-B (NF-kB), reactive oxygen species (ROS), microRNAs (miR-126, miR-29a, miR-101a), and exerkine (FGF21, irisin, FSTL1, and CHI3L1). Therefore, this paper aims to review the specific role and molecular mechanisms of exercise as a potential intervention to ameliorate organ fibrosis. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

Figure 1
<p>The pathogenesis of fibrosis: After tissue injury, immune cells (mainly macrophages) are activated and release cytokines (e.g., IL-4, IL-13, PDGF, TGF-β, etc.). Through signaling pathways such as TGF-β/Smad and Wnt/β-catenin, fibroblasts are transformed into myofibroblasts, and myofibroblasts produce a large amount of ECM, leading to the generation of fibrosis. IL-4: interleukin-4; IL-13: interleukin-13; IL-1β: interleukin-1β; TGF-β: transforming growth factor-β; smad: small mother decapentaplegic protein; PDGF: platelet-derived growth factor-D; ERK: extracellular signal-regulated kinase; p38: p38 mitogen-activated protein kinases; JNK: c-Jun N-terminal kinases; TAK1: transforming growth factor-β (TGF-β)-activated kinase 1; α-SMA: α-smooth muscle actin; COL1: Collagen 1; COL3: Collagen 3; MMP: matrix metallopeptidase; TIMP-1: tissue inhibitor of metal protease1. Created with Figdraw (<a href="http://www.figdraw.com" target="_blank">www.figdraw.com</a>), license ID: TTWOW4366f.</p>
Full article ">Figure 2
<p>The mechanisms of exercise modulation of pulmonary fibrosis. Exercise ameliorates paraquat-induced pulmonary fibrosis by impeding the Wnt/β-catenin pathway, dampening inflammation, oxidative stress, and EMT. In bleomycin-induced pulmonary fibrosis, exercise alleviates fibrosis by enhancing endogenous hydrogen sulfide (H<sub>2</sub>S) synthesis, thereby inhibiting the LRP-6/β-catenin and TGF-β1 signaling pathways or reducing lung inflammation and EMT via the suppression of serotonin (5-HT) and Akt phosphorylation. Additionally, in silica-induced silicosis, exercise attenuates lung fibrosis by suppressing the TLR4-TNF-α and SRB-NLRP3 pathways and further inhibiting the IL-17A-CXCL5-CXCR2 inflammatory axis. SRB: scavenger receptor B; NLPR3: NOD-like receptor thermal protein domain associated protein 3; TLR4: Toll-like receptor 4; TNF-α: tumor necrosis factor; IL-17A: interleukin-17A; CXCL5: CXC motif chemokine ligand 5; CXCR2: Chemokine (C-X-C motif) Receptor 2; 5-HT: serotonin; AKT: protein kinase B; LRP-6: low-density lipoprotein receptor-related proteins; H<sub>2</sub>S: hydrogen sulfide; TGF-β1: transforming growth factor-β1; smad: small mother decapentaplegic protein. Created with Figdraw (<a href="http://www.figdraw.com" target="_blank">www.figdraw.com</a>), license ID: PYPIP737ba.</p>
Full article ">Figure 3
<p>Mechanisms of exercise modulation of renal fibrosis. Exercise exerts its beneficial effects on renal fibrosis through various mechanisms: Exercise suppresses NOX4-dependent ROS production in the kidney, thereby inhibiting the NF-κB/NLPR3 inflammasome pathway, or by targeting Sirt1, which ultimately ameliorates renal fibrosis associated with diabetic nephropathy. Exercise inhibits the TGF-β/Smad pathway or reduces Ang II content, diminishes AT1R and Ang II binding, and inhibits the Ang II-AT1R-TGF-β pathway. These actions contribute to the mitigation of renal fibrosis development in hypertensive conditions. In aging kidneys, exercise improves renal fibrosis by inhibiting the TGF-β1/TAK1/MKK3/p38 MAPK signaling pathway, enhancing autophagy activity, and delaying the epithelial–mesenchymal transition, or activating PPAR α to reduce oxidative stress, inflammation, and lipid accumulation by inhibiting the expression of miR-21 and miR-34a. TGF-β1: transforming growth factor-β1; TAK1: transforming growth factor-β (TGF-β)-activated kinase 1; MKK3: mitogen-activated protein kinase (MAPK) kinase; p38MAPK: p38 mitogen-activated protein kinase; NOX4: NADPH oxidase 4; ROS: reactive oxygen species; NF-κB: nuclear factor kappa-B; NLPR3: NOD-like receptor thermal protein domain associated protein 3; AngⅡ: angiotensin II; AT1R: Ang II-angiotensin II type I receptor; α-SMA: α-smooth muscle actin; CTGF: connective tissue growth factor; Sirt1: silent information regulator 1; H<sub>2</sub>S: hydrogen sulfide; PGC-1α: peroxisome proliferator-activated receptor gamma coactivator 1-α; miR-21: microRNA-21; miR-34a: microRNA-34a. Created with Figdraw (<a href="http://www.figdraw.com" target="_blank">www.figdraw.com</a>), license ID: TRPRT70cb4.</p>
Full article ">Figure 4
<p>Mechanisms of exercise modulation of myocardial fibrosis. Aging, myocardial infarction, metabolic diseases, and hypertension can all lead to myocardial fibrosis. Aging: Exercise reduces collagen deposition by enhancing MMP-2 activity and decreasing the expression of fibrosis-associated factors (TGF-β1, TIMP-1, COL-I). It also restores endogenous H<sub>2</sub>S levels or inhibits the FGF-2/uPA/MMP-2 signaling pathway. Metabolic diseases: Exercise inhibits the TGF-β1/Smad signaling pathway or reduces ROS production, promoting HO-1 expression and inhibiting fibrosis-related factors. Myocardial infarction: Exercise inhibits TGF-β1 signaling through the NRG-1/ErbB signaling pathway or by upregulating miR-29a, miR-101a, and FGF-21 expression. In addition, exercise can also secrete several myokines such as irisin, CHI3L1, and FSTL1 to ameliorate myocardial fibrosis after myocardial infarction. Hypertension: Exercise attenuates myocardial fibrosis by inhibiting the LOXL-2/TGF-β signaling pathway and the expression of AT1R and FGF23, or by promoting the expression of ccdc80tide and AMPKα1. MMP: matrix metallopeptidase; TGF-β1: transforming growth factor-β1; TIMP-1: tissue inhibitor of metal protease1; H2S: hydrogen sulfide; COL-Ⅰ: Collagen 1; FGF-2: fibroblast growth factor 2; uPA: urokinase-type plasminogen activator; NRG1: Neuregulin 1; FGF-21: fibroblast Growth Factor 21; miR-29a: microRNA-29a; miR-101a: microRNA-101a; smad: small mother decapentaplegic protein; ROS: reactive oxygen species; CTGF: connective tissue growth factor; HO-1: heme oxygenase 1; JAK: janus kinase 2; STAT3: signal transducer and activator of transcription 3; AMPKα1: AMP-activated protein kinase α1; Sirt1: silent information regulator 1; PGC-1α: peroxisome proliferator-activated receptor gamma coactivator 1-α; AT1R: Ang II-angiotensin II type I receptor; FGF-23: fibroblast Growth Factor 23; LOXL2: Lysyl oxidase-like 2; miR-34a: microRNA-34a; miR-486a-5p: microRNA-486a-5p; miR-29: microRNA-29; miR-133: microRNA-133; TNF-α: tumor necrosis factor; NF-kB: nuclear factor kappa-B; AKT: protein kinase B; PI3K: phosphoinositide 3-kinase; ALCAT1: lysocardiolipin acyltransferase-1. Created with Figdraw (<a href="http://www.figdraw.com" target="_blank">www.figdraw.com</a>), license ID: PRPRW1c841.</p>
Full article ">Figure 5
<p>Antifibrotic mechanisms of exercise: Exercise exerts its anti-fibrotic effects by directly or indirectly (secreting exerkines or targeting microRNAs) affecting multiple signaling pathways associated with fibrogenesis. Created with Figdraw (<a href="http://www.figdraw.com" target="_blank">www.figdraw.com</a>), license ID: IYAUA656e8.</p>
Full article ">
13 pages, 832 KiB  
Article
Biomarkers of Extracellular Matrix Fragments in Patients with Psoriasis
by Mila Broby Johansen, Signe Holm Nielsen, Helena Port, Tanja Todberg, Marianne Bengtson Løvendorf and Lone Skov
Int. J. Mol. Sci. 2025, 26(1), 261; https://doi.org/10.3390/ijms26010261 - 30 Dec 2024
Viewed by 294
Abstract
Blood-based extracellular matrix (ECM) fragments have been identified as potential pharmacologic biomarkers in spondyloarthritis and diagnostic biomarkers in psoriatic arthritis and psoriasis vulgaris. This study aimed to explore whether ECM fragments can differentiate patients with psoriasis from healthy controls (HC) and determine their [...] Read more.
Blood-based extracellular matrix (ECM) fragments have been identified as potential pharmacologic biomarkers in spondyloarthritis and diagnostic biomarkers in psoriatic arthritis and psoriasis vulgaris. This study aimed to explore whether ECM fragments can differentiate patients with psoriasis from healthy controls (HC) and determine their potential as biomarkers for response to treatment in psoriasis. The study population included 59 patients with moderate to severe psoriasis, not receiving systemic anti-psoriatic treatment at inclusion, and 52 HC matched by age, sex, and BMI. An EDTA plasma sample was taken from all subjects at inclusion. Nine patients with psoriasis who initiated treatment with adalimumab after inclusion and responded successfully had an additional EDTA plasma sample taken after three to six months. Twelve ECM fragments were measured using validated ELISAs and Immunodiagnostic Systems automated chemiluminescent assays. C4M, indicating collagen IV degradation, PRO-C3, indicating tissue fibrosis, and PRO-C4, indicating epidermal basement membrane turnover showed significantly elevated levels in psoriasis patients compared with HC (p = 0.005, p = 0.016, and p = 0.018, respectively). Despite successful treatment, adalimumab did not alter C4M, PRO-C3, or PRO-C4 levels. In conclusion, compared with controls, C4M, PRO-C3, and PRO-C4 were elevated in psoriasispatients, but treatment did not modulate these fragments. Full article
Show Figures

Figure 1

Figure 1
<p>Comparison of ECM fragment levels between patients with PSO (PSO) and healthy controls (HC). Plasma levels of 12 ECM fragments were measured in healthy controls, (HC, n = 52), and patients with psoriasis (PSO, n = 59). We observed significantly elevated levels of C4M, assessing collagen IV degradation (<span class="html-italic">p</span> = 0.005), PRO-C3, assessing tissue fibrosis (<span class="html-italic">p</span> = 0.016), and PRO-C4, assessing epidermal basement membrane turnover (<span class="html-italic">p</span> = 0.018), in patients with PSO compared with HC (<b>E</b>,<b>G</b>,<b>H</b>). (<b>A</b>–<b>C</b>): Elastin degradation and neutrophil activity (ELP-3), T-cell activity (C4G), neutrophil activity (CPa9-HNE). (<b>D</b>–<b>F</b>): Catabolic ECM fragments of type III collagen (C3M), type IV collagen (C4M), type VI (C6M). (<b>G</b>–<b>L</b>): Anabolic ECM fragments of type III collagen (PRO-C3), type IV collagen (PRO-C4), type VI collagen (PRO-C6), type VII collagen (PRO-C7), type XVII (PRO-C17), and type XXII collagen (PRO-22). The significance threshold was set at <span class="html-italic">p</span> &lt; 0.05 *, and data are presented as scatterplots with bars and lines at the median.</p>
Full article ">Figure 2
<p>Levels of EMC fragments C4M, PRO-C3, and PRO-C4 before and after successful treatment response in patients with PSO. Plasma levels of three ECM fragments were measured at inclusion and 3–6 months after adalimumab treatment in patients with PSO with a successful treatment response (n = 9). No significant change was observed in C4M, PRO-C3, and PRO-C4 before and after treatment in the group with a successful response. (<b>A</b>): Catabolic ECM fragments of type IV collagen (C4M). (<b>B</b>,<b>C</b>): Anabolic ECM fragments of type III collagen (PRO-C3) and type IV collagen (PRO-C4). The significance threshold was set at <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">
24 pages, 9508 KiB  
Article
From High Protection to Lethal Effect: Diverse Outcomes of Immunization Against Invasive Candidiasis with Different Candida albicans Extracellular Vesicles
by Raquel Martínez-López, Gloria Molero, Claudia Marcela Parra-Giraldo, Matías Sebastián Cabeza, Guillermo Castejón, Carmen García-Durán, Luis Felipe Clemente, María Luisa Hernáez, Concha Gil and Lucía Monteoliva
Int. J. Mol. Sci. 2025, 26(1), 244; https://doi.org/10.3390/ijms26010244 - 30 Dec 2024
Viewed by 294
Abstract
Extracellular vesicles (EVs) from Candida albicans can elicit immune responses, positioning them as promising acellular vaccine candidates. We characterized EVs from an avirulent C. albicans cell wall mutant (ecm33Δ) and evaluated their protective potential against invasive candidiasis. EVs from the yeast [...] Read more.
Extracellular vesicles (EVs) from Candida albicans can elicit immune responses, positioning them as promising acellular vaccine candidates. We characterized EVs from an avirulent C. albicans cell wall mutant (ecm33Δ) and evaluated their protective potential against invasive candidiasis. EVs from the yeast (YEVs) and hyphal (HEVs) forms of the SC5314 wild-type strain were also tested, yielding high survival rates with SC5314 YEV (91%) and ecm33 YEV immunization (64%). Surprisingly, HEV immunization showed a dual effect, resulting in 36% protection but also causing premature death in some mice. Proteomic analyses revealed distinct profiles among the top 100 proteins in the different EVs, which may explain these effects: a shared core of 50 immunogenic proteins such as Pgk1, Cdc19, and Fba1; unique, relevant immunogenic proteins in SC5314 YEVs; and proteins linked to pathogenesis, like Ece1 in SC5314 HEVs. Sera from SC5314 YEV-immunized mice showed the highest IgG2a titers and moderate IL-17, IFN-γ, and TNF-α levels, indicating the importance of both humoral and cellular responses for protection. These findings highlight the distinct immunogenic properties of C. albicans EVs, suggesting their potential in acellular vaccine development while emphasizing the need to carefully evaluate pathogenic risks associated with certain EVs. Full article
(This article belongs to the Special Issue Fungal Pathogen-Host Interactions)
Show Figures

Figure 1

Figure 1
<p>NTA analysis of SC5314 YEVs and <span class="html-italic">ecm33</span> YEVs. TEM images showing spherical electron-dense bilayered structures, typical of extracellular vesicles, are provided for visual reference. The X10, X50, and X90 indicate a 10%, 50%, or 90% of EVs, respectively, with the specified size or smaller.</p>
Full article ">Figure 2
<p>(<b>a</b>) Functional enrichment analysis for proteins identified in SC5314 YEVs (1219 proteins) and <span class="html-italic">ecm33</span> YEVs (1333 proteins). The word clouds represent enriched terms based on the Fungal Database (CGD GO) analysis. These terms correspond to cellular compartments and are derived from Gene Ontology (GO). The size and color intensity of the terms indicate the significance of the enrichment, with larger and darker terms representing lower <span class="html-italic">p</span>-values. (<b>b</b>) Quantitative comparison of proteins present in EVs from both strains. The volcano plot illustrates proteins with significant differences in abundance between EV types (log2(fold change) vs. −log10(q-value)). Proteins more abundant in SC5314 YEVs or ecm33 YEVs are highlighted in blue and pink, respectively. Additionally, functional enrichment analyses were performed on the differentially abundant protein sets, focusing on “cellular component” (<b>top</b>) and “biological process” (<b>bottom</b>). The red asterisks indicate cellular components or biological processes related to the cell wall or cell surface.</p>
Full article ">Figure 3
<p>Vaccination schedule (<b>a</b>) and survival curves (<b>b</b>) of a mouse model of IC, showing the protective effect of immunization with different <span class="html-italic">C. albicans</span> EVs. Mice were immunized with different doses of EVs with adjuvant (Ad) or without adjuvant and subsequently challenged with an intravenous lethal dose of SC5314 (1 × 10<sup>6</sup> cells). Statistical differences between immunization and control groups are marked with an asterisk. * Mantel–Cox test, <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 4
<p>Vaccination schedule (<b>a</b>) and survival curves (<b>b</b>) of a murine model of IC, showing the protective effect achieved with immunization with SC5314 YEVs and <span class="html-italic">ecm33</span> YEVs. Mice were immunized with different doses of EVs with adjuvant (Ad) and subsequently challenged with an intravenous lethal dose of SC5314 (1 × 10<sup>6</sup> cells). Vaccination with live cells (2.5 × 10<sup>6</sup> cells, one dose) of the completely avirulent <span class="html-italic">ecm33</span>Δ mutant was included as a positive vaccination control (<b>a</b>,<b>b</b>). * Mantel–Cox test, <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 5
<p>Vaccination schedule (<b>a</b>) and survival curves (<b>b</b>) of a mouse model of invasive candidiasis, showing the protective effect of immunization with different <span class="html-italic">C. albicans</span> EVs. Mice were immunized with different doses of EVs with adjuvant (Ad) and subsequently challenged with an intravenous lethal dose of SC5314 (5 <span class="html-italic">×</span> 10<sup>5</sup> cells). Mantel–Cox test, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.005, *** <span class="html-italic">p</span> &lt; 0.0005 (black asterisks represent statistically significant differences between the control group and the different EV-immunized groups. Red asterisks represent statistically significant differences in the protection acquired between the different EV-immunized groups).</p>
Full article ">Figure 6
<p>(<b>a</b>) Hierarchical heatmap depicting the relative abundance of each protein across the three different types of EVs, with darker shades of red indicating higher relative abundance (measured by NSAF) (protein names shown represent 1 out of every 22 proteins for clarity). (<b>b</b>) Zoomed-in view on the region of the heatmap with a higher abundance in cell surface proteins in SC5314 YEVs. Proteins described as immunogenic in the Candida Genome Database (CGD) are marked with an asterisk.</p>
Full article ">Figure 7
<p>Venn diagram illustrating the top 100 most abundant proteins in each type of extracellular vesicle (EV). Proteins previously reported as immunogenic in other studies are highlighted in bold. Proteins associated with virulence are shaded.</p>
Full article ">Figure 8
<p>IgG and IgG2a antibody titers. ELISAs with pooled sera obtained prior to infection from immunized mice with SC5314 YEVs (blue), <span class="html-italic">ecm33</span> YEVs (pink), and SC5314 HEVs (green). Protein samples from SC5314 YEVs, SC5314 HEVs, and total cytoplasmic extracts from the wild-type strain SC5314 were used for detection purposes. Statistically significant differences in antibody titers were observed across all groups compared to the control group. Two-way ANOVA test, ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 9
<p>Cytokine profile for each of the pooled sera obtained after complete immunization with the different EVs (SC5314 YEVs (blue), <span class="html-italic">ecm33</span> YEVs (pink), and SC5314 HEVs (green)) but prior to infection (immunized), and 30 days after infection (surviving). Adjuvant group (control group) received only the adjuvant. Statistically significant differences in cytokine levels were observed across all cases compared to the control group. Two-way ANOVA test, *** <span class="html-italic">p</span> &lt;0.001, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">
14 pages, 2011 KiB  
Article
Characterizing Viscoelasticity of Corneal Stromal Models Using Non-Contact Air-Pulse Optical Coherence Elastography (OCE) and Validating Using Ramp–Hold Relaxation Testing
by Yilong Zhang, Zhengshuyi Feng, Zhihong Huang and Chunhui Li
Photonics 2025, 12(1), 24; https://doi.org/10.3390/photonics12010024 (registering DOI) - 30 Dec 2024
Viewed by 190
Abstract
Corneal biomechanical properties are closely related to the cornea’s physiological and pathological conditions, primarily determined by the stromal layer. However, little is known about the influence of corneal cell interaction on the viscoelasticity of the stromal extracellular matrix (ECM). In this study, collagen-based [...] Read more.
Corneal biomechanical properties are closely related to the cornea’s physiological and pathological conditions, primarily determined by the stromal layer. However, little is known about the influence of corneal cell interaction on the viscoelasticity of the stromal extracellular matrix (ECM). In this study, collagen-based hydrogels incorporated with keratocytes were reconstructed as corneal stromal models. Air-pulse optical coherence elastography (OCE) was used to characterize the viscoelastic properties of the corneal models. Plate compression, ramp–hold relaxation testing was performed on the initial corneal models. The findings demonstrated that the elastic modulus increased 5.27, 2.65 and 1.42 kPa, and viscosity increased 0.22, 0.06 and 0.09 Pa·s in the stromal models with initial collagen concentrations of 3, 5, and 7 mg/mL over 7 days. The elastic modulus and viscosity exhibited high correlation coefficients between air-pulse OCE and ramp–hold relaxation testing, with 92.25% and 98.67%, respectively. This study enhances the understanding of the influence of cell–matrix interactions on the corneal viscoelastic properties and validates air-pulse OCE as an accurate method for the mechanical characterization of tissue-engineered materials. Full article
Show Figures

Figure 1

Figure 1
<p>Dome-shaped (blue arrows) and cylindrical shape (green arrows) collagen hydrogels with initial collagen concentrations of 3 mg/mL, 5 mg/mL and 7 mg/mL.</p>
Full article ">Figure 2
<p>Schematic of an air-pulse OCE system for the SAW imaging in stromal model. (<b>A</b>) an air-pulse generation part, and (<b>B</b>): a PhS-OCT system PC: polarization controller; DAQ: data acquisition; NI: national instrument.</p>
Full article ">Figure 3
<p>Signal-processing steps of a 5 mg/mL cell-seeded stromal model on day 1. (<b>A</b>) The central cross-sectional structural image. (<b>B</b>) The structural image of the yellow dashed rectangle. The red and blue solid lines illustrate the boundaries of the analyzed region. (<b>C</b>) The spatial–temporal displacement map of the impulse SAW. (<b>D</b>) The wavenumber–frequency domain map of the impulse SAW. (<b>E</b>) The phase velocity dispersion curve (red solid line) overlaps the grayscale intensity map. (<b>F</b>) fitting the dispersion curve (black solid line) into the Rayleigh wave dispersion model. Red solid line: fitting curve; <span class="html-italic">μ</span><sub>1</sub>: shear modulus; <span class="html-italic">μ</span><sub>2</sub>: shear viscosity; R<sup>2</sup>: coefficient of determination.</p>
Full article ">Figure 4
<p>Experimental apparatus for the plate compression, ramp–hold stress relaxation test. Cylindrical stromal model placed on the testing plate of a Tinius Olsen H5KS material test machine.</p>
Full article ">Figure 5
<p>Light microscopic images of keratocytes in cell-seeded stromal models with initial collagen concentrations of 3 mg/mL (<b>A</b>,<b>B</b>), 5 mg/mL (<b>C</b>,<b>D</b>) and 7 mg/mL (<b>E</b>,<b>F</b>) on day 3 and day 7. Scale bar = 500 μm.</p>
Full article ">Figure 6
<p>Central thickness of cell-seeded (red dashed line) and no-cell (black solid line) stromal models with initial collagen concentrations of 3 mg/mL (<b>A</b>), 5 mg/mL (<b>B</b>) and 7 mg/mL (<b>C</b>) in 7 days.</p>
Full article ">Figure 7
<p>Elastic modulus (<b>A</b>–<b>C</b>) and viscosity (<b>D</b>–<b>F</b>) of no-cell (black solid line) and cell-seeded (red dashed line) stromal models with initial collagen concentrations of 3 mg/mL (<b>A</b>,<b>D</b>), 5 mg/mL (<b>B</b>,<b>E</b>) and 7 mg/mL (<b>C</b>,<b>F</b>).</p>
Full article ">Figure 8
<p>Plate compression, ramp–hold relaxation curves for no-cell stromal models with initial collagen concentrations of 3 mg/mL, 5 mg/mL and 7 mg/mL with corresponding KVFD model fitting.</p>
Full article ">
31 pages, 2005 KiB  
Review
Surface Molecular Markers for the Isolation of Viable Fibroblast Subpopulations in the Female Reproductive Tract: A Comprehensive Review
by Krzysztof Łuszczyński, Michał Komorowski, Marta Soszyńska, Paulina Lewandowska, Robert Zdanowski, Monika Szafarowska, Paweł Kamiński, Marcin Niemcewicz, Jacek Malejczyk, Anna Lutyńska and Aneta Ścieżyńska
Int. J. Mol. Sci. 2025, 26(1), 233; https://doi.org/10.3390/ijms26010233 - 30 Dec 2024
Viewed by 254
Abstract
Advancements in single-cell analyzis technologies, particularly single-cell RNA sequencing (scRNA-seq) and Fluorescence-Activated Cell Sorting (FACS), have enabled the analyzis of cellular diversity by providing resolutions that were not available previously. These methods enable the simultaneous analyzis of thousands of individual transcriptomes, facilitating the [...] Read more.
Advancements in single-cell analyzis technologies, particularly single-cell RNA sequencing (scRNA-seq) and Fluorescence-Activated Cell Sorting (FACS), have enabled the analyzis of cellular diversity by providing resolutions that were not available previously. These methods enable the simultaneous analyzis of thousands of individual transcriptomes, facilitating the classification of cells into distinct subpopulations, based on transcriptomic differences, adding a new level of complexity to biomolecular and medical research. Fibroblasts, despite being one of the most abundant cell types in the human body and forming the structural backbone of tissues and organs, remained poorly characterized for a long time. This is largely due to the high morphological similarity between different types of fibroblasts and the lack of specific markers to identify distinct subpopulations. Once thought to be cells responsible solely for the synthesis of extracellular matrix (ECM) components, fibroblasts are now recognized as active participants in diverse physiological processes, including inflammation and antimicrobial responses. However, defining the molecular profile of fibroblast subpopulations remains a significant challenge. In this comprehensive review, which is based on over two thousand research articles, we focus on the identification and characterization of fibroblast subpopulations and their specific surface markers, with an emphasis on their potential as molecular targets for selective cell isolation. By analyzing surface markers, alongside intra- and extracellular protein profiles, we identified multiple fibroblast subtypes within the female reproductive system. These subtypes exhibit distinct molecular signatures and functional attributes, shaped by their anatomical localization and the surrounding physiological or pathological conditions. Our findings underscore the heterogeneity of fibroblasts and their diverse roles in various biological contexts. This improved understanding of fibroblast subpopulations paves the way for innovative diagnostic and therapeutic strategies, offering the potential for precision targeting of specific fibroblast subsets in clinical applications. Full article
(This article belongs to the Special Issue Molecular Advances in Obstetrical and Gynaecological Disorders)
Show Figures

Figure 1

Figure 1
<p>Flowchart of the selection process for the examined research publications.</p>
Full article ">Figure 2
<p>A graphical summary of various fibroblast subpopulations derived from healthy tissue and their corresponding surface markers. The major fibroblast subpopulations derived from the endometrium were endometrial stromal fibroblasts and differentiated fibroblasts, each characterized by a unique panel of surface markers. Moreover, fibroblasts with a similar profile to endometrial stromal fibroblasts were isolated from menstrual blood. Additionally, two distinct subtypes were isolated from myometrium and uterine ligaments. Created in BioRender.</p>
Full article ">Figure 3
<p>A graphical summary of various fibroblast subpopulations derived from benign gynecological diseases and their corresponding surface markers. The majority of research was conducted on fibroblasts derived from endometriosis tissue. Endometriosis-derived fibroblasts possess a surface marker profile similar to fibroblasts derived from healthy tissue, with a few unique surface markers. Other studied diseases were uterine leiomyoma and pelvic organ prolapse. A couple of unique fibroblast subtypes were described in regard to both of these diseases. Created in BioRender.</p>
Full article ">Figure 4
<p>A graphical summary of numerous cancer-associated fibroblasts derived from gynecological cancers and their corresponding surface markers. Fibroblasts derived from cancer tissue show characteristics of cancer-associated fibroblasts (CAFs), with significant differences in the surface marker panel in comparison to fibroblasts derived from healthy tissue. The most prevalent cancer-associated fibroblast subtypes were myofibroblast CAFs, inflammatory CAFs, and antigen-presenting CAFs; among different tumors, they had similar, but not identical, expression profiles. Moreover, in regard to some tumors, unique cancer-associated fibroblast subpopulations were described. Created in BioRender.</p>
Full article ">
16 pages, 10700 KiB  
Article
Systematic Evaluation of Extracellular Coating Matrix on the Differentiation of Human-Induced Pluripotent Stem Cells to Cortical Neurons
by Siyao Li, Yan Liu, Xianyang Luo and Wei Hong
Int. J. Mol. Sci. 2025, 26(1), 230; https://doi.org/10.3390/ijms26010230 - 30 Dec 2024
Viewed by 222
Abstract
Induced pluripotent stem cell (iPSC)-derived neurons (iNs) have been widely used as models of neurodevelopment and neurodegenerative diseases. Coating cell culture vessels with extracellular matrixes (ECMs) gives structural support and facilitates cell communication and differentiation, ultimately enhances neuronal functions. However, the relevance of [...] Read more.
Induced pluripotent stem cell (iPSC)-derived neurons (iNs) have been widely used as models of neurodevelopment and neurodegenerative diseases. Coating cell culture vessels with extracellular matrixes (ECMs) gives structural support and facilitates cell communication and differentiation, ultimately enhances neuronal functions. However, the relevance of different ECMs to the natural environment and their impact on neuronal differentiation have not been fully characterized. In this study, we report the use of four commonly used extracellular matrixes, poly-D-lysine (PDL), poly-L-ornithine (PLO), Laminin and Matrigel, which we applied to compare the single-coating and double-coating conditions on iNs differentiation and maturation. Using the IncuCyte live-cell imaging system, we found that iNs cultured on single Matrigel- and Laminin-coated vessels have significantly higher density of neurite outgrowth and branch points than PLO or PDL but produce abnormal highly straight neurite outgrowth and larger cell body clumps. All the four double-coating conditions significantly reduced the clumping of neurons, in which the combination of PDL+Matrigel also enhanced neuronal purity. Double coating with PDL+Matrigel also tended to improve dendritic and axonal development and the distribution of pre and postsynaptic markers. These results demonstrate that the extracellular matrix contributes to the differentiation of cultured neurons and that double coating with PDL+Matrigel gives the best outcomes. Our study indicates that neuronal differentiation and maturation can be manipulated, to a certain extent, by adjusting the ECM recipe, and provides important technical guidance for the use of the ECM in neurological studies. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

Figure 1
<p>iNs cultured on Matrigel- and Laminin-coated vessels have a significantly higher density of neurite and branch points than PLO or PDL. iNs were cultured on a 96-well plate coated with a single matrix and their differentiation was monitored using the IncuCyte live-cell imaging system. (<b>A</b>) Representative images of iNs cultured for 17 days. The top panel shows phase-contrast images, the middle panel shows neurons identified by the Incucyte NeuroTrack algorithm (purple), and the bottom panel shows composite images of the identified neurons and phase-contrast field. Scale bars are 100 μm. (<b>B</b>,<b>C</b>) iNs were monitored continuously from day 4 to 17 and images were collected at 24 h intervals. The neurite length and branch points at each time points were quantified using the NeuroTrack algorithm. The values shown represent the mean (±SD) from triplicate wells for each treatment. Single coatings of Laminin (orange) and Matrigel (blue) resulted in significantly higher neurite length and branch points in iNs compared to PDL (purple) and PLO (green).</p>
Full article ">Figure 2
<p>iNs cultured under different double-coating conditions yield similar and robust neurite outgrowth. iNs were cultured on a 96-well plate with double-coating matrixes and their differentiation monitored using the IncuCyte live-cell imaging system. (<b>A</b>) Representative images of iNs cultured for 17 days. The top panel shows phase-contrast images, the middle panel shows neurons identified by the Incucyte NeuroTrack algorithm (purple), and the bottom panel shows composite images of the identified neurons and the phase-contrast field. Scale bars are 100 μm. (<b>B</b>,<b>C</b>) iNs were monitored continuously from day 4 to 17 and images were collected at 24 h intervals. Neurite length and branch points at each time points were quantified using the NeuroTrack algorithm. The values shown represent the mean (±SD) from triplicate wells for each treatment. No significant differences in iNs differentiation were observed between double coatings of PDL+Laminin (purple), PDL+Matrigel (green), PLO+Laminin (orange) and PLO+Matrigel (blue).</p>
Full article ">Figure 3
<p>PDL+Matrigel double coating significantly reduces clumping of induced neurons. Images were collected at 2–4 days intervals from iNs day 4 to 17 and the area of cell body clusters was identified and quantified by the Incucyte NeuroTrack algorithm. (<b>A</b>) Representative images of iNs on day 17 and cell body clusters are masked (yellow). Scale bars are 100 μm. (<b>B</b>) Single-coating comparison shows that Laminin (orange) and Matrigel (blue) led to significantly more cell body clumps than PDL (purple) and PLO (green). (<b>C</b>) Among the four double-coating conditions, PDL+Matrigel (green) resulted in the lowest area of cell body clusters when compared to PDL+Laminin (purple), PLO+Laminin (orange) and PLO+Matrigel (blue). Only the cell body clusters with a size &gt;400 μm<sup>2</sup> were counted. The values represent the mean (±SD) from triplicate wells for each treatment.</p>
Full article ">Figure 4
<p>Comprehensive live-cell comparison of single-coating and double-coating conditions shows that PDL+Matrigel is the optimal substrate for long-term neuronal culture. Images collected at iNs day 10 and 17 (in <a href="#ijms-26-00230-f001" class="html-fig">Figure 1</a>, <a href="#ijms-26-00230-f002" class="html-fig">Figure 2</a> and <a href="#ijms-26-00230-f003" class="html-fig">Figure 3</a>) were statistically compared for their neurite length, neurite branch points and cell body cluster area. (<b>A</b>,<b>B</b>) Neurite length of iNs cultured at day 10 and 17 under four single-coating and four double-coating conditions. (<b>C</b>,<b>D</b>) Neurite branch points of iNs cultured at day 10 and 17 under four single-coating and four double-coating conditions. (<b>E</b>,<b>F</b>) Cell body cluster area of iNs cultured at day 10 and 17 under four single-coating and four double-coating conditions. Data represent the mean values (±SD) from triplicate wells for each condition. Single-coating conditions included Laminin, Matrigel, PDL and PLO. Double-coating conditions included PDL+Matrigel, PDL+Laminin, PLO+Laminin and PLO+Matrigel. Statistical analysis was conducted using one-way ANOVA followed by Tukey’s multiple comparison test. Significant differences are denoted as **** <span class="html-italic">p</span> &lt; 0.0001; *** <span class="html-italic">p</span> &lt; 0.001; * <span class="html-italic">p</span> &lt; 0.05; ns (not significant) <span class="html-italic">p</span> &gt; 0.05.</p>
Full article ">Figure 5
<p>PDL+Matrigel double-coating condition enhances the purity of induced neurons. At 17 days post-induction, iNs were fixed and stained for the mature neuronal marker NeuN (green). Cell nuclei were stained with DAPI (blue). Scale bar: 20 μm. (<b>A</b>,<b>B</b>) Representative immunofluorescence images collected at iNs day 17 show co-localization of NeuN and DAPI. (<b>C</b>) Quantification of NeuN-positive cells versus total cells shows the purity of iNs cultured under different coating conditions. Data represent the mean values (±SD) from triplicate wells for each condition. Single-coating conditions included PDL, PLO, Laminin and Matrigel. Double-coating conditions included PDL+Matrigel, PDL+Laminin, PLO+Laminin and PLO+Matrigel. Statistical analysis was conducted using one-way ANOVA followed by Tukey’s multiple comparison test. Significant differences are denoted as **** <span class="html-italic">p</span> &lt; 0.0001, *** <span class="html-italic">p</span> &lt; 0.001, ** <span class="html-italic">p</span> &lt; 0.01, * <span class="html-italic">p</span> &lt; 0.05, ns (not significant) <span class="html-italic">p</span> &gt; 0.05.</p>
Full article ">Figure 6
<p>Expression of neuronal and synaptic markers tends to be improved in iNs cultured under PDL+Matrigel double-coating condition. At 17 days post-induction, iNs were fixed and stained for an array of neuronal markers. (<b>A</b>) Representative images showing immunostaining for axons (K9JA, red) and dendrites (MAP2, green) under eight coating conditions. (<b>B</b>) Representative images showing immunostaining for presynaptic marker Synapsin-1 (Syn1, red) and postsynaptic marker Postsynaptic Density Protein 95 (PSD-95, green) under eight coating conditions. Scale bar: 20 μm.</p>
Full article ">
19 pages, 4256 KiB  
Article
Sulfated and Phosphorylated Agarose as Biomaterials for a Biomimetic Paradigm for FGF-2 Release
by Aurelien Forget and V. Prasad Shastri
Biomimetics 2025, 10(1), 12; https://doi.org/10.3390/biomimetics10010012 - 30 Dec 2024
Viewed by 270
Abstract
Cardiovascular diseases such as myocardial infarction or limb ischemia are characterized by regression of blood vessels. Local delivery of growth factors (GFs) involved in angiogenesis such as fibroblast blast growth factor-2 (FGF-2) has been shown to trigger collateral neovasculature and might lead to [...] Read more.
Cardiovascular diseases such as myocardial infarction or limb ischemia are characterized by regression of blood vessels. Local delivery of growth factors (GFs) involved in angiogenesis such as fibroblast blast growth factor-2 (FGF-2) has been shown to trigger collateral neovasculature and might lead to a therapeutic strategy. In vivo, heparin, a sulfated polysaccharide present in abundance in the extracellular matrix (ECM), has been shown to function as a local reservoir for FGF-2 by binding FGF-2 and other morphogens and it plays a role in the evolution of GF gradients. To access injectable biomaterials that can mimic such natural electrostatic interactions between soluble signals and macromolecules and mechanically tunable environments, the backbone of agarose, a thermogelling marine–algae-derived polysaccharide, was modified with sulfate, phosphate, and carboxylic moieties and the interaction and release of FGF-2 from these functionalized hydrogels was assessed by ELISA in vitro and CAM assay in ovo. Our findings show that FGF-2 remains active after release, and FGF-2 release profiles can be influenced by sulfated and phosphorylated agarose, and in turn, promote varied blood vessel formation kinetics. These modified agaroses offer a simple approach to mimicking electrostatic interactions experienced by GFs in the extracellular environment and provide a platform to probe the role of these interactions in the modulation of growth factor activity and may find utility as an injectable gel for promoting angiogenesis and as bioinks in 3D bioprinting. Full article
(This article belongs to the Special Issue Biomimetic Drug Delivery Systems 2024)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Chemical structure of the repeat unit of native agarose (NA), carboxylated agarose (CA), sulfated agarose (SA), and phosphorylated agarose (PA). (<b>B</b>) Comparison of the FTIR absorbance spectra of NA and SA. The black arrow points to the vibration of S-O (ν<sub>S = O</sub>) in SA. (<b>C</b>) Comparison of the FTIR absorbance spectra of native agarose (NA) and phosphorylated agarose (PA). The black arrow points to vibrations of POC (ν<sub>POC</sub>) and PO (ν<sub>PO</sub>) in PA. The IR bands at 1158 and 1071 cm<sup>−1</sup> correspond to –C–O–C– and glycosidic linkage [<a href="#B45-biomimetics-10-00012" class="html-bibr">45</a>], while the maxima around 1650 cm<sup>−1</sup> is attributed to polymer-bound water [<a href="#B46-biomimetics-10-00012" class="html-bibr">46</a>].</p>
Full article ">Figure 2
<p>Cumulative H-bond number during the molecular dynamic simulation. Graphs (<b>A</b>–<b>D</b>) show a comparison between H-bonds calculated between two polysaccharide strands and the number of H-bonds between the polysaccharide strands and surrounding water molecules. Graphs (<b>E</b>,<b>F</b>) show a comparison between the interaction potential between strands of the modified agaroses in comparison to native agarose, and how these inter-strand interactions impact H-bonding between the strand and water. Simulations were carried out using the TIP3P water model.</p>
Full article ">Figure 3
<p>(<b>A</b>) Circular dichroism (CD) of 0.1% <span class="html-italic">w</span>/<span class="html-italic">v</span> solution of native agarose (NA), carboxylated agarose (CA), phosphorylated agarose (PA), and sulfonated agarose (SA). (<b>B</b>) Shear modulus (G′) of 2% <span class="html-italic">w</span>/<span class="html-italic">v</span> hydrogels of native agarose (NA), carboxylated agarose (CA), phosphorylated agarose (PA), and sulfated agarose (SA). (<b>C</b>) Scanning electron micrographs of freeze-dried gels prepared from 2% <span class="html-italic">w</span>/<span class="html-italic">v</span> solutions.</p>
Full article ">Figure 4
<p>Mechanism of gelation of the NA (<b>left</b>) depicting the formation of helical bundles due to strong inter-strand H-bond in comparison to SA and PA (<b>right</b>) where the disruption of the helical structures lead to diminished polymer strand interactions and promote H-bonding with water molecules (red) resulting in weak gels.</p>
Full article ">Figure 5
<p>(<b>A</b>) FGF-2 release profile as measured by ELISA from the various modified agarose hydrogels (SA: sulfated agarose; PA: phosphate agarose, CA: carboxylated agarose) showing sustained release for a duration exceeding 1-week with appreciable differences in temporal and cumulative release in comparison to native agarose (NA) (** (<span class="html-italic">p</span> ≤ 0.01)). Data points represent average values with standard deviation (SD). SD bars are only visible in those that exceed size of the size of the symbol. One absorbance unit is 626 pg. All values beyond 10-h were found to be statistically significant with <span class="html-italic">p</span> values of ≤ 0.01 or ≤ 0.001 (see <a href="#biomimetics-10-00012-f0A5" class="html-fig">Figure A5</a> for <span class="html-italic">p</span>-values between various groups and time points) (<b>B</b>) Release profiles during the initial 10-h clearly showing the ability of these hydrogels to sequester FGF-2 to various degree. Release from PA beyond 2 h was found to be statistically significant, from all other conditions, and release from SA was statistically significantly different from CA and NA. (see <a href="#biomimetics-10-00012-f0A5" class="html-fig">Figure A5</a> for <span class="html-italic">p</span>-values between various groups and time points) (<b>C</b>) Release profile beyond 10 h showing linear behavior with similar slop from all hydrogels, suggesting that steady state release of FGF-2 is not impacted by charges characteristic of the hydrogels, implying that dissociation of the FGF-2 from polymer network is the limiting step. (<b>D</b>) The postulated release mechanism of FGF-2 from modified agarose hydrogels. In NA hydrogels (top cartoon) the dense polymer network due to strong interactions between agarose chain limits diffusion, while in the highly charged SA and PA hydrogels (bottom carton) electrostatic sequestration and expulsion dominate.</p>
Full article ">Figure 6
<p>(<b>A</b>) Photographs of vasculature around the different hydrogel on the chorioallantoic membrane (CAM) of a chick egg. (<b>B</b>) Quantification of blood vessels formed around the different hydrogels (native agarose (NA), carboxylated agarose (CA), phosphorylated agarose (PA), and sulfonated agarose (SA)) loaded with FGF-2 and positioned on the CAM over a gelatin mesh. (<b>C</b>–<b>E</b>) Temporal changes in blood vessel numbers showing a higher propensity around SA hydrogels loaded with FGF-2 in comparison to CA and NA, and a similar propensity in comparison to PA hydrogels despite a higher release of FGF-2 from PA hydrogels, suggesting a possible role for sulfonate groups in SA hydrogel in stabilizing FGF-2.</p>
Full article ">Figure A1
<p>FTR spectra of native agarose (NA) and carboxylated agarose (CA). The green arrow indicates the absorption bands at 1158 and 1071 cm<sup>−1</sup> corresponding to –C–O–C– and glycosidic linkage, the peak at 1650 cm<sup>−1</sup> denoted by the black arrow denotes polymer-bound water, and the shoulder at 1750 cm<sup>−1</sup> denoted by the red arrow corresponds to the carbonyl stretching in the carboxylic acid moieties in CA.</p>
Full article ">Figure A2
<p>Visualization of the molecular dynamic simulation at the beginning of the experiment (t = 0 ns) compared to the conformation of the polysaccharide strands at the end of the experiment (t = 15 ns).</p>
Full article ">Figure A3
<p>Zeta potential of the 0.1% <span class="html-italic">w</span>/<span class="html-italic">v</span> solution of native agarose (NA), carboxylated agarose (CA), phosphorylated agarose (PA), and sulfonated agarose (SA).</p>
Full article ">Figure A4
<p>Paired comparison of the FGF-2 release profile over the first 10-h from SA (sulfated agarose) with that from PA (phosphate agarose) (<b>A</b>), CA (carboxylated agarose) (<b>B</b>), and NA (native agarose) (<b>C</b>) hydrogels as measured by ELISA. The plot shows cumulative absorbance as a function of time and the data points represent average values with standard deviation. (* <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01).</p>
Full article ">Figure A5
<p><span class="html-italic">p</span>-values for the cumulative release of FGF-2 at various time points between the various hydrogel groups. Significance was assessed using a student’s <span class="html-italic">t</span>-test (paired, one-tailed). A <span class="html-italic">p</span>-value of ≤0.05 was considered statistically significant (* (<span class="html-italic">p</span> ≤ 0.05), ** (<span class="html-italic">p</span> ≤ 0.01), *** (<span class="html-italic">p</span> ≤ 0.001) and **** (<span class="html-italic">p</span> ≤ 0.0001). Gray boxes represent comparisons that were not statistically significant.</p>
Full article ">
24 pages, 11745 KiB  
Article
Multi-Temporal Energy Management Strategy for Fuel Cell Ships Considering Power Source Lifespan Decay Synergy
by Xingwei Zhou, Xiangguo Yang, Mengni Zhou, Lin Liu, Song Niu, Chaobin Zhou and Yufan Wang
J. Mar. Sci. Eng. 2025, 13(1), 34; https://doi.org/10.3390/jmse13010034 - 29 Dec 2024
Viewed by 465
Abstract
With increasingly stringent maritime environmental regulations, hybrid fuel cell ships have garnered significant attention due to their advantages in low emissions and high efficiency. However, challenges related to the coordinated control of multi-energy systems and fuel cell degradation remain significant barriers to their [...] Read more.
With increasingly stringent maritime environmental regulations, hybrid fuel cell ships have garnered significant attention due to their advantages in low emissions and high efficiency. However, challenges related to the coordinated control of multi-energy systems and fuel cell degradation remain significant barriers to their practical implementation. This paper proposes an innovative multi-timescale energy management strategy that focuses on optimizing the lifespan decay synergy of fuel cells and lithium batteries. The study designs an attention-based CNN-LSTM hybrid model for power prediction and constructs a two-stage optimization framework: The first stage employs Model Predictive Control (MPC) for long-term power planning to optimize equivalent hydrogen consumption, while the second stage focuses on real-time power allocation considering both power source degradation and system operational efficiency. The simulation results demonstrate that compared to single-layer MPC and the Equivalent Consumption Minimization Strategy (ECMS), the proposed method exhibits significant advantages in reducing single-voyage costs, minimizing differences in power source degradation rates, and alleviating power source stress. The overall performance of this strategy approaches the global optimal solution obtained through Dynamic Programming, comprehensively validating its superiority in simultaneously optimizing system economics and durability. Full article
(This article belongs to the Special Issue Advancements in Power Management Systems for Hybrid Electric Vessels)
Show Figures

Figure 1

Figure 1
<p>Power system topology of the hybrid ship.</p>
Full article ">Figure 2
<p>(<b>a</b>) Efficiency; (<b>b</b>) consumption rate.</p>
Full article ">Figure 3
<p>Rint equivalent circuit diagram of battery.</p>
Full article ">Figure 4
<p>Equivalent circuit of supercapacitor.</p>
Full article ">Figure 5
<p>Fuel cell ship load power data.</p>
Full article ">Figure 6
<p>The algorithm hierarchy of the proposed algorithm.</p>
Full article ">Figure 7
<p>Energy management strategy framework.</p>
Full article ">Figure 8
<p>CNN-LSTM based on attention mechanism model framework.</p>
Full article ">Figure 9
<p>Load power prediction results.</p>
Full article ">Figure 10
<p>Model Predictive Control (MPC) principle.</p>
Full article ">Figure 11
<p>Low-pass filtering control strategy.</p>
Full article ">Figure 12
<p>(<b>a</b>) Total fuel cell power; (<b>b</b>) lithium battery power; (<b>c</b>) supercapacitor power; (<b>d</b>) fuel cell power distribution.</p>
Full article ">Figure 13
<p>(<b>a</b>) Lithium battery SOC; (<b>b</b>) supercapacitor SOC.</p>
Full article ">Figure 14
<p>Comparison of life decay rates of dual power sources: (<b>a</b>) Proposed strategy; (<b>b</b>) DP strategy; (<b>c</b>) Single-layer MPC strategy; (<b>d</b>) ECMS strategy.</p>
Full article ">Figure 15
<p>Stress analysis and comparison of power source under different strategies: (<b>a</b>) Proposed strategy; (<b>b</b>) DP strategy; (<b>c</b>) Single-layer MPC strategy; (<b>d</b>) ECMS strategy.</p>
Full article ">
21 pages, 9839 KiB  
Article
Expression of Lumican and Osteopontin in Perivascular Areas of the Glioblastoma Peritumoral Niche and Its Value for Prognosis
by María Dolores Salinas, Pablo Rodriguez, Gonzalo Rubio and Rut Valdor
Int. J. Mol. Sci. 2025, 26(1), 192; https://doi.org/10.3390/ijms26010192 - 29 Dec 2024
Viewed by 313
Abstract
Glioblastoma (GB) is one of the most aggressive and treatment-resistant cancers due to its complex tumor microenvironment (TME). We previously showed that GB progression is dependent on the aberrant induction of chaperone-mediated autophagy (CMA) in pericytes (PCs), which promotes TME immunosuppression through the [...] Read more.
Glioblastoma (GB) is one of the most aggressive and treatment-resistant cancers due to its complex tumor microenvironment (TME). We previously showed that GB progression is dependent on the aberrant induction of chaperone-mediated autophagy (CMA) in pericytes (PCs), which promotes TME immunosuppression through the PC secretome. The secretion of extracellular matrix (ECM) proteins with anti-tumor (Lumican) and pro-tumoral (Osteopontin, OPN) properties was shown to be dependent on the regulation of GB-induced CMA in PCs. As biomarkers are rarely studied in TME, in this work, we aimed to validate Lumican and OPN as prognostic markers in the perivascular areas of the peritumoral niche of a cohort of GB patients. Previously, we had validated their expression in GB xenografted mice presenting GB infiltration (OPN) or GB elimination (Lumican) dependent on competent or deficient CMA PCs, respectively. Then, patient sample classification by GB infiltration into the peritumoral brain parenchyma was related to GB-induced CMA in microvasculature PCs, analyzing the expression of the lysosomal receptor, LAMP-2A. Our results revealed a correlation between GB-induced CMA activity in peritumoral PCs and GB patients’ outcomes, identifying three degrees of severity. The perivascular expression of both immune activation markers, Iba1 and CD68, was related to CMA-dependent PC immune function and determined as useful for efficient GB prognosis. Lumican expression was identified in perivascular areas of patients with less severe outcome and partially co-localizing with PCs presenting low CMA activity, while OPN was primarily found in perivascular areas of patients with poor outcome and partially co-localizing with PCs presenting high CMA activity. Importantly, we found sex differences in the incidence of middle-aged patients, being significantly higher in men but with worse prognosis in women. Our results confirmed that Lumican and OPN in perivascular areas of the GB peritumoral niche are effective predictive biomarkers for evaluating prognosis and monitoring possible therapeutic immune responses dependent on PCs in tumor progression. Full article
(This article belongs to the Special Issue New Wave of Cancer Therapeutics: Challenges and Opportunities)
Show Figures

Figure 1

Figure 1
<p>Expression of ECM proteins Lumican (<b>A</b>) and OPN (<b>B</b>) in GB xenografts from mice presenting GB infiltration or elimination. Right panels (A1 to A2 and B1 to B2) show magnification of microvessels (v) with perivascular positive stain (pointed with arrows). Scale bar: 100 µm. Association study of <span class="html-italic">LAMP2</span> with <span class="html-italic">LUM</span> (<b>C</b>) or <span class="html-italic">SPP1</span> (<b>D</b>) genes in the TCGA-GBM and Rembrandt cohort (GlioVis, <a href="https://gliovis.bioinfo.cnio.es/" target="_blank">https://gliovis.bioinfo.cnio.es/</a> (accessed on 20 December 2024)); * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 2
<p>CMA activity in PCs correlates with patient survival. (<b>A</b>) Representative images of peritumoral areas according to patient classification showing co-localization of puncta pattern expression of LAMP-2A protein (dark brown) with the PC marker α-SMA (pink) in microvessels of GB patients. Samples were classified as severe (highest α-SMA/LAMP-2A co-localization), moderate or mild (basal co-localization) according to the histological evaluation. A1 to A3 shows magnifications of microvessels (v). LAMP-2A co-localizationwith α-SMA<sup>+</sup> cells is marked with arrows. Scale bar: 100 µm. (<b>B</b>) Representative images of PCs marked with PDGFRβ (in red) in microvessels (v) showing co-localization of puncta pattern expression of LAMP-2A protein (in green) in peritumoral areas of severity classified GB patients. B1 to B3 show magnifications of PDGFRβ<sup>+</sup> cells. Positive co-localization (in yellow) is marked with arrows. Nuclei were stained with DAPI (blue). Scale bar: 100 µm. (<b>C</b>) Overall survival shown by Kaplan–Meier curves of the severity classification related to CMA activity in PCs; * <span class="html-italic">p</span> &lt; 0.05: difference between mild and severe; # <span class="html-italic">p</span> &lt; 0.05: difference between mild and moderate. (<b>D</b>) Age distribution by gender of the cohort of GB patients; **** <span class="html-italic">p</span> &lt; 0.0001; ns indicates no significance. (<b>E</b>) Severity classification related to gender in the age range 30–65 years in the cohort of GB patients; **** <span class="html-italic">p</span> &lt; 0.0001; ns indicates no significance.</p>
Full article ">Figure 3
<p>Perivascular CD68 and Iba-1 expression correlate with low CMA in peritumoral PCs and better patient outcome. (<b>A</b>) Expression of the myeloid marker Iba-1 in the peritumoral and perivascular areas (positive perivascular cells pointed with arrows) of the invasion front of GB patients. Images are representative of severe, moderate and mild grades of histopathological severity related to CMA activity in peritumoral PCs. The right corners of the left panels show an amplification of parenchymal positive cells. A1 to A3 panels show the magnification of microvessels (v) with perivascular positive cells (pointed with arrows). Scale bar: 100 µm. Boxplot diagrams of the quantification of Iba-1-positive particles related to the number of cells in the peritumoral (PT) parenchyma (<b>B</b>), expressed as positive particles per mm<sup>2</sup>, and in the perivascular (PV) microenvironment (<b>C</b>), expressed as positive pixels per blood vessel (bv) perimeter. Quantification was performed in at least four fields and in a minimum of 5 blood vessels; * <span class="html-italic">p</span> &lt; 0.05, **** <span class="html-italic">p</span> &lt; 0.0001. (<b>D</b>) Expression of phagocytic activation marker CD68 in the peritumoral and perivascular areas (positive perivascular cells pointed with arrows) of the invasion front of GB patients. Images are representative of patients classified as severe, moderate and mild related to CMA activity in peritumoral PCs. The right corners of the left panels show an amplification of parenchymal positive cells. D1 to D3 panels show magnification of microvessels (v) with perivascular positive cells (indicated with arrows). Scale bar: 100 µm. Boxplot diagrams of the quantification of CD68-positive cells in the peritumoral (PT) area (<b>E</b>), expressed as positive cells per mm<sup>2</sup>, and in the perivascular (PV) microenvironment (<b>F</b>), expressed as positive cells per blood vessel (bv) perimeter. Quantification was performed in at least four fields and in a minimum of 5 blood vessels; * <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. (<b>G</b>) Peritumoral and perivascular correlation between Iba-1- and CD68-positive expression. In total peritumoral parenchyma, Iba-1 and CD68 show a positive correlation (Pearson’s coefficient = 0.5640; ** <span class="html-italic">p</span> = 0.0077), as well as in perivascular areas (Pearson’s coefficient = 0.7827; **** <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 3 Cont.
<p>Perivascular CD68 and Iba-1 expression correlate with low CMA in peritumoral PCs and better patient outcome. (<b>A</b>) Expression of the myeloid marker Iba-1 in the peritumoral and perivascular areas (positive perivascular cells pointed with arrows) of the invasion front of GB patients. Images are representative of severe, moderate and mild grades of histopathological severity related to CMA activity in peritumoral PCs. The right corners of the left panels show an amplification of parenchymal positive cells. A1 to A3 panels show the magnification of microvessels (v) with perivascular positive cells (pointed with arrows). Scale bar: 100 µm. Boxplot diagrams of the quantification of Iba-1-positive particles related to the number of cells in the peritumoral (PT) parenchyma (<b>B</b>), expressed as positive particles per mm<sup>2</sup>, and in the perivascular (PV) microenvironment (<b>C</b>), expressed as positive pixels per blood vessel (bv) perimeter. Quantification was performed in at least four fields and in a minimum of 5 blood vessels; * <span class="html-italic">p</span> &lt; 0.05, **** <span class="html-italic">p</span> &lt; 0.0001. (<b>D</b>) Expression of phagocytic activation marker CD68 in the peritumoral and perivascular areas (positive perivascular cells pointed with arrows) of the invasion front of GB patients. Images are representative of patients classified as severe, moderate and mild related to CMA activity in peritumoral PCs. The right corners of the left panels show an amplification of parenchymal positive cells. D1 to D3 panels show magnification of microvessels (v) with perivascular positive cells (indicated with arrows). Scale bar: 100 µm. Boxplot diagrams of the quantification of CD68-positive cells in the peritumoral (PT) area (<b>E</b>), expressed as positive cells per mm<sup>2</sup>, and in the perivascular (PV) microenvironment (<b>F</b>), expressed as positive cells per blood vessel (bv) perimeter. Quantification was performed in at least four fields and in a minimum of 5 blood vessels; * <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. (<b>G</b>) Peritumoral and perivascular correlation between Iba-1- and CD68-positive expression. In total peritumoral parenchyma, Iba-1 and CD68 show a positive correlation (Pearson’s coefficient = 0.5640; ** <span class="html-italic">p</span> = 0.0077), as well as in perivascular areas (Pearson’s coefficient = 0.7827; **** <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 4
<p>Perivascular Lumican is expressed in peritumoral areas of patients with low CMA activity in PCs. (<b>A</b>) Expression of ECM Lumican in the peritumoral and perivascular areas of the invasion front of GB. Images are representative of severe, moderate and mild grades of histopathological severity related to CMA activity in peritumoral PCs. A1 to A3 panels show magnification of microvessels (v) with perivascular positive stain (pointed with arrows). Scale bar: 100 µm. (<b>B</b>) Boxplot diagrams of the quantification of Lumican-positive particles in the peritumoral (PT) parenchyma, expressed as positive particles per mm<sup>2</sup>, and (<b>C</b>) in the perivascular (PV) microenvironment, expressed as positive pixels per blood vessel (bv) perimeter. Quantification was performed in at least four fields and in a minimum of 5 blood vessels; ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001. (<b>D</b>,<b>E</b>) Representative images of the expression of Lumican (in green) surrounding PCs (marked with αSMA or RGS5 in red) in microvessels (v) in peritumoral areas of GB patients classified by severity. PC co-localization is marked with arrows. Nuclei were stained with DAPI (blue). Scale bar: 100 µm.</p>
Full article ">Figure 5
<p>Perivascular OPN is expressed in peritumoral areas of patients with GB-induced CMA activity. (<b>A</b>) Expression of extracellular protein OPN in the peritumoral and perivascular areas of the invasion front of GB. Images are representative of patients classified as severe, moderate and mild related to CMA activity in peritumoral PCs. A1 to A3 panels show magnification of microvessels (v) with perivascular positive cells (pointed with arrows). Scale bar: 100 µm. (<b>B</b>) Boxplot diagrams of the quantification of OPN-positive cells in the peritumoral (PT) parenchyma, expressed as positive cells per mm<sup>2</sup>, and (<b>C</b>) in the perivascular (PV) microenvironment, expressed as positive area per blood vessel (bv) perimeter. Quantification was performed in at least four fields and in a minimum of 5 blood vessels; ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001. (<b>D</b>) In total peritumoral parenchyma, Lumican and OPN show a positive correlation (Pearson’s r = 0.5797; *** <span class="html-italic">p</span> = 0.0003), whereas in perivascular areas, they correlate negatively (Pearson’s r = −0.5098; ** <span class="html-italic">p</span> = 0.0047). (<b>E</b>,<b>F</b>) Representative images of the expression of OPN (in green) surrounding PCs (marked with αSMA or RGS5 in red) in microvessels (v) in peritumoral areas of GB patients classified by severity. Co-localization with PCs is indicated with arrows. Nuclei were stained with DAPI (blue). Scale bar: 100 µm.</p>
Full article ">Figure 5 Cont.
<p>Perivascular OPN is expressed in peritumoral areas of patients with GB-induced CMA activity. (<b>A</b>) Expression of extracellular protein OPN in the peritumoral and perivascular areas of the invasion front of GB. Images are representative of patients classified as severe, moderate and mild related to CMA activity in peritumoral PCs. A1 to A3 panels show magnification of microvessels (v) with perivascular positive cells (pointed with arrows). Scale bar: 100 µm. (<b>B</b>) Boxplot diagrams of the quantification of OPN-positive cells in the peritumoral (PT) parenchyma, expressed as positive cells per mm<sup>2</sup>, and (<b>C</b>) in the perivascular (PV) microenvironment, expressed as positive area per blood vessel (bv) perimeter. Quantification was performed in at least four fields and in a minimum of 5 blood vessels; ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001. (<b>D</b>) In total peritumoral parenchyma, Lumican and OPN show a positive correlation (Pearson’s r = 0.5797; *** <span class="html-italic">p</span> = 0.0003), whereas in perivascular areas, they correlate negatively (Pearson’s r = −0.5098; ** <span class="html-italic">p</span> = 0.0047). (<b>E</b>,<b>F</b>) Representative images of the expression of OPN (in green) surrounding PCs (marked with αSMA or RGS5 in red) in microvessels (v) in peritumoral areas of GB patients classified by severity. Co-localization with PCs is indicated with arrows. Nuclei were stained with DAPI (blue). Scale bar: 100 µm.</p>
Full article ">
18 pages, 8535 KiB  
Article
Tenascin C-Guided Nanosystem for Precision Delivery of Obeticholic Acid in Liver Fibrosis Therapy
by Yawen Wang, Lei Yang, Qing Xu, Taiyu Liu, Hongliang He, Lisha Liu and Lifang Yin
Pharmaceutics 2025, 17(1), 32; https://doi.org/10.3390/pharmaceutics17010032 - 28 Dec 2024
Viewed by 287
Abstract
Objective: Liver fibrosis, a hallmark of chronic liver diseases, is characterized by excessive extracellular matrix (ECM) deposition and scar tissue formation. Current antifibrotic nanomedicines face significant limitations, including poor penetration into fibrotic tissue, rapid clearance, and suboptimal therapeutic efficacy. The dense fibrotic ECM [...] Read more.
Objective: Liver fibrosis, a hallmark of chronic liver diseases, is characterized by excessive extracellular matrix (ECM) deposition and scar tissue formation. Current antifibrotic nanomedicines face significant limitations, including poor penetration into fibrotic tissue, rapid clearance, and suboptimal therapeutic efficacy. The dense fibrotic ECM acts as a major physiological barrier, necessitating the development of a targeted delivery strategy to achieve effective therapeutic outcomes. Methods: We designed a liposomal delivery system functionalized with the GBI-10 aptamer and encapsulating obeticholic acid (OCA lips@Apt) to enhance selective delivery to fibrotic liver tissue while minimizing systemic toxicity. Results: Both in vitro and in vivo studies demonstrated that the aptamer-modified OCA liposomes effectively treated hepatic fibrosis through dual mechanisms: modulation of abnormal bile acid metabolism and attenuation of inflammation. The targeted delivery system leveraged the overexpression of Tenascin-C (TnC), a key ECM component in fibrotic tissues, for precise localization and enhanced endocytosis via the exposed cationic liposome surface. Conclusions: The OCA lips@Apt nanodrug demonstrated superior therapeutic efficacy with minimal off-target effects, offering a promising strategy to overcome critical barriers in liver fibrosis treatment. By precisely targeting the fibrotic ECM and modulating key pathological pathways, this TnC-guided liposomal delivery system provides a significant advancement in antifibrotic nanomedicine. Full article
(This article belongs to the Section Drug Delivery and Controlled Release)
Show Figures

Figure 1

Figure 1
<p>Preparation and characterization of OCA lips@Apt. (<b>a</b>) TnC expression in liver tissue by Western blot in normal and fibrotic mice. * <span class="html-italic">p</span> &lt; 0.05 compared to control group. (<b>b</b>) Schematic illustration of the preparation of OCA lips and OCA lips@Apt. (<b>c</b>) Average size distribution and TEM images of OCA lips and size change of OCA lips in PBS. (<b>d</b>) Agarose gel electrophoresis of OCA lips and GBI 10 aptamer at different N:P ratios, and Zeta potential of OCA lips and GBI 10 aptamer at different N:P ratios. (<b>e</b>) Average size distribution and TEM images of OCA lips@Apt, and size change of OCA lips in PBS. (<b>f</b>) Agarose gel electrophoresis of OCA lips@Apt at different times in 10% serum. (<b>g</b>) In vitro drug release curves of free OCA, OCA lips, and OCA lips@Apt at pH 7.4 (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 2
<p>In vitro cellular uptake and antifibrotic performance of OCA lips@Apt. (<b>a</b>) Cellular uptake of Dil labeled OCA lips and OCA lips@Apt (red) into AML-12 cells after different incubation durations observed by CLSM. Scale bar = 10 µm. (<b>b</b>) Semi-quantitative analysis from flow cytometry in AML-12 cells incubated with C6 lips and C6 lips@Apt, respectively (<span class="html-italic">n</span> = 3). (<b>c</b>) Cellular uptake of Dil labeled OCA lips and OCA lips@Apt (red) into RAW264.7 cells after different incubation durations observed by CLSM. Scale bar = 30 µm. (<b>d</b>) Semi-quantitative analysis from flow cytometry in RAW 264.7 cells incubated with C6 lips and C6 lips@Apt, respectively (<span class="html-italic">n</span> = 3). (<b>e</b>) RT-qPCR analysis of <span class="html-italic">Fxr</span> mRNA and <span class="html-italic">Cyp7a1</span> mRNA in AML-12 cells after treatment with OCA-contained formulations (<span class="html-italic">n</span> = 3). (<b>f</b>) RT-qPCR analysis of <span class="html-italic">Nf-kb</span> mRNA, <span class="html-italic">Il 6</span> mRNA, <span class="html-italic">Ccl2</span> mRNA, and <span class="html-italic">Nlrp3</span> mRNA in LPS-pretreated RAW264.7 cells after treatment with OCA-contained formulations (<span class="html-italic">n</span> = 3). The statistical analysis in (<b>b</b>,<b>d</b>–<b>f</b>) was performed using a one-way ANOVA with a Turkey 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, and **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 3
<p>In vitro penetration study of OCA lips@Apt and tissue distribution behavior in fibrotic mice. (<b>a</b>) C6 lips and C6 lips@Apt were added to the gel layer and scanned using CLSM Z-stack. Scale bar = 100 μm (<b>b</b>) Schematic illustration of the construction of multicellular ECM-rich 3D multicellular spheroids. (<b>c</b>) In vitro penetration of C6 lips, C6 lips@sApt, and C6 lips@Apt in HepG2/TGF-β1 pre-treated NIH-3T3 cocultured tumor spheroids observed by CLSM, scale bar = 100 µm. Semi-quantification results of the image (white dash line). (<b>d</b>) A total of 24 h after fibrotic mice were intravenously injected with DiR and DiR-loaded liposomes, the fluorescence intensities of vital organs were monitored using an IVIS Spectrum instrument. (<b>e</b>) Fluorescence intensity of each organ was calculated accordingly (<span class="html-italic">n</span> = 3). (<b>f</b>) Representative in vivo IVIS spectrum pictures of fibrotic mice treated with different formulations. ns, not significant. (<b>g</b>) Fluorescence intensity of each mouse was calculated accordingly (red circle). * <span class="html-italic">p</span> = 0.0134 compared with OCA lips@Apt. The statistical significance was assessed using a one-way ANOVA with a Tukey test.</p>
Full article ">Figure 4
<p>In vivo antifibrotic efficacy in CCl4-induced fibrosis model. (<b>a</b>) Schedule of the therapeutic regimen of OCA lips@Apt. (<b>b</b>) Representative photographs of normal and fibrotic livers of mice. Livers were collected at 24 h post-injection and perfused with PBS buffer as well as fixed with 4% paraformaldehyde. (<b>c</b>) Representative images of liver photos, H&amp;E (5×, scale bar = 1mm, 20×, scale bar = 100 μm), Masson (5×, scale bar = 1mm, 20×, scale bar = 100 μm), and Sirius Red (5×, scale bar = 1mm, 20×, scale bar = 100 μm) of liver sections of mice with different treatments. (<b>d</b>,<b>e</b>) Semiquantitative analysis of the area of Masson and Sirius Red staining sections. (<b>f</b>) Liver enzyme assay of AST and ALT of mice with different treatments (<span class="html-italic">n</span> = 3). The statistical significance was performed using a one-way ANOVA with a Tukey test. <span class="html-italic">p</span> values: ns, not significant, * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 5
<p>OCA lips@Apt act as potent FXR agonist in vivo. (<b>a</b>) RT-qPCR assay for monitoring <span class="html-italic">Fxr</span> mRNA, <span class="html-italic">Shp</span> mRNA, and <span class="html-italic">Cyp7al</span> mRNA in livers of fibrotic mice treated with various formulations (<span class="html-italic">n</span> = 3). (<b>b</b>) Levels of serum total bile acids in various groups (<span class="html-italic">n</span> = 3). (<b>c</b>) RT-qPCR assay for monitoring <span class="html-italic">Il6</span> and <span class="html-italic">Nlrp3</span> mRNA in livers of fibrotic mice treated with various formulations (<span class="html-italic">n</span> = 3). (<b>d</b>) RT-qPCR assay for monitoring <span class="html-italic">Tgf-β</span> and <span class="html-italic">Timp-1</span> mRNA in livers of fibrotic mice treated with various formulations (<span class="html-italic">n</span> = 3). The statistical significance was assessed using one-way ANOVA with the Tukey test. <span class="html-italic">p</span> values, ** <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 ">
20 pages, 8938 KiB  
Article
Equivalent Cost Minimization Strategy for Plug-In Hybrid Electric Bus with Consideration of an Inhomogeneous Energy Price and Battery Lifespan
by Di Xue, Haisheng Wang, Junnian Wang, Changyang Guan and Yiru Xia
Sustainability 2025, 17(1), 46; https://doi.org/10.3390/su17010046 - 25 Dec 2024
Viewed by 249
Abstract
The development of energy-saving vehicles is an important measure to deal with environmental pollution and the energy crisis. On this basis, more accurate and efficient energy management strategies can further tap into the energy-saving potential and energy sustainability of vehicles. The equivalent consumption [...] Read more.
The development of energy-saving vehicles is an important measure to deal with environmental pollution and the energy crisis. On this basis, more accurate and efficient energy management strategies can further tap into the energy-saving potential and energy sustainability of vehicles. The equivalent consumption minimization strategy (ECMS) has shown the ability to provide a real-time sub-optimal fuel efficiency performance. However, when taking the different market prices of fuel and electricity cost as well as battery longevity cost into account, this method is not very accurate for total operational economic evaluation. So, as an improved scheme, the instantaneous cost minimization strategy is proposed, where a comprehensive cost function, including the market price of the electricity and fuel as well as the cost of battery aging, is applied as the optimization objective. Simulation results show that the proposed control strategy for series-parallel hybrid electric buses can reduce costs by 41.25% when compared with the conventional engine-driven bus. The approach also impressively improves cost performance over the rule-based strategy and the ECMS. As such, the proposed instantaneous cost minimization strategy is a better choice for hybrid electric vehicle economic evaluation than the other main sub-optimal strategies. Full article
Show Figures

Figure 1

Figure 1
<p>Powertrain layout of HEV.</p>
Full article ">Figure 2
<p>Engine BSFC map.</p>
Full article ">Figure 3
<p>M1 motor efficiency map.</p>
Full article ">Figure 4
<p>M2 motor efficiency map.</p>
Full article ">Figure 5
<p>Correction coefficient curve of rotating mass.</p>
Full article ">Figure 6
<p>Simulink powertrain model.</p>
Full article ">Figure 7
<p>Vehicle operation mode state-flow.</p>
Full article ">Figure 8
<p>Battery capacity loss experimental results.</p>
Full article ">Figure 9
<p>Optimal gear-shifting rules.</p>
Full article ">Figure 10
<p>Engine optimal torque of parallel mode 1 under the different gears.</p>
Full article ">Figure 11
<p>Engine optimal torque of parallel mode 2 under the different gears.</p>
Full article ">Figure 12
<p>China’s urban driving cycle.</p>
Full article ">Figure 13
<p>Engine operating points’ distribution. (<b>a</b>) Engine operation points for RB; (<b>b</b>) engine operation points for ECMS; (<b>c</b>) engine operation points for DP; (<b>d</b>) engine operation points for Min_Cost.</p>
Full article ">Figure 14
<p>Random actual velocity profile.</p>
Full article ">Figure 15
<p>Battery SOC history.</p>
Full article ">
21 pages, 1916 KiB  
Review
The Clinical Application of Gel-Based Composite Scaffolds in Rotator Cuff Repair
by Shebin Tharakan, Michael Hadjiargyrou and Azhar Ilyas
Gels 2025, 11(1), 2; https://doi.org/10.3390/gels11010002 - 24 Dec 2024
Viewed by 371
Abstract
Rotator cuff tears are a common injury that can be treated with or without surgical intervention. Gel-based scaffolds have gained significant attention in the field of tissue engineering, particularly for applications like rotator cuff repair. Scaffolds can be biological, synthetic, or a mixture [...] Read more.
Rotator cuff tears are a common injury that can be treated with or without surgical intervention. Gel-based scaffolds have gained significant attention in the field of tissue engineering, particularly for applications like rotator cuff repair. Scaffolds can be biological, synthetic, or a mixture of both materials. Collagen, a primary constituent of the extracellular matrix (ECM) in musculoskeletal tissues, is one of the most widely used materials for gel-based scaffolds in rotator cuff repair, but other ECM-based and synthetic-based composite scaffolds have also been utilized. These composite scaffolds can be engineered to mimic the biomechanical and biological properties of natural tissues, supporting the healing process and promoting regeneration. Various clinical studies examined the effectiveness of these composite scaffolds with collagen, ECM and synthetic polymers and provided outstanding results with remarkable improvements in range of motion (ROM), strength, and pain. This review explores the material composition, manufacturing process and material properties of gel-based composite scaffolds as well as their clinical outcomes for the treatment of rotator cuff injuries. Full article
(This article belongs to the Special Issue Gel-Based Materials for Biomedical Engineering)
Show Figures

Figure 1

Figure 1
<p>Right shoulder observed from the lateral portal. (<b>A</b>) Demonstrates a large tear at the level of the glenoid. (<b>B</b>) Rotator cuff repair prior to the addition of the synthetic PLLA scaffold. (<b>C</b>) Placement of the scaffold with medial sutures and anchors. Reprinted with permissions from [<a href="#B66-gels-11-00002" class="html-bibr">66</a>].</p>
Full article ">Figure 2
<p>An example of some biological and synthetic materials that can constitute composite scaffold for shoulder augmentation.</p>
Full article ">Figure 3
<p>Ultrasound image of the rotator cuff tears and repairs from representative patients. (<b>A</b>) Rotator cuff tear. a. humeral head; b. defect; c. retracted cuff. (<b>B</b>) Ruptured repair. a. humeral head; b. defect; c. retracted cuff and graft. (<b>C</b>,<b>D</b>) Intact GraftJacket<sup>®</sup> and cuff repair. a. humeral head; b. intact graft; c. suture. Adapted with permission from [<a href="#B91-gels-11-00002" class="html-bibr">91</a>].</p>
Full article ">Figure 4
<p>Histology specimens extracted from sheep infraspinatus tenocytes at the tendon-bone junction were stained with Safranin O, demonstrating healing with a PLGA anchored scaffold. Black brackets indicate interfaces where tissue and bone are not properly integrated. Magnification of 40×. Reprinted with permissions from [<a href="#B73-gels-11-00002" class="html-bibr">73</a>].</p>
Full article ">
12 pages, 6270 KiB  
Article
Distribution and Maturity of Medial Collagen Fibers in Thoracoabdominal Post-Dissection Aortic Aneurysms: A Comparative Study of Marfan and Non-Marfan Patients
by Panagiotis Doukas, Bernhard Hruschka, Cathryn Bassett, Eva Miriam Buhl, Florian Simon, Pepijn Saraber, Michael Johan Jacobs, Christian Uhl, Leon J. Schurgers and Alexander Gombert
Int. J. Mol. Sci. 2025, 26(1), 14; https://doi.org/10.3390/ijms26010014 - 24 Dec 2024
Viewed by 210
Abstract
Thoracoabdominal aortic aneurysms (TAAAs) are rare but serious conditions characterized by dilation of the aorta characterized by remodeling of the vessel wall, with changes in the elastin and collagen content. Individuals with Marfan syndrome have a genetic predisposition for elastic fiber fragmentation and [...] Read more.
Thoracoabdominal aortic aneurysms (TAAAs) are rare but serious conditions characterized by dilation of the aorta characterized by remodeling of the vessel wall, with changes in the elastin and collagen content. Individuals with Marfan syndrome have a genetic predisposition for elastic fiber fragmentation and elastin degradation and are prone to early aneurysm formation and progression. Our objective was to analyze the medial collagen characteristics through histological, polarized light microscopy, and electron microscopy methods across the thoracic and abdominal aorta in twenty-five patients undergoing open surgical repair, including nine with Marfan syndrome. While age at surgery differed significantly between the groups, maximum aortic diameter and aneurysm extent did not. Collagen content increased from thoracic to infrarenal segments in both cohorts, with non-Marfan patients exhibiting higher collagen percentages, notably in the infrarenal aorta (729.3 nm vs. 1068.3 nm, p = 0.02). Both groups predominantly displayed mature collagen fibers, with the suprarenal segment containing the highest proportion of less mature fibers. Electron microscopy revealed comparable collagen fibril diameters across segments irrespective of Marfan status. Our findings underscore non-uniform histological patterns in TAAAs and suggest that ECM remodeling involves mature collagen deposition, albeit with lower collagen content observed in the infrarenal aorta of Marfan patients. Full article
(This article belongs to the Special Issue Arteriogenesis, Angiogenesis and Vascular Remodeling, 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>Intima and media of the infrarenal aorta for non-Marfan (<b>A</b>,<b>C</b>) and Marfan (<b>B</b>,<b>D</b>) patients. (<b>A</b>,<b>B</b>): picrosirius red staining, (<b>C</b>,<b>D</b>): MOVAT pentachrome staining. Red: intima and media, Green: only intima.</p>
Full article ">Figure 2
<p>Total collagen percentage in the aortic media of the different aortic segments for non-Marfan and Marfan patients. <span class="html-italic">p</span>-values calculated with the Dunn-Sidàk post hoc test after Friedman ANOVA.</p>
Full article ">Figure 3
<p>Picrosirius red staining of the different aortic segments for Marfan and non-Marfan patients. The images on the left were taken with conventional microscopy, while those on the right were taken with polarized light microscopy. The media layer is indicated by the green line running across the diameter. Arrows point to the aortic lumen. 40× magnification.</p>
Full article ">Figure 4
<p>Quantification of the collagen fiber types in aortic media, according to level of maturity (red to green—mature to less mature).</p>
Full article ">Figure 5
<p>Collagen fibers in the different aortic segments. 60,000× magnification.</p>
Full article ">
21 pages, 2012 KiB  
Article
Decellularized Green and Brown Macroalgae as Cellulose Matrices for Tissue Engineering
by Caitlin Berry-Kilgour, Indrawati Oey, Jaydee Cabral, Georgina Dowd and Lyn Wise
J. Funct. Biomater. 2024, 15(12), 390; https://doi.org/10.3390/jfb15120390 - 23 Dec 2024
Viewed by 449
Abstract
Scaffolds resembling the extracellular matrix (ECM) provide structural support for cells in the engineering of tissue constructs. Various material sources and fabrication techniques have been employed in scaffold production. Cellulose-based matrices are of interest due to their abundant supply, hydrophilicity, mechanical strength, and [...] Read more.
Scaffolds resembling the extracellular matrix (ECM) provide structural support for cells in the engineering of tissue constructs. Various material sources and fabrication techniques have been employed in scaffold production. Cellulose-based matrices are of interest due to their abundant supply, hydrophilicity, mechanical strength, and biological inertness. Terrestrial and marine plants offer diverse morphologies that can replicate the ECM of various tissues and be isolated through decellularization protocols. In this study, three marine macroalgae species—namely Durvillaea poha, Ulva lactuca, and Ecklonia radiata—were selected for their morphological variation. Low-intensity, chemical treatments were developed for each species to maintain native cellulose structures within the matrices while facilitating the clearance of DNA and pigment. Scaffolds generated from each seaweed species were non-toxic for human dermal fibroblasts but only the fibrous inner layer of those derived from E. radiata supported cell attachment and maturation over the seven days of culture. These findings demonstrate the potential of E. radiata-derived cellulose scaffolds for skin tissue engineering and highlight the influence of macroalgae ECM structures on decellularization efficiency, cellulose matrix properties, and scaffold utility. Full article
(This article belongs to the Special Issue Novel Biomaterials for Tissue Engineering)
Show Figures

Figure 1

Figure 1
<p>Morphological differences between macroalgae: (<b>A</b>) macroscopic images of <span class="html-italic">Durvillaea poha</span> (i), <span class="html-italic">Ulva lactuca</span> (ii), and <span class="html-italic">Ecklonia radiata</span> (iii) illustrate differences in size and morphology; (<b>B</b>) H&amp;E staining of paraffin-embedded sections; and (<b>C</b>) SEM imaging reveals their porous versus fibrous structural composition. Scale bars are as indicated.</p>
Full article ">Figure 2
<p>Structural differences between macroalgae matrices: (<b>A</b>) Images of <span class="html-italic">Durvillaea poha</span> (i), <span class="html-italic">Ulva lactuca</span> (ii), and <span class="html-italic">Ecklonia radiata</span> (iii) matrices demonstrate macrostructure after chemical treatment. (<b>B</b>) H&amp;E staining of paraffin-embedded sections, (<b>C</b>) SEM imaging, and (<b>D</b>) calcofluor white staining of paraffin-embedded sections show porous and/or fibrous compositions. Scale bars are as indicated.</p>
Full article ">Figure 3
<p>Human dermal fibroblasts attach to and distribute through the fibrous layers of <span class="html-italic">Ecklonia radiata</span> but not <span class="html-italic">Ulva lactuca</span> and <span class="html-italic">Durvillaea poha</span> scaffolds. Representative two-dimensional optical slices (i,iii) and three-dimensional confocal Z-stacks (ii,iv) showing BJ/5Ta cells cultured on <span class="html-italic">Durvillaea poha</span> (<b>A</b>), <span class="html-italic">Ulva lactuca</span> (<b>B</b>), and <span class="html-italic">Ecklonia radiata</span> (<b>C</b>) matrices for two (i,ii) or seven (iii,iv) days, after fixation and staining with wheat germ agglutinin (WGA; red) and calcofluor white (CW; blue) to visualize cell membrane glycans and cellulose fibers, respectively. Gold arrows show cells and spheroids that have failed to attach to cellulose fibers. Red arrows show acellular punctate staining. Scale bars are as indicated.</p>
Full article ">Figure 4
<p>Fibroblasts differ in their attachment to and morphology on macroalgae scaffolds. Scanning electron microscopy images showing BJ/5Ta cells cultured on <span class="html-italic">Durvillaea poha</span> (<b>A</b>), <span class="html-italic">Ulva lactuca</span> (<b>B</b>), and <span class="html-italic">Ecklonia radiata</span> (<b>C</b>) matrices for two (i) or seven (ii) days. Gold arrows show cells and spheroids lacking fibroblastic morphology. Scale bars are as indicated.</p>
Full article ">
29 pages, 7062 KiB  
Article
Gram Negative Biofilms: Structural and Functional Responses to Destruction by Antibiotic-Loaded Mixed Polymeric Micelles
by Tsvetozara Damyanova, Rumena Stancheva, Milena N. Leseva, Petya A. Dimitrova, Tsvetelina Paunova-Krasteva, Dayana Borisova, Katya Kamenova, Petar D. Petrov, Ralitsa Veleva, Ivelina Zhivkova, Tanya Topouzova-Hristova, Emi Haladjova and Stoyanka Stoitsova
Microorganisms 2024, 12(12), 2670; https://doi.org/10.3390/microorganisms12122670 - 23 Dec 2024
Viewed by 408
Abstract
Biofilms are a well-known multifactorial virulence factor with a pivotal role in chronic bacterial infections. Their pathogenicity is determined by the combination of strain-specific mechanisms of virulence and the biofilm extracellular matrix (ECM) protecting the bacteria from the host immune defense and the [...] Read more.
Biofilms are a well-known multifactorial virulence factor with a pivotal role in chronic bacterial infections. Their pathogenicity is determined by the combination of strain-specific mechanisms of virulence and the biofilm extracellular matrix (ECM) protecting the bacteria from the host immune defense and the action of antibacterials. The successful antibiofilm agents should combine antibacterial activity and good biocompatibility with the capacity to penetrate through the ECM. The objective of the study is the elaboration of biofilm-ECM-destructive drug delivery systems: mixed polymeric micelles (MPMs) based on a cationic poly(2-(dimethylamino)ethyl methacrylate)-b-poly(ε-caprolactone)-b-poly(2-(dimethylamino)ethyl methacrylate) (PDMAEMA35-b-PCL70-b-PDMAEMA35) and a non-ionic poly(ethylene oxide)-b-poly(propylene oxide)-b-poly(ethylene oxide) (PEO100-b-PPO65-b-PEO100) triblock copolymers, loaded with ciprofloxacin or azithromycin. The MPMs were applied on 24 h pre-formed biofilms of Escherichia coli and Pseudomonas aeruginosa (laboratory strains and clinical isolates). The results showed that the MPMs were able to destruct the biofilms, and the viability experiments supported drug delivery. The biofilm response to the MPMs loaded with the two antibiotics revealed two distinct patterns of action. These were registered on the level of both bacterial cell-structural alterations (demonstrated by scanning electron microscopy) and the interaction with host tissues (ex vivo biofilm infection model on skin samples with tests on nitric oxide and interleukin (IL)-17A production). Full article
(This article belongs to the Special Issue Contemporary Perspectives on Bacterial Virulence Factors)
Show Figures

Figure 1

Figure 1
<p>(<b>a</b>) Hydrodynamic diameter, D<sub>h</sub>, and (<b>b</b>) ζ-potential variations as a function of the micellar concentration of SCPMs and MPMs based on PDMAEMA<sub>35</sub>-PCL<sub>70</sub>-PDMAEMA<sub>35</sub> and Pluronic F127 triblock copolymers. (<b>c</b>) Size distribution curves (<b>d</b>) DLS correlation functions and (<b>e</b>) representative AFM micrograph of MPMs prepared from PDMAEMA<sub>35</sub>-PCL<sub>70</sub>-PDMAEMA<sub>35</sub> and Pluronic F127 triblock copolymers at a molar ratio of 1:1 in the concentration range of 1 to 0.125 mg mL<sup>−1</sup>. The PDI values ranged in the 0.11–0.19 interval. All DLS measurements were performed at 25 °C.</p>
Full article ">Figure 2
<p>Variations of encapsulation efficiency (<b>a</b>) and drug loading content (<b>b</b>) as a function of the composition of SCPMs and MPMs based on PDMAEMA<sub>35</sub>-PCL<sub>70</sub>-PDMAEMA<sub>35</sub> and Pluronic F127 triblock copolymers. The loading was performed at polymer-to-drug mass ratio of 10:1.</p>
Full article ">Figure 3
<p>Hydrodynamic diameter, Dh, (<b>a</b>,<b>c</b>,<b>e</b>) and ζ potential (<b>b</b>,<b>d</b>,<b>f</b>) of empty or loaded with antibiotics MPMs based on PDMAEMA<sub>35</sub>-PCL<sub>70</sub>-PDMAEMA<sub>35</sub> and Pluronic F127 triblock copolymers in the concentration range of 1 to 0.125 mg mL<sup>−1</sup>. Measurements were performed at 25 °C at pH 7. Each data point represents the arithmetic mean ± SD of three separate experiments.</p>
Full article ">Figure 3 Cont.
<p>Hydrodynamic diameter, Dh, (<b>a</b>,<b>c</b>,<b>e</b>) and ζ potential (<b>b</b>,<b>d</b>,<b>f</b>) of empty or loaded with antibiotics MPMs based on PDMAEMA<sub>35</sub>-PCL<sub>70</sub>-PDMAEMA<sub>35</sub> and Pluronic F127 triblock copolymers in the concentration range of 1 to 0.125 mg mL<sup>−1</sup>. Measurements were performed at 25 °C at pH 7. Each data point represents the arithmetic mean ± SD of three separate experiments.</p>
Full article ">Figure 4
<p>Drug release profiles of MPMs based on PDMAEMA<sub>35</sub>-PCL<sub>70</sub>-PDMAEMA<sub>35</sub> and Pluronic F127 triblock copolymers, prepared at a 10:1 polymer-to-drug mass ratio, determined by HPLC. MPMs were formed at molar ratios of 3:1 (<b>a</b>), 1:1 (<b>b</b>), and 1:3 (<b>c</b>). The release was performed at 37 °C in phosphate buffer pH 7.4. Each data point represents the arithmetic mean ± SD of three separate experiments.</p>
Full article ">Figure 5
<p>Cytotoxicity of the SCPMs and MPMs based on PDMAEMA<sub>35</sub>-PCL<sub>70</sub>-PDMAEMA<sub>35</sub> and Pluronic F127 triblock copolymers loaded with CF (<b>a</b>) or AZ (<b>b</b>) at a 10:1 polymer-to-drug mass ratio. The micelles were applied for 4 h in concentrations of 0.5, 0.25, and 0.125 mg mL<sup>−1</sup> onto confluent cultured HaCaT. The results are presented as percentage of the control—cells cultivated parallelly in DMEM. The data are the means of four repeats and are presented as the mean ± SD. Differences between control (DMEM) and treated with micelles cells are accepted as statistically significant (*) when <span class="html-italic">p</span> &lt; 0.05 and (**) when <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 6
<p>Reduction of the biomass of mature 24 h biofilms as a result of treatment for 4 or 24 h with 0.25 mg mL<sup>−1</sup> of empty or antibiotics-loaded MPMs based on PDMAEMA<sub>35</sub>-PCL<sub>70</sub>-PDMAEMA<sub>35</sub> and Pluronic F127 triblock copolymers. The results were calculated as percentage of the biofilm at the start of each experiment. (<b>a</b>) <span class="html-italic">E. coli</span> 25922; (<b>b</b>) <span class="html-italic">P. aeruginosa</span> PAO1. Results for biofilms treated with dH<sub>2</sub>O are included since the micelles were dispersed in dH<sub>2</sub>O. Each data point represents the mean ± SD of six repeats.</p>
Full article ">Figure 7
<p>Viability of the biofilms after treatment for 24 h with 0.25 mg mL<sup>−1</sup> of empty or antibiotics-loaded MPMs based on PDMAEMA<sub>35</sub>-PCL<sub>70</sub>-PDMAEMA<sub>35</sub> and Pluronic F127 triblock copolymers. Viability was estimated by the reduction of resazurin using the Alamar Blue reagent (Invitrogen). The results were calculated as percentage of the untreated control (biofilm cultivated parallelly in M63 medium in the absence of the tested agents). dH<sub>2</sub>O bars are included to show the effect of treatment with dH<sub>2</sub>O alone_ the medium in which the micelles were dispersed. (<b>a</b>) <span class="html-italic">E. coli</span> 25922; (<b>b</b>) <span class="html-italic">P. aeruginosa</span> PAO1. Each data point represents the mean ± SD of six repeats. <span class="html-italic">p</span> &lt; 0.05 (*); <span class="html-italic">p</span> &lt; 0.001 (***), ANOVA test.</p>
Full article ">Figure 8
<p>Reduction of biofilms of pathogenic strains of <span class="html-italic">E. coli</span> treated with empty or antibiotic-loaded MPMs 3:1 (<b>a</b>) and of <span class="html-italic">P. aeruginosa</span> treated with empty or antibiotic-loaded MPMs 1:1 (<b>b</b>). The results were calculated as percentage of the “0” controls, i.e., the amount of biofilms of the strains before the start of the treatments. Each data point represents the mean ± SD of six repeats.</p>
Full article ">Figure 9
<p>Scanning electron microscopy of biofilms of <span class="html-italic">E. coli</span> 25922 (<b>A</b>–<b>H</b>) and <span class="html-italic">P. aeruginosa</span> PAO1 (<b>I</b>–<b>P</b>). Arrows: white—infolds of the cell wall; yellow—outer membrane vesicles; red—tunneling nanotubules. (<b>A</b>) <span class="html-italic">E. coli</span> 48 h control biofilm; (<b>B</b>,<b>E</b>,<b>F</b>,<b>F1</b>) <span class="html-italic">E. coli</span> 24 h biofilm treated for a further 24 h with empty MPMs 3:1; yellow asterisk mark slimy covering of cells in some areas of the treated biofilm. (<b>G</b>,<b>G1</b>) <span class="html-italic">E. coli</span> 24 h biofilm treated for a further 24 h with CF-loaded MPMs 3:1; (<b>H</b>,<b>H1</b>) <span class="html-italic">E. coli</span> 24 h biofilm treated for a further 24 h with AZ-loaded MPMs 3:1. (<b>I</b>,<b>M</b>) <span class="html-italic">P. aeruginosa</span> 48 h control biofilm; (<b>J</b>,<b>N</b>) <span class="html-italic">P. aeruginosa</span> 24 h biofilm treated for a further 24 h with empty MPMs 1:1; (<b>K</b>,<b>O</b>) <span class="html-italic">P. aeruginosa</span> 24 h biofilm treated for a further 24 h with CF-loaded MPMs 1:1; (<b>L</b>,<b>P</b>,<b>P1</b>) <span class="html-italic">P. aeruginosa</span> 24 h biofilm treated for a further 24 h with AZ-loaded MPMs 1:1; white asterisks, cells with extensively blebbed surfaces.</p>
Full article ">Figure 10
<p>Histological sections of skin explants infected with <span class="html-italic">P. aeruginosa</span> PAO1 biofilm. (<b>A</b>) Untreated 24 h ex vivo biofilm. (<b>B</b>,<b>C</b>) Mature 24 h biofilms on skin explants were treated for 24 h with 0.25 mg mL<sup>−1</sup> of MPMs 1:1 loaded with CF (<b>B</b>) or AZ (<b>C</b>). Bar = 10 µm.</p>
Full article ">Figure 11
<p>Effect of MPMs loaded with CF or AZ on NO (<b>a</b>) and IL-17A (<b>b</b>) production in ex vivo murine skin explant <span class="html-italic">P. aeruginosa</span> PAO1 biofilm model. Murine skin explants were infected with <span class="html-italic">P. aeruginosa</span> for 24 h for the development of biofilm. Afterwards the skin explants were treated with 50 µL of either 0.5 or 0.25 mg mL<sup>−1</sup> MPMs loaded with CF or AZ. Control samples, infected or uninfected with <span class="html-italic">P. aeruginosa</span> biofilm, were treated in parallel with either PBS or dH<sub>2</sub>O (the solvent for the MPM samples). Data represents mean ± SD from 3 samples/group * <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 when comparing the biofilm groups to the control PBS one, ANOVA test; ## <span class="html-italic">p</span> &lt; 0.05 when comparing the non-biofilm groups to the control PBS one, ANOVA test.</p>
Full article ">
Back to TopTop