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

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (4,082)

Search Parameters:
Keywords = skin tissue

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
19 pages, 7699 KiB  
Article
Development of New Chitosan-Based Complex with Bioactive Molecules for Regenerative Medicine
by Natasha Maurmann, Gabriela Moraes Machado, Rafaela Hartmann Kasper, Marcos do Couto, Luan Paz, Luiza Oliveira, Juliana Girón Bastidas, Paola Arosi Bottezini, Lucas Machado Notargiacomo, Carlos Arthur Ferreira, Luciano Pighinelli, Caren Serra Bavaresco, Patricia Pranke and Myrian Brew
Future Pharmacol. 2024, 4(4), 873-891; https://doi.org/10.3390/futurepharmacol4040046 (registering DOI) - 16 Dec 2024
Abstract
Background/Objectives: The development of new materials incorporating bioactive molecules for tissue regeneration is a growing area of interest. The objective of this study was to develop a new complex specifically designed for bone and skin tissue engineering, combining chitosan, ascorbic acid-2-magnesium phosphate (ASAP), [...] Read more.
Background/Objectives: The development of new materials incorporating bioactive molecules for tissue regeneration is a growing area of interest. The objective of this study was to develop a new complex specifically designed for bone and skin tissue engineering, combining chitosan, ascorbic acid-2-magnesium phosphate (ASAP), and β-tricalcium phosphate (β-TCP). Methods: Chitosan and the complexes chitosan/ASAP and chitosan/ASAP/β-TCP were prepared in membrane form, macerated to a particulate format, and then subjected to characterization through Fourier transform infrared (FTIR) spectroscopy, optical and scanning electron microscopy (SEM), zeta potential, thermogravimetric analysis (TGA), and differential scanning calorimetry (DSC). Cell viability was evaluated through a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and with fluorescein diacetate (FDA) and propidium iodide (PI) staining in stem cells obtained from deciduous teeth. Statistical analyses were performed using analysis of variance (ANOVA), followed by Tukey’s test. Results: The FTIR results indicated the characteristic bands in the chitosan group and the complexation between chitosan, ASAP, and β-TCP. Microscopic characterization revealed a polydisperse distribution of micrometric particles. Zeta potential measurements demonstrated a reduction in surface charge upon the addition of ASAP and β-TCP to the chitosan matrix. TGA and DSC analyses further indicated complexation between the three components and the successful formation of a cross-linked structure in the chitosan matrix. Stem cells cultured with the particulate biomaterials demonstrated their biocompatibility. Statistical analysis revealed a significant increase in cell viability for the chitosan/ASAP and chitosan/ASAP/β-TCP groups compared to the chitosan control. Conclusions: Therefore, the chitosan/ASAP complex demonstrated potential for skin regeneration, while the chitosan/ASAP/β-TCP formulation showed promise as a biomaterial for bone regeneration due to the presence of β-tricalcium phosphate. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Summary of experiments: (<b>a</b>) schematic presentation of the chemical structure and dissolution of chitosan in an aqueous solution of hydrochloric acid; (<b>b</b>) chemical structure and dissolution of L-ascorbic acid 2-phosphate sesquimagnesium salt hydrate (ASAP) in an aqueous solution of hydrochloric acid; (<b>c</b>) chemical structure and dissolution of beta-tricalcium phosphate (β-TCP) in the aqueous solution of hydrochloric acid; (<b>d</b>) filtration; (<b>e</b>) pasty material; (<b>f</b>) dried membrane; (<b>g</b>) particulate biomaterial obtained after maceration; (<b>h</b>) ultraviolet radiation in a laminar hood to reduce the risk of contamination.</p>
Full article ">Figure 2
<p>FTIR spectrum of the chitosan particles (Chit), chitosan/ascorbic acid-2-magnesium phosphate complex (Chit/ASAP), and the complex with β-tricalcium phosphate (Chit/ASAP/β-TCP).</p>
Full article ">Figure 3
<p>Micrographs of chitosan particles (Chit), chitosan/ascorbic acid-2-magnesium phosphate complex (Chit/ASAP), and the complex with β-tricalcium phosphate (Chit/ASAP/β-TCP): (<b>a</b>) optical microscopy reveals the general morphology of the particles; (<b>b</b>) scanning electron microscopy offers a high-resolution view, highlighting surface details of the particles. The scale bar represents 100 μm, except for in 2000× magnification, which is 10 μm.</p>
Full article ">Figure 3 Cont.
<p>Micrographs of chitosan particles (Chit), chitosan/ascorbic acid-2-magnesium phosphate complex (Chit/ASAP), and the complex with β-tricalcium phosphate (Chit/ASAP/β-TCP): (<b>a</b>) optical microscopy reveals the general morphology of the particles; (<b>b</b>) scanning electron microscopy offers a high-resolution view, highlighting surface details of the particles. The scale bar represents 100 μm, except for in 2000× magnification, which is 10 μm.</p>
Full article ">Figure 4
<p>Zeta potential measurement of chitosan particles (Chit), chitosan/ascorbic acid-2-magnesium phosphate complex (Chit/ASAP), and the complex with β-tricalcium phosphate (Chit/ASAP/β-TCP) in 1 mM NaCl: (<b>a</b>) graphical representation with data expressed as mean ± standard deviation. Different letters indicate significant differences (<span class="html-italic">p</span> ≤ 0.05) by ANOVA, followed by Tukey’s test. (<b>b</b>) Distribution.</p>
Full article ">Figure 5
<p>Thermogravimetric analysis (TGA) of chitosan particles (Chit), chitosan/ascorbic acid-2-magnesium phosphate complex (Chit/ASAP), and the complex with β-tricalcium phosphate (Chit/ASAP/β-TCP).</p>
Full article ">Figure 6
<p>Differential scanning calorimetry (DSC) of chitosan particles (Chit), chitosan/ascorbic acid-2-magnesium phosphate complex (Chit/ASAP), and the complex with β-tricalcium phosphate (Chit/ASAP/β-TCP).</p>
Full article ">Figure 7
<p>Biological test with mesenchymal stem cells (MSCs). (<b>a</b>) Fibroblastoid morphology of cells adhered to in vitro culture plastics, and osteogenic, adipogenic, and chondrogenic differentiation of MSCs, stained with alizarin red S, oil red O, and alcian blue, respectively. The scale bar represents 50 μm. Viability of MSCs 3 days after treatment with particles: (<b>b</b>) MTT assay; (<b>c</b>) staining of live/dead cells with fluorescein diacetate and propidium iodide. The scale bar represents 100 μm. The control corresponds to cells grown directly in the wells of the tissue culture plate; Chit, to 10 mg/mL of chitosan; Chit/ASAP, to 10.3 mg/mL of the complex chitosan with ascorbic acid-2-magnesium phosphate; and Chit/ASAP/TCP, to 20.3 mg/mL of the complex Chit/ASAP with β-tricalcium phosphate. Data expressed as mean ± standard error of the mean. * indicates a statistically significant difference (<span class="html-italic">p</span> &lt; 0.05) in relation to the control by ANOVA followed by Tukey’s test.</p>
Full article ">Figure 7 Cont.
<p>Biological test with mesenchymal stem cells (MSCs). (<b>a</b>) Fibroblastoid morphology of cells adhered to in vitro culture plastics, and osteogenic, adipogenic, and chondrogenic differentiation of MSCs, stained with alizarin red S, oil red O, and alcian blue, respectively. The scale bar represents 50 μm. Viability of MSCs 3 days after treatment with particles: (<b>b</b>) MTT assay; (<b>c</b>) staining of live/dead cells with fluorescein diacetate and propidium iodide. The scale bar represents 100 μm. The control corresponds to cells grown directly in the wells of the tissue culture plate; Chit, to 10 mg/mL of chitosan; Chit/ASAP, to 10.3 mg/mL of the complex chitosan with ascorbic acid-2-magnesium phosphate; and Chit/ASAP/TCP, to 20.3 mg/mL of the complex Chit/ASAP with β-tricalcium phosphate. Data expressed as mean ± standard error of the mean. * indicates a statistically significant difference (<span class="html-italic">p</span> &lt; 0.05) in relation to the control by ANOVA followed by Tukey’s test.</p>
Full article ">
14 pages, 2756 KiB  
Article
Tissue Sources Influence the Morphological and Morphometric Characteristics of Collagen Membranes for Guided Bone Regeneration
by Josefa Alarcón-Apablaza, Karina Godoy-Sánchez, Marcela Jarpa-Parra, Karla Garrido-Miranda and Ramón Fuentes
Polymers 2024, 16(24), 3499; https://doi.org/10.3390/polym16243499 - 16 Dec 2024
Viewed by 76
Abstract
(1) Background: Collagen, a natural polymer, is widely used in the fabrication of membranes for guided bone regeneration (GBR). These membranes are sourced from various tissues, such as skin, pericardium, peritoneum, and tendons, which exhibit differences in regenerative outcomes. Therefore, this study aimed [...] Read more.
(1) Background: Collagen, a natural polymer, is widely used in the fabrication of membranes for guided bone regeneration (GBR). These membranes are sourced from various tissues, such as skin, pericardium, peritoneum, and tendons, which exhibit differences in regenerative outcomes. Therefore, this study aimed to evaluate the morphological and chemical properties of porcine collagen membranes from five different tissue sources: skin, pericardium, dermis, tendons, and peritoneum. (2) Methods: The membrane structure was analyzed using energy-dispersive X-ray spectrometry (EDX), variable pressure scanning electron microscopy (VP-SEM), Fourier transform infrared spectroscopy (FTIR), and thermal stability via thermogravimetric analysis (TGA). The absorption capacity of the membranes for GBR was also assessed using an analytical digital balance. (3) Results: The membranes displayed distinct microstructural features. Skin- and tendon-derived membranes had rough surfaces with nanopores (1.44 ± 1.24 µm and 0.46 ± 0.1 µm, respectively), while pericardium- and dermis-derived membranes exhibited rough surfaces with macropores (78.90 ± 75.89 µm and 64.89 ± 13.15 µm, respectively). The peritoneum-derived membrane featured a rough surface of compact longitudinal fibers with irregular macropores (9.02 ± 3.70 µm). The thickness varied significantly among the membranes, showing differences in absorption capacity. The pericardium membrane exhibited the highest absorption, increasing by more than 10 times its initial mass. In contrast, the skin-derived membrane demonstrated the lowest absorption, increasing by less than 4 times its initial mass. Chemical analysis revealed that all membranes were primarily composed of carbon, nitrogen, and oxygen. Thermogravimetric and differential scanning calorimetry analyses showed no significant compositional differences among the membranes. FTIR spectra confirmed the presence of collagen, with characteristic peaks corresponding to Amide A, B, I, II, and III. (4) Conclusions: The tissue origin of collagen membranes significantly influences their morphological characteristics, which may, in turn, affect their osteogenic properties. These findings provide valuable insights into the selection of collagen membranes for GBR applications. Full article
(This article belongs to the Section Polymer Membranes and Films)
Show Figures

Figure 1

Figure 1
<p>Three-dimensional structures of membranes for guided bone regeneration observed by variable pressure scanning electron microscopy. (D) Diaderm; (J) Jason; (M) Mucoderm; (OP) OSSiX Plus; (VF) Via Flex. (1,3) Membrane surface views; (2) Membrane thickness.</p>
Full article ">Figure 2
<p>Mapping elemental distribution in membranes: (D) Diaderm. (J) Jason membrane. (M) mucoderm. (V) Via Flex. (OP) OSSiX plus. (VF) Via Flex. (Mag: ×250). Blue = carbon; yellow = oxygen; pink = nitrogen; orange = sodium; green = phosphorus.</p>
Full article ">Figure 3
<p>Thermogravimetric analysis TGA-DSC.</p>
Full article ">Figure 4
<p>Characteristic vibrations of the chemical functional groups present in collagen by FT-IR.</p>
Full article ">
9 pages, 290 KiB  
Article
Discordant β-Lactam Susceptibility in Clinical Staphylococcus aureus Isolates: A Molecular and Phenotypical Exploration to Detect the BORSA/MODSA Isolates in Bogotá, Colombia
by Angie Lorena Fonseca-Fernández, María Alejandra Mancera-García, Aura Lucia Leal-Castro, Chad Leidy, Sandra Rincón, Lina P. Carvajal, Jinnethe Reyes and Adriana Marcela Celis Ramírez
Microorganisms 2024, 12(12), 2598; https://doi.org/10.3390/microorganisms12122598 - 16 Dec 2024
Viewed by 127
Abstract
Staphylococcus aureus is a human pathogen responsible for a wide range of diseases, such as skin and soft tissue infections, pneumonia, toxic shock syndrome, and urinary tract infections. Methicillin-resistant S. aureus (MRSA) is a well-known pathogen with consistently high mortality rates. Detecting the [...] Read more.
Staphylococcus aureus is a human pathogen responsible for a wide range of diseases, such as skin and soft tissue infections, pneumonia, toxic shock syndrome, and urinary tract infections. Methicillin-resistant S. aureus (MRSA) is a well-known pathogen with consistently high mortality rates. Detecting the mecA resistance gene and phenotypical profile to β-lactams allows for the differentiation of MRSA from methicillin-susceptible S. aureus (MSSA) isolates. In this study, we characterized 57 S. aureus clinical isolates for β-lactam susceptibility and mecA presence. We classified 52.63% as MRSA and 45.61% as MSSA. However, some isolates evidenced different oxacillin resistance profiles, such as borderline oxacillin-resistant or modified S. aureus (BORSA/MODSA). The cefazolin inoculum effect (CzIE) was established for these samples, emphasizing the relevance of these isolates as a source of therapeutic failure. We also performed the detection of the Panton-Valentine Leucocidin virulence genes as well as the S. aureus spa-type clonality. As expected, spa-types t002 and t008 were the most prevalent clones, demonstrating the success of well-established clones. These findings emphasize the importance of establishing sensitivity profiles, especially in isolates with poor resistance mechanisms, to determine their prevalence and their impact on public health. Full article
(This article belongs to the Section Medical Microbiology)
10 pages, 1794 KiB  
Article
Development of Biofidelic Skin Simulants Based on Fresh Cadaveric Skin Tests
by Gurpreet Singh, Pramod Yadav and Arnab Chanda
Eur. Burn J. 2024, 5(4), 454-463; https://doi.org/10.3390/ebj5040040 (registering DOI) - 16 Dec 2024
Viewed by 144
Abstract
The development of artificial skin that accurately mimics the mechanical properties of human skin is crucial for a wide range of applications, including surgical training for burn injuries, biomechanical testing, and research in sports injuries and ballistics. While traditional materials like gelatin, polydimethylsiloxane [...] Read more.
The development of artificial skin that accurately mimics the mechanical properties of human skin is crucial for a wide range of applications, including surgical training for burn injuries, biomechanical testing, and research in sports injuries and ballistics. While traditional materials like gelatin, polydimethylsiloxane (PDMS), and animal skins (such as porcine and bovine skins) have been used for these purposes, they have inherent limitations in replicating the intricate properties of human skin. In this work, we conducted uniaxial tensile tests on freshly obtained cadaveric skin to analyze its mechanical properties under various loading conditions. The stress–strain data obtained from these tests were then replicated using advanced skin simulants. These skin simulants were specifically formulated using a cost-effective and moldable multi-part silicone-based polymer. This material was chosen for its ability to accurately replicate the mechanical behavior of human skin while also addressing ethical considerations and biosafety concerns. In addition, the non-linear mechanical behavior of the developed skin simulants was characterized using three different hyperelastic curve-fit models (i.e., Neo-Hookean, Mooney–Rivlin, and Yeoh models). Moreover, these innovative simulants offer an ethical and practical alternative to cadaveric skin for use in laboratory and clinical settings. Full article
Show Figures

Figure 1

Figure 1
<p>(<b>a</b>) Removal of cadaveric thigh skin using dermatone, (<b>b</b>) test coupon of cadaveric skin for tensile testing, and (<b>c</b>) fabricated test coupons for candidate skin tissue simulants. (<b>c</b>) reproduced under license CC-BY-4.0 [<a href="#B21-ebj-05-00040" class="html-bibr">21</a>].</p>
Full article ">Figure 2
<p>Uniaxial tensile testing of samples to characterize their mechanical properties.</p>
Full article ">Figure 3
<p>Stress–strain plot of fresh cadaveric skin compared with the lower bound from the literature [<a href="#B28-ebj-05-00040" class="html-bibr">28</a>].</p>
Full article ">Figure 4
<p>Stress–strain plot of uniaxially tested candidate skin tissue simulants compared with the stress–strain results of fresh cadaveric skin.</p>
Full article ">Figure 5
<p>Controlled skin tissue simulants mimicking the stress–strain results of the fresh cadaveric skin.</p>
Full article ">
13 pages, 3729 KiB  
Article
Quasi-Static Mechanical Biomimetics Evaluation of Car Crash Dummy Skin
by Yurun Li, Zhixin Liu, Cuiru Sun, Xiaoya Zheng, Guorui Du, Xiaoshuang Wang, Songchen Wang and Weidong Liu
Biomimetics 2024, 9(12), 762; https://doi.org/10.3390/biomimetics9120762 - 15 Dec 2024
Viewed by 370
Abstract
Accurate replication of soft tissue properties is essential for the development of car crash test dummy skin to ensure the precision of biomechanical injury data. However, the intricacy of multi-layer soft tissue poses challenges in standardizing the development and testing of dummy skin [...] Read more.
Accurate replication of soft tissue properties is essential for the development of car crash test dummy skin to ensure the precision of biomechanical injury data. However, the intricacy of multi-layer soft tissue poses challenges in standardizing the development and testing of dummy skin materials to emulate soft tissue properties. This study presents a comprehensive testing and analysis of the compressive mechanical properties of both single and multi-layered soft tissues and car crash dummy skin materials, aiming to enhance the biofidelity of dummy skin. We presented one-term Ogden hyperelastic models and generalized Maxwell viscoelastic models for single-layer and multi-layer soft tissues, as well as dummy skin materials. The comparative analysis results indicate that the existing dummy skin material fails to fully consider the strain-rate-dependent characteristic of soft tissue. Furthermore, dummy skin materials exhibited ~3 times shorter relaxation times and ~2–3 times lower stress decay rates compared to soft tissues, suggesting a less viscous nature. This study provides an accurate representation of the mechanics of soft tissue and dummy skin under quasi-static compressive loading. The findings are instrumental for the development of novel bionic skin materials or structures to more precisely replicate the biomechanical properties of soft tissues, thereby enhancing the accuracy and reliability of car crash test dummies. Full article
Show Figures

Figure 1

Figure 1
<p>Photo of the porcine soft tissue specimens: (<b>a</b>) skin; (<b>b</b>) subcutaneous tissue; (<b>c</b>) muscle; (<b>d</b>) multi-layer soft tissue; (<b>e</b>) the skin sample under compression.</p>
Full article ">Figure 2
<p>Photo of the dummy skin specimen: (<b>a</b>) the arm skin of a dummy; (<b>b</b>) rubber; (<b>c</b>) sponge; (<b>d</b>) multi-layer dummy skin material.</p>
Full article ">Figure 3
<p>Compression test results of soft tissue subjected to 1.0 mm/s loading. Each type of tissue was tested three times, and the stress–strain curves were fitted with the one-term Ogden model. (<b>a</b>–<b>c</b>) Measured and fitted stress–strain curves of skin, subcutaneous, and muscle tissue specimens. (<b>d</b>) Average stress–strain curves of each tissue type and the multi-layer tissue sample. “subcu” represents “subcutaneous”, “multi” represents multi-layer tissue.</p>
Full article ">Figure 4
<p>Compression test stress–strain curves of the dummy skin specimens with one-term Ogden model fitting curves in red: (<b>a</b>) curves of the sponge material with incompressible one-term Ogden model (red) and compressible multi-term Ogden model fitting curves (blue); (<b>b</b>) curves of the rubber specimen; (<b>c</b>) curves of the three-layer dummy skin specimen.</p>
Full article ">Figure 5
<p>Stress–strain curves comparison between the soft tissue and dummy skin: (<b>a</b>) Single-layer dummy material with single-layer soft tissue from pig back; (<b>b</b>) Full-thickness multi-layer dummy skin with soft tissue from pig back under 1 mm/s and 0.5 mm/s loading rate; (<b>c</b>) Single layer dummy material with single-layer soft tissue from pig butt; (<b>d</b>) Full-thickness multi-layer dummy skin with soft tissue from pig butt under 1 mm/s. “subcu” represents “subcutaneous”.</p>
Full article ">Figure 6
<p>Finite element analysis of dummy skin. (<b>a</b>) Three-layer dummy skin model and z-direction stress distribution at 2.2 s; (<b>b</b>) Comparison of stress–strain curves of single-layer rubber specimen. (<b>c</b>) Comparison of stress–strain curves of single-layer sponge specimen. (<b>d</b>) Comparison of stress–strain curves of three-layer dummy skin.</p>
Full article ">Figure 7
<p>Stress relaxation measurement results of soft tissue and dummy skin with a constant strain of 20%. (<b>a</b>) Stress relaxation curves of soft tissue specimens, where “subcu” represents subcutaneous specimen, and “skin and subcu” represents a skin and subcutaneous bi-layer specimen. (<b>b</b>) Stress relaxation curves of rubber and sponge specimens. (<b>c</b>) Stress relaxation curves of full-thickness multi-layer dummy skin soft tissue specimens.</p>
Full article ">Figure 8
<p>Influence of thickness to the mechanical properties of muscle and rubber material in dummy skin. (<b>a</b>) Stress–strain curves and (<b>c</b>) stress relaxation curves of muscle specimens with 10 mm and 30 mm thickness; (<b>b</b>) stress–strain curves and (<b>d</b>) stress relaxation curves of rubber specimens with 2.20 mm and 5.02 mm thickness.</p>
Full article ">Figure 9
<p>Compression stress–strain curves of porcine soft tissue specimens. (<b>a</b>) Curves of skin specimens; (<b>b</b>) curves of subcutaneous specimens; (<b>c</b>) curves of muscle specimens; (<b>d</b>) curves of average stress–strain values of the four specimens for each type of tissue.</p>
Full article ">
11 pages, 904 KiB  
Article
Skin Tear Treatment with Copaifera multijuga Hayne in Polymeric Hydrogel: A Randomized Clinical Trial
by Camila Castanho Cardinelli, Jéssica Teixeira Gâmba Passos, Valdir Florêncio Veiga-Junior, Beatriz Guitton Renaud Baptista de Oliveira, Elisabete Pereira dos Santos, Guilherme Guilhermino Neto, Karina Chamma Di Piero and Zaida Maria Faria de Freitas
Pharmaceuticals 2024, 17(12), 1691; https://doi.org/10.3390/ph17121691 - 15 Dec 2024
Viewed by 306
Abstract
A double-blind, randomized, and controlled clinical trial with therapeutic intervention was performed at a university hospital in Rio de Janeiro to evaluate whether the addition of Copaifera multijuga Hayne oleoresin to a carboxypolymethylene hydrogel is more effective in skin tear healing than standard [...] Read more.
A double-blind, randomized, and controlled clinical trial with therapeutic intervention was performed at a university hospital in Rio de Janeiro to evaluate whether the addition of Copaifera multijuga Hayne oleoresin to a carboxypolymethylene hydrogel is more effective in skin tear healing than standard treatment. The sample consisted of 84 patients, predominantly men, with a mean age of 67.37 years. These participants were divided into three groups (29 in the intervention group, which received 10% Copaifera oleoresin; 28 in the intervention group, which received 2% Copaifera oleoresin; and 27 in the control group, which received carboxypolymethylene hydrogel). Data were tabulated and analyzed according to the relevant protocols and included only patients who had completed the treatment, while losses were excluded. Weekly follow-ups were conducted to monitor progress. The average healing time differed among the three groups (p > 0.05). There was also a significant difference in healing time between the two intervention groups. Ultimately, CopaibaPolyHy-2 led to significantly faster wound healing than CopaibaPolyHy-10 (p < 0.05). A high increase in granulation and epithelial tissue and a decrease in exudate quantity were observed in the CopaibaPolyHy-2 group. It was not possible to infer whether the wound size reduction differed between the treatments (p > 0.05). At the end of the study, 100% of wounds were healed, with 47,6% healing in week 2 (n = 40). No participants experienced local or serious adverse effects throughout the study period. The current study shows that CopaibaPolyHy-2 is effective, offering a statistically significantly faster healing time, better-quality tissue, and safe treatment for skin tears. Full article
Show Figures

Figure 1

Figure 1
<p>CONSORT diagram.</p>
Full article ">Figure 2
<p>(<b>a</b>) An ST treated with CopaibaPolyHy-10 on the 15th day; (<b>b</b>) the same ST healed on the 32nd day.</p>
Full article ">
23 pages, 5797 KiB  
Article
Ultrasound Examination of Skin, Fasciae and Subcutaneous Tissue: Optimizing Rehabilitation for Secondary Upper Limb Lymphedema
by Carmelo Pirri, Chiara Ferraretto, Nina Pirri, Lara Bonaldo, Raffaele De Caro, Stefano Masiero and Carla Stecco
Diagnostics 2024, 14(24), 2824; https://doi.org/10.3390/diagnostics14242824 - 15 Dec 2024
Viewed by 255
Abstract
Background: Lymphedema represents a frequent cause of disability for patients undergoing oncological treatments and, being a chronic, non-reversible pathology, requires targeted and continuous rehabilitation treatments. To date, the studies available on the use of ultrasound in patients with lymphedema mainly report descriptive data; [...] Read more.
Background: Lymphedema represents a frequent cause of disability for patients undergoing oncological treatments and, being a chronic, non-reversible pathology, requires targeted and continuous rehabilitation treatments. To date, the studies available on the use of ultrasound in patients with lymphedema mainly report descriptive data; therefore, with this study, we wanted to describe in a more objective way the typical ultrasound alterations found in these patients, measuring the thickness of the different superficial structures, and defining subcutis echogenicity. Methods: 14 patients affected by secondary lymphedema of the upper limbs were enrolled in this cross-sectional observational study (12 had breast cancer and 2 with melanoma as their primary diagnosis). All patients were classified as stage II according to the ISL classification. Patients were examined between March and July 2023 with a clinical and an ultrasound evaluation. Ultrasound evaluation was performed following a protocol and took into consideration thickness of the cutis, subcutis, superficial and deep fascia, and subcutis echogenicity. Results: The cutis of the affected limbs was thicker in the distal anterior region of the arm and throughout the anterior region of the forearm. The subcutaneous tissue was thicker in the posterior region of the distal arm and throughout the forearm, including the dorsum of the hand and excluding only the proximal posterior region of the forearm. Fascial structures did not demonstrate statistically significant differences in thickness between pathological and healthy limbs, despite undergoing significant changes from a qualitative point of view (loss of the trilaminar skin appearance and the development of anechoic areas due to fluid accumulation around the hyperechoic adipose lobule). A statistically significant difference in the echogenicity of subcutaneous tissue was found at the distal anterior region of the arm and at the entire anterior forearm. Conclusions: High-resolution ultrasound has been confirmed to be a tool capable of supporting the diagnosis of lymphedema and identifying the most compromised regions of the limb. A tailored rehabilitation plan can be developed based on the non-uniform alterations in subcutaneous tissue, where some areas are affected earlier than others. This compartmentalization should be considered in lymphedema staging and management. Ultrasound may provide early detection of these changes, guiding a more precise therapeutic approach. Full article
(This article belongs to the Special Issue Diagnostic Imaging in Musculoskeletal Diseases)
Show Figures

Figure 1

Figure 1
<p>Representation of the upper limb quadrant division: 1—proximal anteromedial arm; 2—proximal anterolateral arm; 3—distal anteromedial arm; 4—distal anterolateral arm; 5—proximal anteromedial forearm; 6—proximal anterolateral forearm; 7—distal anteromedial forearm; 8—distal anterolateral forearm.</p>
Full article ">Figure 2
<p>(<b>A</b>): Circumferential measurements of the hand were performed using the “figure-of-eight” method, which involves wrapping a millimeter tape in a specific pattern around hand to capture the dimensions accurately. (<b>B</b>): For the forearm, circumferential measurements were taken every 5 cm, using a millimeter tape to ensure precision and tissue texture across all measurements.</p>
Full article ">Figure 3
<p>(<b>A</b>) Normal US image of the healthy limb, showing the preserved trilaminar structure of the skin, the normal structure of subcutaneous tissue with superficial fascia, and normal deep fascia. (<b>B</b>) US appearance of the limb affected by chronic lymphedema, demonstrating a preserved trilaminar structure of the skin with increased thickness compared to the healthy limb. The subcutaneous tissue exhibits hyperecheoic regions, while the superficial fascia remains well defined, despite the tissue change. The deep fascia is identifiable. These findings reflected the structural remodeling characteristics of the different stages of chronic lymphedema.</p>
Full article ">Figure 4
<p>Ultrasound measurements of skin thickness (epidermis and dermis) taken from various regions and levels of the affected upper limb (PAT) and the healthy upper limb. The figure highlights the levels/regions where a statistically significant difference in thickness was found between the two limbs. These significant differences indicate a marked increase in skin thickness in the pathological limb, underscoring the impact of lymphedema on tissue structure across specific levels/regions of the upper limb.</p>
Full article ">Figure 5
<p>Ultrasound measurements of subcutaneous tissue thickness taken from different regions and levels of both the affected upper limb (PAT) and the healthy limb. The figure highlights the levels/regions where statistically significant differences in thickness were observed between the pathological and healthy limbs. These findings underscore the localized nature of tissue alterations in lymphedema and their varying impact across different anatomical regions.</p>
Full article ">Figure 6
<p>Ultrasound measurements of the superficial fascia thickness taken across different regions and levels of the affected upper limb (PAT) and the healthy limb. No statistically significant differences in thickness were found between the pathological and healthy limbs in any of the levels/regions.</p>
Full article ">Figure 7
<p>Ultrasound measurements of deep fascia thickness were conducted across various regions and levels of both the affected upper limb (PAT) and the healthy limb. No statistically significant differences in thickness were found between the two limbs.</p>
Full article ">Figure 8
<p>Echogenicity of the subcutaneous tissue measured across different regions and levels of both the affected upper limb (PAT) and the healthy limb. The figure highlights the regions and levels where statistically significant differences in echogenicity were observed between pathological and healthy limbs. These differences indicate localized changes in tissue composition due to lymphedema, particularly in regions/levels where increased echogenicity was detected.</p>
Full article ">
11 pages, 2370 KiB  
Article
Exploring the Link Between Psoriasis and Adipose Tissue: One Amplifies the Other
by Roberta Di Caprio, Ersilia Nigro, Eugenia Veronica Di Brizzi, Dario Buononato, Marta Mallardo, Vittorio Tancredi, Aurora Daniele and Anna Balato
Int. J. Mol. Sci. 2024, 25(24), 13435; https://doi.org/10.3390/ijms252413435 - 15 Dec 2024
Viewed by 248
Abstract
Psoriasis and obesity, while distinct, are inter-related inflammatory conditions. Adipose tissue (AT)-derived mediators could be pathogenically active in triggering and/or amplifying psoriatic skin inflammation and, vice versa, skin inflammation could drive increased adiposity that triggers the development of several chronic conditions. Gaining insight [...] Read more.
Psoriasis and obesity, while distinct, are inter-related inflammatory conditions. Adipose tissue (AT)-derived mediators could be pathogenically active in triggering and/or amplifying psoriatic skin inflammation and, vice versa, skin inflammation could drive increased adiposity that triggers the development of several chronic conditions. Gaining insight into their intricate relationship could be essential for effective management and treatment. The aim of this study was to determine (i) the pathogenic role of psoriasis-signature cytokines in contributing to AT metabolism and (ii) the role of AT-derived mediators in triggering and/or amplifying skin inflammation. For this reason, firstly, whole AT was treated with IL-17 and TNF-α, alone or in combination, to investigate their effects on the expression and production of adipokines and inflammatory factors. IL-17 and TNF-α were able to induce an additive or synergistic effect on AT-derived mediators. In order to assess the effects on the skin of inflamed AT by psoriasis-signature cytokines, ex vivo skin organ culture was performed and an increase in several inflammatory mediators was observed. These findings confirm that psoriasis and obesity amplify each other’s inflammatory processes and understanding this mutual exacerbation could lead to more effective therapeutic strategies that address both skin inflammation and AT metabolism. Full article
Show Figures

Figure 1

Figure 1
<p>Radar plots comparing the effects of IL-17 and TNF-α on AT secretory profiles. Five categories of proteins are represented: adipocytokines (blue), angiogenesis-related factors (orange), immunity-related mediators (gray), migration-associated proteins (yellow) and glucose homeostasis regulators (light blue). IL-17 induced a moderate increase in adipocytokines and glucose homeostasis proteins, while TNF-α robustly enhanced proteins across all categories, particularly those involved in immunity and migration.</p>
Full article ">Figure 2
<p>Protein levels of adiponectin (<b>a</b>), leptin (<b>b</b>) and resistin (<b>c</b>) in AT supernatants after stimulation with IL-17, TNF-α or their combination. Data are expressed as mean ± standard deviation (SD); statistical significance is indicated for comparisons between conditions (* <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 3
<p>Effect of IL-17 and TNF-α on cytokine gene expression in AT fragments and corresponding skin OCs. Panels (<b>a</b>,<b>c</b>,<b>e</b>,<b>g</b>,<b>i</b>) show gene expression in AT stimulated with IL-17, TNF-α or their combination, while panels (<b>b</b>,<b>d</b>,<b>f</b>,<b>h</b>,<b>j</b>) depict cytokine expression in skin OCs treated with supernatants derived from stimulated AT. Cytokines analyzed include IL-6 (<b>a</b>,<b>b</b>), IL-8 (<b>c</b>,<b>d</b>), IL-23 (<b>e</b>,<b>f</b>), IL-36γ (<b>g</b>,<b>h</b>) and IL-10 (<b>i</b>,<b>j</b>). Statistical analysis was performed with respect to controls. Data are displayed as mean ± SD of triplicates pooled from three independent experiments. Mann–Whitney test was used to calculate significant differences. * <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.</p>
Full article ">Figure 4
<p>Illustration of AT in vitro model.</p>
Full article ">Figure 5
<p>Illustration of ex vivo healthy skin organ culture.</p>
Full article ">
10 pages, 538 KiB  
Article
Risk Identification and Mitigation of Skin and Soft Tissue Infections in Military Training Environments
by Rebecca Suhr, Amy Peart, Brian Vesely, Michael Waller, Andrew Trudgian, Christopher Peatey and Jessica Chellappah
Trop. Med. Infect. Dis. 2024, 9(12), 306; https://doi.org/10.3390/tropicalmed9120306 - 14 Dec 2024
Viewed by 396
Abstract
Objective: Staphylococcus aureus (SA), including methicillin-resistant strains (MRSAs), is a major cause of skin and soft tissue infections (SSTIs) in military populations. This study investigated SSTI incidence and SA carriage in a military training site over 16 weeks using a prospective observational cohort [...] Read more.
Objective: Staphylococcus aureus (SA), including methicillin-resistant strains (MRSAs), is a major cause of skin and soft tissue infections (SSTIs) in military populations. This study investigated SSTI incidence and SA carriage in a military training site over 16 weeks using a prospective observational cohort design. Methods: Two training cohorts provided pre- and post-training self-collected swabs for bacterial carriage, and environmental swabs from accommodations, personal items, and training facilities. Hygiene awareness and practices were assessed through questionnaires. Bacteria were identified using culture, mass spectrometry (MALDI-TOF), and genomic sequencing. Results: Nasal carriage of SA increased from 19% to 49% by the end of training. SSTIs requiring treatment occurred in 16% of participants. Steam cleaning reduced but did not eliminate SA on personal bed linen. Additionally, 40% of participants had poor knowledge of antibacterial cleaning practices and wound management. Conclusions: Increased SA carriage was linked to human-to-human transmission in close-quarter military training environments. Implications for Public Health: Improved personal hygiene training, wound management education, and monitored cleaning protocols are essential to mitigate SSTI risks in communal military training environments. Full article
Show Figures

Figure 1

Figure 1
<p>Comparing pre- and post-training samples of sites with <span class="html-italic">Staphylococcus aureus</span> (SA) present (* denotes significant difference with <span class="html-italic">p</span>-value ˂ 0.05).</p>
Full article ">
18 pages, 2161 KiB  
Article
Syndecan-1 and E-Cadherin Expression in Canine Cutaneous Squamous Cell Carcinoma
by Rita Files, Cláudia Cardoso, Justina Prada, Filipe Silva and Isabel Pires
Vet. Sci. 2024, 11(12), 652; https://doi.org/10.3390/vetsci11120652 - 14 Dec 2024
Viewed by 250
Abstract
Cutaneous squamous cell carcinoma (CSCC) in dogs is a locally invasive tumor that typically occurs in areas of poorly pigmented skin due to sun exposure. Identifying new biomarkers, such as syndecan-1 (CD138) and E-cadherin, is fundamental for tumor diagnosis and prognosis. Dysregulation of [...] Read more.
Cutaneous squamous cell carcinoma (CSCC) in dogs is a locally invasive tumor that typically occurs in areas of poorly pigmented skin due to sun exposure. Identifying new biomarkers, such as syndecan-1 (CD138) and E-cadherin, is fundamental for tumor diagnosis and prognosis. Dysregulation of syndecan-1, expressed in epithelial tissue, fibroblasts, and plasma cells, is associated with poor prognosis in several types of cancer. Similarly, E-cadherin, which plays a crucial role in cell adhesion and epithelial functionality, is also linked to adverse outcomes. This study evaluated the expression of syndecan-1 and E-cadherin in 47 cases of canine cutaneous squamous cell carcinoma. The results showed that the intensity of syndecan-1 decreased with increasing tumor aggressiveness, and its presence in the stroma was significantly associated with tumor grade. E-cadherin also demonstrated a decrease in intensity with increasing malignancy. However, the association between syndecan-1 and E-cadherin was not statistically significant. E-cadherin reduction and stromal syndecan-1 positivity seem to be associated with tumor aggressiveness in canine cutaneous squamous cell carcinoma. Further studies are needed to explore their roles in tumor progression. Full article
Show Figures

Figure 1

Figure 1
<p>Histological grade of malignancy.</p>
Full article ">Figure 2
<p>Syndecan-1 labeling intensity in tumors with different histological grades of malignancy.</p>
Full article ">Figure 3
<p>Syndecan-1 stromal immunoexpression in tumors with different histological grades of malignancy.</p>
Full article ">Figure 4
<p>Syndecan-1 immunoexpression in canine cutaneous squamous cell carcinoma. (<b>A</b>) Membranous labeling with moderate intensity (2) in a well-differentiated tumor (Grade I); (<b>B</b>) Membranous and cytoplasmatic expression with moderate intensity (2) in a well-differentiated tumor (Grade I); (<b>C</b>) Week cytoplasmatic and nuclear labeling in moderately differentiated tumor (Grade II); (<b>D</b>) Stroma immunoexpression in a poorly differentiated tumor (Grade III).</p>
Full article ">Figure 5
<p>E-cadherin labeling intensity in tumors with different histological grades of malignancy.</p>
Full article ">Figure 6
<p>E-cadherin immunoexpression in canine cutaneous squamous cell carcinoma. (<b>A</b>) Membranous labeling with strong intensity (3) in a well-differentiated tumor (Grade I); (<b>B</b>) Membranous and cytoplasmatic expression with strong intensity (3) in a moderately differentiated tumor (Grade II); (<b>C</b>) Moderate membranous labeling in poorly differentiated tumor (Grade III); (<b>D</b>) Weak membranous labeling in poorly differentiated tumor (Grade III).</p>
Full article ">
16 pages, 10610 KiB  
Article
Enhanced Wound Healing and Autogenesis Through Lentiviral Transfection of Adipose-Derived Stem Cells Combined with Dermal Substitute
by Shiqi Wang, Dinghui Gao, Mingyu Li, Qian Wang, Xuanyu Du and Siming Yuan
Biomedicines 2024, 12(12), 2844; https://doi.org/10.3390/biomedicines12122844 - 13 Dec 2024
Viewed by 497
Abstract
Background: Burns and chronic ulcers may cause severe skin loss, leading to critical health issues like shock, infection, sepsis, and multiple organ failure. Effective healing of full-thickness wounds may be challenging, with traditional methods facing limitations due to tissue shortage, infection, and lack [...] Read more.
Background: Burns and chronic ulcers may cause severe skin loss, leading to critical health issues like shock, infection, sepsis, and multiple organ failure. Effective healing of full-thickness wounds may be challenging, with traditional methods facing limitations due to tissue shortage, infection, and lack of structural support. Methods: This study explored the combined use of gene transfection and dermal substitutes to improve wound healing. We used the DGTM (genes: DNP63A, GRHL2, TFAP2A, and MYC) factors to transfect adipose-derived stem cells (ADSCs), inducing their differentiation into keratinocytes. These transfected ADSCs were then incorporated into Pelnac® dermal substitutes to enhance vascularization and cellular proliferation for better healing outcomes. Results: Gene transfer using DGTM factors successfully induced keratinocyte differentiation in ADSCs. The application of these differentiated cells with Pelnac® dermal substitute to dermal wounds in mice resulted in the formation of skin tissue with a normal epidermal layer and proper collagen organization. This method alleviates the tediousness of the multiple transfection steps in previous protocols and the safety issues caused by using viral transfection reagents directly on the wound. Additionally, the inclusion of dermal substitutes addressed the lack of collagen and elastic fibers, promoting the formation of tissue resembling healthy skin rather than scar tissue. Conclusion: Integrating DGTM factor-transfected ADSCs with dermal substitutes represents a novel strategy for enhancing the healing of full-thickness wounds. Further research and clinical trials are warranted to optimize and validate this innovative approach for broader clinical applications. Full article
(This article belongs to the Section Biomedical Engineering and Materials)
Show Figures

Figure 1

Figure 1
<p>Diagrammatic summary of this study.</p>
Full article ">Figure 2
<p>Identification of ADSCs. (<b>A</b>) A Morphology of human ADSCs at 12 h and 72 h; scale bar: 100 μm. (<b>B</b>) Flow cytometry detection of ADSCs. ADSCs were positive for the markers CD73, CD90, and CD105 (blue peaks) and negative for the markers CD34 and CD45 (red peaks). The table summarizes the percentage of cells expressing each marker (mean ± SEM). (<b>C</b>) ADSC lipogenicity assay; scale bar: 100 μm. (<b>D</b>) ADSC osteogenicity assay; scale bar: 100 μm.</p>
Full article ">Figure 3
<p>Identification of ADSCs<sup>DGTM+</sup>. (<b>A</b>) Statistical graph of RT-PCR detection of the expression of transfected genes <span class="html-italic">GRHL</span>, <span class="html-italic">TFAP2</span>, <span class="html-italic">MYC</span>, and <span class="html-italic">TP63</span> in ADSCs<sup>DGTM+</sup>, ***, <span class="html-italic">p</span> &lt; 0.001. (<b>B</b>) Representative graph of immunohistochemistry detection of the expression of the keratinocyte-specific marker KRT14 in ADSCs<sup>DGTM+</sup> and ADSCs<sup>DGTM−</sup>. (<b>C</b>) WB detection of keratinocyte-specific markers KRT14 and CDH1 expression in ADSC, ADSCs<sup>DGTM+</sup> and ADSCs<sup>DGTM-</sup>. (<b>D</b>) Representative fluorescent staining of expression of keratinocyte-specific markers KRT14, CY3 (Pelnac<sup>®</sup> staining), and DAPI in the ADSCs + Pelnac<sup>®</sup> group, ADSCs<sup>DGTM+</sup> + Pelnac<sup>®</sup> group and ADSCs<sup>DGTM-</sup> + Pelnac<sup>®</sup> group.</p>
Full article ">Figure 4
<p>Construction of dermal substitute–ADSCs<sup>DGTM+</sup> complexes. (<b>A</b>) Observation of dermal substitute–ADSCs<sup>DGTM+</sup> complexes under inverted fluorescence microscope; scale bar: 200 μm. (<b>B</b>) Observation of dermal substitute–ADSCs<sup>DGTM+</sup> complexes under scanning electron microscope at different magnifications. The red arrow in the middle image points to the pore. (<b>C</b>,<b>D</b>) Observation of the proliferation of ADSCs<sup>DGTM+</sup> grown in Pelnac<sup>®</sup> by culturing ADSCs<sup>DGTM+</sup> alone within 9 days under an inverted fluorescence microscope and statistical graphs; scale bar: 200 μm.</p>
Full article ">Figure 5
<p>Dermal substitute–ADSCs<sup>DGTM+</sup> complexes fill full-thickness defective wounds. (<b>A</b>) Flowchart of animal experiments. (<b>B</b>) Representative images of skin regeneration. Mice wounds were divided into the following groups: blank, Pelnac<sup>®</sup> ADSCs + Pelnac<sup>®</sup>, and ADSCs<sup>DGTM+</sup> + Pelnac<sup>®</sup>. (<b>C</b>) Temporal variation of skin regeneration. (<b>D</b>) HE staining of mice in each group of the wound. (<b>E</b>) MASSON staining of mice in each group of the wound. (<b>F</b>) Statistical graph of the percentage of wound healing over time for each group. (<b>G</b>) Statistical graph of the thickness of re-epithelialization of the wound in each group. (<b>H</b>) Statistical graph of the proportion of wound collagen in each group. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, n = 4.</p>
Full article ">Figure 6
<p>Dermal substitute–ADSCs<sup>DGTM+</sup> complexes promote wound cell proliferation and revascularization. (<b>A</b>) Representative immunofluorescence staining of CD31, Ki67, and DAPI in the whole layer defect wounds of neoplastic skin of each wound group. (<b>B</b>) Immunofluorescence staining of CD31 in neoplastic whole skin defect wounds in each group. (<b>C</b>) Immunofluorescence staining of Ki67 in neoplastic whole skin defect wounds in each group; ** <span class="html-italic">p</span> &lt; 0.01, n = 4.</p>
Full article ">Figure 7
<p>Dermal substitute–ADSCs<sup>DGTM+</sup> complexes involved in the filling of full-thickness defective skin. (<b>A</b>) Representative fluorescent staining of KRT14, HLA-ABC, and DAPI in the wounds of mice in the ADSCs + Pelnac<sup>®</sup> and ADSCs<sup>DGTM+</sup> + Pelnac<sup>®</sup> groups. (<b>B</b>,<b>C</b>) Representative plots of average fluorescence intensity of different channels in the ADSCs<sup>DGTM+</sup> + Pelnac<sup>®</sup> group and ADSCs + Pelnac<sup>®</sup> group.</p>
Full article ">
21 pages, 8603 KiB  
Article
Cellular and Molecular Effects of the Bruck Syndrome-Associated Mutation in the PLOD2 Gene
by Olga I. Bolshakova, Evgenia M. Latypova, Artem E. Komissarov, Alexandra D. Slobodina, Elena V. Ryabova, Elena Yu. Varfolomeeva, Olga E. Agranovich, Sergey F. Batkin and Svetlana V. Sarantseva
Int. J. Mol. Sci. 2024, 25(24), 13379; https://doi.org/10.3390/ijms252413379 - 13 Dec 2024
Viewed by 247
Abstract
Bruck syndrome is a rare autosomal recessive disorder characterized by increased bone fragility and joint contractures similar to those in arthrogryposis and is known to be associated with mutations in the FKBP10 (FKBP prolyl isomerase 10) and PLOD2 (Procollagen-Lysine,2-Oxoglutarate 5-Dioxygenase [...] Read more.
Bruck syndrome is a rare autosomal recessive disorder characterized by increased bone fragility and joint contractures similar to those in arthrogryposis and is known to be associated with mutations in the FKBP10 (FKBP prolyl isomerase 10) and PLOD2 (Procollagen-Lysine,2-Oxoglutarate 5-Dioxygenase 2) genes. These genes encode endoplasmic reticulum proteins that play an important role in the biosynthesis of type I collagen, which in turn affects the structure and strength of connective tissues and bones in the body. Mutations are associated with disturbances in both the primary collagen chain and its post-translational formation, but the mechanism by which mutations lead to Bruck syndrome phenotypes has not been determined, not only because of the small number of patients who come to the attention of researchers but also because of the lack of disease models. In our work, we investigated the cellular effects of two forms of the wild-type PLOD2 gene, as well as the PLOD2 gene with homozygous mutation c.1885A>G (p.Thr629Ala). The synthesized genetic constructs were transfected into HEK293 cell line and human skin fibroblasts (DF2 line). The localization of PLOD2 protein in cells and the effects caused by the expression of different isoforms—long, short, and long with mutation—were analyzed. In addition, the results of the transcriptome analysis of a patient with Bruck syndrome, in whom this mutation was detected, are presented. Full article
(This article belongs to the Special Issue New Sights: Genetic Advances and Challenges in Rare Diseases)
Show Figures

Figure 1

Figure 1
<p>Schematic representation of genetic constructs CMV—CMV promoter: (<b>A</b>) wt <span class="html-italic">PLOD2</span> (<span class="html-italic">LH2a</span>) expression plasmid, (<b>B</b>) wt <span class="html-italic">PLOD2</span> (<span class="html-italic">LH2b</span>) expression plasmid, and (<b>C</b>) mutant Thr629Ala allele <span class="html-italic">PLOD2</span> expression plasmid.</p>
Full article ">Figure 2
<p>Transfection efficiency of a plasmid with a <span class="html-italic">GFP</span> reporter. Flow cytometry analysis of transfection efficacy of HEK293 (<b>left</b>) and DF2 (<b>right</b>) cells in 24 h (<b>A</b>). Visualization of HEK293 (<b>left</b>) and DF2 (<b>right</b>) cells in 48 h using the EVOS FL Auto Imaging System (<b>B</b>). Green – PLOD2 fused to GFP. Scale bar—200 µm.</p>
Full article ">Figure 3
<p>Confocal microscopy images of HEK293 cells transfected with three different constructs, including GFP-LH2a (<b>B</b>), GFP-LH2b (<b>C</b>), and GFP-LH2b with mutation Thr629Ala (<b>D</b>) and wild-type without constructs (<b>A</b>). In Figure (<b>D</b>), the arrow shows a change in the morphology of the cell with expression of the LH2b (Thr629Ala). The left column (green) shows the images of GFP-tagged protein PLOD2, the middle column (red) shows the images of ER, and the right column shows the overlaid images of the left and middle columns. Scale bar—25 µm.</p>
Full article ">Figure 4
<p>Visualization of DF2 cells transfected with three different constructs, including <span class="html-italic">GFP-LH2a</span> (<b>A</b>), <span class="html-italic">GFP-LH2b</span> (<b>B</b>), and <span class="html-italic">GFP-LH2b</span> with mutation Thr629Ala (<b>C</b>) using the EVOS FL Auto Imaging System. The middle column (red) shows the images of ER, and the right column shows the overlaid images of the left and middle columns. Scale bar—100 µm.</p>
Full article ">Figure 5
<p>Cell viability assays of HEK293 (<b>A</b>) and DF2 (<b>B</b>) 24, 48, and 120 h after transfection. Cell adhesion rate in each group of the HEK293 cells at 30, 60, and 90 min after transfection (<b>C</b>,<b>D</b>). Scale bar—1000 µm; mean ± SEM, Student’s <span class="html-italic">t</span>-test. * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01, *** <span class="html-italic">p</span> ≤ 0.001. N = 6 (<b>A</b>,<b>B</b>) and N = 12 (<b>C</b>).</p>
Full article ">Figure 6
<p>Cell migration assay of DF2. Scale bar—1000 µm. N = 4.</p>
Full article ">Figure 7
<p>Cell migration assay of HEK293. Scale bar—1000 µm. N = 6.</p>
Full article ">Figure 8
<p>(<b>A</b>) Volcano plot showing −log10 of adjusted <span class="html-italic">p</span>-value vs. log2FoldChange. Blue dots represent downregulated differentially expressed genes (DEGs) and red dots represent upregulated DEGs. Enrichment analysis of human phenotype of (<b>B</b>) downregulated DEGs and (<b>C</b>) upregulated DEGs.</p>
Full article ">Figure 9
<p>GO function and pathway enrichment analysis of down- and up-regulated DEGs. (<b>A</b>) BP of down- and (<b>B</b>) up-regulated DEGs, (<b>C</b>) MF of down- and (<b>D</b>) up-regulated DEGs, and (<b>E</b>) CC of down- and (<b>F</b>) up-regulated DEGs.</p>
Full article ">
15 pages, 6049 KiB  
Article
Slit1 Promotes Hypertrophic Scar Formation Through the TGF-β Signaling Pathway
by Hui Song Cui, Ya Xin Zheng, Yoon Soo Cho, Yu Mi Ro, Kibum Jeon, So Young Joo and Cheong Hoon Seo
Medicina 2024, 60(12), 2051; https://doi.org/10.3390/medicina60122051 - 12 Dec 2024
Viewed by 454
Abstract
Background and objectives: Slit1 is a secreted protein that is closely related to cell movement and adhesion. Few studies related to fibrosis exist, and the preponderance of current research is confined to the proliferation and differentiation of neural systems. Hypertrophic scars (HTSs) are [...] Read more.
Background and objectives: Slit1 is a secreted protein that is closely related to cell movement and adhesion. Few studies related to fibrosis exist, and the preponderance of current research is confined to the proliferation and differentiation of neural systems. Hypertrophic scars (HTSs) are delineated by an overproduction of the extracellular matrix (ECM) by activated fibroblasts, leading to anomalous fibrosis, which is a severe sequela of burns. However, the functionality of Slit1 in HTS formation remains unknown. We aimed to investigate whether Slit1 regulates fibroblasts through a fibrosis-related mechanism derived from post-burn HTS tissues and normal patient tissues. Methods: Human normal fibroblasts (HNFs) and hypertrophic scar fibroblasts (HTSFs) were extracted from normal skin and post-burn HTS tissues, with settings grouped according to the patient of origin. Cell proliferation was evaluated using a CellTiter-Glo Luminescent Cell Viability Assay Kit. Cell migration experiments were carried out using a μ-Dish insert system. Protein and mRNA expression levels were quantified by Western blot and quantitative real-time polymerase chain reaction. Results: We found increased expressions of Slit1 in HTS tissues and HTSFs compared to normal tissues and HNFs. The treatment of human recombinant Slit1 protein (rSlit1) within HNFs promoted cell proliferation and differentiation, leading to an upregulation in ECM components such as α-SMA, type I and III collagen, and fibronectin. The treatment of rSlit1 in HNFs facilitated cell migration, concurrent with enhanced levels of N-cadherin and vimentin, and a diminished expression of E-cadherin. Treatment with rSlit1 resulted in the phosphorylation of SMAD pathway proteins, including SMAD2, SMAD3, and SMAD1/5/8, and non-SMAD pathway proteins, including TAK1, JNK1, ERK1/2, and p38, in HNFs. Conclusions: Exogenous Slit1 potentiates the epithelial–mesenchymal transition and upregulates SMAD and non-SMAD signaling pathways in HNFs, leading to the development of HTS, suggesting that Slit1 is a promising new target for the treatment of post-burn HTS. Full article
(This article belongs to the Special Issue Burn Injuries and Burn Rehabilitation)
Show Figures

Figure 1

Figure 1
<p>Tissue morphology and expression of Slit1 in tissues and fibroblasts. (<b>A</b>) H&amp;E staining in normal skin and hypertrophic scar (HTS). The thickness of the epidermis in HTS appears to be greater than that of normal skin. The arrow marked out the epithelial layer of tissue. Images were acquired at ×10 magnification, scale bar = 50 μm. (<b>B</b>,<b>C</b>) Significantly increased levels of both mRNA and protein of Slit1 were observed in HTS tissue compared to those in normal tissues. ** <span class="html-italic">p</span> &lt; 0.01, vs. Normal. (<b>D</b>,<b>E</b>) Significantly increased levels of both mRNA and protein of Slit1 were observed in HTSFs compared to those in HNFs. HNFs and HTSFs were extracted from normal skin tissues and post-burn HTS tissues obtained from the same patients. ** <span class="html-italic">p</span> &lt; 0.01, vs. HNFs. Data represent the mean ± SD; n = 3.</p>
Full article ">Figure 2
<p>Effects of rSlit1 treatment on the proliferation and differentiation of HNFs. (<b>A</b>) Significantly increased proliferation of HNFs was observed following treatment with 10 and 100 ng/mL compared to DPBS-treated cells. (<b>B</b>,<b>C</b>) Significantly increased levels of both mRNA and protein of α-SMA (<span class="html-italic">ACTA2</span>) were observed in HNFs treated with 10 and 100 ng/mL of rSlit1 compared to DPBS-treated cells. DPBS was used as the control. * <span class="html-italic">p</span> &lt; 0.05, vs. DPBS. Data represent the mean ± SD; n = 3.</p>
Full article ">Figure 3
<p>Effects of rSlit1 treatment on the expression of ECM components in HNFs. Significant increases of both mRNA and protein levels of (<b>A</b>,<b>B</b>) type Ⅰ collagen (<span class="html-italic">COL1AⅠ</span>), (<b>C</b>,<b>D</b>) type Ⅲ collagen (<span class="html-italic">COL3AⅠ</span>), and (<b>E</b>,<b>F</b>) fibronectin (<span class="html-italic">FN1</span>) were observed in HNFs treated with 10 and 100 ng/mL of rSlit1 compared to DPBS-treated cells. DPBS was used as the control. * <span class="html-italic">p</span> &lt; 0.05, vs. DPBS. Data represent the mean ± SD; n = 3.</p>
Full article ">Figure 4
<p>Effects of rSlit1 treatment on the EMT phenotype of HNFs. The mRNA and protein levels exhibited significant increases in the expression of (<b>A</b>,<b>B</b>) vimentin (<span class="html-italic">VIM</span>) and (<b>C</b>,<b>D</b>) N-cadherin (<span class="html-italic">CDH2</span>), whereas a notable decrease in (<b>E</b>,<b>F</b>) E-cadherin (<span class="html-italic">CDH1</span>) expression was observed in HNFs treated with 10 and 100 ng/mL rSlit1, compared to those treated with DPBS. (<b>G</b>,<b>H</b>) Cell imaging demonstrated enhanced migration of HNFs treated with rSlit1 at concentrations of 10 and 100 ng/mL compared to the DPBS-treated controls. Enlarged images belong to the green box. * <span class="html-italic">p</span> &lt; 0.05, vs. DPBS. Data represent the mean ± SD; n = 3.</p>
Full article ">Figure 5
<p>Effects of rSlit1 treatment on expression of SMAD signaling in HNFs. Significantly increased phosphorylated protein expression of (<b>A</b>,<b>B</b>) SMAD2, (<b>A</b>,<b>C</b>) SMAD, and (<b>A</b>,<b>D</b>) SMAD1/5/8 was observed in HNFs treated with 10 and 100 ng/mL rSlit1, compared to DPBS-treated cells. DPBS was used as the control. * <span class="html-italic">p</span> &lt; 0.05, vs. DPBS. Data represent the mean ± SD; n = 3.</p>
Full article ">Figure 6
<p>Effects of rSlit1 treatment on expression of non-SMAD signaling in HNFs. Significantly increased phosphorylated protein expression of (<b>A</b>,<b>B</b>) TAK1, (<b>A</b>,<b>C</b>) JNK1, (<b>A</b>,<b>D</b>) ERK1/2, and (<b>A</b>,<b>E</b>) p38 was observed in HNFs treated with 10 and 100 ng/mL rSlit1 compared to DPBS-treated cells. DPBS was used as the control. * <span class="html-italic">p</span> &lt; 0.05, vs. DPBS. Data represent the mean ± SD; n = 3.</p>
Full article ">
21 pages, 35452 KiB  
Article
Assessment of Agrimonia eupatoria L. and Lipophosphonoxin (DR-6180) Combination for Wound Repair: Bridging the Gap Between Phytomedicine and Organic Chemistry
by Miriam Kaňuchová, Veronika Brindza Lachová, Kateřina Bogdanová, Jana Sabová, Petra Bonová, Tomáš Vasilenko, Ivan Kováč, Martin Novotný, Petra Mitrengová, Nitjawan Sahatsapan, Matúš Čoma, Emil Švajdlenka, Milan Kolář, Peter Bohuš, Pavel Mučaji, Robert Zajíček, Dominik Rejman and Peter Gál
Biomolecules 2024, 14(12), 1590; https://doi.org/10.3390/biom14121590 - 12 Dec 2024
Viewed by 341
Abstract
Agrimonia eupatoria L. (AE) has a rich tradition of use in wound healing improvement across various cultures worldwide. In previous studies, we revealed that Agrimonia eupatoria L. water extract (AE) possesses a rich polyphenolic composition, displaying remarkable antioxidant properties. Our investigations also demonstrated [...] Read more.
Agrimonia eupatoria L. (AE) has a rich tradition of use in wound healing improvement across various cultures worldwide. In previous studies, we revealed that Agrimonia eupatoria L. water extract (AE) possesses a rich polyphenolic composition, displaying remarkable antioxidant properties. Our investigations also demonstrated that lipophosphonoxin (LPPO) exhibited antibacterial efficacy in vitro while preserving the proliferation and differentiation of fibroblasts and keratinocytes. Building upon our prior findings, in this study, we intended to examine whether a combination of AE and LPPO could enhance skin wound healing while retaining antibacterial attributes. The antibacterial activity of AE/LPPO against Staphylococcus aureus was evaluated, alongside its effects on fibroblast-to-myofibroblast transition, the formation of extracellular matrix (ECM), and endothelial cells and keratinocyte proliferation/phenotype. We also investigated AE/LPPO’s impact on TGF-β1 and VEGF-A signaling in keratinocytes/fibroblasts and endothelial cells, respectively. Additionally, wound healing progression in rats was examined through macroscopic observation and histological analysis. Our results indicate that AE/LPPO promotes myofibroblast-like phenotypic changes and augments ECM deposition. Clinically relevant, the AE/LPPO did not disrupt TGF-β1 and VEGF-A signaling and accelerated wound closure in rats. Notably, while AE and LPPO individually exhibited antibacterial activity, their combination did not lead to synergism, rather decreasing antibacterial activity, warranting further examination. These findings underscore substantial wound healing improvement facilitated by AE/LPPO, requiring further exploration in animal models closer to human physiology. Full article
(This article belongs to the Section Natural and Bio-derived Molecules)
Show Figures

Figure 1

Figure 1
<p>High-performance liquid chromatography (HPLC) analysis of the <span class="html-italic">Agrimonia eupatoria</span> L. (AE) water extract and the second-generation lipophosphonoxin (LPPO) DR-6180. (<b>A</b>) Analysis of AE (blue) and LPPO (red) mixture. (<b>B</b>) Analysis of LPPO (blue), showing two isomers represented by two distinct peaks.</p>
Full article ">Figure 2
<p>MTS assay of HaCaT keratinocytes (<b>A</b>–<b>C</b>), human dermal fibroblasts (HDF) (<b>D</b>–<b>F</b>), and human dermal microvascular vein endothelial cells (HMVEC-d) (<b>G</b>–<b>I</b>) in the presence of <span class="html-italic">Agrimonia eupatoria</span> L. (AE) extract, lipophosphonoxin (LPPO) DR-6180, and combination of AE and LPPO. TGF-β1 was used as the positive control in HaCaT and HDF, while VEGF-A was used in HMVEC-d. The figures were generated using GraphPad Prism software (GraphPad Software, San Diego, CA, USA), which was also utilized to logarithmically transform the concentration values and generate fitting curves to visualize the data.</p>
Full article ">Figure 3
<p>Wound healing (2D migration) assay of HaCaT keratinocytes (<b>A</b>) and human dermal microvascular vein endothelial cells—HMVEC-d (<b>B</b>) in the presence of <span class="html-italic">Agrimonia eupatoria</span> L. (AE) extract, lipophosphonoxin (LPPO) DR-6180, and combination of AE and LPPO. TGF-β1 was used as the positive control in HaCaT, while VEGF-A was used in HMVEC-d (Magnification 100×).</p>
Full article ">Figure 4
<p>Western blot (WB) analysis of HaCaT keratinocytes (<b>A</b>), human dermal fibroblasts—HDF (<b>B</b>), and human dermal microvascular vein endothelial cells—HMVEC-d (<b>C</b>) in the presence of <span class="html-italic">Agrimonia eupatoria</span> L. (AE) extract, lipophosphonoxin (LPPO) DR-6180, and combination of AE and LPPO. TGF-β1 was used as the positive control in HaCaT and HDF, while VEGF-A was used in HMVEC-d.</p>
Full article ">Figure 5
<p>Immunofluorescence of HaCaT keratinocytes (<b>A</b>) and human dermal fibroblasts—HDF (<b>B</b>) in the presence of <span class="html-italic">Agrimonia eupatoria</span> L. (AE) extract, lipophosphonoxin (LPPO) DR-6180, and combination of AE and LPPO. TGF-β1 was used as the positive. Magnification 200×.</p>
Full article ">Figure 6
<p>Antimicrobial effect of AE + LPPO (DR-6180) combination tested by checkerboard microdilution assay in 69-well microtiter plate. Colors indicate wells with positive bacterial growth (yellow color for growth control (GC), and AE and DR-6180 alone; green for the combinations). The numbers next to the green areas are ∑FICs (FIC<sub>LPPO</sub> + FIC<sub>AE</sub>) values. ∑FIC &gt;1 to &lt; 2 is evaluated as indifference, and ∑FIC ≥ 2 is evaluated as antagonism.</p>
Full article ">Figure 7
<p>Wounds on rats from control (untreated), LavaSurg-treated (positive control, moist healing), and combination of <span class="html-italic">Agrimonia eupatoria</span> L. (AE) extract and lipophosphonoxin (LPPO) DR-6180 on day 14 post surgery. Scale bar = 1 cm.</p>
Full article ">Figure 8
<p>Histological assessment of wounds (hematoxylin and eosin) control (untreated), LavaSurg-treated (positive control, moist healing), and combination of <span class="html-italic">Agrimonia eupatoria</span> L. (AE) extract and lipophosphonoxin (LPPO) DR-6180 on day 14 post surgery. Scale bar = 100 µm.</p>
Full article ">
10 pages, 2757 KiB  
Article
Expression of αv Integrin in Feline Injection-Site Sarcoma (FISS): Preliminary Investigations
by Andrea Cappelleri, Eleonora Brambilla, Lavinia E. Chiti, Alessia Trapletti, Gaia B. M. Bianchi, Mauro Di Giancamillo, Valeria Grieco and Chiara Giudice
Animals 2024, 14(24), 3588; https://doi.org/10.3390/ani14243588 - 12 Dec 2024
Viewed by 322
Abstract
Feline injection-site sarcomas (FISSs) are malignant skin tumors of mesenchymal origin arising at local post-vaccination (or injection) sites. In recent years, a fluorescence imaging technique based on probes targeting αvβ3 integrin has been effectively applied for the surgical complete resection [...] Read more.
Feline injection-site sarcomas (FISSs) are malignant skin tumors of mesenchymal origin arising at local post-vaccination (or injection) sites. In recent years, a fluorescence imaging technique based on probes targeting αvβ3 integrin has been effectively applied for the surgical complete resection of the tumor. In our study, we investigated the utility of a commercially available anti-αv integrin polyclonal antibody for the histopathological evaluation of FISS’s surgical excision margins. We collected 10 formalin-fixed paraffin-embedded (FFPE) feline excisional biopsies with a histopathological diagnosis of FISS (7 fibrosarcomas and 3 pleomorphic sarcomas) and wide margin tissue, along with one subcutaneous injection-site granuloma and 6 osteosarcomas. Samples were processed for histology, and slides were stained for IHC with the anti-αv integrin antibody. Immunostained slides were evaluated for the cellular localization and intensity of the staining in different neoplastic and non-neoplastic cell populations. Neoplastic and non-neoplastic spindle cells had cytoplasmic positivity in all fibrosarcomas. Inflammatory cells, including macrophages of the injection-site granuloma, were negative. Multinucleated giant cells in the pleomorphic sarcomas had an intense membranous positivity. Although the anti-αv integrin antibody was ineffective for the histopathological evaluation of surgical excision margins, the membranous localization of αv integrin in multinucleated giant cells of pleomorphic sarcomas suggests that it plays a role in the oncogenesis of this FISS variant. Full article
(This article belongs to the Section Companion Animals)
Show Figures

Figure 1

Figure 1
<p>FISS, fibrosarcoma. Moderate cytoplasmic positivity for α<sub>v</sub> integrin. Note the presence of at least 2 mitotic figures in the field (arrowheads). IHC for α<sub>v</sub> integrin, 400×.</p>
Full article ">Figure 2
<p>FISS, pleomorphic sarcoma. (<b>a</b>) Spindle neoplastic cells are mostly negative for α<sub>v</sub> integrin, whereas round and multinucleated giant neoplastic cells display intense membranous labeling. (<b>b</b>) Detail of 2 voluminous multinucleated giant neoplastic cells displaying strong membranous labeling. IHC for α<sub>v</sub> integrin, 400×.</p>
Full article ">Figure 3
<p>FISS, margin of fibrosarcoma. Fibroblasts have diffuse, mild-to-moderate cytoplasmic positivity for α<sub>v</sub> integrin, whereas fibrocytes are mildly stained or negative (arrowhead). IHC for α<sub>v</sub> integrin, 400×.</p>
Full article ">Figure 4
<p>Injection-site granuloma. Macrophages and other inflammatory cells are negative for α<sub>v</sub> integrin. IHC for α<sub>v</sub> integrin, 400×.</p>
Full article ">
Back to TopTop