[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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (348)

Search Parameters:
Keywords = cutaneous application

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
28 pages, 335 KiB  
Review
Topical and Intralesional Treatments for Skin Metastases and Locoregionally Advanced Melanoma
by María Criado-Otero, María Navedo-de las Heras and Elia Samaniego-González
Cancers 2025, 17(1), 67; https://doi.org/10.3390/cancers17010067 - 29 Dec 2024
Viewed by 278
Abstract
Cutaneous melanoma is a malignant neoplasm with local and distant metastatic potential. When feasible, surgery is the first line of treatment in locoregionally advanced disease. Topical and intralesional treatments can be an alternative second-line treatment. The aim of this article was to perform [...] Read more.
Cutaneous melanoma is a malignant neoplasm with local and distant metastatic potential. When feasible, surgery is the first line of treatment in locoregionally advanced disease. Topical and intralesional treatments can be an alternative second-line treatment. The aim of this article was to perform a narrative review of the most widely used topical and intralesional treatments for locoregionally advanced melanoma. Diphenciprone, imiquimod and 5-florouracil were included as topical treatments and bacillus Calmette-Guerin, interleukin 2, rose bengal, talimogene laherparepvec and electrochemotherapy were included as intralesional treatments. Brief comments on other alternatives in development such as interferon-alpha, interleukin-12, ipilimumab and intralesional daromun are presented. Topical treatments generally have higher response rates in epidermal metastases than in deeper metastases. In addition, the larger the lesions, the worse they tend to respond to local treatments. Some reports show that combining certain systemic treatments and topical or intralesional therapies can improve response rates. It has also been described in a few papers that non-injected lesions may respond after the application of a local therapy in distant skin-metastases. Many of these intralesional treatments are being combined in different investigations with systemic immunotherapies, with the aim of obtaining synergic responses in those patients with refractory disease. Full article
(This article belongs to the Special Issue Topical and Intralesional Immunotherapy for Skin Cancer)
12 pages, 5422 KiB  
Article
Could Residents Adequately Assess the Severity of Skin Lesions in Mycosis Fungoides/Sézary Syndrome? Evaluation of Interrater Agreement and Interrater Reliability of mSWAT
by Hanna Cisoń, Alina Jankowska-Konsur and Rafał Białynicki-Birula
J. Clin. Med. 2025, 14(1), 75; https://doi.org/10.3390/jcm14010075 - 27 Dec 2024
Viewed by 234
Abstract
Background/Objectives: Cutaneous T-cell lymphoma (CTCL), including Mycosis fungoides (MF) and Sézary syndrome (SS), is a challenging-to-diagnose lymphoproliferative malignancy characterized by T-cell dysfunction and progressive cutaneous and extra cutaneous involvement. Disease severity assessment in CTCL is crucial for guiding treatment. This study aims [...] Read more.
Background/Objectives: Cutaneous T-cell lymphoma (CTCL), including Mycosis fungoides (MF) and Sézary syndrome (SS), is a challenging-to-diagnose lymphoproliferative malignancy characterized by T-cell dysfunction and progressive cutaneous and extra cutaneous involvement. Disease severity assessment in CTCL is crucial for guiding treatment. This study aims to evaluate the interrater agreement and interrater reliability of mSWAT among dermatology residents and identify lesion types most prone to scoring variability. Methods: Sixteen dermatology residents with varied experience levels assessed 14 patients with confirmed MF/SS diagnoses. Using mSWAT, residents independently scored lesions severity on a standardized set of patient’s photos. The results were compared with reference mSWAT scores provided by an experienced clinician. Descriptive statistics and the Shapiro–Wilk test were applied to evaluate data distributions, while Student’s t-test assessed score deviations from reference values. Furthemore, we conducted a pilot the high frequency ultrasound (HFUS) study on a single patient, whose mSWAT score and photographs are also presented in the manuscript. Results: Significant discrepancies were observed in 64.29% of cases (9/14), with tumors and infiltrative lesions in erythrodermic SS patients posing particular scoring challenges. Misclassification of tumors as patches or plaques was a frequent issue, leading to underestimations in mSWAT scores. Residents’ assessments of infiltrative lesions were also notably inconsistent. Conclusions: This study highlights significant interobserver variability in mSWAT scoring among less experienced dermatology residents, particularly with tumor and erythrodermic lesions. Findings underscore the need for enhanced training and standardized scoring protocols to improve mSWAT reliability. Similar to other comparable indices, such as PASI, the mSWAT should be employed consistently by the same physician during each assessment to systematically monitor and evaluate the skin condition of a patient under observation. However, broader application requires the acquisition of sufficient experience. The study suggests the use of the HFUS as an objective method of assessment of the skin lesion infiltration in MF/SS patients. Full article
(This article belongs to the Section Dermatology)
Show Figures

Figure 1

Figure 1
<p>The mSWAT questionnaire, originally available in English, was utilized in its Polish version in our study to accommodate Polish-speaking residents. In the questionnaire is shown data of our patients (P1), (the English version) indicated in red color, and mSWAT = 145. The values in parentheses represent the range of assessments provided by the resident physicians. For legal reasons some body regions were not assessed: head, buttocks, groin. Feet were free of lesions in all patients. These cases were stated: N/A.</p>
Full article ">Figure 2
<p>One of our patients (P1) exhibited the most pronounced changes in the assessment of infiltrative and erythematous lesions, with statistically significant differences observed in mSWAT evaluations. Reference mSWAT was 145, but mSWAT estimated by residents was from 9 to 110 (refer to <a href="#jcm-14-00075-f003" class="html-fig">Figure 3</a> and <a href="#jcm-14-00075-f004" class="html-fig">Figure 4</a> as well as <a href="#jcm-14-00075-t001" class="html-table">Table 1</a> and <a href="#jcm-14-00075-t002" class="html-table">Table 2</a>).</p>
Full article ">Figure 3
<p>The HFUS (22.5 MHz) picture of the infiltration in our patients (P1)—the broad band hypoechogenic zone interpreted as infiltration under epidermis; terms in alphabetical order: <b>D</b>—dermis; <b>EZ</b>—entrance zone; <b>F</b>—fascia; <b>H</b>—hair; <b>HZ</b>—hypoechogenic zone; <b>M</b>—membrane.</p>
Full article ">Figure 4
<p>Differences between the mSWAT score carried out by residents compared to the reference value.</p>
Full article ">
19 pages, 665 KiB  
Review
Treatment and Prevention of HPV-Associated Skin Tumors by HPV Vaccination
by Thomas Meyer and Eggert Stockfleth
Vaccines 2024, 12(12), 1439; https://doi.org/10.3390/vaccines12121439 - 20 Dec 2024
Viewed by 341
Abstract
HPV-associated dermatological diseases include benign lesions like cutaneous warts and external genital warts. In addition, HPV infection is associated with the development of epithelial skin cancers, in particular cutaneous squamous cell carcinoma (cSCC). In contrast to anogenital and oropharyngeal cancers caused by mucosal [...] Read more.
HPV-associated dermatological diseases include benign lesions like cutaneous warts and external genital warts. In addition, HPV infection is associated with the development of epithelial skin cancers, in particular cutaneous squamous cell carcinoma (cSCC). In contrast to anogenital and oropharyngeal cancers caused by mucosal HPV types of genus alpha papillomavirus, cSCC-associated HPV types belong to the genus beta papillomavirus. Currently available HPV vaccines that target mucosal HPV types associated with anogenital cancer and genital warts are type-specific and provide no cross-protection against beta HPV. When implementing vaccination to beta HPV to prevent skin tumors, it must be considered that acquisition of these HPV types occurs early in childhood and that the risk for cSCC increases with growing age and decreasing immune surveillance. Thus, individuals considered for beta HPV vaccination usually have pre-existing infection and are largely immunocompromised. On the other hand, worldwide increasing incidence rates of epithelial skin cancer reflect an urgent need for skin cancer prevention measures. Based on the pathogenic involvement of beta HPV, vaccination may represent a promising prevention strategy. Indeed, various procedures of prophylactic and therapeutic vaccination have been developed, and some of them have shown efficiency in animal models. Thus far, however, none of these vaccine candidates has been approved for application in humans. Full article
(This article belongs to the Section Human Papillomavirus Vaccines)
Show Figures

Figure 1

Figure 1
<p>Interventions available and under development for mucosal and cutaneous HPV infections. Prophylactic vaccination with licensed L1-VLPs and screening by cytology and HPV tests are established methods to prevent or monitor mucosal HPV infection. Although not approved, L1-VLPs may also be used to treat pre-existing infection. Prophylactic vaccination targeting beta HPVs and therapeutic vaccination against mucosal and cutaneous HPVs are still under development. Antiviral compounds against HPV proteins are presently not on hand and the development is still in an experimental stage.</p>
Full article ">
19 pages, 6832 KiB  
Article
Optimization and Standardization of Stable De-Epidermized Dermis (DED) Models for Functional Evaluation of Cutaneous Cell Therapies
by Xi Chen, Corinne Scaletta, Zhifeng Liao, Alexis Laurent, Lee Ann Applegate and Nathalie Hirt-Burri
Bioengineering 2024, 11(12), 1297; https://doi.org/10.3390/bioengineering11121297 - 20 Dec 2024
Viewed by 398
Abstract
The human skin is a remarkable organ capable of extensive regeneration, especially after severe injuries such as burns and related wounds. The de-epidermized dermis (DED) model has become a valuable in vitro tool for skin regeneration studies, particularly for testing the mechanism of [...] Read more.
The human skin is a remarkable organ capable of extensive regeneration, especially after severe injuries such as burns and related wounds. The de-epidermized dermis (DED) model has become a valuable in vitro tool for skin regeneration studies, particularly for testing the mechanism of action and the efficacy of clinical cutaneous cell therapies. To further improve the quality and robustness of these applications, our study focused on optimizing and standardizing DED tissue preparation and storage, enhancing its effectiveness for clinical testing. Therefore, we optimized the air-liquid interfacial culture medium composition by simplifying the historical formulation without compromising keratinocyte (therapeutic cell model) viability or proliferation. Furthermore, we investigated the impacts of adding burn wound exudates in the model by focusing on cell behavior for enhanced translational significance. The results revealed notable differences in keratinocyte adhesion and proliferation between burn wound exudates collected at the early stages and late stages of acute patient treatment, providing new information on a possible therapeutic window to apply cell therapies on burn patients. Generally, this study reported a robust method for the preclinical in vitro assessment of keratinocyte-based cutaneous cell therapies using DED models. Overall, the study underscored the importance of using in vitro models with enhanced translational relevance to better predict the clinical effects of cutaneous cell therapies in burn patient populations. Full article
(This article belongs to the Section Biomedical Engineering and Biomaterials)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>(<b>A</b>) Abdominal tissue was treated to remove the adipose tissue with surgical scissors and was cut into strips. (<b>B</b>) Tissue strips were transferred to 50 mL Falcon tubes, which were filled with NaCl 1 M. After a 24 h/37 °C incubation phase, the epidermis was separated from the dermis with forceps. Each tissue strip was then cut into individual samples of ~1.5 cm<sup>2</sup> and placed into Falcon tubes with 1× PBS + 1% P/S. The solution was changed 2–3 times before processing for long-term storage. (<b>C</b>) Description of the DED model with the air–liquid interface. A sterile perforated metal support was positioned at the bottom of a 6-well plate. The DED was first incubated in complete culture medium for at least 2 h and carefully transferred onto the support, papillary side up. Selected culture media (~4 mL/well) were added to ensure nutrient perfusion. A 6 mm glass insert was gently placed in the center of the DED, allowing for 100–200 µL of cell seeding. Constructs (i.e., DED + cells) were maintained for 4 days at 37 °C, 5% CO<sub>2</sub>. Then, the glass inserts were removed, and the constructs were incubated for 7 more days. Cellular presence and surface repartition were assessed by MTT staining of the whole construct and by H&amp;E staining of 7 µm histological sections. DED, de-epidermized dermis; PBS, phosphate-buffered saline; MTT, 3-(4,5-dimethylthiazol-2-yl) 2,5-diphenyltetrazolium bromide.</p>
Full article ">Figure 2
<p>Macroscopic images of an MTT assay for whole DED tissue samples, where media combinations were compared. The top medium (T) was used to fill the glass insert positioned on the DED. The bottom medium (B) was the DED culture medium. (<b>A</b>) Serum-free medium on both top and bottom. (<b>B</b>) mGreen’s medium on the bottom, and serum-free medium on the top. (<b>C</b>) Control sample with PBS on the bottom and complete medium on the top. (<b>D</b>) Serum-free medium is on the bottom, with complete medium on the top. (<b>E</b>) mGreen’s medium on the bottom, with complete medium on the top. DED, de-epidermized dermis; MTT, 3-(4,5-dimethylthiazol-2-yl) 2,5-diphenyltetrazolium bromide.</p>
Full article ">Figure 3
<p>The effect of the conservation temperature on DED prior to recellularization with HaCaT cells (i.e., with mGreen’s medium for the air–liquid culture) was evaluated by MTT staining and histological analysis of H&amp;E tissue sections. The MTT assay stains viable and metabolically active keratinocytes and reveals the cell distribution on the DED macroscopically. Histological H&amp;E staining allows for morphological analysis of the stratified epidermal layer with respect to cellular adhesion and migration within the dermal structure. The figure shows DED cross-sections embedded in paraffin, cut at 7 µm, and stained with H&amp;E. Storage was performed at (<b>A</b>) 4 °C for six weeks; (<b>B</b>) −20 °C for six weeks; (<b>C</b>) −80 °C for six weeks; (<b>D</b>) 4 °C for five years. (<b>E</b>) Control group with PBS alone and no nutritive media, stored at 4 °C for six weeks. Upper right corners = MTT staining in macroscopic imaging. Scale bars = 50 µm. DED, de-epidermized dermis; MTT, 3-(4,5-dimethylthiazol-2-yl) 2,5-diphenyltetrazolium bromide; PBS, phosphate-buffered saline.</p>
Full article ">Figure 4
<p>Results of culture media composition screening. (<b>C</b>) Control group. (<b>1</b>–<b>10</b>) Medium formula N.1–N.10. (<b>G</b>) mGreen’s medium. DED was evaluated macroscopically by MTT and histological analysis of H&amp;E tissue sections. The MTT assay stains viable and metabolically active keratinocytes and reveals the cell distribution on the DED macroscopically. Histological H&amp;E staining allows for morphological analysis of the stratified epidermal layer with respect to cellular adhesion and migration within the dermal structure. H&amp;E results and MTT results (i.e., upper right corners). The absence of specific medium components is represented by dark gray highlighting. Scale bars = 100 µm. C, complete medium; CT, cholera toxin; E, EGF; EGF, epidermal growth factor; G, mGreen’s medium; H, hydrocortisone; I, insulin; MTT, 3-(4,5-dimethylthiazol-2-yl) 2,5-diphenyltetrazolium bromide.</p>
Full article ">Figure 5
<p>Immunohistochemical analysis of collagen IV, laminin 1, and K14 on DED constructs maintained in mGreen’s medium and the new simplified medium N°8. (<b>A</b>) Control group. (<b>B</b>) New simplified medium N°8. (<b>C</b>) mGreen’s medium. (<b>D</b>) Human skin control group. Scale bars = 20 µm. DED, de-epidermized dermis.</p>
Full article ">Figure 6
<p>H&amp;E staining and immunohistochemical analysis of collagen IV, laminin 1, and K14 of the DED constructs maintained with different burn wound exudates. (<b>A</b>) Early collection exudate group. (<b>B</b>) Late collection exudate group. Upper right corners = macroscopic images of tissues stained with MTT. Scale bars = 20 µm. DED, de-epidermized dermis.</p>
Full article ">
18 pages, 9926 KiB  
Article
Cannabidiol-Loaded Lipid Nanoparticles Incorporated in Polyvinyl Alcohol and Sodium Alginate Hydrogel Scaffold for Enhancing Cell Migration and Accelerating Wound Healing
by Sarawut Lapmanee, Sakkarin Bhubhanil, Natthawut Charoenphon, Anjaree Inchan, Phichaporn Bunwatcharaphansakun, Mattaka Khongkow and Katawut Namdee
Gels 2024, 10(12), 843; https://doi.org/10.3390/gels10120843 - 20 Dec 2024
Viewed by 541
Abstract
Chronic wounds represent a persistent clinical challenge due to prolonged inflammation and impaired tissue repair mechanisms. Cannabidiol (CBD), recognized for its anti-inflammatory and pro-healing properties, shows therapeutic promise in wound care. However, its delivery via lipid nanoparticles (LNPs) remains challenging due to CBD’s [...] Read more.
Chronic wounds represent a persistent clinical challenge due to prolonged inflammation and impaired tissue repair mechanisms. Cannabidiol (CBD), recognized for its anti-inflammatory and pro-healing properties, shows therapeutic promise in wound care. However, its delivery via lipid nanoparticles (LNPs) remains challenging due to CBD’s inherent instability and low bioavailability. This study developed and characterized a novel hydrogel scaffold composed of CBD-loaded LNPs (CBD/LNPs) integrated into a polyvinyl alcohol (PVA) and sodium alginate (SA) matrix, designed to enhance wound repair and mitigate inflammation. The characteristics of the hydrogel scaffold were observed including the degree of swelling and LNPs’ release profiles. Furthermore, in the results, CBD/LNPs displayed enhanced stability and reduced cytotoxicity compared to unencapsulated CBD. In vitro assays demonstrated that CBD/LNPs significantly promoted fibroblast migration in gap-closure wound models and reduced intracellular reactive oxygen species, supporting their potential as a biocompatible and efficacious agent for cellular repair and oxidative stress attenuation. In vivo experiments using adult male Wistar rats with aseptic cutaneous wounds revealed that treatment with CBD/LNP-PVA/SA hydrogel scaffold significantly accelerated wound closure relative to blank hydrogel controls, demonstrating a substantial reduction in the wound area over time. Histological analysis confirms notable improvements in skin morphology in wounds treated with CBD/LNP-PVA/SA hydrogel scaffold with evidence of accelerated epithelialization, enhanced collagen deposition, and increased dermal thickness and vascularization. Additionally, skin histology showed a more organized epidermal layer and reduced inflammatory cell infiltration in CBD/LNP-PVA/SA hydrogel scaffold-treated wounds, corresponding to a 35% increase in the wound closure rate by day 28 post-treatment. These findings suggest that CBD/LNP-PVA/SA hydrogel scaffolds facilitate inflammation resolution and structural wound healing through localized, sustained CBD delivery. The dual anti-inflammatory and wound-healing effects position CBD/LNP-PVA/SA hydrogel scaffold as a promising approach for chronic wound management. Future investigations are warranted to elucidate the mechanistic pathways by which CBD modulates the skin architecture and to explore its translational applications in clinical wound care. Full article
(This article belongs to the Special Issue Synthesis and Applications of Hydrogels (2nd Edition))
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Swelling behavior of polyvinyl alcohol and sodium alginate (PVA/SA) hydrogel scaffold. Degree of swelling observed in hydrogel compositions containing varying percentages of sodium alginate (SA 25%, 50%, 75%, and 100%) and swelling ratio percentage measured at different time points (0, 15, 30, and 45 min). Significant differences were observed, with ** <span class="html-italic">p</span>  &lt;  0.01 and *** <span class="html-italic">p</span>  &lt;  0.001 compared to SA 0% at the starting point (0 min), as presented in the graph.</p>
Full article ">Figure 2
<p>Release behavior of polyvinyl alcohol and sodium alginate (PVA/SA) hydrogel scaffold. (<b>A</b>) The degree of release was evaluated in hydrogel compositions containing varying percentages of sodium alginate (SA) incorporated with CBD/LNPs, including formulations with 25%, 50%, 75%, and 100% SA. (<b>B</b>) Release percentages measured at different time points (0 min, 15 min, 30 min, 1 h, 2 h, 4 h, 8 h, and 24 h). Significant differences were observed, with * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span>  &lt;  0.01, and *** <span class="html-italic">p</span>  &lt;  0.001 compared to SA 25%, as presented in the graph.</p>
Full article ">Figure 3
<p>Characteristics, cytotoxicity, and antioxidant activity of cannabidiol-loaded lipid nanoparticles (CBD/LNPs). (<b>A</b>) Morphology of CBD/LNPs as demonstrated by Transmission Electron Microscopy (TEM); (<b>B</b>) Cell viability relative to the control in fibroblast-cultured cells exposed to CBD/LNPs; (<b>C</b>) Radical oxidative stress levels induced by hydrogen peroxide exposure were measured by the fluorescent intensity of dichlorofluorescein (DCF) relative to the control group, referring to untreated cells exposed only to the medium without any treatment. The analysis was conducted in fibroblast cultures treated with CBD/LNPs. Data are expressed as mean  ±  SEM (<span class="html-italic">n</span> = 4). * <span class="html-italic">p</span>  &lt;  0.05, ** <span class="html-italic">p</span>  &lt;  0.01, and *** <span class="html-italic">p</span>  &lt;  0.001 compared to the blank group.</p>
Full article ">Figure 4
<p>Effects of cannabidiol-loaded lipid nanoparticles (CBD/LNPs) on wound healing in fibroblast-cultured cells. (<b>A</b>) Human dermal fibroblast cells were treated with 50 ppm CBD/LNPs or 100 ng/mL FGF (positive control). A scratch wound assay was monitored at 0, 24, and 48 h post-scratch and compared to untreated control cells. (<b>B</b>) Percentage of wound gap closure at 24 and 48 h, calculated relative to the initial scratch width (T<sub>0</sub>). Data are expressed as mean  ±  SEM (<span class="html-italic">n</span>  =  3). *** <span class="html-italic">p</span>  &lt;  0.001 compared to the control group.</p>
Full article ">Figure 5
<p>Effects of cannabidiol-loaded lipid nanoparticles incorporated in polyvinyl alcohol and sodium alginate (CBD/LNP-PVA/SA) hydrogel scaffold on wound healing in rat skin. (<b>A</b>) Time-dependent progression of wound closure in rats treated with PVA/SA hydrogel (Blank) and CBD/LNP-PVA/SA hydrogel. (<b>B</b>) Percentage of wound area contraction calculated on days 3, 7, 14, 21, and 28 post-treatment. Data are expressed as mean  ±  SEM (<span class="html-italic">n</span> = 3 per time point). * <span class="html-italic">p</span>  &lt;  0.05 and ** <span class="html-italic">p</span>  &lt;  0.01 compared to the blank group on the respective day.</p>
Full article ">Figure 6
<p>Effects of cannabidiol-loaded lipid nanoparticles incorporated into polyvinyl alcohol and sodium alginate (CBD/LNP-PVA/SA) hydrogel scaffold on histopathological changes in rat skin on days 3, 7, 14, 21, and 28 post-wounding, as assessed by H&amp;E and Masson’s trichrome staining. Photographs show wound sections from rats treated with PVA/SA (Blank) and CBD/LNP-PVA/SA hydrogel scaffold. Hematoxylin-stained nucleic acids and nuclei, eosin-stained proteins, and collagen are visualized in blue–green. Scale bar = 200 μm.</p>
Full article ">
24 pages, 766 KiB  
Review
Melanoma’s New Frontier: Exploring the Latest Advances in Blood-Based Biomarkers for Melanoma
by Ivana Prkačin, Mislav Mokos, Nikola Ferara and Mirna Šitum
Cancers 2024, 16(24), 4219; https://doi.org/10.3390/cancers16244219 - 18 Dec 2024
Viewed by 435
Abstract
Melanoma is one of the most malignant cancers, and the global incidence of cutaneous melanoma is increasing. While melanomas are highly prone to metastasize if diagnosed late, early detection and treatment significantly reduce the risk of mortality. Identifying patients at higher risk of [...] Read more.
Melanoma is one of the most malignant cancers, and the global incidence of cutaneous melanoma is increasing. While melanomas are highly prone to metastasize if diagnosed late, early detection and treatment significantly reduce the risk of mortality. Identifying patients at higher risk of metastasis, who might benefit from early adjuvant therapies, is particularly important, especially with the advent of new melanoma treatments. Therefore, there is a pressing need to develop additional prognostic biomarkers for melanoma to improve early stratification of patients and accurately identify high-risk subgroups, ultimately enabling more effective personalized treatments. Recent advances in melanoma therapy, including targeted treatments and immunotherapy, have underscored the importance of biomarkers in determining prognosis and predicting treatment response. The clinical application of these markers holds the potential for significant advancements in melanoma management. Various tumor-derived genetic, proteomic, and cellular components are continuously released into the bloodstream of cancer patients. These molecules, including circulating tumor DNA and RNA, proteins, tumor cells, and immune cells, are emerging as practical and precise liquid biomarkers for cancer. In the current era of effective molecular-targeted therapies and immunotherapies, there is an urgent need to integrate these circulating biomarkers into clinical practice to facilitate personalized treatment. This review highlights recent discoveries in circulating melanoma biomarkers, explores the challenges and potentials of emerging technologies for liquid biomarker discovery, and discusses future directions in melanoma biomarker research. Full article
(This article belongs to the Section Cancer Biomarkers)
Show Figures

Figure 1

Figure 1
<p>Peripheral blood centrifugation by density gradient with components of each layer. Abbreviations: cfDNA: cell-free DNA; cfRNA: cell-free RNA. Created in BioRender. Mokos, M. (2024) <a href="https://BioRender.com/x74l463" target="_blank">https://BioRender.com/x74l463</a>.</p>
Full article ">
21 pages, 1174 KiB  
Review
Advances in Encapsulating Marine Bioactive Compounds Using Nanostructured Lipid Carriers (NLCs) and Solid Lipid Nanoparticles (SLNs) for Health Applications
by Rita Favas, Hugo Almeida, Andreia F. Peixoto, Domingos Ferreira and Ana C. Silva
Pharmaceutics 2024, 16(12), 1517; https://doi.org/10.3390/pharmaceutics16121517 - 25 Nov 2024
Viewed by 484
Abstract
As life expectancy rises and modern lifestyles improve, there is an increasing focus on health, disease prevention, and enhancing physical appearance. Consumers are more aware of the benefits of natural ingredients in healthcare products while also being mindful of sustainability challenges. Consequently, marine [...] Read more.
As life expectancy rises and modern lifestyles improve, there is an increasing focus on health, disease prevention, and enhancing physical appearance. Consumers are more aware of the benefits of natural ingredients in healthcare products while also being mindful of sustainability challenges. Consequently, marine bioactive compounds have gained popularity as ingredients in cosmetics and food supplements due to their diverse beneficial properties. Nonetheless, the use of some of these compounds is restricted by their low stability and poor aqueous solubility, necessitating solutions to overcome these limitations. In this context, lipid nanoparticles, such as solid lipid nanoparticles (SLNs) and nanostructured lipid carriers (NLCs), have been investigated for their potential to protect and improve the absorption of molecules through various routes, including oral and cutaneous. Numerous studies have shown that nanoencapsulating these compounds and incorporating them into cosmetics and food supplements can be effective. However, this application remains unregulated at the global level and is not currently addressed by existing legislation. Additional in vivo studies in both animals and humans are necessary to fully assess safety concerns. Full article
(This article belongs to the Topic New Challenges in the Cosmetics Industry)
Show Figures

Figure 1

Figure 1
<p>Different types of SLNs: (<b>A</b>) Homogenous matrix, (<b>B</b>) drug-enriched shell, and (<b>C</b>) drug-enriched core.</p>
Full article ">Figure 2
<p>Different types of NLCs: (<b>A</b>) Imperfect, (<b>B</b>) amorphous, and (<b>C</b>) multiple.</p>
Full article ">
11 pages, 6363 KiB  
Review
Advances in Tumor-Infiltrating Lymphocyte (TIL) as a Prognostic Factor and for Treating Invasive Cutaneous Melanoma
by Gabriel Alves Freiria de Oliveira, Daniel Arcuschin de Oliveira, Melissa Yoshimi Sakamoto Maeda Nisimoto, Rafael Rubinho, Heitor Carvalho Gomes, Luciana Cavalheiro Marti and Renato Santos de Oliveira Filho
Int. J. Mol. Sci. 2024, 25(23), 12596; https://doi.org/10.3390/ijms252312596 - 23 Nov 2024
Viewed by 725
Abstract
Invasive cutaneous melanoma is responsible for about 5% of skin tumors yet is liable for nearly 70% of skin cancer-related deaths. Despite notable advancements over the past decade, including immunotherapies and targeted treatments, more than half of invasive melanoma patients ultimately succumb to [...] Read more.
Invasive cutaneous melanoma is responsible for about 5% of skin tumors yet is liable for nearly 70% of skin cancer-related deaths. Despite notable advancements over the past decade, including immunotherapies and targeted treatments, more than half of invasive melanoma patients ultimately succumb to the disease due to therapeutic resistance. To overcome this obstacle, strategies such as combining immunotherapies with targeted drugs or adding epigenetic therapies have been investigated. Tumor-infiltrating lymphocytes (TILs) therapy has emerged as a promising option for patients whose disease continues to progress despite standard treatments. This article aims to introduce TIL therapy and review recent outcomes in melanoma prognosis in its application for melanoma management. Full article
(This article belongs to the Special Issue Melanoma: Molecular Mechanisms and Therapy)
Show Figures

Figure 1

Figure 1
<p>Schematic illustration of the interaction between tumor cells and immune cells. (<b>A</b>) Tumor cells promote a regulatory environment that impairs the immune system’s ability to act against them. (<b>B</b>) Immune cells overcome the tumor environment and destroy the tumor cells. The cells are represented by colors such as: light green (CD4–Th), dark green (CD4–Treg), light blue (CD8–Tc), grey (CD8 exhausted anergic or senescent). The soluble factors are represented by forms and colors such as: TGF-β, PGE-2, IL-10 (pink dots), Perforin and granzymes (blue dots), GM-CSF (orange triangles), IFN-γ, TNF-α and IL-2 (green triangles).</p>
Full article ">Figure 2
<p>Schematic demonstration of the TIL process and therapy. The cells are represented by colors such as: light pink (TILs), brown (tumor cells), violet (feeder cells) and the stimulatory antibody was represented by blue (anti-CD3).</p>
Full article ">Figure 3
<p>(<b>A</b>) Flowchart of articles selection. (<b>B</b>) Distribution of articles by subject and type.</p>
Full article ">
18 pages, 4004 KiB  
Article
Toxicity and Dermatokinetic Analysis of Ibrutinib in Human Skin Models
by Maria Victória Souto-Silva, Elizabete C. I. Bispo, Lucas F. F. Albuquerque, Stefhani Barcelos, Emãnuella M. Garcez, Luana S. Quilici, Florêncio Figueiredo Cavalcanti Neto, Eliza Carla Barroso Duarte, Jankerle N. Boeloni, Felipe Saldanha-Araujo, Guilherme M. Gelfuso and Juliana Lott Carvalho
Pharmaceutics 2024, 16(11), 1377; https://doi.org/10.3390/pharmaceutics16111377 - 26 Oct 2024
Viewed by 856
Abstract
Background/Objectives: Ibrutinib (IBR) is a tyrosine kinase inhibitor under investigation in preclinical and clinical settings as an alternative treatment for melanoma. Nevertheless, the limited oral bioavailability of IBR and the need for high doses of the drug to kill melanoma cells are major [...] Read more.
Background/Objectives: Ibrutinib (IBR) is a tyrosine kinase inhibitor under investigation in preclinical and clinical settings as an alternative treatment for melanoma. Nevertheless, the limited oral bioavailability of IBR and the need for high doses of the drug to kill melanoma cells are major drawbacks for this purpose. Considering that melanoma is restricted to the skin at early stages, the topical application of IBR might constitute an effective and safer administration route. In this study, we determined IBR’s toxicity and dermatokinetics using human primary cells and human organotypic skin explant cultures (hOSECs). Methods: After demonstrating that human primary fibroblasts and keratinocytes present IBR target genes, the cytotoxicity of the drug was determined using the MTT and annexin V/PI staining assays. IBR toxicity in the skin was assessed using the TTC assay, and the irritation potential was established using histological assessment. Finally, IBR cutaneous permeation was assessed ex vivo to determine the drug dermatokinetics. Results: Our findings reveal that IBR exerts dose-dependent toxicity towards skin cells, presenting an IC50 in the same range as melanoma cells. The topical application of the drug successfully reduced irritation and toxicity in the skin, and the drug was shown to successfully permeate the stratum corneum and reach the viable skin layers in therapeutic concentrations. Conclusions: Overall, our data encourage the topical application of IBR to treat melanoma, paving the way for future studies in this theme. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Experimental design. This study included 4 different phases. 1. qRT-PCR of IBR target genes was performed to evaluate the expression of these genes in primary human skin and melanoma cells. 2. Human primary keratinocytes and fibroblasts were isolated and treated with increasing concentrations of IBR to carry out MTT metabolization and apoptosis detection assays. Melanoma cells were also investigated. 3. hOSEC models were used to determine the toxicity and irritation profile of IBR applied topically and in the tissue culture media at different concentrations. The TTC metabolization assay was performed, and the skin irritation score was determined using histology. 4. Assessment of IBR dermatokinetics in hOSECs. Drug permeation was determined after 12 and 24 h of IBR application using HPLC.</p>
Full article ">Figure 2
<p>Human organotypic skin explant cultures. Skin fragments obtained from healthy donors were processed and cut into circular 6 mm fragments. The explants were maintained in an air–liquid interface for up to 7 days.</p>
Full article ">Figure 3
<p>qRT-PCR of IBR target genes in primary cells from the skin. The mRNA levels of EGFR (<b>A</b>), JAK3 (<b>B</b>), BTK (<b>C</b>), ITK (<b>D</b>), and ERBB2 (<b>E</b>) were determined in human primary fibroblasts (HFIB) and keratinocytes (KCS). The median Ct values obtained from SKMEL-28 cells were used as a reference. All real-time PCR reactions were performed in technical duplicate. The relative fold change was obtained using the formula 2−ΔΔCt, using GAPDH to normalize sample loading. No comparisons between the groups resulted in statistically significant differences. The data were normalized to SKMEL-28 mRNA levels and are represented with the mean ± standard error of the mean (SEM). The data were analyzed using the ANOVA and Tukey post hoc test.</p>
Full article ">Figure 4
<p>Cytotoxicity of IBR towards human primary skin cells. Human primary skin fibroblasts (<b>A</b>), keratinocytes (<b>B</b>), and SKMEL-28 cells (<b>C</b>) were treated with increasing doses of IBR for 48 h and had their viability assessed using the MTT assay. (<b>D</b>) Dose-response cytotoxicity of IBR to the different cell types tested. The data obtained from the MTT assay were used to determine the IC<sub>50</sub> and IC<sub>25</sub> of IBR for each cell type (<b>E</b>). The data are presented as the mean ± SEM. The data were analyzed using the ANOVA followed by Tukey’s post hoc test for multiple comparisons. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 5
<p>Apoptotic induction by IBR in human primary skin cells. Human primary skin fibroblasts (<b>A</b>), keratinocytes (<b>B</b>), and SKMEL-28 cells (<b>C</b>) were treated with the IC<sub>25</sub>, IC<sub>50</sub>, as well as high (100 µM) IBR doses for 48 h and had their viability assessed using annexin V and PI staining. Representative dot plots are shown on the left. The data are presented as the mean ± SEM. The data were analyzed using the ANOVA followed by Tukey’s post hoc test for multiple comparisons. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 6
<p>Irritation potential of IBR in hOSEC models. Human organotypic skin explant cultures were treated with increasing doses of IBR. The drug was either added in the tissue culture medium (<b>A</b>) or topically administered (<b>B</b>). After 48 h, tissue metabolic viability was assessed using the TTC assay. The data are presented as the mean ± SEM. The data were analyzed using the ANOVA followed by Tukey’s post hoc test for multiple comparisons. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001; and # <span class="html-italic">p</span> &lt; 0.0001 compared to all other groups.</p>
Full article ">Figure 7
<p>Histological observations of ex vivo skin samples treated with IBR that was added into the tissue culture medium. (<b>A</b>) Control group with slight peri-nuclear edema (white arrow) in the epidermis. (<b>B</b>) Group treated with 10 µM presenting mild peri-nuclear edema (white arrow) in the epidermis. (<b>C</b>) Group treated with 25 µM presenting mild peri-nuclear edema (white arrow) in the epidermis. (<b>D</b>) Group treated with 50 µM presenting mild peri-nuclear edema (white arrow) in the epidermis. (<b>E</b>) Group treated with 100 µM presenting mild peri-nuclear edema (white arrow) in the epidermis. (<b>F</b>) Group treated with DMSO presenting nuclear pyknosis (white arrowhead). (<b>G</b>) Group treated with SDS presenting necrotic skin and dermal–epidermal junction separation (asterisk). Scale bar: 100 µm. Masson’s trichrome.</p>
Full article ">Figure 8
<p>Histological observations of ex vivo skin samples topically treated with IBR. (<b>A</b>) Control group with slight peri-nuclear edema (white arrow) in the epidermis. (<b>B</b>) Group treated with 10 µM presenting mild peri-nuclear edema (white arrow) in the epidermis. (<b>C</b>) Group treated with 25 µM presenting mild peri-nuclear edema (white arrow) in the epidermis. (<b>D</b>) Group treated with 50 µM presenting mild peri-nuclear edema (white arrow) in the epidermis. (<b>E</b>) Group treated with 100 µM presenting mild peri-nuclear edema (white arrow) in the epidermis. (<b>F</b>) Group treated with DMSO presenting nuclear pyknosis (white arrow). (<b>G</b>) Group treated with SDS presenting necrotic skin, nuclear pyknosis (white arrowhead), and dermal–epidermal junction separation (asterisk). Scale bar: 100 µm. Masson’s trichrome.</p>
Full article ">Figure 9
<p>IBR dermatokinetics analysis in human skin. IBR was topically administered in human skin samples. The amount of drug recovered from the stratum corneum and remaining skin after 12 and 24 h was determined using the HPLC method. The data are presented as the mean ± SEM. The data were analyzed using a two-way ANOVA followed by Sidak’s post hoc test for multiple comparisons; * <span class="html-italic">p</span> &lt; 0.05; **** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">
24 pages, 5361 KiB  
Review
Nutritional and Microbial Strategies for Treating Acne, Alopecia, and Atopic Dermatitis
by Alejandro Borrego-Ruiz and Juan J. Borrego
Nutrients 2024, 16(20), 3559; https://doi.org/10.3390/nu16203559 - 20 Oct 2024
Viewed by 3362
Abstract
Background/Objectives: Diet is one of the major determinants of the composition and function of the gut microbiome, and diverse studies have established directional connections between gut microbiome dysbiosis and skin dyshomeostasis. Furthermore, a significant link between the gut and certain skin-related disorders has [...] Read more.
Background/Objectives: Diet is one of the major determinants of the composition and function of the gut microbiome, and diverse studies have established directional connections between gut microbiome dysbiosis and skin dyshomeostasis. Furthermore, a significant link between the gut and certain skin-related disorders has been reported. This work reviews the mechanisms underlying the relationship between nutritional factors, gut microbiome, and certain skin diseases such as acne vulgaris, alopecia, and atopic dermatitis. In addition, it explores how the modulation of the gut microbiome and human skin through diet and various microbial strategies, including probiotics, synbiotics, postbiotics, and fecal microbiota transplantation, may serve as future treatments for skin diseases, possibly replacing traditional methods such as antibiotic, topical corticosteroid, and laser therapies. Results: The adequate intake of certain foods can promote a balanced gut microbiome, potentially reducing skin inflammation and improving overall skin health, while poor dietary choices may lead to worse outcomes by disrupting gut homeostasis. In this regard, diets high in antioxidants, fiber, and phytonutrients appear to be beneficial for enhancing skin health and preventing associated comorbidities. In addition, the administration of probiotics, synbiotics, and postbiotics in the treatment of cutaneous diseases has been shown to restore skin dyshomeostasis and to improve the symptoms of the reviewed skin conditions. Conclusions: Consuming a healthy, plant-based diet can reduce skin inflammation and enhance overall skin health. Although the application of probiotics, synbiotics, and postbiotics has demonstrated promise in modulating inflammation, enhancing tissue regeneration, and inhibiting pathogenic colonization, further research is required. Full article
(This article belongs to the Section Prebiotics and Probiotics)
Show Figures

Figure 1

Figure 1
<p>Hypothetical relationship between gut microbiome dysbiosis, the Western diet, and acne (based on Rygula et al. [<a href="#B44-nutrients-16-03559" class="html-bibr">44</a>] and Melnik et al. [<a href="#B45-nutrients-16-03559" class="html-bibr">45</a>]).</p>
Full article ">Figure 2
<p>Hypothetical relationship between gut microbiome dysbiosis, low-protein diet, and alopecia vulgaris via the JAK-STAT pathway (based on Simakou et al. [<a href="#B51-nutrients-16-03559" class="html-bibr">51</a>]).</p>
Full article ">Figure 3
<p>Hypothetical relationship between gut microbiome dysbiosis, the Western diet, and atopic dermatitis (based on Moniaga et al. [<a href="#B62-nutrients-16-03559" class="html-bibr">62</a>], Kim et al. [<a href="#B63-nutrients-16-03559" class="html-bibr">63</a>], and Guo et al. [<a href="#B64-nutrients-16-03559" class="html-bibr">64</a>]).</p>
Full article ">Figure 4
<p>Mechanisms of probiotics and prebiotics involved in the improvement of skin diseases (red arrows represent inhibition processes). Probiotics can restore gut homeostasis by improving GM dysbiosis and repairing intestinal mucosal damage. Additionally, probiotics mitigate skin damage by inhibiting oxidative stress, inflammatory response, and immune homeostasis. Prebiotics, in turn, enhance probiotics growth.</p>
Full article ">
17 pages, 2324 KiB  
Article
Local Inflammatory and Systemic Antibody Responses Initiated by a First Intradermal Administration of Autogenous Salmonella-Killed Vaccines and Their Components in Pullets
by Jossie M. Santamaria, Chrysta N. Beck and Gisela F. Erf
Vaccines 2024, 12(10), 1159; https://doi.org/10.3390/vaccines12101159 - 11 Oct 2024
Cited by 1 | Viewed by 872
Abstract
Vaccination strategies are used to manage Salmonella in chickens. Salmonella-killed vaccines are considered safer since they are inactivated. However, little is known regarding the cellular immune activities at the site of vaccine administration of Salmonella-killed vaccines. The growing feather (GF) cutaneous [...] Read more.
Vaccination strategies are used to manage Salmonella in chickens. Salmonella-killed vaccines are considered safer since they are inactivated. However, little is known regarding the cellular immune activities at the site of vaccine administration of Salmonella-killed vaccines. The growing feather (GF) cutaneous test has been shown to be an effective bioassay to monitor local tissue/cellular responses. We assessed local and systemic antibody responses initiated by intradermal injection of Salmonella-killed vaccines into GF-pulps of 14–15-week-old pullets. Treatments consisted of two autogenous Salmonella-killed vaccines (SV1 and SV2), S. Enteritidis (SE) lipopolysaccharide (SE-LPS), and the water-oil-water (WOW) emulsion vehicle. GF-pulps were collected before (0 h) and at 6, 24, 48, and 72 h post-GF-pulp injection for leukocyte population analysis, while heparinized blood samples were collected before (0 d) and at 3, 5, 7, 10, 14, 21, and 28 d after GF-pulp injections to assess plasma levels (a.u.) of SE-specific IgM, avian IgY (IgG), and IgA antibodies using an ELISA. Injection of GF-pulps with SV1, SV2, or SE-LPS, all in a WOW vehicle, initiated inflammatory responses characterized by the recruitment of heterophils, monocytes/macrophages, and a few lymphocytes. The WOW vehicle emulsion alone recruited more lymphocytes than vaccines or SE-LPS. The SV1 and SV2 vaccines stimulated Salmonella-specific IgM and IgA early, while IgG levels were greatly elevated later during the primary response. Overall, SV1 and SV2 stimulated a heterophil and macrophage-dominated local inflammatory- and SE-specific humoral response with an isotype switch from IgM to IgG, characteristic of a T-dependent primary antibody response. This study provides comprehensive information on innate and adaptive immune responses to autogenous Salmonella-killed vaccines and their components that will find application in the management of Salmonella in poultry. Full article
(This article belongs to the Special Issue Veterinary Vaccines and Host Immune Responses)
Show Figures

Figure 1

Figure 1
<p>T cell infiltration profiles in response to injection of autogenous <span class="html-italic">Salmonella</span>-killed vaccines (SV1 and SV2), <span class="html-italic">S</span>. Enteritidis lipopolysaccharide (SE-LPS), or vehicle into the pulp of growing feathers. Twenty-four growing feathers (GF) of 14 to 15 wk old Light-brown Leghorn (LBL) pullets from (<b>A</b>) Trial 2-PHL and (<b>B</b>) Trial 3-Farm were injected with 10 µL of SV1, SV2, SE-LPS, or vehicle (water-oil-water emulsion). Injected GFs from each chicken were collected before injection (0 h) and at 6, 24, 48, and 72 h post-GF injection for leukocyte population analysis. Pulp cell suspensions were prepared from each GF, immunofluorescently stained with fluorescence-conjugated mouse monoclonal antibody (Southern Biotech) to chicken CD3 (T cells), and the percentage of CD3+ pulp cells was determined by fluorescence-based flow cytometry. Data shown are mean ± SEM. For each Trial, <span class="html-italic">n</span> = 4 pullets for SV1, SV2, and LPS, and <span class="html-italic">n</span> = 3 for the vehicle. Due to interactions involving the Trial, two-way ANOVA was conducted for each Trial. Student <span class="html-italic">t</span>-test multiple means comparisons were conducted to identify Treatment (Trt) and Time (h) differences. a–c: for each time point, treatment means without a common letter are different (<span class="html-italic">p</span> &lt; 0.05); w–z: for each treatment, means at each time-point without a common letter are different (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 2
<p>B cell infiltration profiles in response to injection of autogenous <span class="html-italic">Salmonella</span>-killed vaccines (SV1 and SV2), <span class="html-italic">S.</span> Enteritidis lipopolysaccharide (SE-LPS), or vehicle into the pulp of growing feathers. Forty-eight growing feathers (GF) of 14 to 15 wk old Light-brown Leghorn (LBL) pullets from Trial 2-PHL and Trial 3-Farm were injected with 10 µL of SV1, SV2, SE-LPS, or vehicle (water-oil-water emulsion). Injected GFs from each chicken were collected before injection (0 h) and at 6, 24, 48, and 72 h post-GF injection for leukocyte population analysis. Pulp cell suspensions were prepared from each GF, immunofluorescently stained with fluorescence-conjugated mouse monoclonal antibody (Southern Biotech) to chicken Bu-1 (B cells), and the percentage of Bu-1+ pulp cells determined by fluorescence-based flow cytometry. Data shown are mean ± SEM. For each Trial, <span class="html-italic">n</span> = 8 pullets for SV1, SV2, and LPS, and <span class="html-italic">n</span> = 6 for the vehicle. Student <span class="html-italic">t</span>-test multiple means comparisons were conducted to identify Treatment (Trt) and Time (h) differences. a–c: for each time point, treatment means without a common letter are different (<span class="html-italic">p</span> &lt; 0.05); x–z: for each treatment, time means without a common letter are different (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 3
<p><span class="html-italic">Salmonella</span> Enteritidis specific IgM levels in plasma after injection of autogenous <span class="html-italic">Salmonella</span>-killed vaccines (SV1 and SV2), <span class="html-italic">S.</span> Enteritidis lipopolysaccharide (SE-LPS), or vehicle into the pulp of growing feathers. SE-specific IgM levels in the plasma of 14–15 wk old Light-brown Leghorn (LBL) pullets were measured after intradermal injection of SV1, SV2, SE-LPS, or vehicle (water-oil-water emulsion) into the pulp of growing feathers. Data shown were pooled across three Trials. Heparinized blood (1.5 mL) was collected before (0 d) and at 3, 5, 7, 10, 14, 21, and 28 d post-pulp injection. Data are means ± SEM; <span class="html-italic">n</span> = 12 pullets for SV1, SV2, and SE-LPS and <span class="html-italic">n</span> = 6 for vehicle (V). Two-way repeated measures ANOVA; Student <span class="html-italic">t</span>-test multiple means comparison-test was used to determine Treatment (Trt) and Time (Day) differences. a,b: for each time point, treatment means without a common letter are different (<span class="html-italic">p</span> &lt; 0.05); w–z: for each treatment, time means without a common letter are different (<span class="html-italic">p</span> &lt; 0.05). Note: the total SV1 and SV2 immunization doses for Trial 1 were 0.140 mL/bird, and for Trial 2 &amp; 3, 0.165 mL/bird. The total SE-LPS dose was 5 µg/bird for all Trials.</p>
Full article ">Figure 4
<p><span class="html-italic">Salmonella</span> Enteritidis specific IgG levels in plasma after injection of autogenous <span class="html-italic">Salmonella</span>-killed vaccines (SV1 and SV2), <span class="html-italic">S</span>. Enteritidis lipopolysaccharide (SE-LPS), or vehicle into the pulp of growing feathers. SE-specific IgG levels in the plasma of 14–15 wk old Light-brown Leghorn (LBL) pullets were measured after intradermal injection of SV1, SV2, SE-LPS, or vehicle (water-oil-water emulsion) into the pulp of growing feathers. Data shown were pooled across three Trials. Heparinized blood (1.5 mL) was collected before (0 d) and at 3, 5, 7, 10, 14, 21, and 28 d post-pulp injection. Data are means ± SEM; <span class="html-italic">n</span> = 12 pullets for SV1, SV2, and SE-LPS and <span class="html-italic">n</span> = 6 for vehicle (V). Two-way repeated measures ANOVA; Student <span class="html-italic">t</span>-test multiple means comparison-test was used to determine Treatment (Trt) and Time (Day) differences. a,b: for each time point, treatment means without a common letter are different (<span class="html-italic">p</span> &lt; 0.05); w–z: for each treatment, time means without a common letter are different (<span class="html-italic">p</span> &lt; 0.05). Note: the total SV1 and SV2 immunization doses for Trial 1 were 0.140 mL/bird, and for Trial 2 &amp; 3, 0.165 mL/bird. The total SE-LPS dose was 5 µg/bird for all Trials.</p>
Full article ">Figure 5
<p><span class="html-italic">Salmonella</span> Enteritidis specific IgA levels in plasma after injection of autogenous <span class="html-italic">Salmonella</span>-killed vaccines (SV1 and SV2), <span class="html-italic">S.</span> Enteritidis lipopolysaccharide (SE-LPS), or vehicle into the pulp of growing feathers. SE-specific IgA levels in the plasma of 14–15 wk old Light-brown Leghorn (LBL) pullets were measured after intradermal injection of SV1, SV2, SE-LPS, or vehicle (water-oil-water emulsion) into the pulp of growing feathers. Data shown were pooled across three trials. Heparinized blood (1.5 mL) was collected before (0 d) and at 3, 5, 7, 10, 14, 21, and 28 d post-pulp injection. Data are means ± SEM; <span class="html-italic">n</span> = 12 pullets for SV1, SV2, and SE-LPS and <span class="html-italic">n</span> = 6 for vehicle (V). Two-way repeated measures ANOVA; Student <span class="html-italic">t</span>-test multiple means comparison-test was used to determine Treatment (Trt) and Time (Day) differences. a–c: for each time point, treatment means without a common letter are different (<span class="html-italic">p</span> &lt; 0.05); w–z: for each treatment, time means without a common letter are different (<span class="html-italic">p</span> &lt; 0.05). Note: the total SV1 and SV2 immunization doses for Trial 1 were 0.140 mL/bird, and for Trial 2 &amp; 3, 0.165 mL/bird. The total SE-LPS dose was 5 µg/bird for all Trials.</p>
Full article ">
11 pages, 996 KiB  
Review
Morphological Variability of the Sural Nerve and Its Clinical Significance
by Weronika Marcinkowska, Nicol Zielinska, Bartłomiej Szewczyk, Piotr Łabętowicz, Mariola Głowacka and Łukasz Olewnik
J. Clin. Med. 2024, 13(20), 6055; https://doi.org/10.3390/jcm13206055 - 11 Oct 2024
Viewed by 791
Abstract
The sural nerve provides sensory innervation to the skin on the distal posterolateral third of the lower extremity. The morphological variants are characterized by high variability. However, it most commonly arises from a union of the medial sural cutaneous nerve and the peroneal [...] Read more.
The sural nerve provides sensory innervation to the skin on the distal posterolateral third of the lower extremity. The morphological variants are characterized by high variability. However, it most commonly arises from a union of the medial sural cutaneous nerve and the peroneal communicating branch of the common fibular nerve. This article overviews the anatomical and clinical significance of the sural nerve. Despite the remarkable development of genetic diagnostics, sural nerve biopsy is still a very important tool to diagnose peripheral neuropathies such as diabetic, vascular and inflammatory neuropathies. Furthermore, the sural nerve is also commonly transplanted due to its characteristics. Such a procedure is applicable in cases of segmental nerve loss, but it is also used to restore potency in patients after radical prostatectomy. The knowledge of anatomical variants of the sural nerve is also crucial as it allows to minimize its damage during surgical procedures. Furthermore, during an ankle surgery, a nerve block can be used to complement anesthesia. The major aim of this work is to review contributions of the sural nerve to physiological and pathophysiological processes. Full article
(This article belongs to the Section Orthopedics)
Show Figures

Figure 1

Figure 1
<p>(<b>a</b>) The small saphenous vein and the sural nerve; Henry Vandyke Carter, public domain, via Wikimedia Commons [<a href="#B17-jcm-13-06055" class="html-bibr">17</a>]; and (<b>b</b>) the most common formation of the sural nerve; Robert Steele MS DO, via Wikimedia Commons [<a href="#B18-jcm-13-06055" class="html-bibr">18</a>].</p>
Full article ">
10 pages, 1413 KiB  
Article
Characterizing CD133 and NANOG Expression in Melanoma: Associations with Histological and Epidemiological Parameters
by Adrian-Horațiu Sabău, Raluca Niculescu, Iuliu-Gabriel Cocuz, Andreea-Cătălina Tinca, Andreea Raluca Szöke, Bianca Andreea Lazar, Diana Maria Chiorean, Corina Eugenia Budin, Alexandru Nicușor Tomuț and Ovidiu Simion Cotoi
Medicina 2024, 60(10), 1658; https://doi.org/10.3390/medicina60101658 - 10 Oct 2024
Viewed by 874
Abstract
Background/Objectives: Melanoma is an aggressive skin malignancy, and the majority of deaths associated with melanoma result from malignant skin lesions. Our study aims to evaluate the expression of the markers CD133 and NANOG, associated with tumor stem cells, and to analyze their [...] Read more.
Background/Objectives: Melanoma is an aggressive skin malignancy, and the majority of deaths associated with melanoma result from malignant skin lesions. Our study aims to evaluate the expression of the markers CD133 and NANOG, associated with tumor stem cells, and to analyze their link with epidemiological and histological parameters, thus contributing to early diagnosis and the development of targeted therapies. Methods: We performed a retrospective study in the Mureș Clinical County Hospital, Romania, which included 66 cases of melanoma: 50 primary cutaneous melanomas, 10 metastases, and 6 local recurrences. CD133 and NANOG marker expression was assessed by immunohistochemistry and quantified using the H score. Statistical analyses were applied to determine the correlations between marker expression and clinicopathological parameters. Results: CD133 expression was identified in six cases (12%) of primary melanoma, with a mean H-Score of 29, and was associated with an increased Breslow index and a higher number of mitoses. NANOG expression was positive in 30 cases (60%) of primary melanoma, with a median H-Score of 15 and with increased expression observed in cases with pagetoid migration and lesions in situ. In metastases, eight cases (80%) were positive for NANOG and four (40%) for CD133. Local recurrences showed positive expression for NANOG in four cases (66%). Conclusions: The expression of CD133 and NANOG markers highlights the role of tumor stem cells in melanoma progression. Early identification of these markers could improve diagnosis and treatment, including the application of targeted therapies. Full article
(This article belongs to the Special Issue Advances in the Diagnosis, Prevention and Treatment of Skin Tumors)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>–<b>C</b>) A case of primary cutaneous nodular melanoma, stage pT4b (Breslow index greater than 4 mm and surface ulceration). (<b>A</b>) The HE stain (×20) reveals tumor proliferation with a nodular architecture, composed of epithelioid cells with eosinophilic cytoplasm and enlarged, pleomorphic nuclei. (<b>B</b>) Immunohistochemical reaction with anti-CD133 antibodies (×20); focally positive (H-Score 5 × 3 + 5 × 2 + 5 × 1 = 30). (<b>C</b>) Immunohistochemical reaction with anti-NANOG antibodies (×20); negative (H-Score 0). (<b>D</b>–<b>F</b>) A case of pleural melanoma metastasis. (<b>D</b>) Cytoblock in HE stain (×20) shows normal or reactive mesothelial cells, inflammatory cells, predominantly lymphocytes and plasma cells, along with isolated tumor cells with eosinophilic cytoplasm, irregular, pleomorphic nuclei, and prominent nucleolus. (<b>E</b>) Immunohistochemical reaction with anti-CD133 antibodies (×20); focally positive at the membrane level (H-Score 0 × 3 + 5 × 2 + 5 × 1 = 15). (<b>F</b>) Immunohistochemical reaction with anti-NANOG antibodies (×20); focally positive at the nuclear level (H-Score 30 × 3 + 10 × 2 + 5 × 1 = 115).</p>
Full article ">
24 pages, 6303 KiB  
Article
Optimization of the Treatment of Squamous Cell Carcinoma Cells by Combining Photodynamic Therapy with Cold Atmospheric Plasma
by Sigrid Karrer, Petra Unger, Nina Spindler, Rolf-Markus Szeimies, Anja Katrin Bosserhoff, Mark Berneburg and Stephanie Arndt
Int. J. Mol. Sci. 2024, 25(19), 10808; https://doi.org/10.3390/ijms251910808 - 8 Oct 2024
Viewed by 1017
Abstract
Actinic keratosis (AK) is characterized by a reddish or occasionally skin-toned rough patch on sun-damaged skin, and it is regarded as a precursor to squamous cell carcinoma (SCC). Photodynamic therapy (PDT), utilizing 5-aminolevulinic acid (ALA) along with red light, is a recognized treatment [...] Read more.
Actinic keratosis (AK) is characterized by a reddish or occasionally skin-toned rough patch on sun-damaged skin, and it is regarded as a precursor to squamous cell carcinoma (SCC). Photodynamic therapy (PDT), utilizing 5-aminolevulinic acid (ALA) along with red light, is a recognized treatment option for AK that is limited by the penetration depth of light and the distribution of the photosensitizer into the skin. Cold atmospheric plasma (CAP) is a partially ionized gas with permeability-enhancing and anti-cancer properties. This study analyzed, in vitro, whether a combined treatment of CAP and ALA-PDT may improve the efficacy of the treatment. In addition, the effect of the application sequence of ALA and CAP was investigated using in vitro assays and the molecular characterization of human oral SCC cell lines (SCC-9, SCC-15, SCC-111), human cutaneous SCC cell lines (SCL-1, SCL-2, A431), and normal human epidermal keratinocytes (HEKn). The anti-tumor effect was determined by migration, invasion, and apoptosis assays and supported the improved efficacy of ALA-PDT in combination with CAP. However, the application sequence ALA-CAP–red light seems to be more efficacious than CAP-ALA–red light, which is probably due to increased intracellular ROS levels when ALA is applied first, followed by CAP and red light treatment. Furthermore, the expression of apoptosis- and senescence-related molecules (caspase-3, -6, -9, p16INK4a, p21CIP1) was increased, and different genes of the junctional network (ZO-1, CX31, CLDN1, CTNNB1) were induced after the combined treatment of CAP plus ALA-PDT. HEKn, however, were much less affected than SCC cells. Overall, the results show that CAP may improve the anti-tumor effects of conventional ALA-PDT on SCC cells. Whether this combined application is successful in treating AK in vivo has to be carefully examined in follow-up studies. Full article
(This article belongs to the Special Issue Molecular Aspects of Photodynamic Therapy)
Show Figures

Figure 1

Figure 1
<p>Onset and progression of squamous cell carcinoma (SCC). Early actinic keratosis (AK) is mainly caused by chronic ultraviolet (UV) light exposure. UV radiation induces a mutation of the tumor suppressor gene <span class="html-italic">tp53</span>, which is considered to be one cause of the development of AK. As a result, atypical keratinocytes proliferate in an uncontrolled manner, and apoptosis is reduced. AK is regarded as a potential precursor of squamous cell carcinoma (SCC). (red arrow: reduction; green arrow: induction). Created with BioRender.com.</p>
Full article ">Figure 2
<p>(<b>a</b>) Metabolic activity curves of cutaneous SCC cell lines (SCL-1, SCL-2, A431), oral SCC cell lines (SCC-9, SCC-15, SCC-111), and HEKn cells were generated 24 h after ALA incubation for 3 h followed by red light treatment (100 J/cm<sup>2</sup>; 160 mW/cm<sup>2</sup>). The ALA concentration at which approximately 70–80% of cells are metabolically active was defined as the MA<sub>75</sub> value and served as the individual ALA concentration for cell culture examinations. (<b>b</b>) The numbers of living cells of (a–c) cutaneous SCC cells (SCL-1, SCL-2, A431), (d–f) oral SCC cells (SCC-9, SCC-15, SCC-111), and (g) HEKn cells were determined 24 h after treatments (CAP, ALA-PDT, CAP-ALA–red light, ALA-CAP–red light) and in untreated cells (ctrl.) using Acridine Orange (AO)/Propidium Iodide (PI) staining and were analyzed using LUNA-FL™ in an automated fluorescence cell counting mode The results are the means of a single experiment performed in duplicate.</p>
Full article ">Figure 2 Cont.
<p>(<b>a</b>) Metabolic activity curves of cutaneous SCC cell lines (SCL-1, SCL-2, A431), oral SCC cell lines (SCC-9, SCC-15, SCC-111), and HEKn cells were generated 24 h after ALA incubation for 3 h followed by red light treatment (100 J/cm<sup>2</sup>; 160 mW/cm<sup>2</sup>). The ALA concentration at which approximately 70–80% of cells are metabolically active was defined as the MA<sub>75</sub> value and served as the individual ALA concentration for cell culture examinations. (<b>b</b>) The numbers of living cells of (a–c) cutaneous SCC cells (SCL-1, SCL-2, A431), (d–f) oral SCC cells (SCC-9, SCC-15, SCC-111), and (g) HEKn cells were determined 24 h after treatments (CAP, ALA-PDT, CAP-ALA–red light, ALA-CAP–red light) and in untreated cells (ctrl.) using Acridine Orange (AO)/Propidium Iodide (PI) staining and were analyzed using LUNA-FL™ in an automated fluorescence cell counting mode The results are the means of a single experiment performed in duplicate.</p>
Full article ">Figure 3
<p>Determination of DCF in arbitrary units [a.u.] as indicator of intracellular ROS levels in untreated SCC and HEKn cells using fluorogenic DCFH-DA assay. Intracellular ROS levels in cutaneous SCC cells (SCL-1, SCL-2, A431) and oral SCC cells (SCC-9, SCC-15, SCC-11) in comparison to non-tumorigenic HEKn cells. The results are the means of three independent measurements. Statistical analysis: Ordinary one-way ANOVA with Tukey’s multiple comparison test was carried out to compare the means of HEKn and SCC cell lines. **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 4
<p>The determination of DCF as an indicator of intracellular ROS levels in treated SCC and HEKn cells using a fluorogenic DCFH-DA assay. DCF (x-fold to ctrl.) was determined in cutaneous SCC cell lines (<b>a</b>) SCL-1, (<b>b</b>) SCL-2, and (<b>c</b>) A431; in oral SCC cell lines (<b>d</b>) SCC-9, (<b>e</b>) SCC-15 and (<b>f</b>) SCC-111; and in (<b>g</b>) normal HEKn after CAP treatment, ALA-PDT and after combined treatments (CAP-ALA–red light, ALA-CAP–red light). Statistical analysis: Ordinary one-way ANOVA with Bonferroni’s multiple comparison test was carried out to compare the mean of ALA-PDT with the results of the combined CAP-ALA–red light and ALA-CAP–red light treatment. * <span class="html-italic">p</span> ≤ 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>Migration and invasion of SCC cells after conventional ALA-PDT and after combined treatments (CAP-ALA–red light, ALA-CAP–red light). (<b>a</b>) Boyden Chamber migration assay. Exemplary overview of stained 8 µm pore filter membranes of migrated cutaneous SCC cells (SCL-1, SCL-2, A431) and oral SCC cells (SCC-9, SCC-15, SCC-111). (<b>b</b>) Boyden Chamber invasion assay. Exemplary overview of stained 8 µm pore filter membranes of matrigel-invaded cutaneous SCC cells (SCL-1, SCL-2, A431) and oral SCC cells (SCC-9, SCC-15, SCC-111). Quantification and statistical examination of (<b>c</b>) migrated and (<b>d</b>) invaded SCC cells. Each experiment was carried out in triplicates. Of each experiment, at least five representative images per group were taken at 20-fold magnification, and the cells per field of view were counted and summarized. Statistical analysis: Ordinary one-way ANOVA with Bonferroni’s multiple comparison test was used with * <span class="html-italic">p</span> ≤ 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 to indicate the mean differences within the conventional ALA-PDT and the combined treatments (CAP-ALA–red light, ALA-CAP–red light).</p>
Full article ">Figure 5 Cont.
<p>Migration and invasion of SCC cells after conventional ALA-PDT and after combined treatments (CAP-ALA–red light, ALA-CAP–red light). (<b>a</b>) Boyden Chamber migration assay. Exemplary overview of stained 8 µm pore filter membranes of migrated cutaneous SCC cells (SCL-1, SCL-2, A431) and oral SCC cells (SCC-9, SCC-15, SCC-111). (<b>b</b>) Boyden Chamber invasion assay. Exemplary overview of stained 8 µm pore filter membranes of matrigel-invaded cutaneous SCC cells (SCL-1, SCL-2, A431) and oral SCC cells (SCC-9, SCC-15, SCC-111). Quantification and statistical examination of (<b>c</b>) migrated and (<b>d</b>) invaded SCC cells. Each experiment was carried out in triplicates. Of each experiment, at least five representative images per group were taken at 20-fold magnification, and the cells per field of view were counted and summarized. Statistical analysis: Ordinary one-way ANOVA with Bonferroni’s multiple comparison test was used with * <span class="html-italic">p</span> ≤ 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 to indicate the mean differences within the conventional ALA-PDT and the combined treatments (CAP-ALA–red light, ALA-CAP–red light).</p>
Full article ">Figure 5 Cont.
<p>Migration and invasion of SCC cells after conventional ALA-PDT and after combined treatments (CAP-ALA–red light, ALA-CAP–red light). (<b>a</b>) Boyden Chamber migration assay. Exemplary overview of stained 8 µm pore filter membranes of migrated cutaneous SCC cells (SCL-1, SCL-2, A431) and oral SCC cells (SCC-9, SCC-15, SCC-111). (<b>b</b>) Boyden Chamber invasion assay. Exemplary overview of stained 8 µm pore filter membranes of matrigel-invaded cutaneous SCC cells (SCL-1, SCL-2, A431) and oral SCC cells (SCC-9, SCC-15, SCC-111). Quantification and statistical examination of (<b>c</b>) migrated and (<b>d</b>) invaded SCC cells. Each experiment was carried out in triplicates. Of each experiment, at least five representative images per group were taken at 20-fold magnification, and the cells per field of view were counted and summarized. Statistical analysis: Ordinary one-way ANOVA with Bonferroni’s multiple comparison test was used with * <span class="html-italic">p</span> ≤ 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 to indicate the mean differences within the conventional ALA-PDT and the combined treatments (CAP-ALA–red light, ALA-CAP–red light).</p>
Full article ">Figure 6
<p>Annexin V/PI double-staining assay was performed 48 h after different treatments (CAP, ALA-PDT, CAP-ALA–red light, and ALA-CAP–red light) and compared to untreated control cells (ctrl.). Apoptosis (early and late apoptosis), necrosis, and the number of live cells were analyzed in oral SCC cell lines (<b>a</b>) SCC-9, (<b>b</b>) SCC-15 and (<b>c</b>) SCC-111; cutaneous SCC cell lines (<b>d</b>) SCL-1, (<b>e</b>) SCL-2 and (<b>f</b>) A431; and (<b>g</b>) HEKn cells. The graphs present the percentage (mean ± SD) of the cells in the region among the total cells from three independent experiments in duplicate. Statistical analysis: Ordinary one-way ANOVA with Bonferroni’s multiple comparison test was used with * <span class="html-italic">p</span> ≤ 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 to indicate the mean differences within the conventional ALA-PDT and the combined treatment groups (CAP-ALA–red light and ALA-CAP–red light).</p>
Full article ">Figure 7
<p>Expression of apoptosis- and senescence-related molecules on the mRNA level after different treatments (CAP, ALA-PDT, CAP-ALA–red light and ALA-CAP–red light) and in untreated cells (ctrl.). (<b>a</b>–<b>c</b>) mRNA expression (caspase-3, -7, -9, p16<sup>INK4a</sup>, p21<sup>CIP1</sup>) in cutaneous SCC cell lines (SCL-1, SCL-2, A431), (<b>d</b>–<b>f</b>) in oral SCC cell lines, and (<b>g</b>) HEKn was measured 24 h after treatment. Statistical analysis: Ordinary one-way ANOVA with Bonferroni’s multiple comparison test was carried out to compare the mean of conventional ALA-PDT with combined treatments (CAP-ALA–red light, ALA–CAP-red light). * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 8
<p>Expression of genes of the junctional network after different treatments (CAP, ALA-PDT, CAP-ALA–red light, or ALA-CAP–red light) and in untreated cells (ctrl.). (<b>a</b>–<b>c</b>) mRNA expression (CLDN1, ZO-1, CTNNB1, CX31) in cutaneous SCC cell lines (SCL-1, SCL-2, A431), (<b>d</b>–<b>f</b>) in oral SCC cell lines, and (<b>g</b>) in HEKn was measured 24 h after treatment. Statistical analysis: Ordinary one-way ANOVA with Bonferroni’s multiple comparison test was carried out to compare the mean of conventional ALA-PDT with combined treatments (CAP-ALA–red light, ALA-CAP–red light). * <span class="html-italic">p</span> ≤ 0.05, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 8 Cont.
<p>Expression of genes of the junctional network after different treatments (CAP, ALA-PDT, CAP-ALA–red light, or ALA-CAP–red light) and in untreated cells (ctrl.). (<b>a</b>–<b>c</b>) mRNA expression (CLDN1, ZO-1, CTNNB1, CX31) in cutaneous SCC cell lines (SCL-1, SCL-2, A431), (<b>d</b>–<b>f</b>) in oral SCC cell lines, and (<b>g</b>) in HEKn was measured 24 h after treatment. Statistical analysis: Ordinary one-way ANOVA with Bonferroni’s multiple comparison test was carried out to compare the mean of conventional ALA-PDT with combined treatments (CAP-ALA–red light, ALA-CAP–red light). * <span class="html-italic">p</span> ≤ 0.05, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">
18 pages, 1036 KiB  
Review
Assessing the Impact of ChatGPT in Dermatology: A Comprehensive Rapid Review
by Polat Goktas and Andrzej Grzybowski
J. Clin. Med. 2024, 13(19), 5909; https://doi.org/10.3390/jcm13195909 - 3 Oct 2024
Viewed by 2349
Abstract
Background/Objectives: The use of artificial intelligence (AI) in dermatology is expanding rapidly, with ChatGPT, a large language model (LLM) from OpenAI, showing promise in patient education, clinical decision-making, and teledermatology. Despite its potential, the ethical, clinical, and practical implications of its application [...] Read more.
Background/Objectives: The use of artificial intelligence (AI) in dermatology is expanding rapidly, with ChatGPT, a large language model (LLM) from OpenAI, showing promise in patient education, clinical decision-making, and teledermatology. Despite its potential, the ethical, clinical, and practical implications of its application remain insufficiently explored. This study aims to evaluate the effectiveness, challenges, and future prospects of ChatGPT in dermatology, focusing on clinical applications, patient interactions, and medical writing. ChatGPT was selected due to its broad adoption, extensive validation, and strong performance in dermatology-related tasks. Methods: A thorough literature review was conducted, focusing on publications related to ChatGPT and dermatology. The search included articles in English from November 2022 to August 2024, as this period captures the most recent developments following the launch of ChatGPT in November 2022, ensuring that the review includes the latest advancements and discussions on its role in dermatology. Studies were chosen based on their relevance to clinical applications, patient interactions, and ethical issues. Descriptive metrics, such as average accuracy scores and reliability percentages, were used to summarize study characteristics, and key findings were analyzed. Results: ChatGPT has shown significant potential in passing dermatology specialty exams and providing reliable responses to patient queries, especially for common dermatological conditions. However, it faces limitations in diagnosing complex cases like cutaneous neoplasms, and concerns about the accuracy and completeness of its information persist. Ethical issues, including data privacy, algorithmic bias, and the need for transparent guidelines, were identified as critical challenges. Conclusions: While ChatGPT has the potential to significantly enhance dermatological practice, particularly in patient education and teledermatology, its integration must be cautious, addressing ethical concerns and complementing, rather than replacing, dermatologist expertise. Future research should refine ChatGPT’s diagnostic capabilities, mitigate biases, and develop comprehensive clinical guidelines. Full article
(This article belongs to the Section Dermatology)
Show Figures

Figure 1

Figure 1
<p>Overview of ChatGPT’s core applications in dermatology, highlighting its roles in patient education and interaction, professional education, clinical decision, and medical writing supports.</p>
Full article ">Figure 2
<p>Key future directions for the integration of ChatGPT into dermatology.</p>
Full article ">
Back to TopTop