[go: up one dir, main page]

 
 
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

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
remove_circle_outline

Search Results (3,312)

Search Parameters:
Keywords = skin cancer

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
24 pages, 2012 KiB  
Review
A Review of the Benefits of the Sustainable Utilization of Shrimp Waste to Produce Novel Foods and the Impact on Human Health
by Ioannis Fotodimas, Zacharias Ioannou and Grigorios Kanlis
Sustainability 2024, 16(16), 6909; https://doi.org/10.3390/su16166909 (registering DOI) - 12 Aug 2024
Abstract
In recent years, there has been an increase in the industrial processing of shrimp, aiming to cover the increasing demand for shrimp products for human consumption, and, consequently, an increase in shrimp by-products as shrimp waste. This waste includes the cephalothoraxes, heads, shells, [...] Read more.
In recent years, there has been an increase in the industrial processing of shrimp, aiming to cover the increasing demand for shrimp products for human consumption, and, consequently, an increase in shrimp by-products as shrimp waste. This waste includes the cephalothoraxes, heads, shells, tails, pleopods, and exoskeleton appendages of processed shrimps. The appropriate method for the enzymatic hydrolysis of shrimp waste can recover its bioactive substances, including carotenoids. Thus, these xanthophylls and carotenes are of high financial interest and have high antioxidant, anti-inflammatory, and anti-cancer activities. Therefore, these substances can be incorporated into fish feed as ingredients that improve fish health and simultaneously lead to the production of aquaculture fishes similar in coloration to the wild ones. Thus, the consumption of such novel food acts as a preventive factor for human health. In this regard, β-carotene has antioxidant and fat-soluble activities owing to vitamin A sufficiency and has an anti-cancer effect, too. Canthaxanthin can be used as a product for personal care and as a natural tanning agent for human skin. Zeaxanthin and lutein have positive effects on various eye and heart diseases, neuronal damage, human skin diseases, and certain types of cancer. Astaxanthin also has anti-diabetic and anti-obesity properties. Therefore, the purpose of this review is to highlight the sustainable utilization of shrimp waste via enzymatic hydrolysis, the benefits of a fish diet enriched with astaxanthin, the consumption of fish enriched with carotenoids, and the effects of carotenoids on human health. The problem of shrimp waste disposal affects the environment, does not contribute to sustainable development, and is directly related to the phenomenon of environmental change. Full article
(This article belongs to the Special Issue Sustainable Food Management in the Era of Climate Change)
Show Figures

Figure 1

Figure 1
<p>Management and processing of raw shrimp waste before performing enzymatic hydrolysis.</p>
Full article ">Figure 2
<p>Description of the enzymatic hydrolysis method with the use of proteolytic enzymes for the recovery of bioactive components from raw shrimp waste.</p>
Full article ">Figure 3
<p>Description of the production of fish feeds with astaxanthin and its benefit to farmed fish.</p>
Full article ">Figure 4
<p>Carotenoids and their impact on human health [<a href="#B19-sustainability-16-06909" class="html-bibr">19</a>,<a href="#B108-sustainability-16-06909" class="html-bibr">108</a>,<a href="#B109-sustainability-16-06909" class="html-bibr">109</a>,<a href="#B110-sustainability-16-06909" class="html-bibr">110</a>,<a href="#B111-sustainability-16-06909" class="html-bibr">111</a>,<a href="#B113-sustainability-16-06909" class="html-bibr">113</a>,<a href="#B114-sustainability-16-06909" class="html-bibr">114</a>,<a href="#B115-sustainability-16-06909" class="html-bibr">115</a>,<a href="#B116-sustainability-16-06909" class="html-bibr">116</a>,<a href="#B117-sustainability-16-06909" class="html-bibr">117</a>,<a href="#B118-sustainability-16-06909" class="html-bibr">118</a>,<a href="#B119-sustainability-16-06909" class="html-bibr">119</a>,<a href="#B120-sustainability-16-06909" class="html-bibr">120</a>,<a href="#B121-sustainability-16-06909" class="html-bibr">121</a>,<a href="#B122-sustainability-16-06909" class="html-bibr">122</a>,<a href="#B123-sustainability-16-06909" class="html-bibr">123</a>,<a href="#B124-sustainability-16-06909" class="html-bibr">124</a>,<a href="#B125-sustainability-16-06909" class="html-bibr">125</a>,<a href="#B128-sustainability-16-06909" class="html-bibr">128</a>].</p>
Full article ">
11 pages, 5130 KiB  
Article
Understanding the Importance of Daily Imaging in the Treatment of Non-Melanoma Skin Cancer with Image-Guided Superficial Radiation Therapy
by Jeffrey B. Stricker, Janine Hopkins, Aaron S. Farberg and Peyton M. Harris
Dermato 2024, 4(3), 86-96; https://doi.org/10.3390/dermato4030010 (registering DOI) - 12 Aug 2024
Abstract
Image-guided superficial radiation therapy (IGSRT) combines superficial radiation therapy (SRT) with full dermal visualization (FDV) via high-resolution dermal ultrasound (HRDUS) for the treatment of non-melanoma skin cancer (NMSC). The gold standard for IGSRT delivery includes a comprehensive cancer care model with support for [...] Read more.
Image-guided superficial radiation therapy (IGSRT) combines superficial radiation therapy (SRT) with full dermal visualization (FDV) via high-resolution dermal ultrasound (HRDUS) for the treatment of non-melanoma skin cancer (NMSC). The gold standard for IGSRT delivery includes a comprehensive cancer care model with support for dermatologists from a multidisciplinary team. When delivered in this model, IGSRT can achieve cure rates of 99% for treatment of NMSC. This paper focuses on the benefits of HRDUS used in conjunction with SRT for NMSC. Medical records from 7 dermatology clinics of 883 patients with 1507 cases of NMSC treated with IGSRT between 2017 and 2018 were retrospectively reviewed. In total, 92% of the NMSC lesions showed daily depth fluctuations, 60.32% of lesions did not require changes during therapy, and nearly 40% of lesions required at least one compensatory change during therapy. In total, 83% of NMSC lesions were labeled as high risk based on the 2024 NCCN guidelines. Increasing and decreasing tumor depth measurements during IGSRT inform dermatologists when adaptive changes in energy (kV), TDF, and dose will result in more efficacy and less toxicity, respectively. Full article
Show Figures

Figure 1

Figure 1
<p>HRDUS imaging confirms tumor location when clinical presentation of nodular basal cell carcinoma is subtle or challenging.</p>
Full article ">Figure 2
<p>Lesion depth fluctuations during IGSRT mid-treatment, in moderately differentiated SCC.</p>
Full article ">Figure 3
<p>Increasing repopulation during IGSRT, nodular basal cell carcinoma.</p>
Full article ">
16 pages, 1908 KiB  
Article
Synthesis of Second-Generation Analogs of Temporin-SHa Peptide Having Broad-Spectrum Antibacterial and Anticancer Effects
by Arif Iftikhar Khan, Shahzad Nazir, Muhammad Nadeem ul Haque, Rukesh Maharjan, Farooq-Ahmad Khan, Hamza Olleik, Elise Courvoisier-Dezord, Marc Maresca and Farzana Shaheen
Antibiotics 2024, 13(8), 758; https://doi.org/10.3390/antibiotics13080758 (registering DOI) - 11 Aug 2024
Viewed by 317
Abstract
Antimicrobial peptides (AMPs) are a promising class of therapeutic alternatives with broad-spectrum activity against resistant pathogens. Small AMPs like temporin-SHa (1) and its first-generation analog [G10a]-SHa (2) possess notable efficacy against Gram-positive and Gram-negative bacteria. In an effort to [...] Read more.
Antimicrobial peptides (AMPs) are a promising class of therapeutic alternatives with broad-spectrum activity against resistant pathogens. Small AMPs like temporin-SHa (1) and its first-generation analog [G10a]-SHa (2) possess notable efficacy against Gram-positive and Gram-negative bacteria. In an effort to further improve this antimicrobial activity, second-generation analogs of 1 were synthesised by replacing the natural glycine residue at position-10 of the parent molecule with atypical amino acids, such as D-Phenylalanine, D-Tyrosine and (2-Naphthyl)-D-alanine, to study the effect of hydrophobicity on antimicrobial efficacy. The resultant analogs (36) emerged as broad-spectrum antibacterial agents. Notably, the [G10K]-SHa analog (4), having a lysine substitution, demonstrated a 4-fold increase in activity against Gram-negative (Enterobacter cloacae DSM 30054) and Gram-positive (Enterococcus faecalis DSM 2570) bacteria relative to the parent peptide (1). Among all analogs, [G10f]-SHa peptide (3), featuring a D-Phe substitution, showed the most potent anticancer activity against lung cancer (A549), skin cancer (MNT-1), prostate cancer (PC-3), pancreatic cancer (MiaPaCa-2) and breast cancer (MCF-7) cells, achieving an IC50 value in the range of 3.6–6.8 µM; however, it was also found to be cytotoxic against normal cell lines as compared to [G10K]-SHa (4). Peptide 4 also possessed good anticancer activity but was found to be less cytotoxic against normal cell lines as compared to 1 and 3. These findings underscore the potential of second-generation temporin-SHa analogs, especially analog 4, as promising leads to develop new broad-spectrum antibacterial and anticancer agents. Full article
(This article belongs to the Section Antimicrobial Peptides)
Show Figures

Figure 1

Figure 1
<p>UPLC profiles of the synthesised peptides; (<b>A</b>) temporin-SHa; (<b>B</b>) [G10a]-SHa; (<b>C</b>) [G10f]-SHa; (<b>D</b>) [G10y]-SHa; (<b>E</b>) [G10n]-SHa and (<b>F</b>) [G10K]-SHa.</p>
Full article ">Figure 2
<p>Circular dichroism of temporin SHa, [G10a]-Sha, and newly synthesised second-generation analogs of [G10a]-SHa in 20 mM SDS.</p>
Full article ">Figure 3
<p>Antiproliferative effect of SHa derivatives on human cancer cells. The antiproliferative effect of SHa derivatives was measured on dividing cancer cells, as explained in <a href="#sec4-antibiotics-13-00758" class="html-sec">Section 4</a> (Temporin-SHa (<b>1</b>): open black circles, [G10a]-SHa (<b>2</b>): closed red circles, [G10f]-SHa (<b>2</b>): closed green squares, [G10K]-SHa (<b>3</b>): closed black diamonds, [G10n]-SHa (<b>4</b>): inverted open purple triangles, [G10y]-SHa (<b>5</b>): closed blue triangles). Results are expressed as a percentage of cell proliferation, the untreated cells giving 100% proliferation (means ± SD, <span class="html-italic">n</span> = 3).</p>
Full article ">Figure 4
<p>Antiproliferative effect of SHa derivatives on human normal/non cancerous cells. The antiproliferative effect of SHa derivatives was measured on dividing normal cells, as explained in <a href="#sec4-antibiotics-13-00758" class="html-sec">Section 4</a> (temporin-SHa (<b>1</b>): open black circles, [G10a]-SHa (<b>2</b>): closed red circles, [G10f]-SHa (<b>2</b>): closed green squares, [G10K]-SHa (<b>3</b>): closed black diamonds, [G10n]-SHa (<b>4</b>): inverted open purple triangles, [G10y]-SHa (<b>5</b>): closed blue triangles). Results are expressed as a percentage of cell proliferation, the untreated cells giving 100% proliferation (means ± SD, <span class="html-italic">n</span> = 3).</p>
Full article ">Figure 5
<p>Cytotoxic effect of SHa derivatives on human normal and cancer lung cells. The cytotoxic effect of temporin-SHa (<b>1</b>) derivatives was measured on confluent/non-dividing cells, as explained in <a href="#sec4-antibiotics-13-00758" class="html-sec">Section 4</a>, using human lung cancer (A549 cells) and normal cells (BEAS-2B cells); ([G10a]-SHa (<b>2</b>): closed red circles, [G10f]-SHa (<b>2</b>): closed green squares, [G10K]-SHa (<b>3</b>): closed black diamonds, [G10n]-SHa (<b>4</b>): inverted open purple triangles, [G10y]-SHa (<b>5</b>): closed blue triangles). Results are expressed as a percentage of cell proliferation, the untreated cells giving 100% proliferation (means ± SD, <span class="html-italic">n</span> = 3).</p>
Full article ">Scheme 1
<p>Synthesis and structure of temporin-SHa (<b>1</b>), its first-generation [G10a]-SHa peptide (<b>2</b>) and newly synthesised second-generation analogs (<b>3</b>–<b>6</b>).</p>
Full article ">
20 pages, 5455 KiB  
Article
Selectivity Screening and Structure–Cytotoxic Activity Observations of Selected Oleanolic Acid (OA)-Type Saponins from the Amaranthaceae Family on a Wiade Panel of Human Cancer Cell Lines
by Karolina Grabowska, Agnieszka Galanty, Łukasz Pecio, Anna Stojakowska, Janusz Malarz, Paweł Żmudzki, Paweł Zagrodzki and Irma Podolak
Molecules 2024, 29(16), 3794; https://doi.org/10.3390/molecules29163794 (registering DOI) - 10 Aug 2024
Viewed by 290
Abstract
Plants from the Amaranthaceae family are a source of oleanolic acid (OA)-type saponins with cytotoxic activity. Two known OA-type saponins, calenduloside E and chikusetsusaponin IVa, were isolated from the roots of Chenopodium strictum Roth. Their structures were confirmed using MS and NMR techniques. [...] Read more.
Plants from the Amaranthaceae family are a source of oleanolic acid (OA)-type saponins with cytotoxic activity. Two known OA-type saponins, calenduloside E and chikusetsusaponin IVa, were isolated from the roots of Chenopodium strictum Roth. Their structures were confirmed using MS and NMR techniques. This constitutes the inaugural report of the saponins in Ch. strictum. Both the isolated saponins and structurally similar compounds, momordin Ic and OA, were compared for their cytotoxicity against various cancer and normal cell lines (including skin, breast, thyroid, gastrointestinal, and prostate panels). Their effects were dose- and time-dependent, varying with the specific cell line and compound structure. A chemometric approach demonstrated the effects of the compounds on the cell lines. The study discusses the structure–activity observations. The key structural elements for potent cytotoxic activity included the free carboxyl group 28COOH in the sapogenin structure (OA) and the presence of a sugar moiety. The monodesmosides with glucuronic acid (GlcA) at the C3 position of OA were generally more cytotoxic than bidesmosides or OA alone. The addition of xylose in the sugar chain modified the activity towards the cancer cells depending on the specific cell line. OA-type saponins with GlcA (particularly calenduloside E and momordin Ic) represent a promising avenue for further investigation as potential anticancer agents. Full article
Show Figures

Figure 1

Figure 1
<p>Structures of compounds: calenduloside E (CE), chikusetsusaponin IVa (ChIVa), momordin Ic (MIc), and oleanolic acid.</p>
Full article ">Figure 2
<p>The cytotoxic effects of analyzed compounds: caleduloside E (CE), chikusetsusaponin IVa (ChIVa), momordin Ic (Mic), and oleanolic acid (OA) on cancerous (FTC133, 8505C, and TPC-1) and normal (Nthy-ori 3) thyroid cells. Values are presented as the mean ± SD (standard deviation). Results from the MTT viability assay after 24 h incubation with tested substances. Results are presented as the mean ± SD. The values significantly different from the control (untreated group) are indicated by * for <span class="html-italic">p</span> ˂ 0.05.</p>
Full article ">Figure 3
<p>The cytotoxic effects of analyzed compounds: caleduloside E (CE), chikusetsusaponin IVa (ChIVa), momordin Ic (Mic), and oleanolic acid on cancerous (DLD1, HCT-116, Caco2, HepG2, and Ht29) and normal (CCD841CoN) colon cells. Results from the MTT viability assay after 24 h incubation. Results are presented as the mean ± SD. The values significantly different from the control (untreated group) are indicated by * for <span class="html-italic">p</span> ˂ 0.05.</p>
Full article ">Figure 4
<p>The score scatterplot of hPCA model (cell lines were depicted).</p>
Full article ">Figure 5
<p>Dendrogram of similarity between different cell lines. Method of grouping: single linkage; function of the distance: Euclidean distance (subsequently identified clusters were marked with subsequent letters; further explanation in the text).</p>
Full article ">Figure 6
<p>The most important structure–activity correlations for cytotoxic activity of analyzed saponins. Abbreviations: ↑—increased activity; ≈—similar activity; ↓—decreased activity; calenduloside E (CE); chikusetsusaponin IVa (ChIVa); momordin Ic (MIc); oleanolic acid (OA).</p>
Full article ">
24 pages, 4377 KiB  
Article
Feasibility Study on the Use of Infrared Cameras for Skin Cancer Detection under a Proposed Data Degradation Model
by Ricardo F. Soto and Sebastián E. Godoy
Sensors 2024, 24(16), 5152; https://doi.org/10.3390/s24165152 - 9 Aug 2024
Viewed by 402
Abstract
Infrared thermography is considered a useful technique for diagnosing several skin pathologies but it has not been widely adopted mainly due to its high cost. Here, we investigate the feasibility of using low-cost infrared cameras with microbolometer technology for detecting skin cancer. For [...] Read more.
Infrared thermography is considered a useful technique for diagnosing several skin pathologies but it has not been widely adopted mainly due to its high cost. Here, we investigate the feasibility of using low-cost infrared cameras with microbolometer technology for detecting skin cancer. For this purpose, we collected infrared data from volunteer subjects using a high-cost/high-quality infrared camera. We propose a degradation model to assess the use of lower-cost imagers in such a task. The degradation model was validated by mimicking video acquisition with the low-cost cameras, using data originally captured with a medium-cost camera. The outcome of the proposed model was then compared with the infrared video obtained with actual cameras, achieving an average Pearson correlation coefficient of more than 0.9271. Therefore, the model successfully transfers the behavior of cameras with poorer characteristics to videos acquired with higher-quality cameras. Using the proposed model, we simulated the acquisition of patient data with three different lower-cost cameras, namely, Xenics Gobi-640, Opgal Therm-App, and Seek Thermal CompactPRO. The degraded data were used to evaluate the performance of a skin cancer detection algorithm. The Xenics and Opgal cameras achieved accuracies of 84.33% and 84.20%, respectively, and sensitivities of 83.03% and 83.23%, respectively. These values closely matched those from the non-degraded data, indicating that employing these lower-cost cameras is appropriate for skin cancer detection. The Seek camera achieved an accuracy of 82.13% and a sensitivity of 79.77%. Based on these results, we conclude that this camera is appropriate for less critical applications. Full article
(This article belongs to the Special Issue Sensors and Devices for Biomedical Image Processing)
Show Figures

Figure 1

Figure 1
<p>Example of a plastic marker used to select the region of interest. The region of interest is indicated in red, and the suspicious lesion is highlighted in blue.</p>
Full article ">Figure 2
<p>Detection scheme used to evaluate the feasibility of using different IR cameras in detecting skin cancer using active thermography.</p>
Full article ">Figure 3
<p>Schematic of the proposed degradation model, which addresses 3 areas: temporal, spatial, and thermal resolution. Giving rise to a process composed of 5 stages.</p>
Full article ">Figure 4
<p>Samples of the measurement behaviors from different IR cameras on a blackbody stabilized at 40 °C in a room with controlled ambient temperature at 20 °C using an AC unit. The order of measurements taken with the different cameras is as follows: (<b>a</b>) Xenics Gobi-640, (<b>b</b>) Opgal Therm-App and (<b>c</b>) Seek Thermal CompactPRO.</p>
Full article ">Figure 5
<p>Sample of the results of the jump correction produced by the NUC in the Xenics Gobi-640 camera. (<b>a</b>) Uncorrected measurements; (<b>b</b>) corrected measurements.</p>
Full article ">Figure 6
<p>Sample of the results of the jump correction produced by the NUC in the Seek Thermal CompactPRO camera. (<b>a</b>) Uncorrected measurements; (<b>b</b>) corrected measurements.</p>
Full article ">Figure 7
<p>Sample of the degradation performed on a high-quality video to mimic Opgal Therm-App camera features. (<b>a</b>) Image captured with the Xenics Gobi-640 camera, (<b>b</b>) image captured at the same instant of time and same area with the Opgal Therm-App camera, (<b>c</b>) Xenics image adapted to Opgal camera features, (<b>d</b>,<b>e</b>) correspond to representative TRCs of the video acquired with the Xenics and Opgal cameras, respectively. (<b>f</b>) TRC from the adaptation.</p>
Full article ">Figure 8
<p>Sample of the degradation performed on a high-quality video to mimic Seek Thermal CompactPRO camera features. (<b>a</b>) Image captured with the Xenics Gobi-640 camera, (<b>b</b>) image captured at the same instant of time and same area with the Seek Thermal CompactPRO camera, (<b>c</b>) Xenics image adapted to Seek camera features, (<b>d</b>,<b>e</b>) correspond to representative TRCs of the video acquired with the Xenics and Seek cameras, respectively. (<b>f</b>) TRC from the adaptation.</p>
Full article ">Figure A1
<p>Sample results of the degradation of an IR video to the characteristics of different simulated cameras in the case of a benign lesion. The top row shows an image from the video and the bottom row shows characteristic TRCs of the mole and non-mole areas. (<b>a</b>,<b>e</b>) Correspond to high-quality data captured with the QmagiQ camera; (<b>b</b>,<b>f</b>) data adapted to Xenics Gobi-640; (<b>c</b>,<b>g</b>) data adapted to Opgal Therm-App; (<b>d</b>,<b>h</b>) data adapted to Seek Thermal CompactPRO.</p>
Full article ">Figure A2
<p>Sample results of the degradation of an IR video to the characteristics of different simulated cameras in a case of a malignant lesion, diagnosed as MM. The top row shows an image from the video and the bottom row shows characteristic TRCs of the mole and non-mole areas. (<b>a</b>,<b>e</b>) Correspond to high-quality data captured with the QmagiQ camera; (<b>b</b>,<b>f</b>) data adapted to Xenics Gobi-640; (<b>c</b>,<b>g</b>) data adapted to Opgal Therm-App; (<b>d</b>,<b>h</b>) data adapted to Seek Thermal CompactPRO.</p>
Full article ">Figure A3
<p>Sample results of the degradation of an IR video to the characteristics of different simulated cameras in the case of a malignant lesion, diagnosed as BCC. The top row shows an image from the video and the bottom row shows characteristic TRCs of the mole and non-mole areas. (<b>a</b>,<b>e</b>) Correspond to high-quality data captured with the QmagiQ camera; (<b>b</b>,<b>f</b>) data adapted to Xenics Gobi-640; (<b>c</b>,<b>g</b>) data adapted to Opgal Therm-App; (<b>d</b>,<b>h</b>) data adapted to Seek Thermal CompactPRO.</p>
Full article ">Figure A4
<p>Sample results of the degradation of an IR video to the characteristics of different simulated cameras in the case of a malignant lesion, diagnosed as SCC. The top row shows an image from the video and the bottom row shows characteristic TRCs of the mole and non-mole areas. (<b>a</b>,<b>e</b>) Correspond to high-quality data captured with the QmagiQ camera; (<b>b</b>,<b>f</b>) data adapted to Xenics Gobi-640; (<b>c</b>,<b>g</b>) data adapted to Opgal Therm-App; (<b>d</b>,<b>h</b>) data adapted to Seek Thermal CompactPRO.</p>
Full article ">
15 pages, 3316 KiB  
Article
In Vitro Effect of Epigallocatechin Gallate on Heme Synthesis Pathway and Protoporphyrin IX Production
by Daniela León, María Elena Reyes, Helga Weber, Álvaro Gutiérrez, Claudio Tapia, Ramón Silva, Tamara Viscarra, Kurt Buchegger, Carmen Ili and Priscilla Brebi
Int. J. Mol. Sci. 2024, 25(16), 8683; https://doi.org/10.3390/ijms25168683 - 9 Aug 2024
Viewed by 226
Abstract
Photodynamic therapy (PDT) treats nonmelanoma skin cancer. PDT kills cells through reactive oxygen species (ROS), generated by interaction among cellular O2, photosensitizer and specific light. Protoporphyrin IX (PpIX) is a photosensitizer produced from methyl aminolevulinate (MAL) by heme group synthesis (HGS) pathway. [...] Read more.
Photodynamic therapy (PDT) treats nonmelanoma skin cancer. PDT kills cells through reactive oxygen species (ROS), generated by interaction among cellular O2, photosensitizer and specific light. Protoporphyrin IX (PpIX) is a photosensitizer produced from methyl aminolevulinate (MAL) by heme group synthesis (HGS) pathway. In PDT-resistant cells, PDT efficacy has been improved by addition of epigallocatechin gallate (EGCG). Therefore, the aim of this work is to evaluate the effect of EGCG properties over MAL-TFD and PpIX production on A-431 cell line. EGCG’s role over cell proliferation (flow cytometry and wound healing assay) and clonogenic capability (clonogenic assay) was evaluated in A-431 cell line, while the effect of EGCG over MAL-PDT was determined by cell viability assay (MTT), PpIX and ROS detection (flow cytometry), intracellular iron quantification and gene expression of HGS enzymes (RT-qPCR). Low concentrations of EGCG (<50 µM) did not have an antiproliferative effect over A-431 cells; however, EGCG inhibited clonogenic cell capability. Furthermore, EGCG (<50 µM) improved MAL-PDT cytotoxicity, increasing PpIX and ROS levels, exerting a positive influence on PpIX synthesis, decreasing intracellular iron concentration and modifying HGS enzyme gene expression such as PGB (upregulated) and FECH (downregulated). EGCG inhibits clonogenic capability and modulates PpIX synthesis, enhancing PDT efficacy in resistant cells. Full article
(This article belongs to the Special Issue Molecular Aspects of Photodynamic Therapy)
Show Figures

Figure 1

Figure 1
<p><b>A-431 cell viability evaluated by MTT assay 24 h after EGCG treatments</b>. Each experimental group was compared with its respective control (NT). Values of <span class="html-italic">p</span> &lt; 0.05 were considered statistically significant. Data are expressed as mean ± SD of three biological replicates.</p>
Full article ">Figure 2
<p><b>Proliferation assay of A-431 cells incubated with 10 µM, 20 µM and 40 µM EGCG for 4 h.</b> Each experimental group was compared with its respective control (NT). Values of <span class="html-italic">p</span> &lt; 0.05 were considered statistically significant. ** <span class="html-italic">p</span> &lt; 0.01. Data are expressed as mean ± SD of three biological replicates.</p>
Full article ">Figure 3
<p><b>Wound healing assay of A-431 cells incubated with 10 µM and 40 µM EGCG.</b> (<b>A</b>) Representative images of wound healing assay using an objective with 20× magnification. (<b>B</b>) Percentage of wound closure. Each experimental group was compared with its respective control (NT). Values of <span class="html-italic">p</span> &lt; 0.05 were considered statistically significant. Data are expressed as mean ± SD of three biological replicates.</p>
Full article ">Figure 4
<p><b>Clonogenic assay of A-431 cells incubated with 10 µM, 20 µM and 40 µM EGCG.</b> (<b>A</b>) Colony formation. (<b>B</b>) Percentage of colonies. Each experimental group was compared with its respective control (NT). Values of <span class="html-italic">p</span> &lt; 0.05 were considered statistically significant. ** <span class="html-italic">p</span> &lt; 0.01. Data are expressed as mean ± SD of three biological replicates.</p>
Full article ">Figure 5
<p><b>A-431 cell viability evaluated by MTT assay 24 h after treatments.</b> (<b>A</b>) Intrinsic resistance to MAL-PDT of A-431 cells. (<b>B</b>) Effect of MAL-PDT + EGCG combination. Values of <span class="html-italic">p</span> &lt; 0.05 were considered statistically significant. ** <span class="html-italic">p</span> &lt; 0.01. Data are expressed as mean ± SD of three biological replicates.</p>
Full article ">Figure 6
<p><b>PpIX levels in A-431 cells detected by flow cytometry</b>. (<b>A</b>) Cells positive for PpIX production. (<b>B</b>) Average fluorescence intensity of PpIX. (<b>C</b>) Representative flow cytometry charts: (<b>a</b>) autofluorescence control, (<b>b</b>) MAL–PDT, (<b>c</b>) MAL–PDT–EGCG 10 µM, (<b>d</b>) MAL–PDT–EGCG 20 µM and (<b>e</b>) MAL–PDT–EGCG 40 µM. Quadrants: Q3 (PpIX and ROS negative cells), Q4: PpIX positive cells, Q1: ROS positive cells, Q2 (PpIX and ROS positive cells). Values of <span class="html-italic">p</span> &lt; 0.05 were considered statistically significant. ** <span class="html-italic">p</span> &lt; 0.01. Data express mean ± SD of three biological replicates. a.u. = arbitrary units.</p>
Full article ">Figure 7
<p><b>ROS levels in A-431 cells detected by flow cytometry.</b> (<b>A</b>) Mean fluorescence intensity (MFI) of ROS in light-free controls of A-431 cells. (<b>B</b>) MFI of ROS in A-431 cells treated with MAL 2 mM and EGCG (10, 20 or 40 µM), with red light (630 nm, 4 J/cm<sup>2</sup>). (<b>C</b>) Representative histogram graphs: (<b>a</b>) autofluorescence control, (<b>b</b>) MAL-PDT, (<b>c</b>) MAL-PDT-EGCG 10 µM, (<b>d</b>) MAL-PDT-EGCG 20 µM and (<b>e</b>) MAL-PDT-EGCG 40 µM. Values of <span class="html-italic">p</span> &lt; 0.05 were considered statistically significant. *** <span class="html-italic">p</span> &lt; 0.001. Data are expressed as mean ± SD of three biological replicates. a.u. = arbitrary units.</p>
Full article ">Figure 8
<p><b>Gene expression of relevant heme synthesis pathway enzymes in A-431 cells exposed to EGCG for 4 h.</b> (<b>A</b>) PGB gene. (<b>B</b>) FECH gene. Values of * <span class="html-italic">p</span> &lt; 0.05 were considered statistically significant. ** <span class="html-italic">p</span> &lt; 0.01. Data are expressed as mean ± SD of three biological replicates.</p>
Full article ">Figure 9
<p><b>Concentration of intracellular iron in A-431 cells exposed to EGCG for 4 h.</b> Values of <span class="html-italic">p</span> &lt; 0.05 were considered statistically significant. * <span class="html-italic">p</span> &lt; 0.05. Data are expressed as mean ± SD of three biological replicates.</p>
Full article ">
11 pages, 670 KiB  
Review
The Role of the Vitamin D Receptor in the Epidermal Stem Cell Response to Wounding
by Daniel D. Bikle
Receptors 2024, 3(3), 397-407; https://doi.org/10.3390/receptors3030019 - 9 Aug 2024
Viewed by 366
Abstract
Chronic skin wounds are estimated to affect 6.5 million patients in the US, at a cost of over USD 25 billion. Efforts to prevent and/or treat such wounds will result in reduced morbidity and economic losses. This project is focused on the role [...] Read more.
Chronic skin wounds are estimated to affect 6.5 million patients in the US, at a cost of over USD 25 billion. Efforts to prevent and/or treat such wounds will result in reduced morbidity and economic losses. This project is focused on the role of vitamin D signaling in the epidermis in the control of stem cell (SC) activation and function during the initial response to the wounding of the skin, a response that, if defective, contributes to poor wound healing or cancer. In this review, I first describe the anatomy of the skin, focusing first on the epidermis, describing the different cell layers which in a spatial way also represent the differentiation process of the interfollicular epidermis (IFE) as it undergoes continuous regeneration. I then describe the other components of the skin, particularly the hair follicle (HF), which undergoes a cyclic pattern of regeneration. Adult SCs residing in these regenerative tissues play essential roles in the maintenance of these tissues. However, when the skin is wounded, the progeny of SCs from all regions of the HF and IFE contribute to the healing process by changing their initial cell fate to take on an epithelial genotype/phenotype to heal the wound. Although earlier lineage tracing studies helped to define the contributions SCs from the different niches made to wound healing, scRNAseq studies have demonstrated a considerably more nuanced picture. The role of vitamin D signaling will be introduced by reviewing the unique role played by the epidermal keratinocyte first in producing vitamin D and then in metabolizing it into its active form 1,25(OH)2D. 1,25(OH)2D is the principal ligand for the vitamin D receptor (VDR), a transcription factor that helps to mediate the genomic changes in the stem cells in their response to wounding. In these actions, the VDR is regulated by coregulators, of which the steroid receptor coactivator complexes SRC 2 and 3 and the mediator complex (MED) play essential roles. The VDR generally acts in association with other transcription factors such as p63 and β-catenin that can colocalize with the VDR in the genes it regulates. Although much remains to be understood, the role of the VDR in the stem cell response to wounding is clearly essential and quite different from its classic roles in regulating calcium metabolism, although calcium is essential for the actions of vitamin D signaling in the skin. Full article
Show Figures

Figure 1

Figure 1
<p><b>The four layers of the epidermis.</b> The basal layer of the epidermis (stratum basale) contains the stem cells and transient amplifying cells from which cells in the upper layers are derived. These cells express the keratins K5 and K14 and markers of proliferation such as β-catenin (CTNNB), cyclin D1, and GLI1. As these cells migrate into the spinous layer, they begin the production of different keratins, namely K1 and K10, as well as precursors and enzymes involved in cornified envelope formation, namely involucrin and transglutaminase-K. Further migration into the stratum granulosum is marked by the expression of filaggrin and loricrin, proteins also contributing to the cornified envelope. These cells also express enzymes involved in lipid production, lipids that are packaged into lamellar bodies and subsequently injected into the intercellular spaces between the stratum granulosum and stratum corneum to waterproof the permeability barrier. Moreover, lamellar bodies also contain antimicrobial peptides such as cathelicidin produced in the stratum granulosum, providing protection against invasive organisms. VDR and CYP27B1 expression is highest in the stratum basale. Also shown in this figure is the asymmetric distribution of two major coregulators of VDR action—Med1, which is expressed primarily in the stratum basale and spinosum, facilitates the VDR’s regulation of proliferation and early stages of differentiation; SRC3, found in highest concentration in the stratum granulosum, facilitates the VDR’s regulation of terminal differentiation.</p>
Full article ">Figure 2
<p><b>The hair follicle and its stem cell niches.</b> The hair follicle can be divided into the infundibulum merging into the IFE, the junctional zone in the upper portion of the isthmus which separates the infundibulum and isthmus, the bulge region below the isthmus, and the hair germ adjacent to the dermal papilla in the dermis. The sebaceous gland attaches to the hair shaft in the junctional zone/isthmus. These regions contain different stem cell niches. Several markers of the different stem cell niches are shown. The IFE in this cartoon is marked by Lgr6, which is also expressed in other stem cell niches including the sebaceous gland and isthmus. Lrig1 and Plet1 are markers of stem cells in the isthmus and sebaceous gland. Gli1 is a marker for the stem cells in the upper portion of the bulge, with CD34 and Krt15 also marking the bulge stem cells. Lgr5 marks cells of the lower bulge and hair germ. See the text for a more complete description of these stem cell markers.</p>
Full article ">
10 pages, 1030 KiB  
Case Report
Unveiling a Dermatological Rarity: The Enigma of Vulvar Intraepithelial Neoplasia Grade III (HSIL) and the Role of p53 in Its Development
by Piotr Brzeziński, Igor Feszak, Beatriz Di Martino Ortiz, Sylwia Feszak, Piotr Kawczak and Tomasz Bączek
Biomedicines 2024, 12(8), 1799; https://doi.org/10.3390/biomedicines12081799 - 8 Aug 2024
Viewed by 275
Abstract
Vulvar intraepithelial neoplasia, also known as VIN, is a non-invasive squamous lesion and precursor of squamous cell carcinoma (SCC) of the vulva. There is no screening test for vulvar intraepithelial neoplasia. Diagnosis of VIN is made clinically and confirmed with a biopsy. We [...] Read more.
Vulvar intraepithelial neoplasia, also known as VIN, is a non-invasive squamous lesion and precursor of squamous cell carcinoma (SCC) of the vulva. There is no screening test for vulvar intraepithelial neoplasia. Diagnosis of VIN is made clinically and confirmed with a biopsy. We describe a 66-year-old woman with a condyloma-like tumour located in the skin on the vestibule of the vagina. A biopsy sample was taken from the nodule. The definitive diagnosis is supported by the histological examination (VIN III) and immunohistochemical examination of p16(+), p53(+), and a few cell nuclei. The case provides information on the importance of multidisciplinary cooperation. Lifelong surveillance is essential since the resection of individual lesions does not guarantee the prevention of invasive cancer. Full article
(This article belongs to the Special Issue p53 Signaling in Cancer Progression and Therapy)
Show Figures

Figure 1

Figure 1
<p>The p53 protein signalling and regulation; ATM (ataxia telangiectasia mutated); ATR (ataxia telangiectasia and Rad3-related); CHK1 (Checkpoint Kinase 1); CHK2 (Checkpoint Kinase 2); MDM2 (E3 ubiquitin protein ligase); ARF (Alternate Reading Frame); +P (Phosphorylation), based on Ref. [<a href="#B7-biomedicines-12-01799" class="html-bibr">7</a>].</p>
Full article ">Figure 2
<p>Histological findings in the patient’s biopsy sample: (<b>a</b>) epidermal acanthosis, agranulocytosis, and parakeratosis; (<b>b</b>) epidermal keratinocytes show loss of cell maturation and polarity that affects the full thickness of the epithelium, the basal membrane is not exceeded, and there are images of atypical mitosis typically arrested in metaphase; (<b>c</b>) positive p16 in intraepidermal atypical cells.</p>
Full article ">
11 pages, 1586 KiB  
Article
Nano-Sized Graphene Oxide Attenuates Ovalbumin/Alum-Induced Skin Inflammation by Down-Regulating Th2 Immune Responses in Balb/c Mice
by Hyun Jung Park, Sung Won Lee, Luc Van Kaer, Suklyun Hong and Seokmann Hong
Biomolecules 2024, 14(8), 962; https://doi.org/10.3390/biom14080962 - 7 Aug 2024
Viewed by 272
Abstract
Graphene oxide (GO), a carbon-based material with oxygen-containing functional groups, can be applied in biomedicine for drug delivery, cancer therapy, and tissue regeneration. We have previously shown that nanoscale-sized graphene oxide (NGO), an oxidized graphene derivative, exhibits effective anti-inflammatory activity in a murine [...] Read more.
Graphene oxide (GO), a carbon-based material with oxygen-containing functional groups, can be applied in biomedicine for drug delivery, cancer therapy, and tissue regeneration. We have previously shown that nanoscale-sized graphene oxide (NGO), an oxidized graphene derivative, exhibits effective anti-inflammatory activity in a murine model of sepsis mediated by T helper (Th)1-promoting cytokines such as IFNγ and TNFα. However, whether NGO influences Th2-induced skin inflammation remains unclear. To address this issue, we employed an ovalbumin (OVA) plus aluminum hydroxide (Alum)-induced Th2-mediated skin inflammation model in conjunction with OVA-specific DO11.10 T cell receptor transgenic Balb/c mice. In vivo NGO injection upon OVA/Alum sensitization down-regulated OVA-elicited antigen-specific Th2 cells and GATA3-expressing Th2-type regulatory T cells. Next, we examined the effect of NGO injection on OVA/Alum-induced atopic dermatitis (AD)-like skin inflammation. NGO-injected mice exhibited significantly decreased Th2 disease phenotypes (e.g., a lower clinical score, decreased epidermal thickness and Th2 cell differentiation, and fewer infiltrated mast cells and basophils in skin lesions) compared with vehicle-injected control mice. Overall, our results suggest that NGOs are promising therapeutic materials for treating allergic diseases such as AD. Full article
(This article belongs to the Special Issue Nanomaterials and Their Applications in Biomedicine)
Show Figures

Figure 1

Figure 1
<p>NGO injection suppresses Th2 cell differentiation after OVA/Alum immunization. (<b>A</b>) 4Get/DO11.10 TCR Tg Balb/c mice were immunized i.p. with Alum (1 mg) plus OVA<sub>323–339</sub> peptide (100 µg) after i.v. injection of either NGO (50 µg) or PBS once a week for 3 weeks. One week after the last injection (at 21 days after immunization), splenocytes were prepared from both PBS- and NGO-treated mice. (<b>B</b>,<b>C</b>) The frequency of IL4(GFP)- (<b>B</b>) and IFNγ-expressing (<b>C</b>) populations in splenic KJ1-26<sup>+</sup>CD4<sup>+</sup> T cells from each group was determined by flow cytometry. (<b>B</b>) (left) Representative FACS plots; (right) summary figure. (<b>D</b>) The expression of Ki-67 on splenic KJ1-26<sup>+</sup>CD4<sup>+</sup> T cells was evaluated by flow cytometry. (left) Representative FACS plots; (right) summary figure. (<b>E</b>) The expression of CD44 on splenic KJ1-26<sup>+</sup>CD4<sup>+</sup> T cells was evaluated by flow cytometry. The mean values ± SD (<span class="html-italic">n</span> = 4 per group in the experiment; one-way ANOVA; * <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). One representative experiment of two experiments is shown. ns, not significant.</p>
Full article ">Figure 2
<p>NGO injection suppresses Th2-type Treg cell differentiation after OVA/Alum immunization. (<b>A</b>,<b>B</b>) DO11.10 TCR Tg Balb/c mice were immunized i.p. with Alum (1 mg) plus OVA<sub>323–339</sub> peptide (100 µg) after i.v. injection of either NGO (50 µg) or PBS once a week for 3 weeks. One week after the last injection (at 21 days after immunization), splenocytes were prepared from both PBS- or NGO-treated mice. (<b>A</b>) The intracellular expression of Foxp3 in splenic KJ1-26<sup>+</sup>CD4<sup>+</sup> T cells from each group was analyzed by flow cytometry. (left) Representative FACS plots; (right) summary figure. (<b>B</b>) (left) the frequency of GATA3-expressing cells among splenic Treg cells (KJ1-26<sup>+</sup>CD4<sup>+</sup>Foxp3<sup>+</sup>) was plotted; (right) summary figure. The mean values ± SD (<span class="html-italic">n</span> = 4 per group in the experiment; one-way ANOVA; *** <span class="html-italic">p</span> &lt; 0.001). One representative experiment of two experiments is shown.</p>
Full article ">Figure 3
<p>NGO injection suppresses OVA-induced skin inflammation in DO11.10 TCR Tg Balb/c mice. (<b>A</b>) DO11.10 TCR Tg Balb/c mice were immunized i.p. with Alum (1 mg) plus OVA<sub>323–339</sub> peptide (100 µg) after i.v. injection of either NGO (50 µg) or PBS once a week for 3 weeks. One week after the last injection (at 21 days after immunization), mice were exposed to an OVA patch for 1 week. (<b>B</b>) The clinical symptoms were measured after patch removal (at 28 days after immunization). (<b>C</b>) Skins were prepared from these mice after patch removal. Skin lesions were sectioned and stained with H&amp;E. The epidermal thickness was measured in 10 random high-power fields (400×) per sampled lesion. (<b>D</b>) The numbers of skin basophils and mast cells from each group were determined by flow cytometry. (<b>E</b>) The intracellular expression of IFNγ and IL4 in splenic KJ1-26<sup>+</sup>CD4<sup>+</sup> T cells from each group was analyzed by flow cytometry. The mean values ± SD (<span class="html-italic">n</span> = 4 per group in the experiment; one-way ANOVA; * <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). One representative experiment of two experiments is shown. ns, not significant.</p>
Full article ">
8 pages, 687 KiB  
Case Report
Gefitinib-Induced Severe Dermatological Adverse Reactions: A Case Report and Pharmacogenetic Profile
by Mariana Vieira Morau, Cecilia Souto Seguin, Mauricio Wesley Perroud Junior, Carolina Dagli-Hernandez, Eder de Carvalho Pincinato and Patricia Moriel
Pharmaceuticals 2024, 17(8), 1040; https://doi.org/10.3390/ph17081040 - 7 Aug 2024
Viewed by 264
Abstract
Gefitinib is a selective inhibitor of the epidermal growth factor receptor that is used to treat advanced and metastatic non-small cell lung cancer (NSCLC). Dermatological adverse reactions are most commonly associated with gefitinib treatment. The cause of adverse reactions in individuals is multifactorial. [...] Read more.
Gefitinib is a selective inhibitor of the epidermal growth factor receptor that is used to treat advanced and metastatic non-small cell lung cancer (NSCLC). Dermatological adverse reactions are most commonly associated with gefitinib treatment. The cause of adverse reactions in individuals is multifactorial. Pharmacogenetics is an effective tool to detect such adverse reactions. This case report describes a female patient with NSCLC who was administered gefitinib at a dose of 250 mg/day. However, due to severe adverse dermatological reactions, the treatment was interrupted for 15 d and antibiotic therapy was administered to manage the skin rashes, maculopapular rashes, and hyperpigmentation. Treatment adherence was adequate, and no drug interactions were detected. A pharmacogenetic analysis revealed homozygosity in the ATP-binding cassette (ABC)-B1 rs1128503 (c.1236A>G), heterozygosity in ABCG2 rs2231142 (c.421G>T) and rs2622604 (c.-20+614T>C), and a non-functional variant of the cytochrome P450 family 3, subfamily A, member 5 (CYP3A5). The relationship between altered genetic variants and the presence of adverse reactions induced by gefitinib is still controversial. Overall, this case report highlights the importance of continuing to study pharmacogenetics as predictors of adverse drug reactions. Full article
(This article belongs to the Special Issue Pharmacogenomics for Precision Medicine)
Show Figures

Figure 1

Figure 1
<p>Adverse reactions induced by gefitinib (250 mg/day) in a 55-year-old woman. (<b>a</b>) Mild cutaneous adverse reactions on the face after 4 weeks of gefitinib treatment. (<b>b</b>) Severe cutaneous adverse reactions on the face after 10 weeks of gefitinib treatment. (<b>c</b>) Appearance of pustules and papules on the left side of the cheek after 10 weeks of gefitinib treatment. (<b>d</b>) Cutaneous adverse reactions on the face 7 d after interrupting gefitinib treatment and starting oral tetracycline (1 g twice daily) treatment. (<b>e</b>) Left side of the face 7 d after interrupting gefitinib treatment and starting oral tetracycline (1 g twice daily) treatment. (<b>f</b>) Right side of the face 7 d after interrupting gefitinib treatment and starting oral tetracycline (1 g twice daily) treatment. (<b>g</b>) Cutaneous adverse reactions on the face 15 d after interrupting gefitinib treatment and starting oral tetracycline treatment. (<b>h</b>) Left side of the face 15 d after interrupting gefitinib treatment and starting oral tetracycline treatment. (<b>i</b>) Right side of the face 15 d after interrupting gefitinib treatment and starting oral tetracycline treatment. (<b>j</b>) The face showed a serious adverse reaction 8 months after the re-introduction of gefitinib. (<b>k</b>) Patient abdomen exhibited mild adverse reactions 8 months after the re-introduction of gefitinib. (<b>l</b>) Right side of the costal region exhibited adverse reactions after the re-introduction of gefitinib. (<b>m</b>) Left side of the costal region exhibited adverse reactions after the re-introduction of gefitinib.</p>
Full article ">
15 pages, 934 KiB  
Review
Keratins 6, 16, and 17 in Health and Disease: A Summary of Recent Findings
by Daniil D. Romashin, Tatiana V. Tolstova, Alexandra M. Varshaver, Peter M. Kozhin, Alexander L. Rusanov and Natalia G. Luzgina
Curr. Issues Mol. Biol. 2024, 46(8), 8627-8641; https://doi.org/10.3390/cimb46080508 - 6 Aug 2024
Viewed by 417
Abstract
Keratins 6, 16, and 17 occupy unique positions within the keratin family. These proteins are not commonly found in the healthy, intact epidermis, but their expression increases in response to damage, inflammation, and hereditary skin conditions, as well as cancerous cell transformations and [...] Read more.
Keratins 6, 16, and 17 occupy unique positions within the keratin family. These proteins are not commonly found in the healthy, intact epidermis, but their expression increases in response to damage, inflammation, and hereditary skin conditions, as well as cancerous cell transformations and tumor growth. As a result, there is an active investigation into the potential use of these proteins as biomarkers for different pathologies. Recent studies have revealed the role of these keratins in regulating keratinocyte migration, proliferation, and growth, and more recently, their nuclear functions, including their role in maintaining nuclear structure and responding to DNA damage, have also been identified. This review aims to summarize the latest research on keratins 6, 16, and 17, their regulation in the epidermis, and their potential use as biomarkers in various skin conditions. Full article
(This article belongs to the Special Issue Latest Review Papers in Molecular Biology 2024)
Show Figures

Figure 1

Figure 1
<p>Keratin 17 structure and assembly. (<b>A</b>) A schematic structure of keratin 17 showing non-helical domains at the N-terminus and C-terminus ends, with a central helical rod domain. K17’s structure consists of four alpha-helical domains (1A, 1B, 2A, and 2B), separated by three non-helical linkers (L1, L12, and L2). K17 also has two motifs, the helix initiation (him) and termination (hit) motifs, located at the ends of domains 1A and 2B, respectively. (<b>B</b>) A depiction of the alignment of the acidic type I keratins K17 and K16 along with the basic keratin K6, arranged in parallel to create a heterodimer. These heterodimers align in an antiparallel and staggered fashion to form a tetramer.</p>
Full article ">Figure 2
<p>The cell functions of K6, K16, and K17. Damaged keratinocytes release various DAMPs, which induce the expression of K6/16/17 by activating the AP-1, NFkB, Nrf2, MAPK (ERK1/2 and p38) pathways. Altered expression of K17 promotes the secretion of Th1 cytokines (IFNγ, IL17A, and IL22) that maintain K6/16/17 expression through an autoimmune feedback loop. Keratin 17 regulates cell growth and proliferation by binding to the 14-3-3σ/AKT/mTOR pathway and STAT3 phosphorylation. When located in the nucleus, keratin 17 is involved in the DNA damage response by interacting with various DNA damage response proteins, including Aire, hnRNPK, H2AX, DNA-PKcs, 53BP1, and HMGN2. Keratin 16 modulates the secretion of DAMPs through the MAPK and EGFR signaling pathways and plays a role in maintaining the redox balance by interacting with the Nrf2 signaling pathway. Keratins K6 and K16 also play a role in regulating the morphology and functions of mitochondria. Keratin 6 is involved in regulating cell adhesion by directly interacting with myosin IIA and desmosomal proteins, which provide the mechanical properties necessary for wound healing.</p>
Full article ">
23 pages, 5564 KiB  
Review
Innate Immune Cells in Melanoma: Implications for Immunotherapy
by Marialuisa Trocchia, Annagioia Ventrici, Luca Modestino, Leonardo Cristinziano, Anne Lise Ferrara, Francesco Palestra, Stefania Loffredo, Mariaelena Capone, Gabriele Madonna, Marilena Romanelli, Paolo Antonio Ascierto and Maria Rosaria Galdiero
Int. J. Mol. Sci. 2024, 25(15), 8523; https://doi.org/10.3390/ijms25158523 - 5 Aug 2024
Viewed by 325
Abstract
The innate immune system, composed of neutrophils, basophils, eosinophils, myeloid-derived suppressor cells (MDSCs), macrophages, dendritic cells (DCs), mast cells (MCs), and innate lymphoid cells (ILCs), is the first line of defense. Growing evidence demonstrates the crucial role of innate immunity in tumor initiation [...] Read more.
The innate immune system, composed of neutrophils, basophils, eosinophils, myeloid-derived suppressor cells (MDSCs), macrophages, dendritic cells (DCs), mast cells (MCs), and innate lymphoid cells (ILCs), is the first line of defense. Growing evidence demonstrates the crucial role of innate immunity in tumor initiation and progression. Several studies support the idea that innate immunity, through the release of pro- and/or anti-inflammatory cytokines and tumor growth factors, plays a significant role in the pathogenesis, progression, and prognosis of cutaneous malignant melanoma (MM). Cutaneous melanoma is the most common skin cancer, with an incidence that rapidly increased in recent decades. Melanoma is a highly immunogenic tumor, due to its high mutational burden. The metastatic form retains a high mortality. The advent of immunotherapy revolutionized the therapeutic approach to this tumor and significantly ameliorated the patients’ clinical outcome. In this review, we will recapitulate the multiple roles of innate immune cells in melanoma and the related implications for immunotherapy. Full article
Show Figures

Figure 1

Figure 1
<p>Melanoma is a malignant tumor, which develops from an alteration in epidermal melanocytes. One of the main risk factors is overall lifetime exposure to solar light and sunburn frequency. Unprotected exposure to UVA and UVB rays damages the DNA of skin cells, causing genetic defects or mutations that can lead to skin cancer and premature aging (<b>a</b>). Increasing data suggest that innate immunity has a role in affecting the tumor microenvironment (TME) and cancer patients’ clinical outcomes. Innate immune cells exhibit amazing adaptability, acquiring both pro- and anti-tumorigenic roles depending on different factors present in the TME (<b>b</b>). The primary treatment option for cutaneous melanoma is surgery. Among the treatments used against melanoma, it is also possible to find chemotherapy and radiotherapy. Melanoma cells are particularly sensitive to radiation. Until a few years ago, chemotherapy was the only weapon available in advanced disease but, today, it plays a minor role. In recent years, targeted and immunotherapies have shown promise in treating advanced melanomas. Despite these considerable gains, most people become resistant. Understanding the methods by which melanomas gain resistance is critical for developing customized medicines that take into account each patient’s unique genetic and immunological features (<b>c</b>).</p>
Full article ">Figure 2
<p>The innate immune system, composed of MDSCs, macrophages, neutrophils, DCs, MCs, basophils, eosinophils, and ILCs, is the first line of defense. Innate immune cells are characterized by a surprising plasticity and can release both pro- and antitumorigenic molecules depending on factors present in the TME. Arrows indicate protumorigenic (red ones) or antitumorigenic (green ones) effects and molecules produced by melanoma or innate immune cells within the TME.</p>
Full article ">Figure 3
<p>Immune cells respond to different immunogens in a dynamic and specialized manner because of their developmental diversity and phenotypic flexibility. Patrolling monocytes increase NK cell resistance to metastasis by releasing IL-15, which induces IFN-γ production. They also maintain NK cell activation by maintaining high levels of NK-cell-activating receptors and low levels of NK cell inhibitory receptors. Macrophages can activate and recruit NK cells, thus increasing their resistance to metastases. Neutrophils release neutrophils extracellular traps (NETs), which prevents the migration of tumor cells and promotes cytotoxicity towards neoplastic cells. Additionally, neutrophils promote metastasis by preventing NK cell activation basophils and eosinophils can produce various protumor signals through the release of angiogenic molecules such as vascular endothelial growth factor (VEGF) A and B. Eosinophils also display antitumorigenic activity. MDSCs are essential for the development of tumors. The interaction of CCL5 with CCR5 stimulates the growth, invasion, angiogenesis, and recruitment of immune cells into the TME. The known mediators of the immunosuppressive actions of MDSC are ARG-1, ROS, PD-L1, and NO. MCs can also produce a range of cytokines, such as TNF-α, IL-1, IL-4, IL-8, IL-6, MCP-3, and MCP-4, which can help suppress the growth of tumors by triggering apoptosis. Tumor vascularization can be promoted by MCs by the secretion of angiogenic molecules such as VEGF-A, IL-8, FGF-2, VEGF-C, MMP-2, and MMP-9.</p>
Full article ">Figure 4
<p>ILC and DCs cross-talk in melanoma microenvironment. Proinflammatory cytokines expressed by DCs trigger the production of PDGF and GM-CSF by ILC1 cells, which, in turn, promotes tumor angiogenesis. ILC2 induces the recruitment of eosinophils to the lung metastatic niche, through the production of IL-5. Melanoma secretes IL-12, which causes endothelial cells to upregulate ICAM and VCAM, thus attracting NKp46+ ILC3 to the tumor bed and activating the vasculature. By promoting leukocyte infiltration through endothelial activation, NKp46+ ILC3 act against melanoma. Melanoma secreting CCL21 attracts CCR6<sup>+</sup> (CD4<sup>+</sup>) ILC3, which interact with fibroblastic reticular cells (FRC) to produce lymphoid-like stroma, generating a tolerogenic tumor environment. ILC3 are activated by DC-derived IL-23, through the expression of RORγt. Thus, when DC-derived IL-23 is produced in the melanoma microenvironment, ILC3s are activated and contribute to the protection against melanoma.</p>
Full article ">
19 pages, 1048 KiB  
Review
Micro RNA Dysregulation in Keratinocyte Carcinomas: Clinical Evidence, Functional Impact, and Future Directions
by Jessica Conley, Benjamin Genenger, Bruce Ashford and Marie Ranson
Int. J. Mol. Sci. 2024, 25(15), 8493; https://doi.org/10.3390/ijms25158493 - 3 Aug 2024
Viewed by 470
Abstract
The keratinocyte carcinomas, basal cell carcinoma (BCC), and cutaneous squamous cell carcinoma (cSCC), are the most common cancers in humans. Recently, an increasing body of literature has investigated the role of miRNAs in keratinocyte carcinoma pathogenesis, progression and their use as therapeutic agents [...] Read more.
The keratinocyte carcinomas, basal cell carcinoma (BCC), and cutaneous squamous cell carcinoma (cSCC), are the most common cancers in humans. Recently, an increasing body of literature has investigated the role of miRNAs in keratinocyte carcinoma pathogenesis, progression and their use as therapeutic agents and targets, or biomarkers. However, there is very little consistency in the literature regarding the identity of and/or role of individual miRNAs in cSCC (and to a lesser extent BCC) biology. miRNA analyses that combine clinical evidence with experimental elucidation of targets and functional impact provide far more compelling evidence than studies purely based on clinical findings or bioinformatic analyses. In this study, we review the clinical evidence associated with miRNA dysregulation in KCs, assessing the quality of validation evidence provided, identify gaps, and provide recommendations for future studies based on relevant studies that investigated miRNA levels in human cSCC and BCC. Furthermore, we demonstrate how miRNAs contribute to the regulation of a diverse network of cellular functions, and that large-scale changes in tumor cell biology can be attributed to miRNA dysregulation. We highlight the need for further studies investigating the role of miRNAs as communicators between different cell types in the tumor microenvironment. Finally, we explore the clinical benefits of miRNAs as biomarkers of keratinocyte carcinoma prognosis and treatment. Full article
(This article belongs to the Special Issue Immunological and Molecular Networks in the Skin and Skin Diseases)
Show Figures

Figure 1

Figure 1
<p>Consensus of dysregulated miRNAs in cSCC. (<b>A</b>) Conflicting evidence leaves the deregulation status of several miRNAs unclear. The intersections of Venn diagram represent miRNAs whose differential expression trend has been conflictingly reported in two or more studies. (<b>B</b>) The table presents dysregulated/non-differentially expressed miRNAs based on the consensus of two or more studies regarding their dysregulation status. Only comparisons of cSCC to healthy skin were included in these analyses.</p>
Full article ">Figure 2
<p>Consensus of upregulated and downregulated miRNAs in BCC. (<b>A</b>) Conflicting evidence leaves the deregulation status of several miRNAs unclear. The intersections of the Venn diagram represent miRNAs whose differential expression trend has been conflictingly reported in two or more studies. (<b>B</b>) The table presents up- and downregulated miRNAs based on the consensus of two or more studies regarding their dysregulation status. Only comparisons of BCC to healthy skin were included in these analyses.</p>
Full article ">
16 pages, 3367 KiB  
Article
The Invisible Fraction within Melanin Capable of Absorbing UV Light and with Fluorescent Properties: Is It Lacking Consideration?
by Aaliyah Flake and Koen Vercruysse
Int. J. Mol. Sci. 2024, 25(15), 8490; https://doi.org/10.3390/ijms25158490 - 3 Aug 2024
Viewed by 454
Abstract
Expanding on earlier observations, we show that many melanin materials, in vitro synthesized from a wide range of precursors, can be fractionated into a dark-colored precipitate and a near-colorless, dispersible fraction. The dispersible fractions exhibited absorbance in the UVA and UVB range of [...] Read more.
Expanding on earlier observations, we show that many melanin materials, in vitro synthesized from a wide range of precursors, can be fractionated into a dark-colored precipitate and a near-colorless, dispersible fraction. The dispersible fractions exhibited absorbance in the UVA and UVB range of the electromagnetic spectrum, but none in the visible range. In addition, fluorescent properties were associated with all dispersible fractions obtained. FT-IR spectroscopic analyses were performed to compare both types of fractions. Overall, it appears that some of the properties associated with melanin (UV absorbance, fluorescence) may not necessarily reside in the dark-colored portion of melanin, but in a colorless fraction of the material. It remains to be seen whether any of these in vitro observations have any relevance in vivo. However, we raise the possibility that the presence of a colorless fraction within melanin materials and their associated properties may have received inadequate attention. Given the important association between melanin, UV protection, and skin cancer, it is worthwhile to consider this additional aspect of melanin chemistry. Full article
(This article belongs to the Special Issue Melanin and Other Pigments: Function, Synthesis and Characterization)
Show Figures

Figure 1

Figure 1
<p>Chemical structures of the precursors used in the synthesis of MN-like materials.</p>
Full article ">Figure 2
<p>Changes in AUC values of dialyzed reaction mixtures following precipitation with varying amounts of La<sup>3+</sup>. (<b>a</b>) Reactions involving L-DOPA, dopamine, norepinephrine, epinephrine, and DHI. (<b>b</b>) Reactions involving catechol, pyrogallol, 3,4-dihydroxybenzoic acid, and caffeic acid.</p>
Full article ">Figure 2 Cont.
<p>Changes in AUC values of dialyzed reaction mixtures following precipitation with varying amounts of La<sup>3+</sup>. (<b>a</b>) Reactions involving L-DOPA, dopamine, norepinephrine, epinephrine, and DHI. (<b>b</b>) Reactions involving catechol, pyrogallol, 3,4-dihydroxybenzoic acid, and caffeic acid.</p>
Full article ">Figure 3
<p>UV-Vis profiles of the F<sub>disp</sub> fractions obtained from select precursors used in this study. The fractions were dispersed in water at a concentration of 100 μg/mL. (<b>a</b>) Reactions involving L-DOPA, dopamine, norepinephrine, epinephrine. (<b>b</b>) Reactions involving catechol, pyrogallol, 3,4-dihydroxybenzoic acid, and caffeic acid.</p>
Full article ">Figure 3 Cont.
<p>UV-Vis profiles of the F<sub>disp</sub> fractions obtained from select precursors used in this study. The fractions were dispersed in water at a concentration of 100 μg/mL. (<b>a</b>) Reactions involving L-DOPA, dopamine, norepinephrine, epinephrine. (<b>b</b>) Reactions involving catechol, pyrogallol, 3,4-dihydroxybenzoic acid, and caffeic acid.</p>
Full article ">Figure 4
<p>Concentration-dependent fluorescence of the F<sub>disp</sub> fractions obtained from select precursors used in this study. (<b>a</b>) Reactions involving L-DOPA, dopamine, norepinephrine, epinephrine. (<b>b</b>) Reactions involving catechol, pyrogallol, 3,4-dihydroxybenzoic acid, and caffeic acid.</p>
Full article ">Figure 5
<p>FT-IR spectra of the F<sub>disp</sub>, F<sub>prec</sub>, and HCl-washed F<sub>prec</sub> fractions obtained from (<b>a</b>) L-DOPA, (<b>b</b>) dopamine, (<b>c</b>) norepinephrine, (<b>d</b>) epinephrine, (<b>e</b>) catechol, (<b>f</b>) pyrogallol, (<b>g</b>) 3,4-dihydroxybenzoic acid, (<b>h</b>) caffeic acid, (<b>i</b>) DHI, and (<b>j</b>) serotonin. For comparison purposes, all profiles are normalized for their absorbance at 1600 cm<sup>−1</sup>.</p>
Full article ">Figure 5 Cont.
<p>FT-IR spectra of the F<sub>disp</sub>, F<sub>prec</sub>, and HCl-washed F<sub>prec</sub> fractions obtained from (<b>a</b>) L-DOPA, (<b>b</b>) dopamine, (<b>c</b>) norepinephrine, (<b>d</b>) epinephrine, (<b>e</b>) catechol, (<b>f</b>) pyrogallol, (<b>g</b>) 3,4-dihydroxybenzoic acid, (<b>h</b>) caffeic acid, (<b>i</b>) DHI, and (<b>j</b>) serotonin. For comparison purposes, all profiles are normalized for their absorbance at 1600 cm<sup>−1</sup>.</p>
Full article ">Figure 5 Cont.
<p>FT-IR spectra of the F<sub>disp</sub>, F<sub>prec</sub>, and HCl-washed F<sub>prec</sub> fractions obtained from (<b>a</b>) L-DOPA, (<b>b</b>) dopamine, (<b>c</b>) norepinephrine, (<b>d</b>) epinephrine, (<b>e</b>) catechol, (<b>f</b>) pyrogallol, (<b>g</b>) 3,4-dihydroxybenzoic acid, (<b>h</b>) caffeic acid, (<b>i</b>) DHI, and (<b>j</b>) serotonin. For comparison purposes, all profiles are normalized for their absorbance at 1600 cm<sup>−1</sup>.</p>
Full article ">
28 pages, 2311 KiB  
Review
Isothermal Technologies for HPV Detection: Current Trends and Future Perspectives
by Elda A. Flores-Contreras, Everardo González-González, Gerardo de Jesús Trujillo-Rodríguez, Iram P. Rodríguez-Sánchez, Jesús Ancer-Rodríguez, Antonio Alí Pérez-Maya, Salomon Alvarez-Cuevas, Margarita L. Martinez-Fierro, Iván A. Marino-Martínez and Idalia Garza-Veloz
Pathogens 2024, 13(8), 653; https://doi.org/10.3390/pathogens13080653 - 2 Aug 2024
Viewed by 452
Abstract
The human papillomavirus (HPV) is a non-enveloped DNA virus transmitted through skin-to-skin contact that infects epithelial and mucosal tissue. It has over 200 known genotypes, classified by their pathogenicity as high-risk and low-risk categories. High-risk HPV genotypes are associated with the development of [...] Read more.
The human papillomavirus (HPV) is a non-enveloped DNA virus transmitted through skin-to-skin contact that infects epithelial and mucosal tissue. It has over 200 known genotypes, classified by their pathogenicity as high-risk and low-risk categories. High-risk HPV genotypes are associated with the development of different types of cancers, including cervical cancer, which is a leading cause of mortality in women. In clinical practice and the market, the principal tests used to detect HPV are based on cytology, hybrid detection, and qPCR. However, these methodologies may not be ideal for the required timely diagnosis. Tests have been developed based on isothermal nucleic acid amplification tests (INAATs) as alternatives. These tests offer multiple advantages over the qPCR, such as not requiring specialized laboratories, highly trained personnel, or expensive equipment like thermocyclers. This review analyzes the different INAATs applied for the detection of HPV, considering the specific characteristics of each test, including the HPV genotypes, gene target, the limit of detection (LOD), detection methods, and detection time. Additionally, we discuss the tests available on the market that are approved by the Food and Drug Administration (FDA). Finally, we address the challenges and potential solutions for the large-scale implementation of INAATs, particularly in rural or underserved areas. Full article
(This article belongs to the Special Issue Advances in HPV Diagnostic Testing—Improving Access and Accuracy)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>A summary of the global health problem caused by HPV and the methodologies used for its detection for subsequent management. (<b>A</b>) HPV is responsible for causing different types of cancer in the world population, with cervical cancer being the most prevalent. In 2022, it is estimated that 600,000 new cases of cervical cancer were diagnosed globally, and approximately 350,000 people died because of cervical cancer. (<b>B</b>) There are two main approaches to addressing the cancer problem caused by HPV. The first is the direct detection of HPV, and the second is the detection of the effects of HPV infection on cells or tissues.</p>
Full article ">Figure 2
<p>The global strategy 90-70-90 established by the World Health Organization (WHO) is composed of three essential pillars. Diagnosis is a core point for the strategy, where Isothermal nucleic acid amplification techniques (INAATs) can be a powerful tool in the fight against cervical cancer and HPV [<a href="#B18-pathogens-13-00653" class="html-bibr">18</a>].</p>
Full article ">Figure 3
<p>A graph of the INAATs developed and published in the literature used to detect HPV. The number of tests developed is indicated, and the techniques marked in red indicate a commercial product based on it for the detection of HPV.</p>
Full article ">Figure 4
<p>Representative scheme of the general methodology to carry out INAATs and the platforms with which they are usually combined. (<b>A</b>) The steps to be followed for amplification of the target region of the HPV virus using INAATs are shown. (1) The first step consists of taking a sample of the tissue of interest (mouth, cervical, oropharynx, penis, anus, urine, among others); (2) The sample is subsequently incubated with the components of the amplification reaction (enzymes, buffer primers, and dNTPs) at a constant temperature (ranging from 20 °C to 65 °C) for approx. 60 min. (3) The presence of the virus can be detected by electrophoresis, turbidimetry, fluorescence, or colorimetric methods. (<b>B</b>) Furthermore, INAATs are combined with different platforms such as microfluidic techniques that allow the sample to be loaded into the device (1), to later carry out the amplification reaction on a chip (2) and detect the presence of HPV (using colorimetric or fluorescence methods) (3). Also, the CRISPR-Cas system has been used (1), which detects the target sequence by crRNA (2), causing a cut in the target sequence (3), and activating the Cas enzyme which acts as a ssDNase cutting the probes conjugated with fluorophores and quencher (4), and releasing a fluorescence signal indicating the presence of HPV (5). Likewise, lateral flow assay (LFA) has been used, in which after the amplification reaction, the sample is placed on the strip (1), and is labeled with molecules or antibodies located on the sample pad (2), and through the appearance of one band on the test line and another on the control line, it detects HPV since the labeled sample interacts with the molecules or antibodies fixed in the test (3) and control (4) lines. Also, biosensors that identify HPV’s existence through mass, voltage, or electrochemical current changes.</p>
Full article ">
Back to TopTop