Nothing Special   »   [go: up one dir, main page]

You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (946)

Search Parameters:
Keywords = MEK2

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
18 pages, 10138 KiB  
Article
Micheliolide Alleviates Hepatic Fibrosis by Inhibiting Autophagy in Hepatic Stellate Cells via the TrxR1/2-Mediated ROS/MEK/ERK Pathway
by Yi Liu, Ling Yao, Yuanyuan Liu, Yunheng Yang, Ailing Liang, Honglin He, Yao Lei, Wenfu Cao and Zhiwei Chen
Pharmaceuticals 2025, 18(3), 287; https://doi.org/10.3390/ph18030287 - 20 Feb 2025
Viewed by 228
Abstract
Background: Hepatic fibrosis is a major global health issue without an optimal drug treatment, highlighting the urgent need to find effective therapies. This study aimed to clarify the role and mechanism of micheliolide in treating hepatic fibrosis. Methods: The efficacy of [...] Read more.
Background: Hepatic fibrosis is a major global health issue without an optimal drug treatment, highlighting the urgent need to find effective therapies. This study aimed to clarify the role and mechanism of micheliolide in treating hepatic fibrosis. Methods: The efficacy of MCL was evaluated in a mouse model of CCl4-induced hepatic fibrosis. LX-2 cells were subjected to MCL treatment, and subsequent changes in fibrosis markers, autophagy, and the MEK/ERK pathway were analyzed using transcriptomics and Western blotting. The interaction between MCL and TrxR1 or TrxR2 were validated using cellular thermal shift assays (CETSA) and drug affinity responsive target stability (DARTS) assays. Results: Our findings indicated that MCL significantly alleviated CCl4-induced hepatic fibrosis, improved liver function, and downregulated the expression of fibrosis markers. Additionally, MCL significantly inhibited LX-2 cell activation by suppressing cell proliferation, extracellular matrix (ECM) production, and autophagy, while activating the MEK/ERK pathway. Moreover, MCL elevated intracellular and mitochondrial reactive oxygen species (ROS) levels, reduced mitochondrial membrane potential, and altered mitochondrial morphology. The ROS scavenger N-acetylcysteine (NAC) attenuated MCL-induced MEK/ERK pathway activation and increased collagen type I alpha 1 (COL1A1) and fibronectin (FN) expression. Further analysis confirmed that MCL directly interacts with TrxR1 and TrxR2, leading to the inhibition of their enzymatic activities and the induction of ROS generation. Ultimately, MCL attenuated the fibrotic process and autophagic flux in LX-2 cells. Conclusions: The findings of our study confirmed that MCL has the potential to alleviate hepatic fibrosis, thereby introducing a novel candidate drug and therapeutic strategy for management of this condition. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Effects of MCL on CCl<sub>4</sub>-induced hepatic fibrosis in vivo. (<b>A</b>) Schematic of the experimental procedure for animal treatment. (<b>B</b>) Representative photographs of liver sections were stained with H&amp;E, Masson, and Sirius Red. (<b>C</b>) Statistical analysis of collagen volume fraction based on Masson staining. (<b>D</b>) The liver-to-body weight ratio analysis. (<b>E</b>–<b>G</b>) The serum levels of AST, ALT, and HYP in mice. (<b>H</b>) Western blot analysis of COL1A1 and FN in liver tissue. (<b>I</b>) Histogram showing quantification of bands in (<b>H</b>). <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. control group. ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001 vs. model group.</p>
Full article ">Figure 2
<p>Effects of MCL on LX-2 cell proliferation in vitro. (<b>A</b>) Relative cell viability of LX-2 cells treated with MCL. (<b>B</b>) Analysis of cell proliferation using EdU labeling. (<b>C</b>) The percentage analysis of EdU-positive LX-2 cells. (<b>D</b>) GSEA enrichment plots of proliferation-related gene sets. (<b>E</b>) Analysis of cell cycle using flow cytometry. (<b>F</b>) Graph represents quantitative data of cell cycle distribution. (<b>G</b>) Western blot analysis of CDK1 and c-Myc in LX-2 cells. (<b>H</b>) Histogram showing quantification of bands in (<b>G</b>). * <span class="html-italic">p</span> &lt; 0.05 and *** <span class="html-italic">p</span> &lt; 0.001 vs. 0 μM.</p>
Full article ">Figure 3
<p>Effects of MCL on LX-2 cell activation in vitro. (<b>A</b>) Reactome enrichment analysis of differentially expressed genes. Red arrows: ECM-related processes. (<b>B</b>) The gene sets are associated with ECM. (<b>C</b>) The heatmap shows the relative levels of MMPs and collagen-encoding genes. (<b>D</b>,<b>E</b>) Relative mRNA levels of COL1A1 and FN in LX-2 cells. (<b>F</b>) Western blot analysis of COL1A1 and FN in LX-2 cells. (<b>G</b>) Histogram showing quantification of bands in (<b>F</b>). * <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 vs. 0 μM.</p>
Full article ">Figure 4
<p>Effects of MCL on LX-2 cell autophagy. (<b>A</b>) Western blot analysis of ATG5, LC3B, and p62 in LX-2 cells. (<b>B</b>) Histogram showing quantification of bands in (<b>A</b>). (<b>C</b>) Lysosomes of LX-2 cells as stained with LysoTracker Red. (<b>D</b>) Relative level of fluorescence intensity. ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001 vs. 0 μM.</p>
Full article ">Figure 5
<p>Effects of MCL on the MEK/ERK pathway. (<b>A</b>) Western blot analysis of pMEK1/2, MEK1/2, pERK1/2, and ERK1/2 in LX-2 cells. (<b>B</b>) Histogram showing quantification of bands in (<b>A</b>). * <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 vs. 0 μM. (<b>C</b>) Western blot analysis of pERK1/2, LC3B, p62, COL1A1, and FN in LX-2 cells. (<b>D</b>) Histogram showing quantification of bands in (<b>C</b>). * <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.</p>
Full article ">Figure 6
<p>Effects of MCL on ROS and mitochondria. (<b>A</b>) Flow cytometric measurement of ROS levels in LX-2 cells. (<b>B</b>) Relative total ROS levels in (<b>A</b>). (<b>C</b>) Flow cytometric measurement of ROS levels in mitochondria. (<b>D</b>) Relative mtROS levels in (<b>C</b>). (<b>E</b>) Flow cytometric analysis of mitochondrial membrane potential in LX-2 cells. (<b>F</b>) Morphological observation of mitochondria using confocal microscopy. (<b>G</b>) Western blot analysis of pMEK1/2, MEK1/2, pERK1/2, ERK1/2, LC3B, p62, COL1A1, and FN in LX-2 cells. (<b>H</b>) Histogram showing quantification of bands in (<b>G</b>). * <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.</p>
Full article ">Figure 7
<p>MCL targets TrxR1 and TrxR2 in LX-2 cells. (<b>A</b>) The space-filling and ribbon models of MCL and human TrxR1 complexes. (<b>B</b>) The 2D binding model of MCL and human TrxR1 complexes. (<b>C</b>) Western blot analysis of TrxR1 and TrxR2 in CETSA. (<b>D</b>) Histogram showing quantification of bands in (<b>C</b>). (<b>E</b>) Western blot analysis of TrxR1 and TrxR2 in DARTS. (<b>F</b>) Histogram showing quantification of bands in (<b>E</b>). (<b>G</b>) Western blot analysis of TrxR1 and TrxR2 in LX-2 cells. (<b>H</b>) Histogram showing quantification of bands in (<b>G</b>). (<b>I</b>) The activity of TrxR was detected by a TRFS green probe in LX-2 cells. * <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.</p>
Full article ">Figure 8
<p>Schematic representation of the molecular mechanism by which MCL alleviates hepatic fibrosis by inhibiting autophagy in hepatic stellate cells via the TrxR1/2-mediated ROS/MEK/ERK pathway.</p>
Full article ">
21 pages, 4162 KiB  
Article
Anti-Angiogenic Potential of Marine Streptomyces-Derived Lucknolide A on VEGF/VEGFR2 Signaling in Human Endothelial Cells
by Byeoung-Kyu Choi, Min-Hee Jo, Hee Jae Shin and Sun Joo Park
Molecules 2025, 30(5), 987; https://doi.org/10.3390/molecules30050987 - 20 Feb 2025
Viewed by 221
Abstract
Angiogenesis, primarily driven by the vascular endothelial growth factor (VEGF) and its receptor, the VEGFR, plays a key role in various pathological processes such as cancer progression. Here, we investigated the anti-angiogenic effects of Lucknolide A (LA), a marine Streptomyces-derived compound, and [...] Read more.
Angiogenesis, primarily driven by the vascular endothelial growth factor (VEGF) and its receptor, the VEGFR, plays a key role in various pathological processes such as cancer progression. Here, we investigated the anti-angiogenic effects of Lucknolide A (LA), a marine Streptomyces-derived compound, and evaluated its potential as a VEGFR2 inhibitor. LA selectively inhibited the proliferation of human endothelial cells EA.hy926 and HUVEC while exhibiting minimal effects on normal fibroblasts and various tumor cells. LA induced S-phase cell cycle arrest and apoptosis in EA.hy926 cells, increasing apoptotic markers p53, Bax, and p21 and decreasing the anti-apoptotic protein Bcl-2, with these effects being further enhanced under VEGF stimulation. Additionally, LA suppressed VEGFR2 phosphorylation and its downstream signaling pathways, including Akt/mTOR/p70S6K, MEK/ERK, Src, FAK, and p38 MAPK, which are crucial for endothelial survival and angiogenesis. Molecular docking studies revealed that LA binds to both inactive (DFG-out, PDB: 4ASD) and active (DFG-in, PDB: 3B8R) VEGFR2 conformations, with a significantly stronger affinity for the active state (−107.96 kcal/mol) than the inactive state (−33.56 kcal/mol), suggesting its potential as a VEGFR2 kinase inhibitor. Functionally, LA significantly inhibited VEGF-induced endothelial migration, tube formation, and microvessel sprouting in both in vitro and ex vivo rat aortic ring assays. Additionally, LA reduced tumor-associated tube formation induced by human breast tumor cells (MDA-MB-231), indicating its potential to suppress VEGF-dependent tumor angiogenesis. These findings suggest that LA is a promising selective anti-angiogenic agent with potential therapeutic applications in angiogenesis-related diseases such as cancer. Full article
(This article belongs to the Special Issue Bioactive Compounds: Applications and Benefits for Human Health)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The structure of Lucknolide A (LA). LA was isolated from a marine-derived <span class="html-italic">Streptomyces</span> sp. strain 151KO-065.</p>
Full article ">Figure 2
<p>LA decreases the proliferation of EA.hy926 and HUVEC cells in a dose- and time-dependent manner. (<b>A</b>) EA.hy926, HUVEC, MRC5, A549, Ghost, HepG2, MDA-MB-231, PANC-1, and PC3 cells were treated with various concentrations (0–50 μM) of LA for 72 h. Cell viability was determined by MTT assay. Each value is expressed as the mean ± standard deviation (SD) of three independent experiments. (<b>B</b>) EA.hy926 and (<b>C</b>) HUVEC cells were treated with 0–50 µM LA in the presence or absence of 50 ng/mL of the VEGF for different time intervals (24 h, 72 h, and 120 h). CC<sub>50</sub> (μM) is expressed as the mean ± standard deviation (SD) of three independent experiments. Statistical significance is indicated as * <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. LA-only treated groups were compared to the vehicle control (DMSO), while LA + VEGF-treated groups were compared to the VEGF.</p>
Full article ">Figure 3
<p>LA causes the S-phase cell cycle arrest of EA.hy926 cells in a dose-dependent manner. (<b>A</b>) Representative cell cycle analysis images showing LA-induced S-phase arrest. EA.hy926 cells were treated with 0, 12.5, 25, and 50 μM LA for 72 h in the presence or absence of 50 ng/mL of the VEGF. Cells were fixed in ethanol and stained with propidium iodide. Cells were analyzed using flow cytometry with single histogram statistics. (<b>B</b>) Data are expressed as the mean ± SD of three independent experiments. Statistical significance is indicated as * <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. LA-only treated groups were compared to the vehicle control (DMSO), while LA + VEGF-treated groups were compared to the VEGF-treated condition.</p>
Full article ">Figure 4
<p>LA induces the apoptosis of EA.hy926 cells in a dose-dependent manner. (<b>A</b>) Representative images showing LA-induced apoptosis. EA.hy926 cells were treated with 0, 12.5, 25, and 50 μM LA for 72 h in the presence or absence of 50 ng/mL of the VEGF and analyzed by Annexin V-PI staining. (<b>B</b>) Cells that appeared Annexin V-positive/PI-negative (<b>lower right</b>) are considered early-apoptotic cells, while Annexin V/PI double-positive (<b>upper right</b>) are considered late-apoptotic cells. The percentage of live (<b>lower left</b>), early apoptotic, late-apoptotic, and dead (<b>upper left</b>) cells. Data are expressed as the mean ± SD of triplicate independent experiments. Statistical significance is indicated as * <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. LA-only treated groups were compared to the vehicle control (DMSO), while LA + VEGF-treated groups were compared to the VEGF-treated condition.</p>
Full article ">Figure 5
<p>The effect of LA on the nuclei morphology of EA.hy926 cells by Hoechst 33,258 staining. EA.hy926 cells were cultured with 0, 12.5, 25, and 50 μM of LA for 72 h in the presence or absence of 50 ng/mL of the VEGF. The nuclei were visualized under an inverted fluorescent microscope (40×). Arrows indicate apoptotic nuclei. Scale bar, 100 μm.</p>
Full article ">Figure 6
<p>The effect of LA on the proteins involved in S-phase cell cycle arrest and apoptosis in EA.hy926 cells. Representative western blot images indicating LA-induced S-phase arrest (<b>A</b>) and apoptosis (<b>B</b>). EA.hy926 cells were treated with 0, 12.5, 25, and 50 µM LA for 72 h in the presence or absence of 50 ng/mL of the VEGF. * <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 vehicle DMSO control. (<b>C</b>) LA suppresses the phosphorylation of VEGFR2 and its downstream effectors, (<b>C</b>) Akt/mTOR/p70S6K (<b>D</b>) MEK/ERK (<b>E</b>) Src, FAK, and p38 MAPK. The results were quantified using ImageJ (v1.54f) and are presented as the mean ± SD of three independent experiments. Statistical significance is indicated as * <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. LA-only treated groups were compared to the vehicle control (DMSO), while LA + VEGF-treated groups were compared to the VEGF-treated condition. # <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 DMSO control.</p>
Full article ">Figure 6 Cont.
<p>The effect of LA on the proteins involved in S-phase cell cycle arrest and apoptosis in EA.hy926 cells. Representative western blot images indicating LA-induced S-phase arrest (<b>A</b>) and apoptosis (<b>B</b>). EA.hy926 cells were treated with 0, 12.5, 25, and 50 µM LA for 72 h in the presence or absence of 50 ng/mL of the VEGF. * <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 vehicle DMSO control. (<b>C</b>) LA suppresses the phosphorylation of VEGFR2 and its downstream effectors, (<b>C</b>) Akt/mTOR/p70S6K (<b>D</b>) MEK/ERK (<b>E</b>) Src, FAK, and p38 MAPK. The results were quantified using ImageJ (v1.54f) and are presented as the mean ± SD of three independent experiments. Statistical significance is indicated as * <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. LA-only treated groups were compared to the vehicle control (DMSO), while LA + VEGF-treated groups were compared to the VEGF-treated condition. # <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 DMSO control.</p>
Full article ">Figure 7
<p>LA inhibits the VEGF-induced migration and tube formation of EA.hy926 cells. (<b>A</b>) Wound-healing scratch assays were performed on EA.hy926 cells plated onto Matrigel-coated dishes. After serum starvation, cells were pre-cultured with or without various concentrations of LA for 48 h and then further incubated with LA for 12 h in the presence or absence of 50 ng/mL of the VEGF. A sterile 200 μL pipette tip was used to create a scratch wound. Cell migration was quantified by measuring the gap size in 4 different images, and representative images are shown. Data are expressed as mean ± SD from three independent experiments. (<b>B</b>) EA.hy926 cells were pre-treated with 0, 12.5, 25, and 50 µM LA for 48 h and then further incubated with LA for 12 h in the presence or absence of 50 ng/mL of the VEGF. Tube formation was quantified by measuring the average of 4 different images, and representative images are shown. Data are expressed as the mean ± SD of three independent experiments. Statistical significance is indicated as * <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. LA-only treated groups were compared to the vehicle control (DMSO), while LA + VEGF-treated groups were compared to the VEGF. ## <span class="html-italic">p</span> &lt; 0.01, and ### <span class="html-italic">p</span> &lt; 0.001, compared to the DMSO control. Scale bar, 100 μm.</p>
Full article ">Figure 8
<p>(<b>A</b>) LA inhibits VEGF-induced microvessel sprouting ex vivo. Aortas embedded in Matrigel were treated with LA in the presence or absence of 10 ng/mL of the VEGF. After six days of incubation, microvessel growth from the aortic rings was measured using microscopic images. Data are expressed as the mean ± SD of three independent experiments. * <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 vehicle dimethyl sulfoxide (DMSO) control. (<b>B</b>) LA inhibits the MDA-MB-231 breast tumor cell-stimulated tube formation of EA.hy926 cells. EA.hy926 cells were pre-incubated with LA for 48 h, followed by an additional incubation for 12 h with LA in media containing 50% MDA-MB-231 conditioned media (MDA-CM) or control-conditioned media (CON-CM). Tube formation was quantified by measuring the average of 4 different images, with representative images shown. Data are expressed as the mean ± SD of three independent experiments. Statistical significance is indicated as * <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. LA-only treated groups were compared to the vehicle control (DMSO), while LA + VEGF-treated groups were compared to the VEGF. ## <span class="html-italic">p</span> &lt; 0.01, and ### <span class="html-italic">p</span> &lt; 0.001, compared to the DMSO control. Scale bar, 100 μm.</p>
Full article ">Figure 9
<p>Molecular docking analysis of Lucknolide A (LA) with VEGFR2. (<b>A</b>) Docking conformation of LA in the inactive VEGFR2 (DFG-out) conformation (PDB: 4ASD), showing hydrogen bond interactions with D1046 (Asp), C1045 (Cys), K868 (Lys), and E917 (Glu) within the ATP-binding pocket. (<b>B</b>) The docking conformation of LA in the active VEGFR2 (DFG-in) conformation (PDB: 3B8R), highlighting interactions with D1046 (Asp) and C1045 (Cys). (<b>C</b>) A 2D interaction diagram of LA in the inactive VEGFR2 state, illustrating key molecular interactions. (<b>D</b>) A 2D interaction diagram of LA in the active VEGFR2 state, displaying ligand–receptor contacts. (<b>E</b>) A summary of the binding energy calculations and key interacting residues for LA in inactive (4ASD) and active (3B8R) VEGFR2 conformations. Hydrogen bond interactions and other key binding residues are listed, showing a significantly stronger binding affinity in the active state (−107.9579 kcal/mol) compared to the inactive state (−33.5639 kcal/mol), suggesting a higher binding preference for the active VEGFR2 conformation.</p>
Full article ">
20 pages, 5182 KiB  
Article
Combinational Inhibition of MEK and AKT Synergistically Induces Melanoma Stem Cell Apoptosis and Blocks NRAS Tumor Growth
by Ryyan Alobaidi, Nusrat Islam, Toni Olkey, Yogameenakshi Haribabu, Mathew Shamo, Peter Sykora, Cynthia M. Simbulan-Rosenthal and Dean S. Rosenthal
Cells 2025, 14(4), 248; https://doi.org/10.3390/cells14040248 - 10 Feb 2025
Viewed by 552
Abstract
Malignant melanoma is a lethal skin cancer containing melanoma-initiating cells (MICs), implicated in tumorigenesis, invasion, and drug resistance, and characterized by an elevated expression of stem cell markers, including CD133. siRNA knockdown of CD133 has been previously shown to enhance apoptosis induced by [...] Read more.
Malignant melanoma is a lethal skin cancer containing melanoma-initiating cells (MICs), implicated in tumorigenesis, invasion, and drug resistance, and characterized by an elevated expression of stem cell markers, including CD133. siRNA knockdown of CD133 has been previously shown to enhance apoptosis induced by the MEK inhibitor trametinib in melanoma cells. This study investigates the underlying mechanisms of CD133’s anti-apoptotic activity in patient-derived BAKP melanoma, harboring the difficult-to-treat NRASQ61K driver mutation, after CRISPR-Cas9 CD133 knockout or Doxycycline (Dox)-inducible re-expression of CD133. CD133 knockout in BAKP cells increased trametinib-induced apoptosis by reducing anti-apoptotic p-AKT and p-BAD and increasing pro-apoptotic BAX. Conversely, Dox-induced CD133 expression diminished apoptosis in trametinib-treated cells, coincident with elevated p-AKT, p-BAD, and decreased activation of BAX and caspase-3. However, trametinib in combination with pan-AKT inhibitor capivasertib reduced cell survival as measured by XTT viability assays and apoptosis and colony formation assays, independent of CD133 status. CD133 may therefore activate a survival pathway wherein (1) increased AKT phosphorylation and activation induces (2) BAD phosphorylation and inactivation, which (3) decreases BAX activation, and (4) reduces caspases-3 activity and caspase-mediated PARP cleavage, leading to apoptosis suppression and drug resistance in melanoma. In vivo mouse xenograft studies using Dox-inducible melanoma cells revealed increased rates of tumor growth after induction of CD133 expression in trametinib-treated +Dox mice, an effect which was synergistically suppressed by combination treatment. Targeting nodes of the AKT and MAPK survival pathways with trametinib and capivasertib highlights the potential for combination therapies for NRAS-mutant melanoma stem cells for the development of more effective treatments for patients with high-risk melanoma. Full article
(This article belongs to the Section Cellular Pathology)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>(<b>A</b>) CD133 CRISPR-Cas9 KO increases trametinib-induced apoptosis whereas (<b>B</b>) Doxycycline (Dox)-induced CD133 expression in BAKP cells decreases apoptosis (BAX activation, PARP cleavage, and caspase-3 activation) following trametinib treatment, stabilized by the upregulation of pro-survival pAKT and pBAD in CD133-expressing cells. The cells were incubated with 100 nM trametinib for 48 h, followed by immunoblot analysis with antibodies to cleaved active caspase 3 and its substrate—cleaved PARP, the pro-apoptotic active form of Bax, the anti-apoptotic phosphorylated form of BAD (p-BAD), and the pro-survival phosphorylated active form of AKT (p-AKT Ser473). After normalizing to β-actin, a densitometric analysis comparing the intensities of protein bands relative to bands with the highest intensities is shown in the immunoblots. Scans of whole-gel immunoblots for all the figures are shown in “<a href="#app1-cells-14-00248" class="html-app">Supplementary Materials</a>” (<a href="#app1-cells-14-00248" class="html-app">Figure S1</a>). (<b>C</b>,<b>D</b>) Knockdown of CD133 expression in BAKP CD133-KO cells (<b>C</b>) and upregulation of CD133 expression in inducible BAKP cells in the presence of Dox (+Dox; (<b>D</b>)), as verified by qRT-PCR analysis. <span class="html-italic">p</span> &lt; 0.05 was considered significant; *** and **** represent <span class="html-italic">p</span> &lt; 0.001, and <span class="html-italic">p</span> &lt; 0.0001, respectively.</p>
Full article ">Figure 2
<p>Capivasertib enhances apoptosis in trametinib-treated BAKP (<b>A</b>) and POT (<b>B</b>) melanoma cell lines. Cells were seeded in equal numbers in 6-well plates in triplicates, and they were then treated with trametinib and/or capivasertib. After 48 h of treatment, the cells were subjected to Annexin-APC/SYTOX Blue apoptosis assays. The percentage of total apoptosis (the sum of early and late apoptosis in the lower right and upper right quadrants of the dot plots, respectively) was quantified by flow cytometric analysis. The results are the means <span class="html-italic">±</span> SEM of three replicates of a representative experiment; essentially the same results were obtained in three independent experiments. <span class="html-italic">p</span> &lt; 0.05 was considered significant; *, **, and **** represent <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; 0.0001, respectively. (<b>C</b>) Dot plot data used to generate the bar graphs in (<b>A</b>,<b>B</b>). FL1 and FL2 represent fluorescence channel 1 and fluorescence channel 2, respectively. (<b>D</b>) Representative phase contrast (<b>left</b> panel) and fluorescence (<b>right</b> panel) images of BAKP cells showing the loss of mitochondrial membrane potential in BAKP cells treated with trametinib alone or in combination with capivasertib, but not in control cells or those incubated with capivasertib alone, indicating that apoptosis occurs through a mitochondrial-mediated pathway. Insets in the top left corners show enlargement of representative cells.</p>
Full article ">Figure 3
<p>Effects of CD133 KO (<b>A</b>,<b>C</b>) or induced CD133 expression (<b>B</b>,<b>D</b>) on cell viability after treatment with trametinib and capivasertib, alone or in combination. Cells were plated in equal numbers in 6-well plates in triplicates and then treated for 48 h with trametinib and capivasertib, alone or in combination. The cells were then collected and subjected to XTT cell viability metabolic assays (<b>A</b>,<b>B</b>) and Sytox Blue dye exclusion assays (<b>C</b>,<b>D</b>). The percentage (%) of cell viability was quantified as described in <a href="#sec2-cells-14-00248" class="html-sec">Section 2</a>. The results shown are the means <span class="html-italic">±</span> SEM of three replicates of a representative experiment; essentially the same results were obtained in three independent experiments. <span class="html-italic">p</span> &lt; 0.05 was considered significant. *, **, and **** represent <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; 0.0001, respectively.</p>
Full article ">Figure 4
<p>(<b>A</b>) Annexin flow cytometric assays to assess apoptosis induction after treatment with trametinib and capivasertib alone or in combination. (<b>B</b>) Dot plot of data shown in (<b>A</b>); FL1 and FL2 represent fluorescence channel 1 and fluorescence channel 2, respectively.Equal numbers of BAKP-CD133 KO and control BAKP-SC cells were seeded in 6-well plates in triplicate and then treated for 48 h with trametinib, capivasertib, or in combination. After 48 h, the cells were subjected to Annexin-APC/SYTOX Blue assays. The percentage of total apoptosis was quantified by flow cytometric analysis. The results shown are the mean <span class="html-italic">±</span> SEM of triplicates of a representative experiment; essentially the same results were obtained in three independent experiments. <span class="html-italic">p</span> &lt; 0.05 was considered significant. ** represents <span class="html-italic">p</span> &lt; 0.01; ns represents not significant.</p>
Full article ">Figure 5
<p>Capivasertib in combination with trametinib elicits a maximal apoptotic response in both uninduced and Dox-induced CD133-expressing cells, as assessed by annexin flow cytometric assays (<b>A</b>). CD133 expression slightly reverses this response. Cells were seeded in equal numbers in 6-well plates in triplicates, incubated for 24 h with 1 µg/mL Dox to induce CD133 expression, and then treated for 48 h (<b>A</b>) with trametinib and capivasertib, alone or in combination. Cells were collected after treatment and subjected to Annexin-APC apoptosis assays. The percentage (%) of total apoptosis was quantified by flow cytometric analysis. (<b>B</b>) Dot plot of data shown in <b>A</b>; FL1 and FL2 represent fluorescence channel 1 and fluorescence channel 2, respectively. The results shown are the means <span class="html-italic">±</span> SEM of three replicates of a representative experiment; essentially the same results were obtained in three independent experiments. <span class="html-italic">p</span> &lt; 0.05 was considered significant. *** represents <span class="html-italic">p</span> &lt; 0.001; ns represents not significant.</p>
Full article ">Figure 6
<p>The combination of capivasertib plus trametinib synergistically reduces cell viability and induces apoptosis in melanoma cells. (<b>A</b>) Cells were seeded in equal numbers in 96-well plates in triplicate and then treated for 48 h with trametinib and capivasertib, alone or in combination (trametinib at variable concentrations and capivasertib at 1 µM). Cells were collected and subjected to XTT cell viability assays. (<b>B</b>) Cells were plated in a 6-well plate, exposed to the drugs alone or in combination as above, and then analyzed using flow cytometry after Annexin-APC/SYTOX Blue staining. (<b>A</b>,<b>B</b>) Bliss additivity was calculated and is shown as inverted triangles. Pink shading and unshaded areas represent synergy and antagonism, respectively. The results shown are the means <span class="html-italic">±</span> SEM of three replicates of a representative experiment; essentially the same results were obtained in three independent experiments. <span class="html-italic">p</span> &lt; 0.05 was considered significant. **** represents <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 7
<p>(<b>A</b>) Long-term cell survival (clonogenic) assays reveal that treatments reduce colony formation in BAKP cells, with the combination of trametinib + capivasertib decreasing colony formation to the largest extent. Dox-inducible BAKP cells were incubated for 24 h with Dox, and then exposed to trametinib and capivasertib by themselves or in combination. Then, 48 h after treatment, the cells were replated, allowed to grow for 12 days, fixed, and stained, and the colonies of cells that survived treatment were counted. The results shown are the means <span class="html-italic">±</span> SEM of three replicates of a representative experiment; essentially the same results were obtained in three independent experiments. <span class="html-italic">p</span> &lt; 0.05 was considered significant. *** and **** represent <span class="html-italic">p</span> &lt; 0.001 and <span class="html-italic">p</span> &lt; 0.0001, respectively; ns represents not significant. (<b>B</b>) Images of representative 10 cm-plates with stained colonies reveal that the treatments reduced both colony counts and colony sizes. The insets show representative colony sizes.</p>
Full article ">Figure 8
<p>(<b>A</b>) The immunoblot analysis reveals the effective inhibition of phosphorylation of AKT substrates (p-BAD and p-GSK-3β) by capivasertib by itself or in combination with trametinib. BAKP cells were incubated for 24 h with Dox and then exposed to trametinib or capivasertib alone or in combination with trametinib. Cell lysates were then subjected to immunoblot analysis with antibodies specific for CD133, phospho-BAD, p-GSK-3β, and cleaved caspase-3. Anti-β-actin was used for the confirmation of equal loading. (<b>B</b>) The indirect immunofluorescent analysis with antibodies to CD133 or the cleaved active form of caspase-3 reveals that treatment with a combination of trametinib (100 nM) + capivasertib (1 μM), but not either drug alone, markedly increases apoptotic caspase-3 activation in BAKP cells.</p>
Full article ">Figure 9
<p>CD133 increases tumor growth, which is suppressed by the trametinib + capivasertib treatment in vivo. BAKP-inducible cells were used to induce subcutaneous tumors. (<b>A</b>) Tumor volumes in treated vs. vehicle control mice (+/− Dox). Bliss additivity was calculated in +Dox mice (<b>B</b>) or −Dox mice (<b>C</b>), and is shown as a dashed purple line, while the pink-shaded areas show regions where combination treatment demonstrates synergy and improvement over trametinib alone. (<b>D</b>) No effects on the body mass of mice over the treatment period were observed. (<b>E</b>) The immunoblot analysis of tumor lysates from xenografted mice shows apoptotic caspase-3 cleavage in the combination treatment only. The results shown are the means <span class="html-italic">±</span> SEM of three replicates of a representative experiment; essentially the same results were obtained in three independent experiments. <span class="html-italic">p</span> &lt; 0.05 was considered significant. *, **, and **** represent <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; 0.0001, respectively.</p>
Full article ">Figure 10
<p>PI3K/AKT/Bcl2 and MAPK family pro-survival signaling pathway in CD133+ melanoma stem cells (MICs) and possible therapeutic targets.</p>
Full article ">
6 pages, 636 KiB  
Case Report
Successful Treatment of MEK Inhibitor-Induced Paronychia in Neurofibromatosis with Photodynamic Therapy: A Case Report and Review of the Therapeutic Options
by Francesca Ambrogio, Teresa Perillo, Domenico Bonamonte, Aurora De Marco, Benedetta Tirone, Carmelo Laface, Gerardo Cazzato, Caterina Foti and Edoardo Mortato
J. Clin. Med. 2025, 14(4), 1104; https://doi.org/10.3390/jcm14041104 - 9 Feb 2025
Viewed by 390
Abstract
Background/Objectives: Selumetinib, a MEK1/2 inhibitor, is commonly used for treating neurofibromatosis type 1 (NF1) and is associated with cutaneous side effects such as paronychia and periungual granulomas. These complications can be painful and difficult to manage, often leading to the discontinuation of treatment. [...] Read more.
Background/Objectives: Selumetinib, a MEK1/2 inhibitor, is commonly used for treating neurofibromatosis type 1 (NF1) and is associated with cutaneous side effects such as paronychia and periungual granulomas. These complications can be painful and difficult to manage, often leading to the discontinuation of treatment. The objective of this study was to evaluate the effectiveness of photodynamic therapy (PDT) as a novel treatment for MEKi-induced paronychia in a patient with NF1. Methods: We present a case report of an 18-year-old patient with NF1 who developed painful periungual granulomas on the toenails after 12 months of Selumetinib therapy. PDT was administered using methyl aminolevulinate (METVIX®) as the photosensitizing agent, followed by treatment with a red LED light source (630 nm, 37 J/cm2 for 8 min and 30 s). The patient was followed up for two months post-treatment and then at two years. Results: After a single PDT session, the patient exhibited complete clinical remission of the periungual granulomas and associated pain. No recurrence of the lesions was noted during the two-year follow-up. The patient tolerated the procedure well, reporting only mild discomfort during treatment. Conclusions: PDT appears to be an effective, minimally invasive treatment for Selumetinib-induced paronychia and periungual granulomas. This case demonstrates that PDT can provide a complete resolution of symptoms with a single treatment session, offering an alternative to more invasive procedures. Further studies with larger cohorts are needed to establish PDT as a standard treatment option for this condition. Full article
(This article belongs to the Special Issue Skin Diseases: From Diagnosis to Treatment)
Show Figures

Figure 1

Figure 1
<p>Paronychia with pyogenic periungual granulomas after Selumetinib treatment for neurofibromatosis.</p>
Full article ">Figure 2
<p>After one session of PDT, the lesions were completely remitted.</p>
Full article ">
23 pages, 4635 KiB  
Article
Spontaneous Necrosis of a High-Risk Bladder Tumor Under Immunotherapy for Concurrent Malignant Melanoma: Role of BRAF Mutations and PD-L1 Expression
by Cristian Condoiu, Mihael Musta, Alin Adrian Cumpanas, Razvan Bardan, Vlad Dema, Flavia Zara, Cristian Silviu Suciu, Cristina-Stefania Dumitru, Andreea Ciucurita, Raluca Dumache, Hossam Ismail and Dorin Novacescu
Biomedicines 2025, 13(2), 377; https://doi.org/10.3390/biomedicines13020377 - 5 Feb 2025
Viewed by 666
Abstract
Background: Bladder cancer (BC) is a heterogeneous malignancy, and predicting response to immune checkpoint inhibitors (ICIs) remains a challenge. Herein, we investigate a high-risk bladder tumor, which developed during anti-BRAF/MEK therapy for a concurrent advanced BRAF-V600E-positive malignant melanoma (MM) and subsequently underwent [...] Read more.
Background: Bladder cancer (BC) is a heterogeneous malignancy, and predicting response to immune checkpoint inhibitors (ICIs) remains a challenge. Herein, we investigate a high-risk bladder tumor, which developed during anti-BRAF/MEK therapy for a concurrent advanced BRAF-V600E-positive malignant melanoma (MM) and subsequently underwent complete spontaneous necrosis following Nivolumab immunotherapy, only to recur thereafter while still under the same treatment. This unique scenario provided an opportunity to investigate the roles of BRAF gene mutations in BC pathogenesis, respectively, of PD-L1 expression in immunotherapy response prediction. Methods: We retrospectively analyzed BC specimens obtained via transurethral resection at two critical time-points: prior to the complete spontaneous necrosis under Nivolumab (prenecrosis) and after tumor recurrence postnecrosis (postnecrosis). The BRAF gene mutation status was evaluated using quantitative polymerase chain reaction (qPCR). PD-L1 expression was assessed by immunohistochemistry (IHC), quantified using the combined positive score (CPS), and a cutoff of ≥10 for positivity. Results: Neither pre- nor postnecrosis BC samples harbored BRAF gene mutations. Prenecrosis PD-L1 expression (CPS = 5) indicated a minimal likelihood of response to immunotherapy. However, complete spontaneous necrosis occurred under Nivolumab, followed by recurrence with further reduced PD-L1 expression (CPS = 1). Conclusions: The complete BC regression challenges the conventional role of PD-L1 as a sole predictive biomarker for immunotherapy. This study also highlights the potential role of BRAF/MEK inhibitors in BC oncogenesis and underscores the need for alternative biomarkers, such as tumor mutation burden (TMB) and circulating tumor DNA (ctDNA), to guide treatment selection in BC better. Full article
Show Figures

Figure 1

Figure 1
<p>Imaging at initial presentation, i.e., details from contrast-enhanced computer tomography of thorax and abdomen, arterial phase, demonstrating malignant melanoma clinical stage at diagnosis (pT4b cN2 M1lym): (<b>A</b>) coronal view, primary lesion (red circle) in the right subscapular area; (<b>B</b>) axial view, right axillary adenopathic block (yellow circle); (<b>C</b>) axial view, large upper mediastinal adenopathy (green circle); (<b>D</b>) coronal view, right hilar and peribronchial adenopathic block (blue square).</p>
Full article ">Figure 2
<p>Contrast-enhanced computer tomography scan of the pelvis, axial view, and excretory phase (15 min), showing a voluminous recurrence of the bladder tumor during combined targeted chemotherapy (anti-BRAF/MEK) for malignant melanoma.</p>
Full article ">Figure 3
<p>Microscopic findings in prespontaneous bladder tumor necrosis tissue fragments obtained through transurethral resection (biopsy): (<b>A</b>) 200×, HE, nests of neoplastic urothelial cells in the deep portion of the proliferation, showing focal invasion of the submucosa; (<b>B</b>) 200×, HE, high-grade traits, G2 cellularity, in the superficial portion of the proliferation, with some mitotic activity; (<b>C</b>) 200×, HE, intratumoral vascular elements in the superficial portion of the proliferation; (<b>D</b>) 200×, IHC with anti-PD-L1 (Dako clone 22C3), moderate to intense staining reaction in tumor cells, and isolated infiltrating immune cells (CPS = 5%); (<b>E</b>) 200×, HE, high-grade area with abundant vascularity and tumor-associated inflammatory cells; (<b>F</b>) 200×, IHC with anti-PD-L1 (Dako clone 22C3), moderate to intense staining reaction in tumor cells and tumor-associated inflammatory cells (CPS = 5%).</p>
Full article ">Figure 4
<p>Microscopic findings in postspontaneous bladder tumor necrosis relapse tissue fragments obtained through transurethral resection: (<b>A</b>) 200×, HE, urothelial carcinoma proliferation, invading the basal membrane and superficial submucosa focally, while associating a significant submucosal desmoplastic reaction; (<b>B</b>) 200×, IHC with anti-PD-L1 (Dako clone 22C3), a moderate color reaction in tumor-associated inflammatory cells (CPS = 1); (<b>C</b>) 200×, HE, urothelial carcinoma proliferation, with abundant vascularization and a microcalcification focus; (<b>D</b>) 200×, IHC with anti-PD-L1 (Dako clone 22C3), a very weak color reaction in rare tumor-associated inflammatory cells (CPS = 1); (<b>E</b>) 200×, HE, urothelial carcinoma with a classic papillary-type growth pattern; (<b>F</b>) 200×, IHC with anti-PD-L1 (Dako clone 22C3), a weak color reaction in scarce tumor cells and a few tumor-associated inflammatory cells (CPS = 1).</p>
Full article ">
22 pages, 20383 KiB  
Article
Inhibition of MEK1/2 Signaling Pathway Limits M2 Macrophage Polarization and Interferes in the Dental Socket Repair Process in Mice
by Angélica Cristina Fonseca, Priscila Maria Colavite, Michelle de Campos Soriani Azevedo, Daniela Carignatto Passadori, Jessica Lima Melchiades, Rafael Carneiro Ortiz, Camila Oliveira Rodini, Ana Paula Favaro Trombone and Gustavo Pompermaier Garlet
Biology 2025, 14(2), 107; https://doi.org/10.3390/biology14020107 - 21 Jan 2025
Viewed by 667
Abstract
Dental socket repair theoretically involves a constructive inflammatory immune response, which evolves from an initial M1 prevalence to a subsequent M2 dominance. In this scenario, the MEK1/2 signaling pathway is allegedly involved in M2 polarization. This study aimed to evaluate the impact of [...] Read more.
Dental socket repair theoretically involves a constructive inflammatory immune response, which evolves from an initial M1 prevalence to a subsequent M2 dominance. In this scenario, the MEK1/2 signaling pathway is allegedly involved in M2 polarization. This study aimed to evaluate the impact of MEK1/2 pharmacological inhibition in the local host response and repair outcome. C57Bl/6-WT 8-week-old male mice were submitted to the extraction of the right upper incisor and treated (or not, control group) with MEK1/2 inhibitor PD0325901 (10 mg/kg/24 h/IP, MEK1/2i group) and analyzed at 0, 3, 7, and 14 days using microcomputed tomography, histomorphometry, birefringence, immunohistochemistry, and PCR array analysis. The results demonstrate that MEK1/2 inhibition limits the development of M2 response over time, being associated with lower expression of M2, MSCs, and bone markers, lower levels of growth and osteogenic factors, along with a higher expression of iNOS, IL-1b, IL-6, and TNF-α, as well inflammatory chemokines, indicating a predominantly M1 pro-inflammatory environment. This modulation of local inflammatory immune response is associated with impaired bone formation as demonstrated by microtomographic and histomorphometric data. The results show that MEK1/2 inhibition delays bone repair after tooth extraction, supporting the concept that M2 macrophages are essential elements for host response regulation and proper repair. Full article
Show Figures

Figure 1

Figure 1
<p>Comparative molecular analysis (PCR array) in the dental socket repair process over the periods of 0, 3, 7, and 14 days after tooth extraction between control and MEK1/2i groups. Heat map to quantify the expression of the growth factors (BMPs, TGFβ, VEGFs, and FGFs), extracellular matrix markers (COL1a1, COL1a2, MMPs, TIMPs, and Serpine), bone markers (RUNX2, DMP1, ALPL, PHEX, SOST, CTSK, RANKL, RANK, and OPG), chemokines and their receptors (CCLs, CXCLs, CCRs and CXCRs), mesenchymal stem cell markers (CD34, 105, 106, 116, 146, OCT-4, and NANOG), cytokines (ILs, TNF, and IFNG), and markers associated with macrophage polarization (iNOS, ARG, GAPDH, ACTb, HPRT-1, and FIZZ). * (<span class="html-italic">p</span> &lt; 0.05) indicates statistically significant differences between the MEK1/2i and control groups.</p>
Full article ">Figure 2
<p>Comparative immunohistochemical analysis for anti-Ly6G5B/GR1, anti-CD3-e, anti-CD206, anti-CD16&amp;CD32 and anti-F4/80 antibody in the dental socket repair process over the periods of 0, 3, 7, and 14 days after tooth extraction between control and MEK1/2i groups. Photomicrographs are representative of the middle region of the dental socket, where there is the presence of a blood clot (BC), blood vessels (BV) and preservation of the dental socket cortex (closed arrows) immediately after tooth extraction, with its progressive replacement by a granulation tissue (GT) and beginning of bone neoformation (open arrow). In later periods we see the persistence of connective tissue (thin arrow), bone trabeculae in remodeling (O), and the varied presence of + cells (square). IHC staining: Scanned in the Aperio Scanscope CS instrument with a 40× objective and under the Leica MC170 optical microscope with 40× (Bar = 50 μm) and 100× objectives (Bar = 20.34 μm).</p>
Full article ">Figure 3
<p>Comparative analysis of the quantification of (<b>A</b>) Ly6G5B/Gr1+, (<b>B</b>) CD3-e+, (<b>C</b>) CD206+, (<b>D</b>) CD16&amp;CD32+, and (<b>E</b>) F4/80+ cells in absolute numbers. Different lowercase letters represent a statistically significant difference (<span class="html-italic">p</span> &lt; 0.05) between different time points within the same group; time points within the same group presenting the same letter are not statistically different.</p>
Full article ">Figure 4
<p>Three-dimensional morphological analysis by µCT of the dental socket repair process over the periods of 0, 3, 7, and 14 days after tooth extraction between control and MEK1/2i groups. The geometric figures drawn in the Control group represent the conformation of the dental sockets in different section planes. Coronal, sagittal, and transversal section planes are shown.</p>
Full article ">Figure 5
<p>Comparative analysis of morphological parameters of bone trabecular microarchitecture in the dental socket repair process over the periods of 0, 3, 7, and 14 days after tooth extraction between control and MEK1/2i groups. Bone trabecular analyses (<b>A</b>–<b>F</b>) included: (<b>A</b>) Bone volume (BV), (<b>B</b>) bone fraction in relation to total volume (BV/TV), (<b>C</b>) trabecular thickness (Tb.Th), (<b>D</b>) number of trabeculae (Tb.N), (<b>E</b>) mean distance between trabeculae (Tb.Sp), (<b>F</b>) total tissue volume (TV). The results represent the mean and standard deviation values in each of the analyzed periods. Different lowercase letters represent a statistically significant difference (<span class="html-italic">p</span> &lt; 0.05) between different time points within the same group; time points within the same group presenting the same letter are not statistically different. * (<span class="html-italic">p</span> &lt; 0.05) indicates statistically significant differences between the MEK1/2i group versus control group at the specified time point.</p>
Full article ">Figure 6
<p>Comparative panoramic histological analysis in the dental socket repair process over the periods of 0, 3, 7, and 14 days after tooth extraction between control and MEK1/2i groups. Photomicrographs are representative of the middle region of the dental socket, showing the presence of blood clot (BC), blood vessels (BV) and preservation of the dental socket cortex (closed arrows) immediately after tooth extraction, its progressive replacement by a granulation tissue (GT), and beginning of bone neoformation (open arrow). In later periods we see the persistence of connective tissue (thin arrow), osteoclasts promoting resorption (*), and bone trabeculae in remodeling (O). HE staining: Scanned on the Aperio Scanscope CS device with 10× (Bar = 400 μm) and 40× objectives (Bar = 50 μm).</p>
Full article ">Figure 7
<p>Comparative analysis of the total area density (%) occupied by (<b>A</b>) collagen fibers, (<b>B</b>) fibroblasts, (<b>C</b>) blood vessels, (<b>D</b>) inflammatory cells, (<b>E</b>) bone matrix, (<b>F</b>) osteoblasts, (<b>G</b>) osteoclasts, (<b>H</b>) other structures, (<b>I</b>) blood clot in the dental socket repair process over the periods of 0, 3, 7, and 14 days after tooth extraction between control and MEK1/2i groups. The results represent the mean and standard deviation values of the analyzed period. Different lowercase letters represent a statistically significant difference (<span class="html-italic">p</span> &lt; 0.05) between different time points within the same group; time points within the same group presenting the same letter are not statistically different. * (<span class="html-italic">p</span> &lt; 0.05) indicates statistically significant differences between the MEK1/2i and control group at the specified time point.</p>
Full article ">Figure 8
<p>Comparative birefringence analysis of collagen fibers in the dental socket repair process over the periods of 0, 3, 7, and 14 days after tooth extraction between control and MEK1/2i groups. Photomicrographs are representative of the middle region of dental socket, captured under conventional light and polarized light. Green birefringence color indicates thin fibers; yellow and red colors in the birefringence analysis indicate thick collagen fibers. Picrosirius red coloration; objective of 10×; Bar = 200 μm.</p>
Full article ">Figure 9
<p>Comparative analysis of the total area of collagen fibers ((<b>A</b>)—pixel<sup>2</sup>) and the distinction in percentage of the degree of maturation of collagen fibers in green, yellow, red birefringence colors ((<b>B</b>)—%) in the dental socket repair process over the periods of 0, 3, 7, and 14 days after tooth extraction between control and MEK1/2i groups. Results are presented as the mean (±SEM) of percentage or pixels<sup>2</sup> for each color in the birefringence. Different lowercase letters represent a statistically significant difference (<span class="html-italic">p</span> &lt; 0.05) between different time points within the same group; time points within the same group presenting the same letter are not statistically different. * (<span class="html-italic">p</span> &lt; 0.05) indicates statistically significant differences between the MEK1/2i and control group at the specified time point.</p>
Full article ">
42 pages, 2925 KiB  
Review
Detection of Circulating Tumor DNA in Liquid Biopsy: Current Techniques and Potential Applications in Melanoma
by Clara Martínez-Vila, Cristina Teixido, Francisco Aya, Roberto Martín, Europa Azucena González-Navarro, Llucia Alos, Natalia Castrejon and Ana Arance
Int. J. Mol. Sci. 2025, 26(2), 861; https://doi.org/10.3390/ijms26020861 - 20 Jan 2025
Viewed by 1131
Abstract
The treatment landscape for advanced melanoma has transformed significantly with the advent of BRAF and MEK inhibitors (BRAF/MEKi) targeting BRAFV600 mutations, as well as immune checkpoint inhibitors (ICI) like anti-PD-1 monotherapy or its combinations with anti-CTLA-4 or anti-LAG-3. Despite that, many patients [...] Read more.
The treatment landscape for advanced melanoma has transformed significantly with the advent of BRAF and MEK inhibitors (BRAF/MEKi) targeting BRAFV600 mutations, as well as immune checkpoint inhibitors (ICI) like anti-PD-1 monotherapy or its combinations with anti-CTLA-4 or anti-LAG-3. Despite that, many patients still do not benefit from these treatments at all or develop resistance mechanisms. Therefore, prognostic and predictive biomarkers are needed to identify patients who should switch or escalate their treatment strategies or initiate an intensive follow-up. In melanoma, liquid biopsy has shown promising results, with a potential role in predicting relapse in resected high-risk patients or in disease monitoring during the treatment of advanced disease. Several components in peripheral blood have been analyzed, such as circulating tumor cells (CTCs), cell-free DNA (cfDNA), and circulant tumoral DNA (ctDNA), which have turned out to be particularly promising. To analyze ctDNA in blood, different techniques have proven to be useful, including digital droplet polymerase chain reaction (ddPCR) to detect specific mutations and, more recently, next-generation sequencing (NGS) techniques, which allow analyzing a broader repertoire of the mutation landscape of each patient. In this review, our goal is to update the current understanding of liquid biopsy, focusing on the use of ctDNA as a biological material in the daily clinical management of melanoma patients, in particular those with advanced disease treated with ICI. Full article
(This article belongs to the Special Issue Molecular Basis and Progress of Immunotherapy for Melanoma)
Show Figures

Figure 1

Figure 1
<p>Flow chart on how to use ctDNA in clinical decision-making in melanoma patients.</p>
Full article ">Figure 2
<p>ctDNA in melanoma patients: tumor-derived components in peripheral blood, DNA-based alterations and analytical sensitivity from current techniques for ctDNA analysis.</p>
Full article ">Figure 3
<p>BEAMing, ddPCR and NGS schematic flowchart.</p>
Full article ">
17 pages, 4024 KiB  
Article
Two Cysteines in Raf Kinase Inhibitor Protein Make Differential Contributions to Structural Dynamics In Vitro
by Hyun Sang Cho, Mohammad Faysal Al Mazid, Eun-Young Lee, Md Abu Rayhan, Hyoun Sook Kim, Byung Il Lee and Hye Jin You
Molecules 2025, 30(2), 384; https://doi.org/10.3390/molecules30020384 - 17 Jan 2025
Viewed by 506
Abstract
As a scaffolding protein, Raf kinase binding protein (RKIP) is involved in a variety of cellular pathways, including the Raf–MEK–ERK-cascade. It acts as a negative regulator by binding to its partners, making it an attractive target in the development of therapeutic strategies for [...] Read more.
As a scaffolding protein, Raf kinase binding protein (RKIP) is involved in a variety of cellular pathways, including the Raf–MEK–ERK-cascade. It acts as a negative regulator by binding to its partners, making it an attractive target in the development of therapeutic strategies for cancer. Despite its structural stability as a monomer, RKIP may form a dimer, resulting in the switching of binding partners. It is still unclear how RKIP switches between monomeric and dimeric forms. Here, we identified the role of cysteine 133 in RKIP structural dynamics using recombinant human RKIP (rhRKIP) proteins purified from Escherichia coli BL21(DE3) cells. Mutation of alanine or serine instead of cysteine in RKIP proteins did not affect the biochemical characteristics, while dynamic light scattering and liquid chromatography (LC) quadrupole time-of-flight (Q-TOF) mass spectrometry (MS) suggested distinct peaks in solution, which were identified via LC–MS/MS analyses, and further clarified the role of cysteine in RKIP dimerization. rhRKIP dimer formation was abrogated by a 32-aa peptide mimicking the region between two RKIP proteins for dimerization. In addition, the 32-aa peptide and its short derivatives were investigated for effects on cancer cell viability. Taken together, our findings suggest that it may be possible to regulate RKIP function by controlling its dynamics with reducing agents, which could aid the targeting of cancer cells. Full article
Show Figures

Figure 1

Figure 1
<p>Preparation of recombinant human (rh) RKIP proteins and structural dynamics depending on reducing agents. (<b>A</b>) Purified rhRKIP protein samples obtained by affinity chromatography were analyzed by circular dichroism to examine their secondary structure. (<b>B</b>) rhRKIP proteins were prepared with sample loading buffer, with or without β-mercaptoethanol, and separated by SDS-PAGE. Gels were stained directly with Coomassie Brilliant Blue (<b>left</b>) or analyzed by immunoblotting using an anti-His antibody (<b>right</b>). (<b>C</b>) rhRKIP proteins were analyzed by dynamic light scattering (DLS) for molecular weight and homogeneity. Wild-type (WT) rhRKIP proteins showed two peaks (R1 and R2) corresponding to the molecular weights of the monomer (R1) and dimer or higher-order structures (R2) (<b>left</b>), with peak intensities affected by the presence of DTT (<b>right</b> graph). (<b>D</b>) Relative peak intensities for each measurement are expressed as the mean ± SE from at least three independent experiments. Statistical significance was assessed using the paired Student’s <span class="html-italic">t</span> test (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.0001). (<b>E</b>) The predicted model structure of the RKIP dimer was visualized using UCSF ChimeraX (version 1.9) (<b>top</b>). The dimer interface (highlighted in the red box) was magnified to display detailed molecular interactions at 10 ns, 30 ns, and 50 ns during the 50 ns simulation (<b>bottom</b>). The distances between amino acids at the interfaces were measured at each time point (highlighted in the black box). Gray dotted line for physical contact (closest pair of atoms, predicted to contact physically); blue dotted line for H-bond. (<b>F</b>) The dynamics of the rhRKIP dimer model were calculated every 2 ps over a 50 ns simulation using the GROMACS platform (version 2024.0), three times. The analysis included RMSD, distance, number of hydrogen bonds (H-bonds), interaction energy, potential energy, and total energy. The root mean square deviation (RMSD) was used to assess differences in conformation of the protein dimer models. The distance between molecules within the predicted RKIP dimers was measured in angstroms (Å). Number of H-bonds was analyzed, and interaction energy was determined as the sum of van der Waals and Coulomb energies of the predicted RKIP dimers. The data presented represent the average values of three replicas and reflect the most consistent results obtained from at least three independent simulations using different dimer models.</p>
Full article ">Figure 2
<p>Phylogenetic tree and conservation of RKIP primary structure, and generation and circular dichroism characterization of rhRKIP mutant proteins. (<b>A</b>,<b>B</b>) Amino acid sequences of RKIP (PEBP1, UniProt ID: P30086) from various species were retrieved from the UniProt database (<a href="https://www.uniprot.org/" target="_blank">https://www.uniprot.org/</a>, 3 June 2022) and subjected to alignment and phylogenetic analysis. (<b>B</b>) Amino acid residues 121–180 of RKIP from eight species were aligned, highlighting conserved cysteine residues at positions 133 and 168 and a serine residue at position 153 (arrowheads). (<b>C</b>) Schematic representations of plasmid constructs encoding mutant rhRKIP proteins. (<b>D</b>) Wild-type (WT) and mutant rhRKIP proteins were expressed, purified for in vitro studies, resolved by SDS-PAGE, and visualized using Coomassie Brilliant Blue. (<b>E</b>) Purified rhRKIP WT and mutant proteins were analyzed by circular dichroism to examine their secondary structures. The graph depicts the circular dichroism spectra of seven rhRKIP WT and mutant proteins: black solid line, WT; light brown line, C133A; red line, C168A; green line, C133S; blue line, C168S; violet line, C133A and C168A; pink line, C133S and C168S. (<b>F</b>) Based on the circular dichroism data, the secondary structures of the proteins were categorized into turns, various β-sheet types (left-twisted, relaxed, and right-twisted), and different α-helices (regular and distorted), along with other structural elements. The proportions of these structures within the rhRKIP proteins are represented as pie charts.</p>
Full article ">Figure 3
<p>Validation of rhRKIP dimers by Q-TOF analysis based on cysteine at positions 133 and 168. Purified rhRKIP WT and mutant proteins with substitutions of Cys133 or Cys168 were subjected to Q-TOF analysis to detect high-molecular-weight RKIP dimers in addition to monomeric RKIPs. (<b>A</b>) Mass spectra of all rhRKIP WT and mutant proteins are shown in two regions: ~22 kDa (monomeric state, <b>top</b>) and ~44 kDa (high molecular weight dimer, <b>bottom</b>). The lines in the spectra are as follows: blue, WT; violet, C133A; red, C168A; orange, C133S; green, C168S; turquoise, C133A and C168A; sky blue, C133S and C168S. (<b>B</b>) The relative mass spectra of each rhRKIP protein are presented. The intensities of dimeric proteins relative to monomeric proteins are shown. Red arrows indicate high molecular weight proteins in the mass spectra, where detected.</p>
Full article ">Figure 4
<p>Identification of dimer-specific regions in the RKIP dimer in a cysteine-133-dependent manner using mass spectrometry. A simplified workflow for identifying dimer-specific regions (peptide sequences) in the presence or absence of DTT is shown (<b>top</b>). Purified rhRKIP WT and four mutant proteins were separated by electrophoresis, subjected to in-gel digestion with trypsin with or without DTT, and analyzed by mass spectrometry for peptide identification. With the exception of rhRKIP C133A and C133S mutants, rhRKIP proteins showed DTT-dependent mass peaks and peptides associated with cysteine at position 133 (<b>bottom</b>). Gray arrowheads: trypsin cleavage sites.</p>
Full article ">Figure 5
<p>Inhibition of rhRKIP dimer formation by peptides mimicking the region near cysteine 133. Purified RKIP proteins were analyzed by electrophoresis in the absence of the reducing agent β-mercaptoethanol (<b>A</b>,<b>B</b>). (<b>A</b>) All rhRKIP WT and mutant proteins were separated by electrophoresis and stained with Coomassie Brilliant Blue to visualize protein bands. Data shown are representative results from at least three independent experiments. (<b>B</b>) Purified rhRKIP WT, C133A, and C168A proteins were incubated with peptide-32 for 30 min, then subjected to electrophoresis in the absence of reducing agents and visualized using Coomassie Brilliant Blue. Peptide-32 was added at varying molar ratios to the proteins (50:1, 25:1, 10:1, 5:1, 1:1). Data shown are representative results of at least three independent experiments. (<b>C</b>) Band intensities of dimers, monomers, and peptide-conjugated monomers (top of the monomer band region) were quantified using ImageJ software (version 1.54m). Relative intensities were calculated against the total protein (dimers, monomers, and peptide-conjugated monomers) and expressed as graphs using GraphPad Prism v.9 (GraphPad Software LLC, Boston, MA, USA). Data are presented as the mean ± SE from at least three independent experiments. Statistical significance was assessed using the paired Student’s <span class="html-italic">t</span> test. (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 6
<p>Peptides targeting the region near cysteine 133 may be beneficial for anticancer therapy. (<b>A</b>) Various peptides encompassing cysteine 133 and its neighboring regions were synthesized and analyzed. (<b>B</b>) rhRKIP WT proteins were incubated with various peptides at a molar ratio of 1:50 (rhRKIP:peptide) for 30 min at 37 °C in the absence of any reducing agent and subjected to SDS-PAGE. Gels were either stained directly with Coomassie Brilliant Blue (<b>left</b>) or analyzed by immunoblotting using an anti-RKIP antibody (<b>right</b>). (<b>C</b>) HCT116 cells (3 × 10<sup>3</sup>) were seeded in 96-well plates and incubated for 24 h, followed by treatment with peptides (20 μM) for 48 h. Cell viability was quantified by MTT assay. Peptide 1–22 was soluble only in DMSO, which served as a control. Relative cell viability compared to vehicle alone (PBS or DMSO) is presented as the mean ± SE from at least three independent experiments. Statistical significance was assessed using the paired Student’s <span class="html-italic">t</span> test. (<b>D</b>) HCT116 cells (3 × 10<sup>3</sup>) were seeded in 96-well plates and incubated for 24 h, followed by treatment with peptide 1–22 (20 μM), trametinib, or their combination for 48 h. Cell viability was quantified by MTT assay. Relative cell viability compared to the vehicle is presented as the mean ± SE from at least three independent experiments. Statistical significance was assessed using the paired Student’s <span class="html-italic">t</span> test. (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">
15 pages, 480 KiB  
Article
A Mixed Methods Study of Medication Adherence in Adults with Neurofibromatosis Type 1 (NF1) on a Clinical Trial of Selumetinib
by Millicent S. Curlee, Mary Anne Toledo-Tamula, Melissa Baker, Daniel Wikstrom, Cynthia Harrison, Amanda Rhodes, Margaret Fagan, Cecilia Tibery, Pamela L. Wolters, Brigitte C. Widemann, Andrea M. Gross and Staci Martin
Cancers 2025, 17(2), 295; https://doi.org/10.3390/cancers17020295 - 17 Jan 2025
Viewed by 901
Abstract
Background: Oral therapeutic options for plexiform neurofibromas (PNs) in individuals with neurofibromatosis type 1 (NF1) are receiving attention in clinical research. The MEK inhibitor (MEKi) Selumetinib is FDA-approved in children ages 2+ years with inoperable PNs, and shows activity in adults. Prolonged therapy [...] Read more.
Background: Oral therapeutic options for plexiform neurofibromas (PNs) in individuals with neurofibromatosis type 1 (NF1) are receiving attention in clinical research. The MEK inhibitor (MEKi) Selumetinib is FDA-approved in children ages 2+ years with inoperable PNs, and shows activity in adults. Prolonged therapy with selumetinib is necessary to maintain tumor reduction. Therefore, investigating long-term adherence is vital to understand patterns of adherence over time and its impact on clinical outcomes. Mixed methods research offers rich information about adherence that can inform future intervention trials, and can assist practitioners in addressing medication adherence concerns. Methods: This mixed-method pilot study is the first examination of the feasibility of a technology-based adherence assessment method, the medication events monitoring system (MEMSTM), among individuals with NF1-PN. Adherence was monitored in a small sample of patients (N = 12; mean age = 34.36 years; 58% male) with NF1 and PN across eighteen 28-day treatment cycles. Qualitative data were obtained from individual interviews using inductive and deductive techniques for thematic analysis. Results: The predetermined criterion was met, suggesting that using MEMSTM is feasible despite some challenges with the caps. Depression and overall stress were significantly related to reduced adherence, although these results should be considered hypothesis-generating. Barriers to medication adherence included forgetting and the timing of doses related to eating. Facilitators included consistency, reminders, and social support. Conclusions: This study highlights patient characteristics that may be related to increased risk for nonadherence, as well as challenges with electronic pill caps that should be considered in future clinical trials for NF1-related PN. Results can inform future adherence interventions for adults with NF1 and PNs. Future research with larger samples is needed to fully explore factors related to long-term medication adherence among individuals with NF1. Full article
(This article belongs to the Special Issue Neurofibromatosis)
Show Figures

Figure 1

Figure 1
<p>Adherence rates across three assessment methods.</p>
Full article ">
27 pages, 1830 KiB  
Review
Rat Sarcoma Virus Family Genes in Acute Myeloid Leukemia: Pathogenetic and Clinical Implications
by Shaimaa Khattab, Adriatik Berisha, Natalia Baran and Pier Paolo Piccaluga
Biomedicines 2025, 13(1), 202; https://doi.org/10.3390/biomedicines13010202 - 15 Jan 2025
Viewed by 786
Abstract
Acute myeloid leukemias (AMLs) comprise a group of genetically heterogeneous hematological malignancies that result in the abnormal growth of leukemic cells and halt the maturation process of normal hematopoietic stem cells. Despite using molecular and cytogenetic risk classification to guide treatment decisions, most [...] Read more.
Acute myeloid leukemias (AMLs) comprise a group of genetically heterogeneous hematological malignancies that result in the abnormal growth of leukemic cells and halt the maturation process of normal hematopoietic stem cells. Despite using molecular and cytogenetic risk classification to guide treatment decisions, most AML patients survive for less than five years. A deeper comprehension of the disease’s biology and the use of new, targeted therapy approaches could potentially increase cure rates. RAS oncogene mutations are common in AML patients, being observed in about 15–20% of AML cases. Despite extensive efforts to find targeted therapy for RAS-mutated AMLs, no effective and tolerable RAS inhibitor has received approval for use against AMLs. The frequency of RAS mutations increases in the context of AMLs’ chemoresistance; thus, novel anti-RAS strategies to overcome drug resistance and improve patients’ therapy responses and overall survival are the need of the hour. In this article, we aim to update the current knowledge on the role of RAS mutations and anti-RAS strategies in AML treatments. Full article
(This article belongs to the Special Issue Molecular Research on Acute Myeloid Leukemia (AML) Volume II)
Show Figures

Figure 1

Figure 1
<p>Schematic of Ras signaling and its involvement in myeloid malignancies. Ras proteins play a critical role in mediating intracellular signaling in response to extracellular growth factors by toggling between inactive GDP-bound and active GTP-bound states. Ras activation is initiated when growth factor receptors, upon ligand binding, assemble a complex involving adapter proteins, the phosphatase SHP-2, and guanine nucleotide exchange factors (GEFs) like SOS. These GEFs promote the exchange of GDP for GTP on Ras, leading to its activation. Once activated, Ras-GTP interacts with a variety of downstream signaling proteins, initiating a cascade of signaling events that regulate vital cellular processes such as gene expression (β-catenin, FOXO, NF-κB, p53, ELK1, AP1), translation (eIF4E, S6), and apoptosis (BAD). The Ras signaling pathway is tightly controlled through the hydrolysis of GTP to GDP, a process facilitated by GTPase-activating proteins (GAPs) like p120GAP and neurofibromin, which turn off Ras activity. Mutations in Ras or its regulators are implicated in various malignancies, including myeloid cancers, with specific mutations being noted. GF, growth factor; GEF, guanine nucleotide exchange factor; GAP, p120 GTPase-activating protein; NF1, neurofibromin; MPD, myeloproliferative disorders; PDGFR, platelet-derived growth factor receptor; CMML, chronic myelomonocytic leukemia; CML, chronic myelogenous leukemia; mTOR, mammalian target of rapamycin (adapted from [<a href="#B7-biomedicines-13-00202" class="html-bibr">7</a>]).</p>
Full article ">Figure 2
<p>Prevalence of NRAS and KRAS mutations in AML. Current research indicates that NRAS mutations are significantly more prevalent in AML patients than KRAS mutations. NRAS mutations are found in approximately 11–12% of cases, whereas KRAS mutations are reported in about 5% of cases. The error bars represent the range of the highest and lowest prevalence rates reported in the literature [<a href="#B8-biomedicines-13-00202" class="html-bibr">8</a>,<a href="#B21-biomedicines-13-00202" class="html-bibr">21</a>,<a href="#B22-biomedicines-13-00202" class="html-bibr">22</a>,<a href="#B23-biomedicines-13-00202" class="html-bibr">23</a>,<a href="#B24-biomedicines-13-00202" class="html-bibr">24</a>,<a href="#B25-biomedicines-13-00202" class="html-bibr">25</a>].</p>
Full article ">Figure 3
<p>Schematic Representation of the Most Common Mutations Affecting KRAS and NRAS. This schematic illustrates the most common mutations in KRAS, NRAS, and RRAS2, highlighting the involvement of key functional gene domains (adapted from [<a href="#B79-biomedicines-13-00202" class="html-bibr">79</a>]).</p>
Full article ">Figure 4
<p>Novel and ongoing therapeutic strategies targeting RAS. This figure illustrates key therapeutic approaches for targeting RAS, focusing on direct RAS inhibitors, MEK inhibitors, PI3K inhibitors, and PLK1 inhibitors. The RAS signaling cascade is activated when growth factors bind to receptor tyrosine kinases (RTKs), leading to the exchange of GDP for GTP via GEFs like SOS. Active RAS then triggers downstream signaling through RAF and PI3K, regulating processes like cell proliferation and survival. Targets shown in green are in clinical trials for hematologic diseases, those in blue are FDA-approved for oncology, and preclinical/clinical drugs targeting RAS mutations in solid tumors are marked in black and red. Additional strategies include PROTACs and siRNA. Four essential therapeutic axes are emphasized: direct RAS inhibitors, MEK inhibitors, PI3K inhibitors, and PLK1 inhibitors (adapted from [<a href="#B13-biomedicines-13-00202" class="html-bibr">13</a>]).</p>
Full article ">
41 pages, 3315 KiB  
Review
Molecular Insights on Signaling Cascades in Breast Cancer: A Comprehensive Review
by Venketesh K. Panda, Barnalee Mishra, Samikshya Mahapatra, Biswajit Swain, Diksha Malhotra, Suryendu Saha, Sinjan Khanra, Priyanka Mishra, Sambhunath Majhi, Kavita Kumari, Angitha N. Nath, Swarnali Saha, Sarmistha Jena and Gopal C. Kundu
Cancers 2025, 17(2), 234; https://doi.org/10.3390/cancers17020234 - 13 Jan 2025
Viewed by 1337
Abstract
The complex signaling network within the breast tumor microenvironment is crucial for its growth, metastasis, angiogenesis, therapy escape, stem cell maintenance, and immunomodulation. An array of secretory factors and their receptors activate downstream signaling cascades regulating breast cancer progression and metastasis. Among various [...] Read more.
The complex signaling network within the breast tumor microenvironment is crucial for its growth, metastasis, angiogenesis, therapy escape, stem cell maintenance, and immunomodulation. An array of secretory factors and their receptors activate downstream signaling cascades regulating breast cancer progression and metastasis. Among various signaling pathways, the EGFR, ER, Notch, and Hedgehog signaling pathways have recently been identified as crucial in terms of breast cancer proliferation, survival, differentiation, maintenance of CSCs, and therapy failure. These receptors mediate various downstream signaling pathways such as MAPK, including MEK/ERK signaling pathways that promote common pro-oncogenic signaling, whereas dysregulation of PI3K/Akt, Wnt/β-catenin, and JAK/STAT activates key oncogenic events such as drug resistance, CSC enrichment, and metabolic reprogramming. Additionally, these cascades orchestrate an intricate interplay between stromal cells, immune cells, and tumor cells. Metabolic reprogramming and adaptations contribute to aggressive breast cancer and are unresponsive to therapy. Herein, recent insights into the novel signaling pathways operating within the breast TME that aid in their advancement are emphasized and current developments in practices targeting the breast TME to enhance treatment efficacy are reviewed. Full article
(This article belongs to the Special Issue Cell Migration and Invasion in Cancer)
Show Figures

Figure 1

Figure 1
<p>Various cell signaling cascades involved in breast cancer. The major signaling events, including Wnt/β-catenin, TGF-β, Notch, MAPK, Hedgehog, JAK/STAT, PI3K/Akt/mTOR, and NF-κB pathways, and their involvement in the regulation of tumor progression, survival, and metastasis. Wnt/β-catenin pathway is responsible for enhancing the stemness and promoting chemoresistance. TGF-β-mediated signaling is associated with the growth, invasion, and metastasis of breast cancer. Notch pathway as well as Hedgehog signaling induce chemoresistance and metastasis. JAK/STAT and NF-κB signaling cascades modulate breast cancer growth, invasion, metastasis, and angiogenesis. VEGFR-, HER2-, and EGFR-mediated PI3K/Akt/mTOR, MAPK, and ERK pathways have a key role in the regulation of breast tumor growth, metastasis, angiogenesis, and apoptosis.</p>
Full article ">Figure 2
<p>The key signaling and transcription factors associated with energy metabolism networks that serve as the foundation for metabolic reprogramming in the progression of breast cancer.</p>
Full article ">Figure 3
<p>Culmination of signaling pathways in the tumor stroma microenvironment involving cancer-associated fibroblasts (CAFs), adipocytes, and mast cells. CAFs release growth factors like TNF, SDF-1, VEGF, EGF, FGF2, and IGF, resulting in breast cancer progression. Secretion of several MMPs by CAFs such as MMP1, MMP2, MMP3, MMP7, MMP9, MMP13, and MMP14 are responsible for breast cancer advancement. Cytokines and chemokines released from CAFs include IL4, IL6, IL8, CXCL1, CXCL3, CXCL16, and CXCL8, which enhance breast cancer cell motility and aggressiveness. CAAs secrete inflammatory adipokines such as TNFα, leptin, CCL2, CCL5, and IL6, which modulate breast cancer progression. Mast cells contribute to angiogenesis in breast cancer by releasing growth factors such as VEGF, FGF2, and PDGF, along with proteases such as tryptases and chymases.</p>
Full article ">Figure 4
<p>Schematic representation of various TAM subsets based on their molecular signatures and secretory factors.</p>
Full article ">Figure 5
<p>Illustration depicting molecular crosstalk between TAMs and cancer cells via various signaling pathways in promoting breast tumor development and metastasis.</p>
Full article ">Figure 6
<p>Representative molecular targets in breast cancer, along with the corresponding immunotherapeutic and targeted treatment approach. The GPCR-mediated AKT/PI3K/mTOR signaling pathway is a major target for inhibitors such as alpelisib, buparlisib, capivasertib, ipatasertib, everolimus, and sapanisertib, used to suppress breast tumor growth. Inhibitors including trametinib and ONC201 target the EGFR-activated MEK/ERK signaling pathway. Targeting HER2 and VEGF with their respective monoclonal antibodies, trastuzumab and bevacizumab, leads to the inhibition of cell proliferation and angiogenesis. Tarextumab, a potent inhibitor of the Notch receptor, along with MK0752 and DAPT, that target γ-secretase, inactivate the Notch signaling pathway. Palbociclib, ribociclib, and abemaciclib are the CDK4/6 inhibitors that induce cell cycle arrest by suppression of CDK4/6-mediated signaling. Targeting ER with hormone therapy like tamoxifen and aromatase inhibitors can be a key strategy for treating hormone receptor-positive breast cancer. Immune checkpoint inhibitors such as pembrolizumab, nivolumab, cemiplimab, avelumab, atezolizumab, durvalumab, tremelimumab, and ipilimumab block PD-1, PD-L1, and CTLA-4, respectively, act as effective immunotherapeutic drugs.</p>
Full article ">
23 pages, 4232 KiB  
Article
MYC Overexpression Enhances Sensitivity to MEK Inhibition in Head and Neck Squamous Cell Carcinoma
by Cuicui Yang, Xiaowu Pang, Shaolei Teng, Shamel Wilson, Xinbin Gu and Guiqin Xie
Int. J. Mol. Sci. 2025, 26(2), 588; https://doi.org/10.3390/ijms26020588 - 12 Jan 2025
Viewed by 693
Abstract
MEK inhibitors, such as trametinib, have shown therapeutic potential in head and neck squamous cell carcinoma (HNSCC). However, the factors influencing cancer cell sensitivity and resistance to MEK inhibition remain poorly understood. In our study, we observed that MEK inhibition significantly reduced the [...] Read more.
MEK inhibitors, such as trametinib, have shown therapeutic potential in head and neck squamous cell carcinoma (HNSCC). However, the factors influencing cancer cell sensitivity and resistance to MEK inhibition remain poorly understood. In our study, we observed that MEK inhibition significantly reduced the expression of MYC, a transcription factor critical for the therapeutic response. MYC overexpression markedly enhanced the sensitivity of HNSCC cells to trametinib, as evidenced by delayed wound healing and reduced colony formation. Cell cycle analysis revealed that trametinib induced a G1 phase arrest, whereas MYC overexpression accelerated cell cycle progression, with a reduced induction of p27 and p21 and diminished decreases in E2F1 and phospho-Ser2/5 levels. Flow cytometry and protein analyses demonstrated that MYC overexpression amplified trametinib-induced apoptosis and DNA damage, as evidenced by elevated levels of pro-apoptotic markers (p53, cleaved PARP, and BIM) and γH2AX. In vivo xenograft models confirmed these findings, showing increased sensitivity to trametinib in MYC-overexpressing tumors. Moreover, MEK inhibition increased autophagy in HNSCC cells, a factor critical for therapeutic resistance. Inhibiting trametinib-induced autophagy further enhanced apoptotic cell death. These findings suggest that MYC expression and autophagy play crucial roles in HNSCC’s response to MEK inhibition. Combining trametinib with autophagy inhibition may improve therapeutic outcomes in HNSCC. Full article
(This article belongs to the Special Issue New Wave of Cancer Therapeutics: Challenges and Opportunities)
Show Figures

Figure 1

Figure 1
<p>Multiple anti-cancer effects induced by trametinib through MEK/MAPK inhibition in JHU11 HNSCC cells. Total protein extracts were prepared from JHU11 cells treated with either vehicle or 200 nM trametinib for 48 h. Western blot analysis was performed to detect (<b>A</b>) pERK1/2 (<b>a</b>), total ERK1/2 (<b>b</b>), pEGFR (<b>c</b>), MYC (<b>d</b>), and β-Actin (<b>e</b>). (<b>B</b>) Protein band intensities of (<b>a</b>) pERK/ERK, (<b>b</b>) pEGFR, and (<b>c</b>) MYC were quantified and normalized to the corresponding β-actin to compare vehicle- and trametinib-treated cells. (<b>C</b>,<b>D</b>) γH2A.X (<b>a</b>) and β-Actin (<b>Cb</b>); (<b>E</b>,<b>F</b>) p21 (<b>a</b>), p27 (<b>b</b>), cPARP/PARP (<b>c</b>), BIM (<b>d</b>), p53 (<b>e</b>), and β-Actin (<b>Ef</b>); (<b>G</b>,<b>H</b>) pSer2/5 (<b>a</b>), pSer7 (<b>b</b>), and β-Actin (<b>Gc</b>). Protein band intensities for the ratio of cPARP/PARP, pEGFR, MYC, γH2A.X, p21, p27, BIM, p53, pSer2/5, and pSer7 were normalized to β-actin correspondingly and compared between vehicle-treated and trametinib-treated cells. Data from independent experiments are presented as mean ± SD (n = 3~4). Statistical significance is indicated as follows: * <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 compared to vehicle-treated controls. Abbreviations: Veh: Vehicle, Tram: Trametinib.</p>
Full article ">Figure 2
<p>MYC overexpression renders JHU22 HNSCC cells more sensitive to trametinib. (<b>A</b>) Overall survival percentage of HNSCC patients with (solid line) versus without MYC amplification (dotted line) (<span class="html-italic">p</span> = 0.013). (<b>B</b>) Increased MYC mRNA expression is observed in HNSCC patients with MYC amplification. All data are from the TCGA database (279 samples), Nature 2015, accessed via cBioPortal.org [<a href="#B27-ijms-26-00588" class="html-bibr">27</a>,<a href="#B28-ijms-26-00588" class="html-bibr">28</a>]. (<b>C</b>) Total protein extracts were obtained from JHU22-LV and JHU22-MYC cells treated with either vehicle or 200 nM trametinib for 48 h. Western blot analysis was performed to detect MYC and β-Actin. (<b>D</b>) MYC band intensities were quantified and normalized to the corresponding β-actin to compare vehicle- and trametinib-treated cells (n = 4). (<b>E</b>) A dose-escalation study was conducted to evaluate the effects of increasing concentrations of trametinib on cell viability in JHU22-LV and JHU22-MYC cells, with treatment lasting for 7 days. Images represent two replicates. (<b>F</b>) IC50 values for trametinib (<b>a</b>,<b>b</b>) and erlotinib (<b>c</b>,<b>d</b>) were determined in JHU22-LV and JHU22-MYC cells, respectively. The red dotted line indicates 50% viability, corresponding to the IC50 value. (<b>G</b>) Colony formation assays were performed to assess the proliferative ability of JHU22-LV (<b>a</b>,<b>c</b>) and JHU22-MYC (<b>b</b>,<b>d</b>) cells following treatment with either vehicle or trametinib (200 nM) for 7 days. (<b>H</b>) Bar graphs and statistical analysis of the colony coverage (Total area, % of plate). (<b>I</b>) Wound healing assays were used to examine changes in migratory capacity between JHU22-LV and JHU22-MYC cells, with treatments of either vehicle or trametinib (200 nM) conducted at 0, 6, and 24 h. (<b>J</b>) Line graphs and statistical analysis of wound healing assays. Data from independent experiments are presented as mean ± SD (n = 4). Statistical significance is indicated as follows: * <span class="html-italic">p</span> &lt; 0.05; *** <span class="html-italic">p</span> &lt; 0.001 compared to JHU22-LV vehicle-treated controls. ### <span class="html-italic">p</span> &lt; 0.001 compared to JHU22-MYC vehicle-treated controls. Abbreviations: Veh: Vehicle, Tram: Trametinib.</p>
Full article ">Figure 3
<p>MYC overexpression alters the molecular response of HNSCC cells to trametinib. Total protein extracts were prepared from JHU22-LV and JHU22-MYC cells treated with either vehicle or trametinib (200 nM) for 48 h. (<b>A</b>) Western blot analysis was performed to detect pERK1/2 (<b>a</b>), total ERK1/2 (<b>b</b>), pEGFR (<b>c</b>) and β-Actin (<b>d</b>). (<b>B</b>) The pERK1/2/ERK1/2 ratio (<b>a</b>) or protein band intensity of pEGFR (<b>b</b>) was quantified and normalized to the corresponding β-actin to compare the effects between vehicle-treated and trametinib-treated cells (n = 4). (<b>C</b>) Western blot analysis was also conducted to detect γH2A.X (<b>a</b>), pSer2/5 (<b>b</b>), pSer7 (<b>c</b>), and β-actin (<b>d</b>) in JHU22-LV and JHU22-MYC cells treated with vehicle or trametinib for 48 h. (<b>D</b>) The protein band intensities of γH2A.X (<b>a</b>), pSer2/5 (<b>b</b>), and pSer7 (<b>c</b>) were quantified and normalized to the corresponding β-actin to compare the effects between vehicle-treated and trametinib-treated cells. Data from four independent experiments are presented as mean ± SD (n = 4). Statistical significance is indicated as follows: * <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 compared to JHU22-LV vehicle-treated controls. ### <span class="html-italic">p</span> &lt; 0.001 compared to JHU22-MYC vehicle-treated controls. Abbreviations: Veh: Vehicle, Tram: Trametinib.</p>
Full article ">Figure 4
<p>MYC overexpression in JHU22 HNSCC cells promotes cell cycle progression and apoptotic cell death in response to trametinib. (<b>A</b>) JHU22-LV and JHU22-MYC cells were treated with either vehicle or trametinib (200 nM) for 48 h, and flow cytometric analysis of cell cycle phases was performed following DAPI staining. Blue: G1; Yellow: S Phase; Green: G2/M; Magenta: Sum. (<b>B</b>) The percentage of cells in the G1 or G2/M phase in both JHU22-LV and JHU22-MYC cells was quantified (n = 4). (<b>C</b>) Total protein extracts were obtained from JHU22-LV and JHU22-MYC cells following treatment with either vehicle or trametinib (200 nM) for 48 h. Western blot analysis was conducted to detect p21 (<b>a</b>), p27 (<b>b</b>), E2F1 (<b>c</b>), and β-actin (<b>d</b>). (<b>D</b>) Protein band intensities of p21 (<b>a</b>), p27 (<b>b</b>), and E2F1 (<b>c</b>) were quantified and normalized to the corresponding β-actin to compare the effects between vehicle-treated and trametinib-treated cells (n = 4). (<b>E</b>) JHU22-LV (<b>a</b>,<b>c</b>) and JHU22-MYC (<b>b</b>,<b>d</b>) cells were treated with either vehicle or trametinib (200 nM) for 48 h and then collected for annexin V and PI staining, followed by flow cytometric analysis to assess apoptotic cells. Pseudo color plots are used to indicate cell density or number. (<b>F</b>) The populations of apoptotic cells in both early and late stages were quantified (n = 4). (<b>G</b>) Western blot analysis was performed to detect cPARP/PARP (<b>a</b>), BIM (<b>b</b>), p53 (<b>c</b>), and β-actin (<b>d</b>) in JHU22-LV and JHU22-MYC cells. (<b>H</b>) The cPARP/PARP ratio (<b>a</b>) or protein band intensities of BIM (<b>b</b>) and p53 (<b>c</b>) were quantified relative to the corresponding β-actin to compare the effects between vehicle-treated and trametinib-treated cells. Data from four independent experiments are presented as mean ± SD (n = 4). Statistical significance is indicated as follows: * <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 compared to JHU22-LV vehicle-treated controls. # <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 compared to JHU22-MYC vehicle-treated controls. ns: not significant. Abbreviations: Veh: Vehicle, Tram: Trametinib.</p>
Full article ">Figure 5
<p>Inhibition of trametinib-induced autophagy enhances apoptotic cell death in HNSCC cells. (<b>A</b>) Total protein extracts were prepared from JHU22-LV and JHU22-MYC cells treated with either vehicle or trametinib (200 nM) for 48 h. Western blot analysis was performed to detect LC3bI/II (<b>a</b>), p62 (<b>b</b>), and β-actin (<b>c</b>). (<b>B</b>) Protein band intensities of LC3bII (<b>a</b>) and p62 (<b>b</b>) were quantified relative to the corresponding β-actin to compare the effects between vehicle-treated and trametinib-treated cells (n = 4). (<b>C</b>) JHU22-LV and JHU22-MYC cells were treated with vehicle (<b>a</b>,<b>e</b>), trametinib (<b>b</b>,<b>f</b>; 200 nM), HCQ (<b>c</b>,<b>g</b>; 10 µM), or the combination of trametinib and HCQ (<b>d</b>,<b>h</b>) for 48 h, then collected for annexin V and PI staining and analyzed via flow cytometry to assess early and late-stage apoptosis. (<b>D</b>) The populations of apoptotic cells in both early and late stages were quantified (n = 4). (<b>E</b>) Total protein extracts were prepared from JHU22-LV and JHU22-MYC cells treated with either vehicle, trametinib (200 nM), HCQ (10 µM), or the combination of trametinib and HCQ for 48 h. Western blot analysis was performed to detect cPARP/PARP and β-actin. (<b>F</b>) The ratio of cPARP/PARP was quantified to compare the effects between vehicle-treated and trametinib-treated cells. Data from four independent experiments are presented as mean ± SD (n = 4). Statistical significance is indicated as follows: * <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 compared to JHU22-LV vehicle-treated controls. ## <span class="html-italic">p</span> &lt; 0.01; ### <span class="html-italic">p</span> &lt; 0.001 compared to JHU22-MYC vehicle-treated controls. ns: not significant. Abbreviations: Veh: Vehicle, Tram: Trametinib, HCQ: hydroxychloroquine.</p>
Full article ">Figure 6
<p>Impact of trametinib on xenograft tumors overexpressing MYC in HNSCC. JHU22-LV and JHU22-MYC cells were injected subcutaneously into athymic nude mice to form xenograft tumors (n = 4). (<b>A</b>) Representative images of the gross tumors. (<b>B</b>) Average body weights of the mice were monitored during tumor development. (<b>C</b>) Tumor sizes were measured every 2–3 days following injection and calculated using the formula: length × width<sup>2</sup> × 0.5. (<b>D</b>) Comparison of the average weights of dissected tumors from mice. (<b>E</b>) IHC images showing MYC (JHU22-LV: a1, a2; JHU22-MYC: a3, a4), Ki-67 (JHU22-LV: b1, b2; JHU22-MYC: b3, b4), γH2A.X (JHU22-LV: c1, c2; JHU22-MYC: c3, c4), and cleaved-Casp3 (JHU22-LV: d1, d2; JHU22-MYC: d3, d4) in the HNSCC xenograft tumors treated with Veh or Tram, respectively. Scale bar = 30 µm (insets). (<b>F</b>) Quantification of the IHC staining for MYC (<b>a</b>), Ki-67 (<b>b</b>), γH2A.X (<b>c</b>), and cleaved-Caspase 3 (<b>d</b>). Data from four independent experiments are presented as mean ± SD (n = 4). Statistical significance is indicated as follows: * <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 compared to JHU22-LV vehicle-treated controls. ### <span class="html-italic">p</span> &lt; 0.001 compared to JHU22-MYC vehicle-treated controls. Abbreviations: Veh: Vehicle, Tram: Trametinib, cleaved-Casp3: cleaved-Caspase 3.</p>
Full article ">
18 pages, 3285 KiB  
Article
Sorafenib-Loaded Silica-Containing Redox Nanoparticle Decreases Tumorigenic Potential of Lewis Lung Carcinoma
by Babita Shashni, Hao Thi Tran, Long Binh Vong, Ren-Jei Chung and Yukio Nagasaki
Pharmaceutics 2025, 17(1), 50; https://doi.org/10.3390/pharmaceutics17010050 - 2 Jan 2025
Viewed by 675
Abstract
Background: Orally administered sorafenib has shown limited improvement in overall survival for non-small-cell lung cancer patients, likely due to poor pharmacokinetics and adverse effects, including gastrointestinal toxicity. To address these issues, we developed silica-containing antioxidant nanoparticles (siRNP) as a carrier to enhance the [...] Read more.
Background: Orally administered sorafenib has shown limited improvement in overall survival for non-small-cell lung cancer patients, likely due to poor pharmacokinetics and adverse effects, including gastrointestinal toxicity. To address these issues, we developed silica-containing antioxidant nanoparticles (siRNP) as a carrier to enhance the therapeutic efficacy of lipophilic sorafenib. Methods: Sorafenib was loaded into siRNP via dialysis (sora@siRNP). The therapeutic efficacy and safety of sora@siRNP (20 and 40 mg-sora/kg) were evaluated in a xenograft mouse model of Lewis lung carcinoma (subcutaneous tumors and experimental metastasis) following oral administration. Results: Crosslinking nanosilica in siRNP improved drug stability, enabling 8.9% sorafenib loading and pH resilience. Oral sora@siRNP exhibited dose-dependent tumor growth suppression by downregulating pMEK, outperforming free sorafenib, which showed inconsistent efficacy likely due to formulation variability. Intestinal damage, a major adverse effect of free sorafenib, was significantly reduced with sora@siRNP, attributed to siRNP’s antioxidant property of mitigating oxidative damage. Survival rates in the experimental metastasis model were 66–74% for sorafenib but reached 100% for sora@siRNP, highlighting its superior efficacy and safety. Conclusions: These findings demonstrate that nanosilica-crosslinked antioxidant nanoparticles (siRNP) enhance the stability, delivery efficiency, and safety of lipophilic drugs like sorafenib for oral administration. This platform holds promise for improving therapeutic outcomes in lung cancer while minimizing adverse effects. Full article
Show Figures

Figure 1

Figure 1
<p>Illustration showcasing the chemical structure of silica-containing redox polymer (PEG-<span class="html-italic">b</span>-siPMNT) that self-assembles into silica-containing nanoparticle (siRNP) under aqueous conditions driven by hydrophobic force. In the present study, the anticancer hydrophobic drug sorafenib was loaded into the core of siRNP (sora@siRNP) and evaluated for anticancer efficacy in a mouse model of lung cancer (Lewis lung carcinoma) via oral administration [<a href="#B7-pharmaceutics-17-00050" class="html-bibr">7</a>].</p>
Full article ">Figure 2
<p>Characterization of siRNP and sora@siRNP. (<b>A</b>) DLS-based size (intensity, %) distribution of sorafenib, sora@siRNP, and siRNP. (<b>B</b>) The images of sorafenib in water, sora@siRNP, and siRNP after 24 h of dialysis. (<b>C</b>) The sorafenib encapsulation efficacy (EE) and loading capacity (LC) of siRNP. (<b>D</b>) The hydrodynamic diameter of siRNP at different pH conditions. The values are expressed as the mean ± SD.</p>
Full article ">Figure 3
<p>The anti-tumor efficacy and mechanism of sora@siRNP in LLC-tumor-bearing mouse model. (<b>A</b>) The scheme of in vivo treatment regimen in a subcutaneous xenograft mouse model of LLC. (<b>B</b>) The tumor growth profile (mm<sup>3</sup>). (<b>C</b>) The tumor weight of mice at the experimental end point. (<b>D</b>) Representative images of extracted tumors. (<b>E</b>) HE-stained tumor sections. (<b>F</b>) Immunohistochemical staining of pMEK in the tumor sections. (<b>G</b>) The quantitative graph of pMEK<sup>+</sup> signals of the stained tumor sections. Values expressed as mean ± SEM. “ns” non-significant, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.005, *** <span class="html-italic">p</span> &lt; 0.0005, **** <span class="html-italic">p</span> &lt; 0.0001, Tukey’s multiple comparisons test. Sora(40) vs. Sora@siRNP(40); Sora@siRNP(40) vs. siRNP, * <span class="html-italic">p</span> &lt; 0.05, <span class="html-italic">t</span>-test.</p>
Full article ">Figure 4
<p>The protective effect of siRNP on the intestine. (<b>A</b>) Representative images of the HE, PAS, and TUNEL-stained small intestine sections. (<b>B</b>) The length of villi was measured in the HE-stained intestinal sections. (<b>C</b>) The number of goblet cells/villus measured in the PAS-stained intestinal sections. (<b>D</b>) Apoptotic TUNEL<sup>+</sup>-stained areas (the brown area with arrows) in the intestine. Values are shown as a scatter plot with mean shown as a black bar. “ns” non-significant, ** <span class="html-italic">p</span> &lt; 0.005, *** <span class="html-italic">p</span> &lt; 0.0005, **** <span class="html-italic">p</span> &lt; 0.0001, Tukey’s multiple comparisons test.</p>
Full article ">Figure 5
<p>The protective effect of siRNP in the colon. (<b>A</b>) Representative images of the HE and PAS-stained colon sections, scale bar = 200 μm. (<b>B</b>) The number of goblet cells/10 crypts were measured in the PAS-stained colon sections. Values expressed as a scatter plot with mean shown as a black bar. “ns” non-significant, **** <span class="html-italic">p</span> &lt; 0.0001, Tukey’s multiple comparisons test.</p>
Full article ">
10 pages, 242 KiB  
Article
Identification of the Determinants of Plexiform Neurofibroma Morbidity in Pediatric and Young Adult Neurofibromatosis Type 1 Patients: A Pilot Multivariate Approach
by Biagio de Brons, Britt Dhaenens, Rick van Minkelen and Rianne Oostenbrink
Cancers 2025, 17(1), 123; https://doi.org/10.3390/cancers17010123 - 2 Jan 2025
Viewed by 689
Abstract
Background: Plexiform neurofibromas (PNs) are histologically benign peripheral nerve sheath tumors associated with neurofibromatosis type 1 (NF1) and often lead to significant morbidity due to growth. Management includes watchful waiting, surgery for partial debulking, and, since recently, systemic treatment with MEK inhibitors. However, [...] Read more.
Background: Plexiform neurofibromas (PNs) are histologically benign peripheral nerve sheath tumors associated with neurofibromatosis type 1 (NF1) and often lead to significant morbidity due to growth. Management includes watchful waiting, surgery for partial debulking, and, since recently, systemic treatment with MEK inhibitors. However, due to the scarcity of natural history studies, our understanding of the natural progression of PNs to guide clinicians in deciding in whom and when to intervene is scarce. This study aims to describe the characteristics of NF1 patients with PNs and compare those at high risk for PN progression or experiencing significant morbidity from PN (complex PN) with NF1 patients with PNs of lower complexity. Methods: In this retrospective cohort study using clinical data from hospital records of NF1 patients with PNs seen at the Sophia Children’s Hospital in the Netherlands between 2012 and 2023, we assessed determinants of clinical phenotypes and PN characteristics predictive of outcomes, including PN complexity and the timing of intervention for PN. We assessed the outcomes using logistic regression analysis and Cox regression. Results: Ninety patients with a median age at last evaluation of 15.7 years and a median follow-up duration of 9.8 years were included. Out of 90 individuals with a benign PN, 37 developed plexiform neurofibroma morbidity during follow-up. Older age was (corrected for pathogenic NF1 variant and PN location) significantly associated with plexiform neurofibroma morbidity. Cox regression revealed that craniofacial and trunk PNs were associated with a higher intervention hazard compared to limb PNs. Conclusion: Our pilot multivariate approach identified older age and the location of the PN to be mostly associated with a higher chance of plexiform neurofibroma morbidity and higher intervention hazard. This may contribute to decisions regarding in whom and when to initiate treatment in NF1 patients with PNs. Full article
(This article belongs to the Special Issue Neurofibromatosis)
10 pages, 2636 KiB  
Case Report
Impact of BRAF and MEK Inhibitors on Gingival Hyperplasia in Melanoma Patients—A Case Report
by Tanja Veljovic, Milanko Djuric, Ivana Gusic, Nada Vuckovic, Bojana Ramic and Jelena Mirnic
J. Clin. Med. 2025, 14(1), 65; https://doi.org/10.3390/jcm14010065 - 26 Dec 2024
Viewed by 508
Abstract
Background: Although BRAF inhibitors, such as vemurafenib, produce a marked response in patients with advanced melanoma with a BRAF V600 mutation, they eventually develop resistance to this treatment. To address this issue, vemurafenib is increasingly combined with the MEK inhibitor cobimetinib, leading [...] Read more.
Background: Although BRAF inhibitors, such as vemurafenib, produce a marked response in patients with advanced melanoma with a BRAF V600 mutation, they eventually develop resistance to this treatment. To address this issue, vemurafenib is increasingly combined with the MEK inhibitor cobimetinib, leading to improved response rates and enhanced survival. However, this treatment modality is associated with numerous side effects. We present a case of gingival hyperplasia in a patient treated with vemurafenib, along with the strategy adopted for the management of this condition, and the impact of subsequent cobimetinib administration on its severity. Methods: The 59-year-old male patient in the focus of this report presented at the Department of Periodontology at the Medical Faculty, University of Novi Sad, in 2019, complaining of gingival overgrowth and bleeding. The patient reported persistent gum swelling during the preceding six months, which he ascribed to the use of vemurafenib, 960 mg twice daily, since 2018, when this medication was prescribed as a part of malignant melanoma treatment. Detailed clinical examination revealed significant gingival overgrowth around all present teeth, affecting the vestibular as well as the oral sides. The patient underwent thorough scaling and root planing, followed by the surgical removal of hyperplastic gingiva. After gingivectomy, the patient was scheduled for follow-up visits at one-month intervals. Six months after gingivectomy, vemurafenib dose was reduced to 720 mg twice daily, and cobimetinib was introduced at 60 mg per day. Results: The treatment protocol adopted in this study, combined with cobimetinib administration, stabilized the gingiva condition in this patient. However, due to his overall poor oral hygiene, gingiva remained inflamed and edematous, but was no longer hyperplastic and hyperkeratotic in appearance. Conclusions: This case underscores the importance of recognizing and adequately addressing this complication, as its adverse effect on a patient’s quality of life can potentially compromise treatment protocol adherence. Full article
(This article belongs to the Special Issue Melanoma: Clinical Updates and Perspectives)
Show Figures

Figure 1

Figure 1
<p>Gingival overgrowth during the first visit. The enlarged gums were covered with erosions and a thick, white plaque which appeared to be hyperkeratotic.</p>
Full article ">Figure 2
<p>Histopathological changes in mucosa, pronounced acanthotic squamous cell epithelium with focal parakeratosis. Evidence of chronic inflammatory infiltrate in lamina propria (HEx40).</p>
Full article ">Figure 3
<p>(<b>a</b>) Postoperative photograph of the patient’s mouth, (<b>b</b>) Eliminated hyperplastic gingiva.</p>
Full article ">Figure 4
<p>Tooth deposits and swollen inflamed gums three months after gingivectomy.</p>
Full article ">Figure 5
<p>Photographs of the patient’s mouth after Cotellic was introduced. Inflamed but not hyperplastic gingiva was noted on examination.</p>
Full article ">
Back to TopTop