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

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (90)

Search Parameters:
Keywords = AURKA

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
21 pages, 5787 KiB  
Article
Only Infant MLL-Rearranged Leukemia Is Susceptible to an Inhibition of Polo-like Kinase 1 (PLK-1) by Volasertib
by Jacqueline Fischer, Estelle Erkner, Pia Radszuweit, Thomas Hentrich, Hildegard Keppeler, Fulya Korkmaz, Julia Schulze-Hentrich, Rahel Fitzel, Claudia Lengerke, Dominik Schneidawind and Corina Schneidawind
Int. J. Mol. Sci. 2024, 25(23), 12760; https://doi.org/10.3390/ijms252312760 - 27 Nov 2024
Viewed by 648
Abstract
MLL-rearranged (MLLr) leukemia is characterized by a poor prognosis. Depending on the cell of origin, it differs in the aggressiveness and therapy response. For instance, in adults, volasertib blocking Polo-like kinase 1 (PLK-1) exhibited limited success. Otherwise, PLK-1 characterizes an [...] Read more.
MLL-rearranged (MLLr) leukemia is characterized by a poor prognosis. Depending on the cell of origin, it differs in the aggressiveness and therapy response. For instance, in adults, volasertib blocking Polo-like kinase 1 (PLK-1) exhibited limited success. Otherwise, PLK-1 characterizes an infant MLLr signature, indicating potential sensitivity. By using our CRISPR/Cas9 MLLr model in CD34+ cells from human cord blood (huCB) and bone marrow (huBM) mimicking the infant and adult patient diseases, we were able to shed light on this phenomenon. The PLK-1 mRNA level was significantly increased in our huCB compared to the huBM model, which was underpinned by analyzing infant and adult MLLr leukemia patients. Importantly, the expression levels correlated with a functional response. Volasertib induced a significant dose-dependent decrease in proliferation and cell cycle arrest, most pronounced in the infant model. Mechanistically, upon volasertib treatment, we uncovered negative feedback only in the huBM model by compensatory upregulation of PLK-1 and related genes like AURKA involved in mitosis. Importantly, the poor response could be overcome by a combinatorial strategy with alisertib, an Aurora kinase A inhibitor. Our study emphasizes the importance of considering the cell of origin in therapeutic decision-making and provides the rationale for evaluating volasertib and alisertib in MLLr leukemia. Full article
(This article belongs to the Special Issue Hallmarks of Cancer: Emerging Insights and Innovations)
Show Figures

Figure 1

Figure 1
<p><b>Revealing PLK-1 as a potential promising target in infant MLL-AF9 leukemia.</b> (<b>A</b>) CD34+ HSPCs were isolated from huCB and huBM via Ficoll separation and magnetic cell separation and cultured for 48 h. Thereafter, <span class="html-italic">t(9;11)</span> was induced in cultured HSPCs using the CRISPR/Cas9 system. (<b>B</b>) RNA sequencing of human CRISPR/Cas9 <span class="html-italic">MLL-AF9</span> cells derived from huCB (<span class="html-italic">n</span> = 5) and huBM (<span class="html-italic">n</span> = 5) compared with the respective control cells (huCB/huBM-derived CD34+, <span class="html-italic">n</span> = 4). Student’s <span class="html-italic">t</span>-test. * <span class="html-italic">p</span> &lt; 0.05. ns: not significant <span class="html-italic">p</span> &gt; 0.05. (<b>C</b>) Fold change of <span class="html-italic">PLK-1</span> overexpression in huCB <span class="html-italic">MLL-AF4</span> and <span class="html-italic">MLL-AF9</span> cells (<span class="html-italic">n</span> = 5/<span class="html-italic">n</span> = 5); huBM <span class="html-italic">MLL</span>r cells (<span class="html-italic">n</span> = 5); cell lines THP-1, NOMO-1, SEM, RS4;11, KOPN8, and SKM1 (all <span class="html-italic">n</span> = 3); and infant and adult <span class="html-italic">MLL</span>r leukemia patient samples (<span class="html-italic">n</span> = 3/<span class="html-italic">n</span> = 3) compared to CD34+ huCB/BM control cells (ctrl, <span class="html-italic">n</span> = 4), measured by RT-qPCR. One-way ANOVA. * <span class="html-italic">p</span> &lt; 0.05. ns: not significant <span class="html-italic">p</span> &gt; 0.05. Overview of <span class="html-italic">MLL</span> translocation in the cell lines used. (<b>D</b>) Kaplan–Meier survival curve (<a href="http://www.kmplot.com" target="_blank">www.kmplot.com</a>, 26 August 2024). Higher <span class="html-italic">PLK-1</span> expression levels in AML patients show a trend to worse survival rates. Logrank. <span class="html-italic">p</span> = 0.083. Median survival rates [months]: low <span class="html-italic">PLK-1</span> level 16.4, high <span class="html-italic">PLK-1</span> level 12.1.</p>
Full article ">Figure 2
<p><b><span class="html-italic">MLL</span>r leukemia cells derived from huCB are more susceptible to a PLK-1 inhibition than those from huBM.</b> (<b>A</b>) Cell counts were assessed following treatment with volasertib (50 nM), vehicle control (DMSO), or no treatment (as a baseline control) in huCB and huBM CRISPR/Cas9 <span class="html-italic">MLL</span>r cells (<span class="html-italic">n</span> = 3 each). Relative cell counts were determined using a Neubauer counting chamber after Trypan blue staining and normalized to the vehicle control (DMSO). Right: Significant difference of proliferation between huCB and huBM CRISPR/Cas9 <span class="html-italic">MLL</span>r cells at 72 h after 50 nM volasertib treatment. One-way ANOVA. * <span class="html-italic">p</span> &lt; 0.05. (<b>B</b>) huCB and huBM CRISPR/Cas9 <span class="html-italic">MLL</span>r (<span class="html-italic">n</span> = 3/<span class="html-italic">n</span> = 3) and CD34+ huCB/BM control cells (ctrl, <span class="html-italic">n</span> = 4) were treated with increasing concentrations of volasertib or vehicle control (DMSO) for 72 h. Relative cell count was determined by counting cells in a Neubauer counting chamber after Trypan blue staining, normalized to vehicle control (DMSO). IC50 values: huBM <span class="html-italic">MLL</span>r 43.5 nM, huCM <span class="html-italic">MLL</span>r 17.9 nM, ctrl 35.1 nM. IC50 values of the dose-dependent curves were interpolated from a four-parameter logistic model. (<b>C</b>) Significant difference between IC50 values of huBM <span class="html-italic">MLL</span>r and huCB <span class="html-italic">MLL</span>r cells compared to ctrl and each other. One-way ANOVA. * <span class="html-italic">p</span> &lt; 0.05. ns: not significant <span class="html-italic">p</span> &gt; 0.05. (<b>D</b>) Representative flow cytometric histograms of Annexin V/PI staining to determine the apoptotic effect of 72 h volasertib treatment (DMSO, 50 nM, 100 nM) on huCB and huBM CRISPR/Cas9 <span class="html-italic">MLL</span>r (<span class="html-italic">n</span> = 3/<span class="html-italic">n</span> = 3) and CD34+ huCB/BM control cells (ctrl, <span class="html-italic">n</span> = 4), measured by flow cytometry. On the right, summarized fractions normalized to their own vehicle control (DMSO). One-way ANOVA. * <span class="html-italic">p</span> &lt; 0.05. (<b>E</b>) Left: THP-1 and NOMO-1 (<span class="html-italic">n</span> = 3/<span class="html-italic">n</span> = 3) cells were treated with increasing concentrations of volasertib or vehicle control (DMSO) for 72 h. Relative cell count was determined by counting cells in a Neubauer counting chamber after Trypan blue staining and normalized to vehicle control (DMSO). IC50 values: THP-1 8.0 nM; NOMO-1 could not be determined, as 50% cell death was not achieved (n.c.). IC50 values of the dose-dependent curves were interpolated from a four-parameter logistic model. Significant difference between endpoints (200 nM volasertib treatment) of NOMO-1 and THP-1. One-way ANOVA. * <span class="html-italic">p</span> &lt; 0.05. Right: Representative flow cytometric histograms and summarized distribution of Annexin V/PI staining to determine the apoptotic effect of volasertib treatment (DMSO, 50 nM, 100 nM, 72 h incubation) on NOMO-1 and THP-1 (<span class="html-italic">n</span> = 3/<span class="html-italic">n</span> = 3) cells measured by flow cytometry. On the right, summarized fractions normalized to their own vehicle control (DMSO). One-way ANOVA. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 3
<p><b>Inhibition of PLK-1 leads to reduced viability and mitotic arrest.</b> (<b>A</b>) The 72 h volasertib treatment (DMSO vehicle control, 50 nM, 100 nM) on huBM and huCB CRISPR/Cas9 <span class="html-italic">MLL</span>r cells (<span class="html-italic">n</span> = 3/<span class="html-italic">n</span> = 3) decreased cell viability, measured by AlamarBlue viability assay. Comparison of the reduction in cell viability after treatment with 100 nM volasertib between huBM and huCB CRISPR/Cas9 <span class="html-italic">MLL</span>r cells (<span class="html-italic">n</span> = 3/<span class="html-italic">n</span> = 3) and CD34+ huCB/BM control cells (ctrl, <span class="html-italic">n</span> = 4). One-way ANOVA. * <span class="html-italic">p</span> &lt; 0.05. ns: not significant <span class="html-italic">p</span> &gt; 0.05. (<b>B</b>) Representative (left) and pooled (right) data of BrdU cell cycle analysis of huBM and huCB CRISPR/Cas9 <span class="html-italic">MLL</span>r cells (<span class="html-italic">n</span> = 3/<span class="html-italic">n</span> = 3), CD34+ huCB control cells (ctrl, <span class="html-italic">n</span> = 3), and THP-1, NOMO-1 (<span class="html-italic">n</span> = 3/<span class="html-italic">n</span> = 3) after volasertib treatment for 48 h (DMSO vehicle control, 50 nM, 100 nM). This shows a significant increase in G2/M-phase and a decrease in S-phase in CB <span class="html-italic">MLL</span>r cells. Normalized to respective vehicle control (DMSO). One-way ANOVA. * <span class="html-italic">p</span> &lt; 0.05. ns: not significant <span class="html-italic">p</span> &gt; 0.05. (<b>C</b>) Images show representative morphologies of huBM and huCB CRISPR/Cas9 <span class="html-italic">MLL</span>r cells after volasertib treatment (DMSO vehicle control, 50 nM, 100 nM). Black arrows point at mitotic figures; cells arrested in M-phase. Pappenheim staining.</p>
Full article ">Figure 3 Cont.
<p><b>Inhibition of PLK-1 leads to reduced viability and mitotic arrest.</b> (<b>A</b>) The 72 h volasertib treatment (DMSO vehicle control, 50 nM, 100 nM) on huBM and huCB CRISPR/Cas9 <span class="html-italic">MLL</span>r cells (<span class="html-italic">n</span> = 3/<span class="html-italic">n</span> = 3) decreased cell viability, measured by AlamarBlue viability assay. Comparison of the reduction in cell viability after treatment with 100 nM volasertib between huBM and huCB CRISPR/Cas9 <span class="html-italic">MLL</span>r cells (<span class="html-italic">n</span> = 3/<span class="html-italic">n</span> = 3) and CD34+ huCB/BM control cells (ctrl, <span class="html-italic">n</span> = 4). One-way ANOVA. * <span class="html-italic">p</span> &lt; 0.05. ns: not significant <span class="html-italic">p</span> &gt; 0.05. (<b>B</b>) Representative (left) and pooled (right) data of BrdU cell cycle analysis of huBM and huCB CRISPR/Cas9 <span class="html-italic">MLL</span>r cells (<span class="html-italic">n</span> = 3/<span class="html-italic">n</span> = 3), CD34+ huCB control cells (ctrl, <span class="html-italic">n</span> = 3), and THP-1, NOMO-1 (<span class="html-italic">n</span> = 3/<span class="html-italic">n</span> = 3) after volasertib treatment for 48 h (DMSO vehicle control, 50 nM, 100 nM). This shows a significant increase in G2/M-phase and a decrease in S-phase in CB <span class="html-italic">MLL</span>r cells. Normalized to respective vehicle control (DMSO). One-way ANOVA. * <span class="html-italic">p</span> &lt; 0.05. ns: not significant <span class="html-italic">p</span> &gt; 0.05. (<b>C</b>) Images show representative morphologies of huBM and huCB CRISPR/Cas9 <span class="html-italic">MLL</span>r cells after volasertib treatment (DMSO vehicle control, 50 nM, 100 nM). Black arrows point at mitotic figures; cells arrested in M-phase. Pappenheim staining.</p>
Full article ">Figure 4
<p><b>Transcriptomic analysis</b> revealed a compensatory PLK-1 feedback mechanism only in adult <span class="html-italic">MLL</span>r cells upon volasertib treatment. huBM and huCB CRISPR/Cas9 <span class="html-italic">MLL</span>r cells (<span class="html-italic">n</span> = 4/<span class="html-italic">n</span> = 3) were treated with 50 nM volasertib or vehicle control (DMSO) for 72 h and used for RNA-seq. (<b>A</b>) Analysis revealed in huBM <span class="html-italic">MLL</span>r cells 728 differentially expressed genes (DEGs) and 151 DEGs in huCB <span class="html-italic">MLL</span>r cells after volasertib treatment. Volcano plot of huBM and huCB CRISPR/Cas9 <span class="html-italic">MLL</span>r cells after volasertib treatment highlighting downregulated (blue) and upregulated (red) DEGs. Dotted lines indicate significant thresholds (pFDR ≤ 0.05, |log2(fold-change)| ≥  0.5). (<b>B</b>) log2(fold-change) of DEGs are shown for <span class="html-italic">PLK-1</span> and associated genes, normalized to own vehicle control (DMSO). Black * p_adjust value, white * p_nominal value. huBM <span class="html-italic">MLL</span>r cells show an upregulation of PLK-1-related gene pattern, whereas the pattern of huCB <span class="html-italic">MLL</span>r is downregulated. (<b>C</b>) Interactome of significant altered normalized Reads per Kilobase Millions (nRPKMs) around <span class="html-italic">PLK-1</span> in huBM and huCB CRISPR/Cas9 <span class="html-italic">MLL</span>r cells reveals potential feedback mechanism. Upregulation (red), downregulation (blue). Additional RNA-seq analysis regarding phosphorylation activity and targets around <span class="html-italic">PLK-1</span> are highlighted with red arrows. (<b>D</b>) Fold change of <span class="html-italic">PLK-1</span>, <span class="html-italic">BORA</span>, <span class="html-italic">AURKA,</span> and <span class="html-italic">FOXM1</span> in huBM and huCB CRISPR/Cas9 <span class="html-italic">MLL</span>r cells (<span class="html-italic">n</span> = 4/<span class="html-italic">n</span> = 3), as well as NOMO-1 and THP-1 cells (<span class="html-italic">n</span> = 3/<span class="html-italic">n</span> = 3), after 72 h 50 nM volasertib treatment compared to vehicle control (DMSO) measured by RT-qPCR. One-way ANOVA. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 4 Cont.
<p><b>Transcriptomic analysis</b> revealed a compensatory PLK-1 feedback mechanism only in adult <span class="html-italic">MLL</span>r cells upon volasertib treatment. huBM and huCB CRISPR/Cas9 <span class="html-italic">MLL</span>r cells (<span class="html-italic">n</span> = 4/<span class="html-italic">n</span> = 3) were treated with 50 nM volasertib or vehicle control (DMSO) for 72 h and used for RNA-seq. (<b>A</b>) Analysis revealed in huBM <span class="html-italic">MLL</span>r cells 728 differentially expressed genes (DEGs) and 151 DEGs in huCB <span class="html-italic">MLL</span>r cells after volasertib treatment. Volcano plot of huBM and huCB CRISPR/Cas9 <span class="html-italic">MLL</span>r cells after volasertib treatment highlighting downregulated (blue) and upregulated (red) DEGs. Dotted lines indicate significant thresholds (pFDR ≤ 0.05, |log2(fold-change)| ≥  0.5). (<b>B</b>) log2(fold-change) of DEGs are shown for <span class="html-italic">PLK-1</span> and associated genes, normalized to own vehicle control (DMSO). Black * p_adjust value, white * p_nominal value. huBM <span class="html-italic">MLL</span>r cells show an upregulation of PLK-1-related gene pattern, whereas the pattern of huCB <span class="html-italic">MLL</span>r is downregulated. (<b>C</b>) Interactome of significant altered normalized Reads per Kilobase Millions (nRPKMs) around <span class="html-italic">PLK-1</span> in huBM and huCB CRISPR/Cas9 <span class="html-italic">MLL</span>r cells reveals potential feedback mechanism. Upregulation (red), downregulation (blue). Additional RNA-seq analysis regarding phosphorylation activity and targets around <span class="html-italic">PLK-1</span> are highlighted with red arrows. (<b>D</b>) Fold change of <span class="html-italic">PLK-1</span>, <span class="html-italic">BORA</span>, <span class="html-italic">AURKA,</span> and <span class="html-italic">FOXM1</span> in huBM and huCB CRISPR/Cas9 <span class="html-italic">MLL</span>r cells (<span class="html-italic">n</span> = 4/<span class="html-italic">n</span> = 3), as well as NOMO-1 and THP-1 cells (<span class="html-italic">n</span> = 3/<span class="html-italic">n</span> = 3), after 72 h 50 nM volasertib treatment compared to vehicle control (DMSO) measured by RT-qPCR. One-way ANOVA. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 5
<p><b>Combined treatment of volasertib and alisertib in <span class="html-italic">MLL</span>r cells.</b> (<b>A</b>) huCB <span class="html-italic">MLL</span>r and huBM <span class="html-italic">MLL</span>r (<span class="html-italic">n</span> = 3/<span class="html-italic">n</span> = 3) cells were treated for 72 h with increasing concentrations of volasertib alone, alisertib alone, or in combination. The percentage of viable cells (Annexin V-, PI-) was determined by flow cytometry. In the isobologram, IC50 values were mapped, and the Chou–Talalay method was used to measure the CI for the identification of synergistic effects. CI: huBM <span class="html-italic">MLL</span>r 0.25, huCB <span class="html-italic">MLL</span>r 1.52. (<b>B</b>) Representative (<b>left</b>) and pooled (<b>right</b>) data of BrdU cell cycle analysis of huBM <span class="html-italic">MLL</span>r cells (<span class="html-italic">n</span> = 3) after combinatorial treatment with volasertib and alisertib for 72 h (DMSO vehicle control, 25 nM V + 5 µM A, 100 nM V + 50 µM A). This shows a significant increase in G2/M-phase and apoptotic cells and a decrease in S-phase. Normalized to respective vehicle control (DMSO). One-way ANOVA. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 5 Cont.
<p><b>Combined treatment of volasertib and alisertib in <span class="html-italic">MLL</span>r cells.</b> (<b>A</b>) huCB <span class="html-italic">MLL</span>r and huBM <span class="html-italic">MLL</span>r (<span class="html-italic">n</span> = 3/<span class="html-italic">n</span> = 3) cells were treated for 72 h with increasing concentrations of volasertib alone, alisertib alone, or in combination. The percentage of viable cells (Annexin V-, PI-) was determined by flow cytometry. In the isobologram, IC50 values were mapped, and the Chou–Talalay method was used to measure the CI for the identification of synergistic effects. CI: huBM <span class="html-italic">MLL</span>r 0.25, huCB <span class="html-italic">MLL</span>r 1.52. (<b>B</b>) Representative (<b>left</b>) and pooled (<b>right</b>) data of BrdU cell cycle analysis of huBM <span class="html-italic">MLL</span>r cells (<span class="html-italic">n</span> = 3) after combinatorial treatment with volasertib and alisertib for 72 h (DMSO vehicle control, 25 nM V + 5 µM A, 100 nM V + 50 µM A). This shows a significant increase in G2/M-phase and apoptotic cells and a decrease in S-phase. Normalized to respective vehicle control (DMSO). One-way ANOVA. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">
20 pages, 3077 KiB  
Article
An Integrated Framework to Identify Prognostic Biomarkers and Novel Therapeutic Targets in Hepatocellular Carcinoma-Based Disabilities
by Md. Okibur Rahman, Asim Das, Nazratun Naeem, Jabeen-E-Tahnim, Md. Ali Hossain, Md. Nur Alam, AKM Azad, Salem A. Alyami, Naif Alotaibi, A. S. Al-Moisheer and Mohammod Ali Moni
Biology 2024, 13(12), 966; https://doi.org/10.3390/biology13120966 - 24 Nov 2024
Viewed by 1009
Abstract
Hepatocellular carcinoma (HCC) is one of the most prevalent malignant tumors globally, significantly affecting liver functions, thus necessitating the identification of biomarkers and effective therapeutics to improve HCC-based disabilities. This study aimed to identify prognostic biomarkers, signaling cascades, and candidate drugs for the [...] Read more.
Hepatocellular carcinoma (HCC) is one of the most prevalent malignant tumors globally, significantly affecting liver functions, thus necessitating the identification of biomarkers and effective therapeutics to improve HCC-based disabilities. This study aimed to identify prognostic biomarkers, signaling cascades, and candidate drugs for the treatment of HCC through integrated bioinformatics approaches such as functional enrichment analysis, survival analysis, molecular docking, and simulation. Differential expression and functional enrichment analyses revealed 176 common differentially expressed genes from two microarray datasets, GSE29721 and GSE49515, significantly involved in HCC development and progression. Topological analyses revealed 12 hub genes exhibiting elevated expression in patients with higher tumor stages and grades. Survival analyses indicated that 11 hub genes (CCNB1, AURKA, RACGAP1, CEP55, SMC4, RRM2, PRC1, CKAP2, SMC2, UHRF1, and FANCI) and three transcription factors (E2F1, CREB1, and NFYA) are strongly linked to poor patient survival. Finally, molecular docking and simulation identified seven candidate drugs with stable complexes to their target proteins: tozasertib (−9.8 kcal/mol), tamatinib (−9.6 kcal/mol), ilorasertib (−9.5 kcal/mol), hesperidin (−9.5 kcal/mol), PF−562271 (−9.3 kcal/mol), coumestrol (−8.4 kcal/mol), and clofarabine (−7.7 kcal/mol). These findings suggest that the identified hub genes and TFs could serve as valuable prognostic biomarkers and therapeutic targets for HCC-based disabilities. Full article
(This article belongs to the Special Issue Multi-omics in Oncology: Discovering Novel Biomarkers and Targets)
Show Figures

Figure 1

Figure 1
<p>Distribution of differentially expressed genes (<b>a</b>) volcano plot for GSE29721; and (<b>b</b>) GSE49515; red dots, blue dots, and black dots indicate up-regulated genes, downregulated genes, and not significantly expressed genes in HCC compared to normal tissue, respectively. (<span class="html-italic">p</span> &lt; 0.05, |log2FC| &gt; 1) (<b>c</b>) Venn diagram showing the overlapping CDEGs between the datasets.</p>
Full article ">Figure 2
<p>Enrichment analysis of up-regulated CDEGs (<b>a</b>) the top 5 enriched GO terms of the BP, CC, and MF categories of up-regulated DEGs; (<b>b</b>) the molecular pathways enriched by up-regulated DEGs in HCC patients.</p>
Full article ">Figure 3
<p>Identification of hub genes through protein–protein interaction network (<b>a</b>) PPI network of CDEGs. The nodes in hexagonal shapes represent identified hub genes; (<b>b</b>) the Venn diagram shows the intersection of 12 hub genes from four cytoHubba ranking methods.</p>
Full article ">Figure 4
<p>Expression levels of 12 hub genes in primary liver tumors (<span class="html-italic">n</span> = 371) compared to normal samples (n = 50) from TCGA data.</p>
Full article ">Figure 5
<p>Depiction of expression patterns of three transcription factors: (<b>a</b>) E2F1, (<b>b</b>) NF-YA, and (<b>c</b>) CREB1, highlighting the upregulation of these transcription factors in liver tumors (red) compared to adjacent normal tissues (blue). Sub-figures (<b>d</b>–<b>f</b>) illustrate survival analyses for patients with high expression levels of the TFs E2F1, NF-YA, and CREB1, indicating worse overall survival in liver cancer patients with higher expression levels.</p>
Full article ">Figure 6
<p>Overall survival (OS) analysis of 12 potential hub genes. <span class="html-italic">p</span> &lt; 0.05 indicates a statistically significant difference in mortality between groups. Sub-figures (<b>a</b>–<b>l</b>) demonstrate that patients in the high-expression group experience significantly worse overall survival compared to the low-expression group, except for SMC3 (<b>c</b>), which is not statistically significant (log-rank <span class="html-italic">p</span> = 0.33). HR: hazard ratio of the two groups.</p>
Full article ">Figure 7
<p>Expression of twelve hub genes in hepatocellular carcinoma patients with different tumor grades using TCGA samples. All boxplots illustrate that patients with higher tumor grades exhibit elevated gene expression in different colors.</p>
Full article ">
20 pages, 2121 KiB  
Review
The ncRNA-AURKA Interaction in Hepatocellular Carcinoma: Insights into Oncogenic Pathways, Therapeutic Opportunities, and Future Challenges
by Clarissa Joy C. Garcia, Luca Grisetti, Claudio Tiribelli and Devis Pascut
Life 2024, 14(11), 1430; https://doi.org/10.3390/life14111430 - 6 Nov 2024
Viewed by 952
Abstract
Hepatocellular carcinoma (HCC) represents a major public health concern and ranks among the leading cancer-related mortalities globally. Due to the frequent late-stage diagnosis of HCC, therapeutic options remain limited. Emerging evidence highlights the critical role of non-coding RNAs (ncRNAs) in the regulation of [...] Read more.
Hepatocellular carcinoma (HCC) represents a major public health concern and ranks among the leading cancer-related mortalities globally. Due to the frequent late-stage diagnosis of HCC, therapeutic options remain limited. Emerging evidence highlights the critical role of non-coding RNAs (ncRNAs) in the regulation of Aurora kinase A (AURKA), one of the key hub genes involved in several key cancer pathways. Indeed, the dysregulated interaction between ncRNAs and AURKA contributes to tumor development, progression, and therapeutic resistance. This review delves into the interplay between ncRNAs and AURKA and their role in hepatocarcinogenesis. Recent findings underscore the involvement of the ncRNAs and AURKA axis in tumor development and progression. Furthermore, this review also discusses the clinical significance of targeting ncRNA-AURKA axes, offering new perspectives that could lead to innovative therapeutic strategies aimed at improving outcomes for HCC patients. Full article
Show Figures

Figure 1

Figure 1
<p><b>The multifaceted roles of AURKA.</b> Under physiological conditions (green), AURKA plays a key function in mitotic progression, including G2/M checkpoint release, mitotic spindle formation, organization, and epigenetic regulation. It also contributes to the disassembly of primary cilia, initiation of DNA replication, and regulation of mitochondrial fission and energy production. AURKA interacts with numerous proteins and participates in diverse signaling pathways; thus, its overexpression in cancer leads to the dysregulation of these pathways, driving oncogenic effects. In the context of cancer (red), AURKA enhances cell survival and proliferation, epithelial-mesenchymal transition (EMT), and cancer invasiveness.</p>
Full article ">Figure 2
<p><b>Regulatory ncRNAs: From physiology to pathology.</b> Regulatory ncRNAs are a fraction of the ncRNAs within the human genome. MicroRNAs (miRNAs), long-non-coding RNAs (lncRNAs), and circular RNAs (circRNAs) are the major players involved in regulating gene expression and protein levels and stability in physiological and pathological conditions.</p>
Full article ">Figure 3
<p><b>The regulatory network of AURKA and ncRNAs in HCC</b>. Regulatory ncRNAs influence the expression of AURKA both at the mRNA and protein levels in HCC. MiRNAs targeting AURKA, such as miR-199-3p [<a href="#B96-life-14-01430" class="html-bibr">96</a>], miR-124-3p [<a href="#B92-life-14-01430" class="html-bibr">92</a>], miR-490-3p [<a href="#B89-life-14-01430" class="html-bibr">89</a>], miR-129-3p [<a href="#B99-life-14-01430" class="html-bibr">99</a>], and miR-26a-5p [<a href="#B101-life-14-01430" class="html-bibr">101</a>], are frequently downregulated in HCC. Transcriptomic analyses revealed upregulation of miR-1269b, miR-518d, and miR-6728 and downregulation of miR-139 and miR-4800 [<a href="#B95-life-14-01430" class="html-bibr">95</a>]. AURKA indirectly upregulates miR-21 expression by activating NF-κB signaling. The p50/p65 complex binds to the miR-21 promoter sequence, thus promoting its transcription. In turn, miR-21 represses PTEN, the negative regulator of the PI3K/AKT pathway [<a href="#B24-life-14-01430" class="html-bibr">24</a>]. LncRNAs such as <span class="html-italic">MALAT1</span> [<a href="#B104-life-14-01430" class="html-bibr">104</a>] and <span class="html-italic">TUG1</span> [<a href="#B105-life-14-01430" class="html-bibr">105</a>] and the circRNA circHMGS1 [<a href="#B106-life-14-01430" class="html-bibr">106</a>] positively regulate AURKA expression, while <span class="html-italic">KDM4A-AS1</span> forms a complex with ILF3 to recruit and stabilize AURKA [<a href="#B20-life-14-01430" class="html-bibr">20</a>]. M6A-mediated hypermethylation of <span class="html-italic">TIALD1</span> effectively blocks AURKA protein from lysosomal degradation [<a href="#B107-life-14-01430" class="html-bibr">107</a>].</p>
Full article ">
20 pages, 1417 KiB  
Review
Progress in Precision Medicine for Head and Neck Cancer
by Sanaz Vakili, Amir Barzegar Behrooz, Rachel Whichelo, Alexandra Fernandes, Abdul-Hamid Emwas, Mariusz Jaremko, Jarosław Markowski, Marek J. Los, Saeid Ghavami and Rui Vitorino
Cancers 2024, 16(21), 3716; https://doi.org/10.3390/cancers16213716 - 4 Nov 2024
Viewed by 1925
Abstract
This paper presents a comprehensive comparative analysis of biomarkers for head and neck cancer (HNC), a prevalent but molecularly diverse malignancy. We detail the roles of key proteins and genes in tumourigenesis and progression, emphasizing their diagnostic, prognostic, and therapeutic relevance. Our bioinformatic [...] Read more.
This paper presents a comprehensive comparative analysis of biomarkers for head and neck cancer (HNC), a prevalent but molecularly diverse malignancy. We detail the roles of key proteins and genes in tumourigenesis and progression, emphasizing their diagnostic, prognostic, and therapeutic relevance. Our bioinformatic validation reveals crucial genes such as AURKA, HMGA2, MMP1, PLAU, and SERPINE1, along with microRNAs (miRNA), linked to HNC progression. OncomiRs, including hsa-miR-21-5p, hsa-miR-31-5p, hsa-miR-221-3p, hsa-miR-222-3p, hsa-miR-196a-5p, and hsa-miR-200c-3p, drive tumourigenesis, while tumour-suppressive miRNAs like hsa-miR-375 and hsa-miR-145-5p inhibit it. Notably, hsa-miR-155-3p correlates with survival outcomes in addition to the genes RAI14, S1PR5, OSBPL10, and METTL6, highlighting its prognostic potential. Future directions should focus on leveraging precision medicine, novel therapeutics, and AI integration to advance personalized treatment strategies to optimize patient outcomes in HNC care. Full article
(This article belongs to the Collection Advances in Diagnostics and Treatment of Head and Neck Cancer)
Show Figures

Figure 1

Figure 1
<p>Epidemiological aspects of head and neck cancer (created with BioRender.com).</p>
Full article ">Figure 2
<p>OncomiRs and tumour-suppressive miRNAs: key regulators in head and neck cancer progression. This figure illustrates the dual roles of oncomiRs and tumour-suppressive miRNAs in head and neck cancer. OncomiRs promote tumour growth and metastasis, while tumour-suppressive miRNAs inhibit cancer progression. Together, these miRNAs orchestrate the molecular pathways in cell proliferation, apoptosis, and angiogenesis (Created with BioRender.com).</p>
Full article ">
16 pages, 18536 KiB  
Article
Molecular Landscape of Bladder Cancer: Key Genes, Transcription Factors, and Drug Interactions
by Danishuddin, Md Azizul Haque, Shawez Khan, Jong-Joo Kim and Khurshid Ahmad
Int. J. Mol. Sci. 2024, 25(20), 10997; https://doi.org/10.3390/ijms252010997 - 12 Oct 2024
Viewed by 1677
Abstract
Bladder cancer is among the most prevalent tumors in the urinary system and is known for its high malignancy. Although traditional diagnostic and treatment methods are established, recent research has focused on understanding the molecular mechanisms underlying bladder cancer. The primary objective of [...] Read more.
Bladder cancer is among the most prevalent tumors in the urinary system and is known for its high malignancy. Although traditional diagnostic and treatment methods are established, recent research has focused on understanding the molecular mechanisms underlying bladder cancer. The primary objective of this study is to identify novel diagnostic markers and discover more effective targeted therapies for bladder cancer. This study identified differentially expressed genes (DEGs) between bladder cancer tissues and adjacent normal tissues using data from The Cancer Genome Atlas (TCGA). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were conducted to explore the functional roles of these genes. A protein–protein interaction (PPI) network was also constructed to identify and analyze hub genes within this network. Gene set variation analysis (GSVA) was conducted to investigate the involvement of these genes in various biological processes and pathways. Ten key genes were found to be significantly associated with bladder cancer: IL6, CCNA2, CCNB1, CDK1, PLK1, TOP2A, AURKA, AURKB, FOXM1, and CALML5. GSVA analyses revealed that these genes are involved in a variety of biological processes and signaling pathways, including coagulation, UV-response-down, apoptosis, Notch signaling, and Wnt/beta-catenin signaling. The diagnostic relevance of these genes was validated through ROC curve analysis. Additionally, potential therapeutic drug interactions with these key genes were identified. This study provides valuable insights into key genes and their roles in bladder cancer. The identified genes and their interactions with therapeutic drugs could serve as potential biomarkers, presenting new opportunities for enhancing the diagnosis and prognosis of bladder cancer. Full article
(This article belongs to the Special Issue Integrative Multi-Omics Analysis for Cancer Biomarkers)
Show Figures

Figure 1

Figure 1
<p>Screening and identification of key genes in the TCGA–BLCA dataset. (<b>A</b>) Volcano plots showing differentially expressed genes (DEGs) from the TCGA–BLCA dataset. Upregulated genes are shown in yellow, downregulated genes in violet, and non-significant genes in grey. (<b>B</b>) The top 10 hub genes were selected based on degree centrality using the CytoHubba app in Cytoscape. The color indicates the degree of interaction, with more intense red representing higher interaction, while orange and yellow denote intermediate and lower interaction levels, respectively. (<b>C</b>) Correlation analysis among the hub genes.</p>
Full article ">Figure 2
<p>Top modules within the PPI network. These two modules were identified using the MCODE algorithm in Cytoscape with a K-core value of 4, a node score cutoff of 0.3, and a maximum depth of 100, including their interacting gene partners.</p>
Full article ">Figure 3
<p>ROC curves for the significant gene expression data. The AUC values suggest that the expression analysis of these markers can effectively differentiate between patient groups with different diseases and controls.</p>
Full article ">Figure 4
<p>The box plots display the expression levels of several of the ten hub genes that exhibit statistically significant differential expression between normal and bladder cancer samples (** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001; NS: Not Significant).</p>
Full article ">Figure 5
<p>Gene Ontology analyses of upregulated genes include biological process (BP), cellular component (CC), and molecular function (MF), along with Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways.</p>
Full article ">Figure 6
<p>Gene Ontology analyses of downregulated genes include biological process (BP), cellular component (CC), and molecular function (MF), along with Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways.</p>
Full article ">Figure 7
<p>The relationship between hub genes and disease-related genes. (<b>A</b>) Comparison of expression levels for various disease-related genes between control samples and individuals with bladder cancer. (<b>B</b>) A bubble plot illustrates the Pearson correlations between nine hub genes and disease-related genes.</p>
Full article ">Figure 8
<p>GSVA analysis of high and low expression.</p>
Full article ">Figure 8 Cont.
<p>GSVA analysis of high and low expression.</p>
Full article ">Figure 9
<p>The hub gene–transcription factor (TF) regulatory network. The regulatory network of hub genes and transcription factors (TFs) was obtained using the NetworkAnalyst 3.0 web server. Square nodes represent TFs, and circle nodes stand for hub genes.</p>
Full article ">
15 pages, 1486 KiB  
Review
Contribution of AurkA/TPX2 Overexpression to Chromosomal Imbalances and Cancer
by Federica Polverino, Anna Mastrangelo and Giulia Guarguaglini
Cells 2024, 13(16), 1397; https://doi.org/10.3390/cells13161397 - 22 Aug 2024
Viewed by 1402
Abstract
The AurkA serine/threonine kinase is a key regulator of cell division controlling mitotic entry, centrosome maturation, and chromosome segregation. The microtubule-associated protein TPX2 controls spindle assembly and is the main AurkA regulator, contributing to AurkA activation, localisation, and stabilisation. Since their identification, AurkA [...] Read more.
The AurkA serine/threonine kinase is a key regulator of cell division controlling mitotic entry, centrosome maturation, and chromosome segregation. The microtubule-associated protein TPX2 controls spindle assembly and is the main AurkA regulator, contributing to AurkA activation, localisation, and stabilisation. Since their identification, AurkA and TPX2 have been described as being overexpressed in cancer, with a significant correlation with highly proliferative and aneuploid tumours. Despite the frequent occurrence of AurkA/TPX2 co-overexpression in cancer, the investigation of their involvement in tumorigenesis and cancer therapy resistance mostly arises from studies focusing only on one at the time. Here, we review the existing literature and discuss the mitotic phenotypes described under conditions of AurkA, TPX2, or AurkA/TPX2 overexpression, to build a picture that may help clarify their oncogenic potential through the induction of chromosome instability. We highlight the relevance of the AurkA/TPX2 complex as an oncogenic unit, based on which we discuss recent strategies under development that aim at disrupting the complex as a promising therapeutic perspective. Full article
Show Figures

Figure 1

Figure 1
<p>Mechanisms through which AurkA overexpression can yield chromosome segregation defects and aneuploidy by acting at the level of centrosomes (top-left box), MTs (bottom-left box), and centromeres/KTs (right box) are summarised. Described targets are also indicated.</p>
Full article ">Figure 2
<p>Effects of overexpressing TPX2 alone (<b>A</b>) or the whole AurkA/TPX2 complex (<b>B</b>). PP6 mutations harboured by cancer cells or PP6 depletion yield a cellular effect comparable to AurkA/TPX2 overexpression, by impairing the dephosphorylation of AurkA Thr288 within the AurkA/TPX2 complex (<b>B</b>). The distribution of TPX2, AurkA, and auto-phosphorylated AurkA (p-Aurka, Thr288) on the mitotic spindle is schematised on the left in each panel (a half spindle is sketched; the centrosome is in light blue, chromosomes in brown, the black lines represent MTs).</p>
Full article ">Figure 3
<p>PPI inhibitors described to target the AurkA/TPX2 complex and tested in cultured cells are listed, together with their structures and observed effects. The AurkA hydrophobic pocket binding residues 8–10 of TPX2, as well as the AurkinA, C20/23, and 6h compounds, is shown on the left [<a href="#B114-cells-13-01397" class="html-bibr">114</a>,<a href="#B117-cells-13-01397" class="html-bibr">117</a>,<a href="#B118-cells-13-01397" class="html-bibr">118</a>,<a href="#B119-cells-13-01397" class="html-bibr">119</a>].</p>
Full article ">
18 pages, 14149 KiB  
Article
Aurora Kinase A Inhibition Potentiates Platinum and Radiation Cytotoxicity in Non-Small-Cell Lung Cancer Cells and Induces Expression of Alternative Immune Checkpoints
by Huijie Liu, Ayse Ece Cali Daylan, Jihua Yang, Ankit Tanwar, Alain Borczuk, Dongwei Zhang, Vincent Chau, Shenduo Li, Xuan Ge, Balazs Halmos, Xingxing Zang and Haiying Cheng
Cancers 2024, 16(16), 2805; https://doi.org/10.3390/cancers16162805 - 9 Aug 2024
Cited by 1 | Viewed by 1300
Abstract
Despite major advances in non-small-cell lung cancer (NSCLC) treatment, the five-year survival rates for patients with non-oncogene-driven tumors remain low, necessitating combinatory approaches to improve outcomes. Our prior high-throughput RNAi screening identified Aurora kinase A (AURKA) as a potential key player in cisplatin [...] Read more.
Despite major advances in non-small-cell lung cancer (NSCLC) treatment, the five-year survival rates for patients with non-oncogene-driven tumors remain low, necessitating combinatory approaches to improve outcomes. Our prior high-throughput RNAi screening identified Aurora kinase A (AURKA) as a potential key player in cisplatin resistance. In this study, we investigated AURKA’s role in platinum and radiation sensitivity in multiple NSCLC cell lines and xenograft mouse models, as well as its effect on immune checkpoints, including PD-L1, B7x, B7-H3, and HHLA2. Of 94 NSCLC patient tumor specimens, 91.5% tested positive for AURKA expression, with 34% showing moderate-to-high levels. AURKA expression was upregulated following cisplatin treatment in NSCLC cell lines PC9 and A549. Both AURKA inhibition by alisertib and inducible AURKA knockdown potentiated the cytotoxic effects of cisplatin and radiation, leading to tumor regression in doxycycline-inducible xenograft mice. Co-treated cells exhibited increased DNA double-strand breaks, apoptosis, and senescence. Additionally, AURKA inhibition alone by alisertib increased PD-L1 and B7-H3 expression. In conclusion, our study demonstrates that AURKA inhibition enhances the efficacy of platinum-based chemotherapy in NSCLC cells and modulates the expression of multiple immune checkpoints. Therefore, combinatory regimens with AURKA inhibitors should be strategically designed and further studied within the evolving landscape of chemo-immunotherapy. Full article
Show Figures

Figure 1

Figure 1
<p>AURKA expression in lung cancer. Representative AURKA IHC staining of tissue microarrays.</p>
Full article ">Figure 2
<p>Effect of AURKA knockdown in combination with cisplatin treatment. (<b>A</b>) Aurora A protein expression by Western blot increases with cisplatin treatment. (<b>B</b>) Aurora A protein expression decreases when doxycycline-inducible cell lines are treated with varying amounts of doxycycline as detected by Western blot. (<b>C</b>) Cell viability by MTS assay reveals that doxycycline-inducible Aurora A knockdown leads to decreased cell viability in PC9A8 cell line. Addition of cisplatin to Aurora A knockdown works synergistically to decrease cell viability (results for A549A8 can be found in the <a href="#app1-cancers-16-02805" class="html-app">Supplementary Figure S1</a>). (<b>D</b>) Clonogenic assay reveals that doxycycline-inducible Aurora A knockdown reduces colony formation in the PC9A8 cell line. The addition of cisplatin to Aurora A knockdown works synergistically to decrease tumor cell colony formation (results for A549A8 can be found in the <a href="#app1-cancers-16-02805" class="html-app">Supplementary Figure S1</a>). (<b>E</b>) Xenograft studies reveal that both cisplatin and Aurora A knockdown when used alone slow down tumor growth. When Aurora A knockdown is combined with cisplatin, actual tumor size reduction can be noted. The following notation was used to symbolize statistical significance in the figures: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01. Δ is used to denote a synergistic effect of combined treatments.</p>
Full article ">Figure 3
<p>Inhibition of AURKA sensitizes lung cancer cells to cisplatin and radiation. (<b>A</b>) Western blotting: The expression of phospho-Aurora and total AURKA are shown with and without treatment of MLN8237 in H1703, PC9, A549, and H460 individually. (<b>B</b>) Flow cytometry demonstrating cell cycle stages of PC9 cells without treatment in the upper panel and with MLN8237 treatment in the lower panel. (<b>C</b>) The percentage of PC9 cells in G0-1, S, G2-M, sub-G1 cycle stage per flow cytometry with and without treatment of MLN8237. (<b>D</b>) The cell viability of PC9 cells after being treated with MLN8237 and/or cisplatin. (<b>E</b>) The cell viability of A549 cells after being treated with MLN8237 and/or cisplatin. (<b>F</b>) The surviving fraction of PC9 cells with and without treatment of doxycycline and/or radiation; (<b>G</b>) the surviving fraction of A549 cells with and without treatment of doxycycline and/or radiation; (<b>H</b>) the surviving fraction of PC9 cells with and without treatment of MLN8237 and/or radiation; (<b>I</b>) the surviving fraction of A549 cells with and without treatment of MLN8237 and/or radiation. The following notation was used to symbolize statistical significance in the figures: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01. Δ is used to denote a synergistic effect of combined treatments.</p>
Full article ">Figure 4
<p>Blocking the function of AURKA increases cisplatin-induced DNA double-strand breaks. (<b>A</b>) Expression of AURKA, γ-H2AX, and GADPH in PC9A8 cells and A549A8 with and without cisplatin and doxycycline treatment by Western blot. (<b>B</b>) Expression of AURKA, γ-H2AX, and GADPH in PC9 cells and A549 with and without cisplatin and MLN8237 treatment by Western blot. (<b>C</b>) Expression of AURKA, γ-H2AX, and GADPH in PC9A8 and A549A8 cells with and without radiation and doxycycline treatment by Western blot. (<b>D</b>) Expression of AURKA, γ-H2AX, and GADPH in PC9 and A549 cells with and without radiation and MLN8237 treatment. The ratios of band intensities divided by GAPDH are noted at the top of each protein band.</p>
Full article ">Figure 5
<p>Knockdown of AURKA increases apoptosis induced by cisplatin or radiation. (<b>A</b>) Expressions of C-PARP, AURKA, and GADPH were shown in PC9 and A549 cells with and without the treatment of cisplatin and/or doxycycline for 48 h. (<b>B</b>) Expressions of C-PARP, AURKA, and GADPH were shown in PC9 and A549 cells (with and without the treatment of radiation and/or doxycycline for 48 h). The ratios of band intensities divided by GAPDH are noted at the top of each protein band.</p>
Full article ">Figure 6
<p>Inhibiting AURKA by MLN8237 induces apoptosis and senescence. (<b>A</b>) Western blotting: the expression of cleaved PARP and GADPH in PC9 and A549 cells with and without treatment of cisplatin and/or MLN8237 for 48 h. (<b>B</b>) Representative picture of staining of senescence-associated beta-galactosidase in PC9 and A549 cells with and without treatment of cisplatin and/or MLN8237. (<b>C</b>) Quantitative analysis of staining of senescence-associated beta-galactosidase in PC9 and A549 cells with and without treatment of cisplatin and/or MLN8237.</p>
Full article ">Figure 7
<p>(<b>A</b>) Flow cytometric analysis of PD-L1, B7x, HHLA2, and B7H3 in the A549 cell line after treatment with 1 μM of MLN8237 for 72 h. (<b>B</b>) Flow cytometric analysis of PD-L1, HHLA2, and B7H3 in A549 cell line after treatment with 1 μM of MLN8237 for 72 h. The following notation was used to symbolize statistical significance in the figures: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 and **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">
13 pages, 1968 KiB  
Article
High-Intensity Focused Ultrasound Increases Facial Adipogenesis in a Swine Model via Modulation of Adipose-Derived Stem Cell Cilia
by Kyung-A Byun, Hyoung Moon Kim, Seyeon Oh, Sosorburam Batsukh, Sangsu Lee, Myungjune Oh, Jeongwoo Lee, Ran Lee, Jae Woo Kim, Seung Min Oh, Jisun Kim, Geebum Kim, Hyun Jun Park, Hanbit Hong, Jehyuk Lee, Sang-Hyun An, Sung Suk Oh, Yeon-Seop Jung, Kuk Hui Son and Kyunghee Byun
Int. J. Mol. Sci. 2024, 25(14), 7648; https://doi.org/10.3390/ijms25147648 - 12 Jul 2024
Viewed by 1637
Abstract
Decreased medial cheek fat volume during aging leads to loss of a youthful facial shape. Increasing facial volume by methods such as adipose-derived stem cell (ASC) injection can produce facial rejuvenation. High-intensity focused ultrasound (HIFU) can increase adipogenesis in subcutaneous fat by modulating [...] Read more.
Decreased medial cheek fat volume during aging leads to loss of a youthful facial shape. Increasing facial volume by methods such as adipose-derived stem cell (ASC) injection can produce facial rejuvenation. High-intensity focused ultrasound (HIFU) can increase adipogenesis in subcutaneous fat by modulating cilia on ASCs, which is accompanied by increased HSP70 and decreased NF-κB expression. Thus, we evaluated the effect of HIFU on increasing facial adipogenesis in swine (n = 2) via modulation of ASC cilia. Expression of CD166, an ASC marker, differed by subcutaneous adipose tissue location. CD166 expression in the zygomatic arch (ZA) was significantly higher than that in the subcutaneous adipose tissue in the mandible or lateral temporal areas. HIFU was applied only on the right side of the face, which was compared with the left side, where HIFU was not applied, as a control. HIFU produced a significant increase in HSP70 expression, decreased expression of NF-κB and a cilia disassembly factor (AURKA), and increased expression of a cilia increasing factor (ARL13B) and PPARG and CEBPA, which are the main regulators of adipogenesis. All of these changes were most prominent at the ZA. Facial adipose tissue thickness was also increased by HIFU. Adipose tissue volume, evaluated by magnetic resonance imaging, was increased by HIFU, most prominently in the ZA. In conclusion, HIFU increased ASC marker expression, accompanied by increased HSP70 and decreased NF-κB expression. Additionally, changes in cilia disassembly and length and expression of adipogenesis were observed. These results suggest that HIFU could be used to increase facial volume by modulating adipogenesis. Full article
(This article belongs to the Section Biochemistry)
Show Figures

Figure 1

Figure 1
<p>Regulation of ASC markers by HIFU treatment in various types of subcutaneous adipose tissue. (<b>A</b>) Schematic of swine used to assess HIFU regarding increases in subcutaneous adipose tissue thickness. (<b>B</b>) Schematic of various types of subcutaneous adipose tissue in swine. (<b>C</b>) The mRNA level of <span class="html-italic">CD166</span> in the ZA, LT, MD, and FF was measured by qRT-PCR. (<b>D</b>) The mRNA level of <span class="html-italic">CD166</span> in HIFU-treated areas including the ZA, LT, and MD was measured by qRT-PCR. Data are presented as the mean ± SD of three independent experiments. ***, <span class="html-italic">p</span> &lt; 0.001, control vs. HIFU; <span>$</span>, <span class="html-italic">p</span> &lt; 0.05, <span>$</span><span>$</span>, <span class="html-italic">p</span> &lt; 0.01, and <span>$</span><span>$</span><span>$</span>, <span class="html-italic">p</span> &lt; 0.001, vs. ZA; ##, <span class="html-italic">p</span> &lt; 0.01 and ###, <span class="html-italic">p</span> &lt; 0.001, vs. LT; ††, <span class="html-italic">p</span> &lt; 0.01, vs. MD (Mann–Whitney U test). ASC, adipose-derived stem cell; FF, frontal forehead; HIFU, high-intensity focused ultrasound; LT, lateral temporal area; MD, mandible; qRT-PCR, quantitative reverse transcription–polymerase chain reaction; ZA, zygomatic arch.</p>
Full article ">Figure 2
<p>Regulation of <span class="html-italic">HSP70</span> and <span class="html-italic">NF-κB</span> by HIFU treatment in various types of subcutaneous adipose tissue. (<b>A</b>,<b>B</b>) The mRNA levels of <span class="html-italic">HSP70</span> and <span class="html-italic">NF-κB</span> in HIFU-treated ZA, LT, and MD tissues were measured by qRT-PCR. Data are presented as the mean ± SD of three independent experiments. *, <span class="html-italic">p</span> &lt; 0.05 and **, <span class="html-italic">p</span> &lt; 0.01, control vs. HIFU; <span>$</span>, <span class="html-italic">p</span> &lt; 0.05 and <span>$</span><span>$</span>, <span class="html-italic">p</span> &lt; 0.01, vs. ZA; #, <span class="html-italic">p</span> &lt; 0.05 and ##, <span class="html-italic">p</span> &lt; 0.01, vs. LT (Mann–Whitney U test). HIFU, high-intensity focused ultrasound; HSP70, heat shock protein 70; LT, lateral temporal area; MD, mandible; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; qRT-PCR, quantitative reverse transcription–polymerase chain reaction; ZA, zygomatic arch.</p>
Full article ">Figure 3
<p>Regulation of cilia disassembly-related factors, cilia length increasing factors, and adipogenesis factors by HIFU treatment in various types of subcutaneous adipose tissue. (<b>A</b>,<b>B</b>) The mRNA levels of <span class="html-italic">AURKA</span> and <span class="html-italic">ARL13B</span> in HIFU-treated ZA, LT, and MD tissues were measured by qRT-PCR. (<b>C</b>,<b>D</b>) The mRNA levels of <span class="html-italic">WNT5A</span> and <span class="html-italic">CTNNB1</span> in HIFU-treated ZA, LT, and MD tissues were measured by qRT-PCR. (<b>E</b>,<b>F</b>) The mRNA levels of <span class="html-italic">PPARG</span> and <span class="html-italic">CEBPA</span> in HIFU-treated ZA, LT, and MD tissues were measured by qRT-PCR. Data are presented as the mean ± SD of three independent experiments. *, <span class="html-italic">p</span> &lt; 0.05, **, <span class="html-italic">p</span> &lt; 0.01, and ***, <span class="html-italic">p</span> &lt; 0.001, control vs. HIFU; <span>$</span>, <span class="html-italic">p</span> &lt; 0.05, <span>$</span><span>$</span>, <span class="html-italic">p</span> &lt; 0.01, and <span>$</span><span>$</span><span>$</span>, <span class="html-italic">p</span> &lt; 0.001, vs. ZA; #, <span class="html-italic">p</span> &lt; 0.05 and ##, <span class="html-italic">p</span> &lt; 0.01, vs. LT (Mann–Whitney U test). ARL13B, ADP-ribosylation factor-like protein 13B; AURKA, aurora kinase A; CEBPA, CCAAT/enhancer binding protein α; CTNNB1, catenin beta 1; HIFU, high-intensity focused ultrasound; LT, lateral temporal area; MD, mandible; PPARG, peroxisome proliferator-activated receptor γ; qRT-PCR, quantitative reverse transcription–polymerase chain reaction; WNT5A, Wnt family member 5A; ZA, zygomatic arch.</p>
Full article ">Figure 4
<p>Regulation of adipogenesis by HIFU treatment in various types of subcutaneous adipose tissue. (<b>A</b>–<b>C</b>) The fat layer thickness and adipocyte size in HIFU-treated ZA, LT, and MD tissues were measured by hematoxylin and eosin staining. Scale bar = 1 mm (<b>D</b>) Two-dimensional cross-sectional left (control) and right (HIFU) images of magnetic resonance imaging (MRI) before (day 0) and 28 days after HIFU application (day 28). The red dashed line is an imaginary line to distinguish control and HIFU. (<b>E</b>) The fat volume in HIFU-treated ZA, LT, and MD tissues was measured by MRI. Data are presented as the mean ± SD of three independent experiments. *, <span class="html-italic">p</span> &lt; 0.05 and **, <span class="html-italic">p</span> &lt; 0.01, control vs. HIFU; <span>$</span>, <span class="html-italic">p</span> &lt; 0.05 and <span>$</span><span>$</span>, <span class="html-italic">p</span> &lt; 0.01, vs. ZA; #, <span class="html-italic">p</span> &lt; 0.05, vs. LT; §, <span class="html-italic">p</span> &lt; 0.05, day 0 vs. day 28 (Mann–Whitney U test). HIFU, high-intensity focused ultrasound; LT, lateral temporal area; MD, mandible; ZA, zygomatic arch.</p>
Full article ">
17 pages, 11970 KiB  
Article
Suppressing PD-L1 Expression via AURKA Kinase Inhibition Enhances Natural Killer Cell-Mediated Cytotoxicity against Glioblastoma
by Trang T. T. Nguyen, Qiuqiang Gao, Jeong-Yeon Mun, Zhe Zhu, Chang Shu, Aaron Naim, Meri Rogava, Benjamin Izar, Mike-Andrew Westhoff, Georg Karpel-Massler and Markus D. Siegelin
Cells 2024, 13(13), 1155; https://doi.org/10.3390/cells13131155 - 6 Jul 2024
Viewed by 2112
Abstract
Immunotherapies have shown significant promise as an impactful strategy in cancer treatment. However, in glioblastoma multiforme (GBM), the most prevalent primary brain tumor in adults, these therapies have demonstrated lower efficacy than initially anticipated. Consequently, there is an urgent need for strategies to [...] Read more.
Immunotherapies have shown significant promise as an impactful strategy in cancer treatment. However, in glioblastoma multiforme (GBM), the most prevalent primary brain tumor in adults, these therapies have demonstrated lower efficacy than initially anticipated. Consequently, there is an urgent need for strategies to enhance the effectiveness of immune treatments. AURKA has been identified as a potential drug target for GBM treatment. An analysis of the GBM cell transcriptome following AURKA inhibition revealed a potential influence on the immune system. Our research revealed that AURKA influenced PD-L1 levels in various GBM model systems in vitro and in vivo. Disrupting AURKA function genetically led to reduced PD-L1 levels and increased MHC-I expression in both established and patient-derived xenograft GBM cultures. This process involved both transcriptional and non-transcriptional pathways, partly implicating GSK3β. Interfering with AURKA also enhanced NK-cell-mediated elimination of GBM by reducing PD-L1 expression, as evidenced in rescue experiments. Furthermore, using a mouse model that mimics GBM with patient-derived cells demonstrated that Alisertib decreased PD-L1 expression in living organisms. Combination therapy involving anti-PD-1 treatment and Alisertib significantly prolonged overall survival compared to vehicle treatment. These findings suggest that targeting AURKA could have therapeutic implications for modulating the immune environment within GBM cells. Full article
Show Figures

Figure 1

Figure 1
<p>AURKA stands out as a promising therapeutic target in the context of GBM. (<b>A</b>) The survival curve of patients (wild-type and mutated IDH1) with different levels of AURKA mRNA in the GBM TCGA database is displayed. The cutoff point was determined using GlioVis through maximally selected rank statistics; (<b>B</b>) The TCGA GBM dataset was examined to assess the AURKA expression levels in both normal brain tissue and GBM tissue, revealing mRNA expression levels; (<b>C</b>) GBM22, GBM12, GL261, and astrocyte cells were subjected to escalating doses of Alisertib (ranging from 10 nM to 200 μM) for a period of 72 h, after which the viability of the cells was assessed (<span class="html-italic">n</span> = 4). Statistical significance was assessed by student’s <span class="html-italic">t</span>-test. Data are shown as mean ± SD. **** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 2
<p>Inhibiting AURKA triggers an immune response in models of GBM. (<b>A</b>,<b>B</b>) Parental and chronically Alisertib-treated GBM22 cells underwent microarray analysis, followed by GSEA. An illustration of a volcano plot is presented in (A), where red dots highlight an increase in neutrophil-mediated immunity. Additionally, gene set enrichment analysis is depicted in (<b>B</b>). The data include normalized enrichment scores (NES) and FDR-q values for a sample size of 2; (<b>C</b>) Standard Western blot or protein capillary electrophoresis of GBM22, GBM12, GL261, and U87-EGFRvIII treated with increasing concentration of Alisertib for 24 h. Vinculin or actin is used as a loading control; (<b>D</b>) U251, U87, and U87-EGFRvIII cells were transduced with non-targeting shRNA or shARKA and the whole-cell protein lysates were subjected to standard Western blot; (<b>E</b>,<b>F</b>) U251 and U87-EGFRvIII cells were transduced with non-targeting shRNA or shARKA (shRNA) and labeled with PD-L1 antibody and analyzed by flow cytometry. The quantification is shown in F (<span class="html-italic">n</span> = 3); (<b>G</b>) Shown is the quantification of MHC1 level (MFI) of GBM22 and U251 cells treated with an increasing of Alisertib for 24 h; (<b>H</b>) GBM22, U251, and U87-EGFRvIII cells were transduced with non-targeting shRNA or shARKA and the whole-cell protein lysates were subjected to standard Western blot. Actin is used as a loading control. Statistical significance was assessed by ANOVA with Dunnett’s multiple comparison test. Data are shown as mean ± SD. * <span class="html-italic">p</span> &lt; 0.05, ***/**** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>AURKA regulates PD-L1 levels in part through GSK3β in GBM cells. (<b>A</b>) The protein capillary electrophoresis of GBM22 cells transfected with HA-EV (empty vector) or HA-PD-L1 for 24 h and were treated with increasing concentrations of Alisertib for 24 h. Vinculin was used as a loading control; (<b>B</b>) Real-time PCR analysis of PD-L1 mRNA levels of GBM22, GBM12, U251, U87, and U87-EGFRvIII cells treated with increasing concentration of Alisertib for 24 h (<span class="html-italic">n</span> = 4); (<b>C</b>) The protein capillary electrophoresis of GBM22 cells transfected with HA-EV (empty vector) or HA-PD-L1 for 24 h and were treated with 10 µM Alisertib for 24 h in the presence or absence of 5 µM MG132; (<b>D</b>) The protein capillary electrophoresis of GBM22 cells transfected with HA-EV (empty vector) or HA-PD-L1 for 24 h and were treated with 5 µM Alisertib for 24 h in the presence or absence of 2 µM CHIR 99021, GSK3β inhibitor; (<b>E</b>) GBM22 cells were transduced with non-targeting shRNA or shARKA and the whole-cell protein lysates were subjected to protein capillary electrophoresis; (<b>F</b>) The protein capillary electrophoresis of GBM22 cells transfected with HA-PD-L1 or HA-PD-L1-S3A for 24 h and were treated with increasing concentration of Alisertib for 24 h; (<b>G</b>) The protein capillary electrophoresis of GBM22 cells transfected with Aurora A-D274N, Aurora A-D274A, or Aurora A WT. Statistical significance was assessed by ANOVA with Dunnett’s multiple comparison test. Data are shown as mean ± SD. ** <span class="html-italic">p</span> &lt; 0.01, ***/**** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>Loss of function of AURKA enhances NK-cell mediated reduction in cellular viability of GBM cultures. (<b>A</b>,<b>B</b>) GBM22, U251, U87-EGFRvIII, and U87 cells were treated with increasing concentrations of Alisertib for 48 h, and the NK-92 MI cells (6×) were added for another 24 h. Then, cellular viability was analyzed by Crystal Violet (<span class="html-italic">n</span> = 4). Quantification is shown in B; (<b>C</b>,<b>D</b>) GBM22 and U251 cells were transduced with non-targeting shRNA or shARKA and treated with NK-92 MI cells (6×) for 24 h. Then, cellular viability was analyzed by Crystal Violet (<span class="html-italic">n</span> = 4). Quantification is shown in D. Statistical significance was assessed by ANOVA with Dunnett’s multiple comparison test. Images were captured using a 4x objective lens. A statistically significant difference was observed between GBM cells treated with NK cells alone and GBM cells exposed to NK-92 MI cells in combination with varying concentrations of Alisertib. Data are shown as mean ± SD. ***/**** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>PD-L1 inhibitor reduces GBM cell viability but has minimal effect on NK-92 MI cell-mediated killing. (<b>A</b>) Stable cell lines of empty vector or Flag-PD-L1 over-expressed U251 cells were treated increasing concentrations of Alisertib for 48 h and the NK-92 MI cells (6×) were added for another 24 h. Shown is the quantification of cellular viability of Crystal Violet (<span class="html-italic">n</span> = 4); (<b>B</b>) Western blot of stable cell lines of empty vector or Flag-PD-L1 over-expressed U251 cells. Actin is a loading control; (<b>C</b>) GBM22, U251, and U87-EGFRvIII cell lines were exposed to escalating doses of BMS1166 for a period of 48 h, followed by the addition of NK-92 MI cells (6×) for an additional 24 h. Subsequently, cellular viability was assessed using Crystal Violet staining. The results present the quantification of cellular viability as determined by Crystal Violet assay. Statistical significance was assessed by ANOVA with Dunnett’s multiple comparison test. A statistically significant difference was noted between GBM cells treated with NK cells alone versus GBM cells exposed to NK-92 MI cells in combination with diverse concentrations of BMS1166. Data are shown as mean ± SD. ***/**** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 6
<p>Alisertib impacted the microenvironment in the syngeneic mouse model of GBM. (<b>A</b>,<b>B</b>) GL261 cells were implanted in the right striatum of C57BL/6 mice. Four groups were randomly assigned: vehicle, Alisertib, PD-L1, and a combination of both, three days after the implantation. Mice were treated with Alisertib three times per week and anti-PD-1 every three days. The representative MRI image from each group after 22 days of implantation is shown in A and animal survival is provided in B (Kaplan−Meier-curve). The white dotted lines indicates the location of the tumor; (<b>C</b>,<b>D</b>) Tumors from a previously conducted GBM22 orthotopic xenograft experiment were fixed and stained with PD-L1 antibody. Quantification is shown in D. Scale bar: 50 µM; (<b>E</b>,<b>F</b>) Tumors from A were fixed and stained with antibody-detecting NK-cells (NK1.1). Quantification is shown in F. Scale bar: 30 µM. Statistical significance was assessed by student’s t-test. Data are shown as mean ± SD. **** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">
11 pages, 3015 KiB  
Article
Involvement of Endolysosomes and Aurora Kinase A in the Regulation of Amyloid β Protein Levels in Neurons
by Zahra Afghah, Nabab Khan, Gaurav Datta, Peter W. Halcrow, Jonathan D. Geiger and Xuesong Chen
Int. J. Mol. Sci. 2024, 25(11), 6200; https://doi.org/10.3390/ijms25116200 - 4 Jun 2024
Viewed by 1075
Abstract
Aurora kinase A (AURKA) is a serine/threonine-protein kinase that regulates microtubule organization during neuron migration and neurite formation. Decreased activity of AURKA was found in Alzheimer’s disease (AD) brain samples, but little is known about the role of AURKA in AD pathogenesis. Here, [...] Read more.
Aurora kinase A (AURKA) is a serine/threonine-protein kinase that regulates microtubule organization during neuron migration and neurite formation. Decreased activity of AURKA was found in Alzheimer’s disease (AD) brain samples, but little is known about the role of AURKA in AD pathogenesis. Here, we demonstrate that AURKA is expressed in primary cultured rat neurons, neurons from adult mouse brains, and neurons in postmortem human AD brains. AURKA phosphorylation, which positively correlates with its activity, is reduced in human AD brains. In SH-SY5Y cells, pharmacological activation of AURKA increased AURKA phosphorylation, acidified endolysosomes, decreased the activity of amyloid beta protein (Aβ) generating enzyme β-site amyloid precursor protein cleaving enzyme (BACE-1), increased the activity of the Aβ degrading enzyme cathepsin D, and decreased the intracellular and secreted levels of Aβ. Conversely, pharmacological inhibition of AURKA decreased AURKA phosphorylation, de-acidified endolysosomes, decreased the activity of cathepsin D, and increased intracellular and secreted levels of Aβ. Thus, reduced AURKA activity in AD may contribute to the development of intraneuronal accumulations of Aβ and extracellular amyloid plaque formation. Full article
Show Figures

Figure 1

Figure 1
<p>AURKA is expressed in neurons. (<b>A</b>) AURKA (red) was expressed in primary cultured rat cortical neurons (NeuN, green). Nuclei were stained with DAPI. (<b>B</b>) AURKA (green) was expressed in neurons NeuN (red) in the dentate gyrus (DG), CA1, and CA3 subregions of the hippocampus in the 8-month-old adult C57BL/6J mouse brains. (<b>C</b>) Immunoblots showed that AURKA was expressed in adult mouse brains. (<b>D</b>) Immunoblots showed that AURKA was expressed in the adult rat brain. (<b>E</b>) AURKA (red) was expressed in neurons (NeuN, green) in fresh-frozen postmortem human brain hippocampal samples.</p>
Full article ">Figure 2
<p>Phosphorylated AURKA (active form) is reduced in AD human brains. Confocal images showed that AURKA (red) phosphorylation was significantly decreased in neurons labeled with NeuN (green) in the hippocampus of the human AD brain compared to the control brain (<span class="html-italic">n</span> = 3 repeats using different slides, ** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 3
<p>AURKA affects endolysosome pH. (<b>A</b>) In SH-SY5Y cells, AURKA activator anacardic acid (AA 50 µM for 4 h) significantly increased phosphorylation of AURKA, whereas AURKA inhibitor (MLN8237 1 µM for 4 h) significantly decreased phosphorylation of AURKA compared to the DMSO control group. (<span class="html-italic">n</span> = 3 repeats, ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001). (<b>B</b>) AURKA activator anacardic acid (50 µM for 4 h) increased LysoBrite fluorescence, whereas AURKA inhibitor MLN8237 (1 µM for 4 h) decreased LysoBrite fluorescence in SH-SY5Y cells (<span class="html-italic">n</span> = 3 repeats, ** <span class="html-italic">p</span> &lt; 0.01). (<b>C</b>) Activating AURKA with 5 µM (<span class="html-italic">n</span> = 3 repeats, ** <span class="html-italic">p</span> &lt; 0.01) and 50 µM (<span class="html-italic">n</span> = 3 repeats, ** <span class="html-italic">p</span> &lt; 0.01) of anacardic acid for 4 h significantly decreased endolysosome pH in primary cultured neurons.</p>
Full article ">Figure 4
<p>AURKA affects Aβ. Activating AURKA with anacardic acid (AA, 50 μM for 2 days) significantly decreased intracellular and secreted levels of Aβ<sub>1-40</sub> (<b>A</b>) and Aβ<sub>1-42</sub> (<b>B</b>) in SH-SY5Y cells (<span class="html-italic">n</span> = 4 repeats, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001). Inhibiting AURKA with MLN8237 (1 μM for 2 days) significantly increased intracellular and secreted levels of Aβ<sub>1-40</sub> (<b>C</b>) and Aβ<sub>1-42</sub> (<b>D</b>) in SH-SY5Y cells (<span class="html-italic">n</span> = 4 repeats, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001). (<b>E</b>) MLN8237 (1 µM for 2 days) significantly increased released LDH activity (<span class="html-italic">n</span> = 3 repeats, *** <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 5
<p>AURKA affects BACE-1 and cathepsin D activity. (<b>A</b>) AURKA activator anacardic acid (AA, 50 μM for 2 days), but not AURKA inhibitor MLN8237 (1 μM for 2 days), decreased BACE-1 activity in SH-SY5Y cells (<span class="html-italic">n</span> = 3 repeats, ns: not significant, ** <span class="html-italic">p</span> &lt; 0.01). (<b>B</b>) AURKA activator anacardic acid (AA, 50 μM for 2 days) significantly increased the percentage of cathepsin D-positive endolysosomes identified with LAMP1. AURKA inhibitor MLN8237 (1 μM for 2 days) significantly decreased the percentage of cathepsin D-positive endolysosomes identified with LAMP1 (<span class="html-italic">n</span> = 3 repeats, ** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">
14 pages, 6394 KiB  
Article
Evaluation of the Efficacy of OSU-2S in the Treatment of Non-Small-Cell Lung Cancer and Screening of Potential Targets of Action
by Mengyuan Han, Xiangran Liu, Sendaer Hailati, Nurbiya Nurahmat, Dilihuma Dilimulati, Alhar Baishan, Alifeiye Aikebaier and Wenting Zhou
Pharmaceuticals 2024, 17(5), 582; https://doi.org/10.3390/ph17050582 - 1 May 2024
Viewed by 1585
Abstract
(1) Background: OSU-2S is a derivative of FTY720 and exhibits significant inhibitory effects on various cancer cells. There is currently no research on the mechanism of the impact of OSU-2S on NSCLC development. We analysed and validated the hub genes and pharmacodynamic effects [...] Read more.
(1) Background: OSU-2S is a derivative of FTY720 and exhibits significant inhibitory effects on various cancer cells. There is currently no research on the mechanism of the impact of OSU-2S on NSCLC development. We analysed and validated the hub genes and pharmacodynamic effects of OSU-2S to treat NSCLC. (2) Methods: The hub genes of OSU-2S for the treatment of NSCLC were screened in PharmMapper, genecard, and KM Plotter database by survival and expression analysis. The effect of OSU-2S on hub gene expression was verified by Western blot analysis. The ex vivo and in vivo efficacy of OSU-2S on tumour growth was verified using A549 cells and a xenografted animal model. (3) Results: A total of 7 marker genes for OSU-2S treatment of NSCLC were obtained. AURKA and S1PR1 were screened as hub genes. Significant differences in the expression of AURKA and S1PR1 between normal and lung adenocarcinoma (LUAD) tissues were found in the GEPIA2 database; Western blot showed that OSU-2S could affect p-AURKA and S1PR1 protein expression. OSU-2S significantly inhibited tumour growth in A549 cells and xenografted animal models. (4) Conclusions: Our study confirms the inhibitory effect of OSU-2S on NSCLC, screens and demonstrates its potential targets AURKA(p-AURKA) and S1PR1, and provides a research basis for treating NSCLC with OSU-2S. Full article
(This article belongs to the Topic Research in Pharmacological Therapies)
Show Figures

Figure 1

Figure 1
<p>Structure of FTY720 and OSU-2S.</p>
Full article ">Figure 2
<p>Acquisition of genes associated with lung adenocarcinoma. (<b>A</b>) cytoHubba plugin was used to screen for NSCLC disease genes. (<b>B</b>) Expression corrected PCA analysis of GSE10072 data matrix, dark green represents normal tissue, light yellow represents tumor tissue. (<b>C</b>) Heat map of differentially expressed genes, with red areas showing positive correlation and blue areas showing negative correlation. (<b>D</b>) Differentially expressed gene volcano plot, dark green represents up-regulated genes and light yellow represents down-regulated genes.</p>
Full article ">Figure 3
<p>Screening and analysis of marker genes. (<b>A</b>,<b>B</b>) Venn diagrams of the three intersecting genes, DEGs, OSU-2S targets of action and disease targets, with gene expression box line plots. *** represents <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>Survival and expression analysis of marker gene. (<b>A</b>) KM survival analysis of marker gene. (<b>B</b>) Expression analysis of marker in lung adenocarcinoma and lung squamous carcinoma, light red represents tumour and grey represents normal. * represents <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 5
<p>Single-gene signaling pathway analysis. Single gene GSEA-KEGG pathway enrichment analysis of <span class="html-italic">AURKA</span> and <span class="html-italic">S1PR1</span>.</p>
Full article ">Figure 6
<p>A549 cells were treated with OSU-2S for CCK-8, clone formation assays, and wound-healing assays. (<b>A</b>,<b>B</b>) A549 cells were treated with different concentrations OSU-2S for 24 h before the cell viability was determined via CCK8. (<b>C</b>,<b>D</b>) Crystalline violet staining to analyze colony formation in A549 cells treated by three different concentrations (1.5 × 10<sup>−6</sup>, 3 × 10<sup>−6</sup> and 6 × 10<sup>−6</sup> mol/L) of OSU-2S. All values are expressed as the mean ± SD (n = 3). (<b>E</b>) A549 cells were treated with three different concentrations (1.5 × 10<sup>−6</sup>, 3 × 10<sup>−6</sup> and 6 × 10<sup>−6</sup> mol/L) OSU-2S for 24 h, and the intercellular spacing was observed at different time points (0 h, 12 h, and 24 h). (<b>F</b>) The statistical analyses of the migration ability. All values are expressed as the mean ± SD (n = 3). Scale bar, 50 μm. * represents <span class="html-italic">p</span> &lt; 0.05, *** represents <span class="html-italic">p</span> &lt; 0.001 and **** represents <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 7
<p>Effect of OSU-2S on the expression of <span class="html-italic">AURKA</span>, <span class="html-italic">S1PR1</span> detected by western blotting assay A549 cells were exposed to three different concentrations (1.5 × 10<sup>−6</sup>, 3 × 10<sup>−6</sup>, and 6 × 10<sup>−6</sup>) of OSU-2S for 24 h. (<b>A</b>) Effect of different concentrations of OSU-2S on the expression of <span class="html-italic">AURKA</span> and <span class="html-italic">S1PR1</span> protein. (<b>B</b>–<b>D</b>) The statistical analyses of the protein expression. All values are expressed as the mean  ±  SD (n = 3). * represents <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 8
<p>Anti-tumour effect of OSU-2S on A549 xenograft lung cancer mice. (<b>A</b>) Tumours were isolated subcutaneously from mice after 35 days of treatment (n = 6). (<b>B</b>) Tumour volume was measured weekly during the experiment (n = 6). (<b>C</b>) Tumour weights were measured after 35 days of treatment (n = 6). (<b>D</b>) Weights of mice were measured weekly (n = 6) during treatment with β-hydroxypropyl-cyclodextrin, FTY720, and OSU-2S (2.5 mg/kg, 5 mg/kg, 10 mg/kg). * represents <span class="html-italic">p</span> &lt; 0.05, ** represents <span class="html-italic">p</span> &lt; 0.01 and **** represents <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 9
<p>The effect of OSU-2S on the expression of proliferation and apoptosis markers in tumour tissues. (<b>A</b>) Representative IHC images of VEGF and Ki67 of the tumors. (<b>B</b>,<b>C</b>) Quantification analysis of immunoreactive cells for VEGF and Ki67. Data are presented as mean  ±  SD (n  =  3). Scale bar, 50 μm. ** represents <span class="html-italic">p</span> &lt; 0.01, *** represents <span class="html-italic">p</span> &lt; 0.001 and **** represents <span class="html-italic">p</span> &lt; 0.0001. ns not significant.</p>
Full article ">
16 pages, 1764 KiB  
Article
Utility of Clinical Next Generation Sequencing Tests in KIT/PDGFRA/SDH Wild-Type Gastrointestinal Stromal Tumors
by Ryan A. Denu, Cissimol P. Joseph, Elizabeth S. Urquiola, Precious S. Byrd, Richard K. Yang, Ravin Ratan, Maria Alejandra Zarzour, Anthony P. Conley, Dejka M. Araujo, Vinod Ravi, Elise F. Nassif Haddad, Michael S. Nakazawa, Shreyaskumar Patel, Wei-Lien Wang, Alexander J. Lazar and Neeta Somaiah
Cancers 2024, 16(9), 1707; https://doi.org/10.3390/cancers16091707 - 27 Apr 2024
Cited by 2 | Viewed by 2758
Abstract
Objective: The vast majority of gastrointestinal stromal tumors (GISTs) are driven by activating mutations in KIT, PDGFRA, or components of the succinate dehydrogenase (SDH) complex (SDHA, SDHB, SDHC, and SDHD genes). A small fraction of GISTs lack [...] Read more.
Objective: The vast majority of gastrointestinal stromal tumors (GISTs) are driven by activating mutations in KIT, PDGFRA, or components of the succinate dehydrogenase (SDH) complex (SDHA, SDHB, SDHC, and SDHD genes). A small fraction of GISTs lack alterations in KIT, PDGFRA, and SDH. We aimed to further characterize the clinical and genomic characteristics of these so-called “triple-negative” GISTs. Methods: We extracted clinical and genomic data from patients seen at MD Anderson Cancer Center with a diagnosis of GIST and available clinical next generation sequencing data to identify “triple-negative” patients. Results: Of the 20 patients identified, 11 (55.0%) had gastric, 8 (40.0%) had small intestinal, and 1 (5.0%) had rectal primary sites. In total, 18 patients (90.0%) eventually developed recurrent or metastatic disease, and 8 of these presented with de novo metastatic disease. For the 13 patients with evaluable response to imatinib (e.g., neoadjuvant treatment or for recurrent/metastatic disease), the median PFS with imatinib was 4.4 months (range 0.5–191.8 months). Outcomes varied widely, as some patients rapidly developed progressive disease while others had more indolent disease. Regarding potential genomic drivers, four patients were found to have alterations in the RAS/RAF/MAPK pathway: two with a BRAF V600E mutation and two with NF1 loss-of-function (LOF) mutations (one deletion and one splice site mutation). In addition, we identified two with TP53 LOF mutations, one with NTRK3 fusion (ETV6-NTRK3), one with PTEN deletion, one with FGFR1 gain-of-function (GOF) mutation (K654E), one with CHEK2 LOF mutation (T367fs*), one with Aurora kinase A fusion (AURKA-CSTF1), and one with FANCA deletion. Patients had better responses with molecularly targeted therapies than with imatinib. Conclusions: Triple-negative GISTs comprise a diverse cohort with different driver mutations. Compared to KIT/PDGFRA-mutant GIST, limited benefit was observed with imatinib in triple-negative GIST. In depth molecular profiling can be helpful in identifying driver mutations and guiding therapy. Full article
Show Figures

Figure 1

Figure 1
<p>Clinical characteristics and outcomes in triple-negative GIST. (<b>A</b>) Distribution of age at diagnosis. (<b>B</b>) Distribution of tumor size at diagnosis. (<b>C</b>) Distribution of mitotic count from original biopsy or resected specimen. In (<b>A</b>–<b>C</b>), bars represent means ± SD. (<b>D</b>) Anatomic distribution of triple-negative GIST cases. (<b>E</b>) On the left in colored bars are the initial disease stage. Black and gray bars on the right indicate the percent of patients that remained with localized disease versus those that developed recurrent or metastatic disease. Bars represent percentages plus standard error of proportion.</p>
Full article ">Figure 2
<p>Genomics of triple-negative GIST. (<b>A</b>) Map of the genomic alterations in triple-negative GIST patients. Each row represents a patient. Each column represents a clinical feature (left 3 columns) or gene, as indicated. White boxes indicate that the gene was profiled but that no alteration was found, and gray boxes indicate that the gene was not profiled. (<b>B</b>) Number of somatic mutations detected by clinical sequencing assays. Each dot represents a single tumor, and bars represent mean ± SD. (<b>C</b>) Distribution of the most commonly altered genes in the cohort. (<b>D</b>) Percentage of tumors with each hypothesized driver mutation. In (<b>C</b>,<b>D</b>), percentages plus standard error of proportion are plotted.</p>
Full article ">Figure 3
<p>Response to treatment in triple-negative GIST. (<b>A</b>) Progression-free survival while on imatinib (n = 15 patients) versus molecularly matched treatments (n = 3 patients). <span class="html-italic">p</span> = 0.21. (<b>B</b>) Swimmer’s plot showing timeline of indicated therapies. Purported driver mutation is shown in the column on the left.</p>
Full article ">Figure 4
<p>Survival outcomes in triple-negative GIST. Overall survival (<b>A</b>), recurrence-free survival (<b>B</b>), and progression-free survival (<b>C</b>) of patients with triple- negative GIST. Recurrence-free survival was calculated in patients with initially localized disease from the date of histologic diagnosis to the date of recurrence, death, or the latest follow-up. Progression-free survival was calculated from the start of therapy to the date of recurrence, death, or the latest follow-up.</p>
Full article ">
14 pages, 8324 KiB  
Article
Cross-Reactivity of N6AMT1 Antibodies with Aurora Kinase A: An Example of Antibody-Specific Non-Specificity
by Baiba Brūmele, Evgeniia Serova, Aleksandra Lupp, Mihkel Suija, Margit Mutso and Reet Kurg
Antibodies 2024, 13(2), 33; https://doi.org/10.3390/antib13020033 - 22 Apr 2024
Viewed by 2591
Abstract
Primary antibodies are one of the main tools used in molecular biology research. However, the often-occurring cross-reactivity of primary antibodies complicates accurate data analysis. Our results show that three commercial polyclonal antibodies raised against N-6 adenine-specific DNA methyltransferase 1 (N6AMT1) strongly cross-react with [...] Read more.
Primary antibodies are one of the main tools used in molecular biology research. However, the often-occurring cross-reactivity of primary antibodies complicates accurate data analysis. Our results show that three commercial polyclonal antibodies raised against N-6 adenine-specific DNA methyltransferase 1 (N6AMT1) strongly cross-react with endogenous and recombinant mitosis-associated protein Aurora kinase A (AURKA). The cross-reactivity was verified through immunofluorescence, immunoblot, and immunoprecipitation assays combined with mass spectrometry. N6AMT1 and AURKA are evolutionarily conserved proteins that are vital for cellular processes. Both proteins share the motif ENNPEE, which is unique to only these two proteins. We suggest that N6AMT1 antibodies recognise this motif in N6AMT1 and AURKA proteins and exhibit an example of “specific” non-specificity. This serves as an example of the importance of controls and critical data interpretation in molecular biology research. Full article
Show Figures

Figure 1

Figure 1
<p>The target of the N6AMT1 antibody localises in the cytoplasm during interphase: (<b>A</b>) U2OS cells; (<b>B</b>) N6AMT1 knockout cells ΔN6AMT1#1, processed for immunofluorescence with primary antibodies specific to N6AMT1 (red): I (CQA1550), II (HPA059242), III (6211-1-AP), and IV (PA5-121076); α-tubulin (green) and secondary antibodies conjugated with Alexa-568 and Alexa 488. The cells were then counterstained with DAPI (blue) for DNA labelling. Images were captured using a Zeiss LSM 900 confocal microscope at 63× magnification. Scale bar, 10 μm.</p>
Full article ">Figure 2
<p>The target of the N6AMT1 antibody localises at the centrosomes during mitosis. (<b>A</b>) U2OS cells, mitosis; (<b>B</b>) N6AMT1 knockout cells ΔN6AMT1#1, mitosis, processed for immunofluorescence with primary antibodies specific to N6AMT1 (red): I (CQA1550), II (HPA059242), III (6211-1-AP), and IV (PA5-121076); α-tubulin (green) and secondary antibodies conjugated with Alexa-568 and Alexa 488. The cells were then counterstained with DAPI (blue) for DNA labelling. Images were captured using a Zeiss LSM 900 confocal microscope at 63× magnification. Scale bar, 10 μm.</p>
Full article ">Figure 3
<p>Immunoblotting with N6AMT1 antibodies. Lane 1—U2OS cell line; lane 2—N6AMT1 knockout cell line ΔN6AMT1#1; lane 3—N6AMT1 knockout compensatory cell line ΔN6AMT1# comp. N6AMT1 endogenous level in U2OS cells corresponds to 23 kDa (lane 1); no band at 23 kDa should be observed in knockout cell line ΔN6AMT1#1 (lane 2); recombinant N6AMT1-EGFP size corresponds to 55 kDa (lane 3). Immunoblot was performed with N6AMT1 antibody: (<b>A</b>) I (CQA1550), (<b>B</b>) II (HPA059242), (<b>C</b>) III (16211-1-AP), (<b>D</b>) IV (PA5-121076), and (<b>E</b>) V (ARP45845_P050), and (<b>F</b>) VI (Sc-517120). The band corresponding to endogenous N6AMT1 at 23 kDa is marked with a filled-in arrow, and recombinant N6AMT1-EGFP at 55 kDa is marked with an empty arrow.</p>
Full article ">Figure 4
<p>N6AMT1 polyclonal antibody recognises an unknown protein with a size of 45–50 kDa in U2OS cells. Mitotic cells of (<b>A</b>) U2OS and (<b>B</b>) ΔN6AMT1#1 were seeded, released from the mitotic block and collected at 0 h–33 h time points. Samples were analysed using N6AMT1 antibody I (CQA1550) and GAPDH. (<b>C</b>) Immunoprecipitation analysis in mitotic U2OS cells. Input (lane 1), TRMT112 (lane 2), and N6AMT1 (lane 3) antibodies were used for co-immunoprecipitation. Samples were analysed by immunoblotting with antibodies against N6AMT1. The band corresponding to N6AMT1 at 23 kDa is marked with an arrow, and the additional mitosis-associated band at 45–50 kDa is marked with an asterisk. (<b>D</b>) Experimental scheme of mass spectrometry analysis to identify the unknown protein that the N6AMT1 antibody I (CQA1550) recognises at 45–50 kDa.</p>
Full article ">Figure 5
<p>Multiple N6AMT1 polyclonal antibodies recognise Aurora kinase A in U2OS cells. (<b>A</b>) U2OS cells were transfected with recombinant Aurora kinase A-EGFP fusion protein in which EGFP was fused in the N (EGFP-AURKA, 77 kDa, lane 1) or C terminus (AURKA-EGFP, 85 kDa, lane 3). EGFP plasmid vectors were used as controls (lanes 2 and 4). Samples were analysed by immunoblotting using antibodies against Aurora kinase A, EGFP, and GAPDH. (<b>B</b>–<b>G</b>) Samples were analysed using N6AMT1 antibodies I (CQA1550), II (HPA059242), III (6211-1-AP), IV (PA5-121076), V (ARP45845_P050), and VI (sc-517120).</p>
Full article ">Figure 6
<p>N6AMT1 and Aurora kinase A share motifs ENNPEE and EKVDL. (<b>A</b>) Schematic presentation of N6AMT1 protein with the localisation of EKVDL and ENNPEE motifs. Analysed antibodies immunogen range and production recombinant (R) or synthetic (S) are indicated. The antibody specificity to N6AMT1 and Aurora kinase A is shown as positive (+), negative (−), or not clear (?). (<b>B</b>) N6AMT1 and AURKA share motif ENNPEE. An asterisk (*) indicates positions which have a single, fully conserved residue. A colon (:) indicates conservation between groups of strongly similar properties – scoring &gt; 0.5 in the Gonnet PAM 250 matrix. A period (.) indicates conservation between groups of weakly similar properties – scoring =&lt; 0.5 in the Gonnet PAM 250 matrix.(<b>C</b>) Predicted AlphaFold structure of Aurora kinase A (AF_AFO014965F1) with motif ENNPEE. (<b>D</b>) Crystal structure of N6AMT1-TRMT112 complex with motif ENNPEE (PDB accession 6KMR) (<b>E</b>) N6AMT1 and AURKA share motif EKVDL. An asterisk (*) indicates positions which have a single, fully conserved residue. A : (colon) indicates conservation between groups of strongly similar properties – scoring &gt; 0.5 in the Gonnet PAM 250 matrix. (F) Predicted AlphaFold structure of Aurora kinase A (AF_AFO014965F1) with motif EKVDL. (G) Crystal structure of N6AMT1-TRMT112 complex with motif EKVDL (PDB accession 6KMR).</p>
Full article ">
14 pages, 5032 KiB  
Article
Antiproliferative Activity of N-Acylhydrazone Derivative on Hepatocellular Carcinoma Cells Involves Transcriptional Regulation of Genes Required for G2/M Transition
by Amanda Aparecida Ribeiro Andrade, Fernanda Pauli, Carolina Girotto Pressete, Bruno Zavan, João Adolfo Costa Hanemann, Marta Miyazawa, Rafael Fonseca, Ester Siqueira Caixeta, Julia Louise Moreira Nacif, Alexandre Ferro Aissa, Eliezer J. Barreiro and Marisa Ionta
Biomedicines 2024, 12(4), 892; https://doi.org/10.3390/biomedicines12040892 - 18 Apr 2024
Viewed by 1722
Abstract
Liver cancer is the second leading cause of cancer-related death in males. It is estimated that approximately one million deaths will occur by 2030 due to hepatic cancer. Hepatocellular carcinoma (HCC) is the most prevalent primary liver cancer subtype and is commonly diagnosed [...] Read more.
Liver cancer is the second leading cause of cancer-related death in males. It is estimated that approximately one million deaths will occur by 2030 due to hepatic cancer. Hepatocellular carcinoma (HCC) is the most prevalent primary liver cancer subtype and is commonly diagnosed at an advanced stage. The drug arsenal used in systemic therapy for HCC is very limited. Multikinase inhibitors sorafenib (Nexavar®) and lenvatinib (Lenvima®) have been used as first-line drugs with modest therapeutic effects. In this scenario, it is imperative to search for new therapeutic strategies for HCC. Herein, the antiproliferative activity of N-acylhydrazone derivatives was evaluated on HCC cells (HepG2 and Hep3B), which were chemically planned on the ALL-993 scaffold, a potent inhibitor of vascular endothelial growth factor 2 (VEGFR2). The substances efficiently reduced the viability of HCC cells, and the LASSBio-2052 derivative was the most effective. Further, we demonstrated that LASSBio-2052 treatment induced FOXM1 downregulation, which compromises the transcriptional activation of genes required for G2/M transition, such as AURKA and AURKB, PLK1, and CDK1. In addition, LASSBio-2052 significantly reduced CCNB1 and CCND1 expression in HCC cells. Our findings indicate that LASSBio-2052 is a promising prototype for further in vivo studies. Full article
(This article belongs to the Special Issue Signaling Pathways That Regulate Cell Proliferation and Apoptosis)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Chemical structure of AAL-993 and <span class="html-italic">N</span>-acylhydrazone derivatives (NAH). (<b>B</b>,<b>C</b>) Dose–response curves. HepG2, Hep3B, and primary fibroblasts were treated for 48 h with NAH derivatives. (<b>D</b>) IC<sub>50</sub> values were determined after 48 h treatment. * The IC<sub>50</sub> for <b>LASSBio-2052</b> in human dermal primary fibroblasts (PFB) was estimated visually from the graph once its cytotoxic effect on normal cells was not sufficient to determine the exact IC<sub>50</sub> value.</p>
Full article ">Figure 2
<p>(<b>A</b>,<b>B</b>) Cell viability rate was determined in HepG2 (<b>C</b>) and Hep3B (<b>D</b>) cultures at 0, 24, and 48 h after treatment. (<b>C</b>,<b>D</b>) Representative images obtained by phase microscopy showing morphological aspects of cells. (<b>E</b>,<b>F</b>) Representative images from the clonogenic assay. The cells were treated for 48 h and recovered in fresh medium for 12 days. (<b>G</b>,<b>H</b>) Quantification of the number of colonies relative to control DMSO. *** <span class="html-italic">p &lt;</span> 0.001 according to ANOVA followed by a Dunnett post-test.</p>
Full article ">Figure 3
<p>(<b>A</b>,<b>B</b>) Representative histograms obtained by flow cytometry after 48 h treatment with <b>LASSBio-2052</b>. (<b>C</b>,<b>D</b>) Cell cycle analysis. *** <span class="html-italic">p &lt;</span> 0.001, ** <span class="html-italic">p &lt;</span> 0.01 according to ANOVA followed by a Dunnett post-test.</p>
Full article ">Figure 4
<p>Gene expression profiles determined by RT-qPCR after 24 h treatment. (<b>A</b>) HepG2. (<b>B</b>) Hep3B. **** <span class="html-italic">p</span> &lt; 0.0001, ** <span class="html-italic">p</span> &lt; 0.01, * <span class="html-italic">p</span> &lt; 0.05 according to ANOVA followed by a Dunnett post-test.</p>
Full article ">Figure 5
<p>(<b>A</b>,<b>C</b>) Representative dot plots from the annexin assay. Cells were treated for 48 h with <b>LASSBio-2052</b>. (<b>B</b>,<b>D</b>) Determination of apoptotic cells considering the cell population positive for the Annexin V assay. * <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 according to ANOVA followed by a Dunnett post-test.</p>
Full article ">Figure 6
<p>Reduced expression of genes downregulated by <b>LASSBio-2052</b> is associated with improved overall survival in patients with hepatocellular carcinoma. Overall survival probability analysis using samples from Liver Hepatocellular Carcinoma (TCGA, Firehose Legacy, study ID: “lihc_tcga”). (<b>A</b>–<b>G</b>) Single gene analysis. (<b>H</b>) Gene signature made with the average of all genes downregulated by <b>LASSBio-2052</b>. (<b>I</b>–<b>K</b>) <span class="html-italic">CDKN1A</span> analysis using the whole dataset (<b>I</b>), patients with <span class="html-italic">TP53</span> not mutated (<b>J</b>), and patients with <span class="html-italic">TP53</span> mutation (<b>K</b>). Log-rank <span class="html-italic">p</span>-values are presented.</p>
Full article ">
20 pages, 14537 KiB  
Article
Identification of AURKA as a Biomarker Associated with Cuproptosis and Ferroptosis in HNSCC
by Xiao Jia, Jiao Tian, Yueyue Fu, Yiqi Wang, Yang Yang, Mengzhou Zhang, Cheng Yang and Yijin Liu
Int. J. Mol. Sci. 2024, 25(8), 4372; https://doi.org/10.3390/ijms25084372 - 16 Apr 2024
Cited by 2 | Viewed by 2141
Abstract
Cuproptosis and ferroptosis represent copper- and iron-dependent forms of cell death, respectively, and both are known to play pivotal roles in head and neck squamous cell carcinoma (HNSCC). However, few studies have explored the prognostic signatures related to cuproptosis and ferroptosis in HNSCC. [...] Read more.
Cuproptosis and ferroptosis represent copper- and iron-dependent forms of cell death, respectively, and both are known to play pivotal roles in head and neck squamous cell carcinoma (HNSCC). However, few studies have explored the prognostic signatures related to cuproptosis and ferroptosis in HNSCC. Our objective was to construct a prognostic model based on genes associated with cuproptosis and ferroptosis. We randomly assigned 502 HSNCC samples from The Cancer Genome Atlas (TCGA) into training and testing sets. Pearson correlation analysis was utilized to identify cuproptosis-associated ferroptosis genes in the training set. Cox proportional hazards (COX) regression and least absolute shrinkage operator (LASSO) were employed to construct the prognostic model. The performance of the prognostic model was internally validated using single-factor COX regression, multifactor COX regression, Kaplan–Meier analysis, principal component analysis (PCA), and receiver operating curve (ROC) analysis. Additionally, we obtained 97 samples from the Gene Expression Omnibus (GEO) database for external validation. The constructed model, based on 12 cuproptosis-associated ferroptosis genes, proved to be an independent predictor of HNSCC prognosis. Among these genes, the increased expression of aurora kinase A (AURKA) has been implicated in various cancers. To further investigate, we employed small interfering RNAs (siRNAs) to knock down AURKA expression and conducted functional experiments. The results demonstrated that AURKA knockdown significantly inhibited the proliferation and migration of HNSCC cells (Cal27 and CNE2). Therefore, AURKA may serve as a potential biomarker in HNSCC. Full article
(This article belongs to the Special Issue Iron Metabolism and Toxicity)
Show Figures

Figure 1

Figure 1
<p>Construction of the risk model based on cuproptosis-associated ferroptosis genes in HNSCC. (<b>A</b>) Correlation networks explain the interactions between cuproptosis genes in HNSCC: red represents positive correlations; blue represents negative correlations. (<b>B</b>) Clustered heatmap of the expression of 150 cuproptosis-associated ferroptosis genes in HNSCC. (<b>C</b>) One-way regression analysis of cuproptosis-associated ferroptosis genes in HNSCC. (<b>D</b>,<b>E</b>) LASSO regression analysis with cuproptosis-associated ferroptosis genes in HNSCC. Coefficient profiles were drawn based on (log λ) sequences and the value of lambda. Min was defined based on 10-fold cross-validation, where the optimal λ yielded 12 cuproptosis-associated ferroptosis genes.</p>
Full article ">Figure 2
<p>The risk model was constructed based on the TCGA training set. (<b>A</b>) The plot of the distribution of patient risk rating points in the TCGA training population. The red curve represents those in the high-risk group and the green represents those in the low-risk group in the TCGA testing population. (<b>B</b>) The scatterplot of patient survival status in the TCGA training population. (<b>C</b>) The Kaplan–Meier survival plots for patients in the TCGA training set population. (<b>D</b>) The ROC curve analysis of predictive performance of assessment risk prognostic model in the TCGA training set population. (<b>E</b>) The clustered heatmap of the expression of the twelve cuproptosis-associated ferroptosis genes modeled in two groups of people at high and low risk. (<b>F</b>) In the TCGA training set, PCA analyses assess the discriminatory ability of our constructed prognostic model.</p>
Full article ">Figure 3
<p>Validation of the risk model in HNSCC. (<b>A</b>) The plot of the distribution of patient risk rating points in the TCGA testing population. The red curve represents those in the high-risk group and the green represents those in the low-risk group in the TCGA testing population. (<b>B</b>) The Kaplan–Meier survival plots for patients in the TCGA testing set population. (<b>C</b>) The ROC curves analysis of predictive performance of assessment risk prognostic model in the TCGA testing set population. (<b>D</b>) The plot of the distribution of patient risk rating points in the GEO validation database. The red curve represents those in the high-risk group and the blue represents those in the low-risk group in the GEO validation database. (<b>E</b>) The Kaplan–Meier survival plots for patients in the GEO validation database. (<b>F</b>) The ROC curves analysis of predictive performance of assessment risk prognostic model in the GEO validation database.</p>
Full article ">Figure 4
<p>Validation of the predictive performance of our risk prognostic model constructed on the basis of 12 cuproptosis-associated ferroptosis genes in HNSCC. (<b>A</b>) One-way regression analyses were performed to validate the predictive performance of the risk prognostic model we constructed. (<b>B</b>) Multifactorial regression analyses were performed to validate the predictive performance of the risk prognostic model we constructed. (<b>C</b>) The prognostic nomogram graph for a given patient was assessed using the risk prognostic model we constructed (** <span class="html-italic">p</span>-value &lt; 0.01, *** <span class="html-italic">p</span>-value &lt; 0.001). The red numbers in the column line graph represent the overall score and predicted 1-year survival, 3-year survival, and 5-year survival for a given patient, respectively. (<b>D</b>) The calibration plot for a given patient was assessed using the risk prognostic model we constructed.</p>
Full article ">Figure 5
<p>The correlation between the risk model and clinical characteristics. (<b>A</b>) Heatmap depicting the relationship between risk model and clinical characteristics. The risk prognostic model correlates with TNM staging. (<b>B</b>) T-staging characteristics of the risk prognostic model. (<b>C</b>) Risk score grouping characteristics of the risk prognostic model. (<b>D</b>) Tumor N grading characteristics of the risk prognostic model. (<b>E</b>) Kaplan–Meier survival curve analysis of female patients. (<b>F</b>) Kaplan–Meier survival curve analysis of patients with stage N-stage 2–3. (<b>G</b>) Kaplan–Meier survival curve analysis of patients with tumor stage 3–4. (<b>H</b>) Kaplan–Meier survival curve analysis of patients with tumor T-stage 1–2. (<b>I</b>) Kaplan–Meier survival curve analysis of patients with tumor T-stage 3–4. (<b>J</b>) Kaplan–Meier survival curve analysis of M0 patients. * <span class="html-italic">p</span>-value &lt; 0.05.</p>
Full article ">Figure 5 Cont.
<p>The correlation between the risk model and clinical characteristics. (<b>A</b>) Heatmap depicting the relationship between risk model and clinical characteristics. The risk prognostic model correlates with TNM staging. (<b>B</b>) T-staging characteristics of the risk prognostic model. (<b>C</b>) Risk score grouping characteristics of the risk prognostic model. (<b>D</b>) Tumor N grading characteristics of the risk prognostic model. (<b>E</b>) Kaplan–Meier survival curve analysis of female patients. (<b>F</b>) Kaplan–Meier survival curve analysis of patients with stage N-stage 2–3. (<b>G</b>) Kaplan–Meier survival curve analysis of patients with tumor stage 3–4. (<b>H</b>) Kaplan–Meier survival curve analysis of patients with tumor T-stage 1–2. (<b>I</b>) Kaplan–Meier survival curve analysis of patients with tumor T-stage 3–4. (<b>J</b>) Kaplan–Meier survival curve analysis of M0 patients. * <span class="html-italic">p</span>-value &lt; 0.05.</p>
Full article ">Figure 6
<p>The correlation between the risk model and immunological characteristics. (<b>A</b>) The histograms of the immunological function analysis of the two populations in the high-risk and low-risk groups. (<b>B</b>) The histograms of the immune cell analysis of the two populations in the high-risk and low-risk groups. (<b>C</b>) The results of the analysis of the difference in the immune checkpoint “CD40LG” between the high-risk and low-risk groups. (<b>D</b>) The results of the analysis of the difference in the immune checkpoint “BLTA” between the high-risk and low-risk groups. (<b>E</b>) The results of the analysis of the difference in the immune checkpoint “ADORO2A” between the high-risk and low-risk groups. (<b>F</b>) The results of the difference analysis of immune checkpoint “CD244” between the high-risk and low-risk groups. (<b>G</b>) The results of the difference analysis of immune checkpoint “CD44” between the high-risk and low-risk groups. (<b>H</b>) The results of the difference analysis of immune checkpoint “CD27” between the high-risk and low-risk groups. * <span class="html-italic">p</span>-value &lt; 0.05, ** <span class="html-italic">p</span>-value &lt; 0.01, *** <span class="html-italic">p</span>-value &lt; 0.001, **** <span class="html-italic">p</span>-value &lt; 0.0001, ns: not significant.</p>
Full article ">Figure 7
<p>The correlation between the risk model and immunological characteristics. (<b>A</b>) Immune infiltration characteristics of the population in the high- and low-risk groups were calculated based on seven algorithms. (<b>B</b>) TIDE scores of people in the high- and low-risk groups. (<b>C</b>) Characteristics of the first 15 TMB mutations in the population of the high-risk group. (<b>D</b>) Characteristics of the first 15 TMB mutations in the population of the low-risk group. (<b>E</b>) Results of KM survival analyses for populations with different TMB scores. (<b>F</b>) Results of KM survival analysis for different scores in different subgroup populations. *** <span class="html-italic">p</span>-value &lt; 0.001.</p>
Full article ">Figure 8
<p>The correlation between the risk model and biological function characteristics. (<b>A</b>,<b>B</b>) The bar and bubble plots of the results of GO enrichment analyses in the at-risk population. (<b>C</b>,<b>D</b>) The bar and bubble plots of KEGG enrichment analysis results for the risk population. (<b>E</b>,<b>F</b>) The GSEA enrichment analysis results for the at-risk population.</p>
Full article ">Figure 9
<p>Identification of potential biomarkers in constructed model. (<b>A</b>) The RNA expression analysis of AUKRA, CAV1, and CDKN2A in normal tissue and HNSCC samples. * <span class="html-italic">p</span>-value &lt; 0.05 (<b>B</b>) The analysis of immunohistochemical results of AUKRA, CAV1, and CDKN2A in normal tissue and HNSCC samples.</p>
Full article ">Figure 10
<p>Inhibition of AURKA suppresses the proliferation and migration capabilities of HNSCC cells. (<b>A</b>,<b>B</b>) RT-qPCR analysis demonstrates the efficiency of AURKA knockdown in Cal27 and CNE2 cell lines. (<b>C</b>,<b>D</b>) The CCK8 cell proliferation assay after AURKA knockdown in Cal27 and CNE2 cell lines. (<b>E</b>,<b>F</b>) The cell scratch test results showed that the AURKA knockdown impedes the cellular migration of Cal27 and CNE2 cells (scale bar: 50 μm). The data are presented as relative cell migration (%) at 0, 24, or 48 h. **** <span class="html-italic">p</span>-value &lt; 0.0001.</p>
Full article ">
Back to TopTop