[go: up one dir, main page]

 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (105)

Search Parameters:
Keywords = anoikis

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
24 pages, 3741 KiB  
Article
Protein Structure Inspired Discovery of a Novel Inducer of Anoikis in Human Melanoma
by Fangfang Qiao, Thomas Andrew Binkowski, Irene Broughan, Weining Chen, Amarnath Natarajan, Gary E. Schiltz, Karl A. Scheidt, Wayne F. Anderson and Raymond Bergan
Cancers 2024, 16(18), 3177; https://doi.org/10.3390/cancers16183177 (registering DOI) - 17 Sep 2024
Abstract
Drug discovery historically starts with an established function, either that of compounds or proteins. This can hamper discovery of novel therapeutics. As structure determines function, we hypothesized that unique 3D protein structures constitute primary data that can inform novel discovery. Using a computationally [...] Read more.
Drug discovery historically starts with an established function, either that of compounds or proteins. This can hamper discovery of novel therapeutics. As structure determines function, we hypothesized that unique 3D protein structures constitute primary data that can inform novel discovery. Using a computationally intensive physics-based analytical platform operating at supercomputing speeds, we probed a high-resolution protein X-ray crystallographic library developed by us. For each of the eight identified novel 3D structures, we analyzed binding of sixty million compounds. Top-ranking compounds were acquired and screened for efficacy against breast, prostate, colon, or lung cancer, and for toxicity on normal human bone marrow stem cells, both using eight-day colony formation assays. Effective and non-toxic compounds segregated to two pockets. One compound, Dxr2-017, exhibited selective anti-melanoma activity in the NCI-60 cell line screen. In eight-day assays, Dxr2-017 had an IC50 of 12 nM against melanoma cells, while concentrations over 2100-fold higher had minimal stem cell toxicity. Dxr2-017 induced anoikis, a unique form of programmed cell death in need of targeted therapeutics. Our findings demonstrate proof-of-concept that protein structures represent high-value primary data to support the discovery of novel acting therapeutics. This approach is widely applicable. Full article
(This article belongs to the Section Molecular Cancer Biology)
Show Figures

Figure 1

Figure 1
<p>Computational pipeline schema for evaluating compound binding.</p>
Full article ">Figure 2
<p>Schema for identification and characterization of 3D protein structures with the potential to bind therapeutically active small molecules.</p>
Full article ">Figure 3
<p>Proteins that contain pockets structurally suited to binding drug-like small molecules. The surfaces of potential binding pockets are depicted, as are ribbon structures of proximal portions of the protein. The proteins are: 1-deoxy-D-xylulose 5-phosphate reductoisomerase (Dxr; Dxr1 (magenta) and Dxr2 (green) binding pockets), ß-ketoacyl acyl carrier protein reductase (FabG), 3-phosphoshikimate 1-carboxyvinyltransferase (EPSP synthase), dihydrofolate synthase (FolC; FolC1 (orange) and FolC2 (light green) binding pockets), hypoxanthine-guanine phosphoribosyltransferase (HGPRT), and glucose-1-phosphate thymidylyltransferase (TYLT).</p>
Full article ">Figure 4
<p>Structures of the acquired compounds.</p>
Full article ">Figure 5
<p>Effect of 5FU on normal bone marrow. (<b>A</b>) Effect of 5FU on eight- and fourteen-day human stem cell hematopoietic colony formation. (<b>B</b>) Effect of 5FU on eight-day colony formation for human stem cells and HT29 colon cancer cells. Data are the mean ± SEM of N = 4 and N = 2 replicates for stem and HT29 cells, respectively.</p>
Full article ">Figure 6
<p>Effect of compounds on cancer cell and bone marrow colony formation. The effect of denoted compounds on eight-day cancer cell colony formation (<b>A</b>,<b>C</b>,<b>E</b>,<b>G</b>). The effect of denoted compounds on eight- and fourteen-day bone marrow colony formation (<b>B</b>,<b>D</b>,<b>F</b>,<b>H</b>). Data are mean ± SD (N = 2 replicates), with similar findings in separate experiments (also N = 2 replicates).</p>
Full article ">Figure 7
<p>The predicted poses of bound Dxr2-017 and FolC2-001. The poses of FolC2-001 (<b>A</b>) and Dxr2-017 (<b>B</b>) bound to their respective FolC2 (yellow) and Dxr2 (green) binding surfaces are depicted, as are ribbon structures of proximal portions of the protein.</p>
Full article ">Figure 8
<p>Dxr2-017 inhibits melanoma cell growth through induction of anoikis. (<b>A</b>) Inhibition of melanoma cell growth. Cells were treated with different concentrations of Dxr2-017, and formation of colonies at eight days is depicted. Data are expressed as the percent of untreated control and are the mean ± SEM of three separate experiments. Each experiment was conducted with N = 3 replicates. (<b>B</b>) No effect on cell cycle progression. M14 and SK-MEL-5 cells were treated for eight days with 20 nM and 50 nM of Dxr2-017, respectively, and the phase of the cell cycle was determined by flow cytometry. Control cells were treated with DMSO vehicle only. Representative histograms are depicted. Graphical data are the mean ± SEM (N = 3). * Denotes <span class="html-italic">p</span>-value ≤ 0.05. (<b>C</b>,<b>D</b>) Transition to floating cells. Cells were treated for three days with different Dxr2-017 concentrations. (<b>C</b>) Depicted are representative light photomicrographs at 20X. The scale bar is 150 µm. Blue and green arrows denote adherent and floating cells, respectively. (<b>D</b>) Adherent and floating cells were quantified from captured images. Data are the mean ± SEM (N = 4), expressed as a percentage to total cells. * Denotes <span class="html-italic">p</span>-value ≤ 0.05 compared to respective floating or attached cells. (<b>E</b>) Cleaved caspase 3 is induced in floating cells. Cells were treated for 3 days, and cell lysate from only adherent cells, or from adherent and floating cells combined, was probed by Western blot for cleaved caspase 3. (<b>F</b>) Dxr2-017 decreases cadherin. Cell lysate from only adherent cells or from adherent and floating cells combined was probed by Western blot for pan-cadherin. All experiments were repeated at separate times, yielding similar results. Original western blots are presented in File S2.</p>
Full article ">
14 pages, 32468 KiB  
Article
Anoikis-Related Long Non-Coding RNA Signatures to Predict Prognosis and Immune Infiltration of Gastric Cancer
by Wen-Jun Meng, Jia-Min Guo, Li Huang, Yao-Yu Zhang, Yue-Ting Zhu, Lian-Sha Tang, Jia-Ling Wang, Hong-Shuai Li and Ji-Yan Liu
Bioengineering 2024, 11(9), 893; https://doi.org/10.3390/bioengineering11090893 - 5 Sep 2024
Viewed by 486
Abstract
Anoikis is a distinct type of programmed cell death and a unique mechanism for tumor progress. However, its exact function in gastric cancer (GC) remains unknown. This study aims to investigate the function of anoikis-related lncRNA (ar-lncRNA) in the prognosis of GC and [...] Read more.
Anoikis is a distinct type of programmed cell death and a unique mechanism for tumor progress. However, its exact function in gastric cancer (GC) remains unknown. This study aims to investigate the function of anoikis-related lncRNA (ar-lncRNA) in the prognosis of GC and its immunological infiltration. The ar-lncRNAs were derived from RNA sequencing data and associated clinical information obtained from The Cancer Genome Atlas. Pearson correlation analysis, differential screening, LASSO and Cox regression were utilized to identify the typical ar-lncRNAs with prognostic significance, and the corresponding risk model was constructed, respectively. Comprehensive methods were employed to assess the clinical characteristics of the prediction model, ensuring the accuracy of the prediction results. Further analysis was conducted on the relationship between immune microenvironment and risk features, and sensitivity predictions were made about anticancer medicines. A risk model was built according to seven selected ar-lncRNAs. The model was validated and the calibration plots were highly consistent in validating nomogram predictions. Further analyses revealed the great accuracy of the model and its ability to serve as a stand-alone GC prognostic factor. We subsequently disclosed that high-risk groups display significant enrichment in pathways related to tumors and the immune system. Additionally, in tumor immunoassays, notable variations in immune infiltrates and checkpoints were noted between different risk groups. This study proposes, for the first time, that prognostic signatures of ar-lncRNA can be established in GC. These signatures accurately predict the prognosis of GC and offer potential biomarkers, suggesting new avenues for basic research, prognosis prediction and personalized diagnosis and treatment of GC. Full article
(This article belongs to the Special Issue Computational Biology and Biostatistics for Public Health)
Show Figures

Figure 1

Figure 1
<p>Ar-lncRNAs’ identification and expression in GC. (<b>A</b>) The network that connects ar-lncRNAs to ARGs. (<b>B</b>) A volcano plot of ARGs with differential expression. (<b>C</b>) Heatmap of top 20 ar-lncRNAs with upregulated and downregulated expressions. Red dot: significant upregulated gene. Green dot: significant downregulated gene. Black dot: insignificant gene.</p>
Full article ">Figure 2
<p>Extraction of the predictive signature of ar-lncRNAs in GC. (<b>A</b>) Uni-Cox regression was used to identify the lncRNAs associated with prognosis. (<b>B</b>) Expressions of the retrieved lncRNAs in the heatmap. (<b>C</b>) LncRNAs were selected for the LASSO model by 10-fold cross-validation. (<b>D</b>) The diagram of the LASSO regression analysis. (<b>E</b>) The Sankey diagram illustrating the relationship between ARGs and lncRNAs. *** <span class="html-italic">p</span> &lt; 0.001, ** <span class="html-italic">p</span> &lt; 0.01, * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 3
<p>Prognostic values of the seven ar-lncRNAs in the train, test and whole sets, corresponding to the high- and low-risk groups. (<b>A</b>–<b>C</b>) Display of the ar-lncRNA model according to risk ratings. (<b>D</b>–<b>F</b>) Time and status of survival between high- and low-risk groups. (<b>G</b>–<b>I</b>) The seven ar-lncRNAs’ heatmap expression. (<b>J</b>–<b>L</b>) Kaplan–Meier curves of survival probability. (<b>M</b>) Probability of survival classified by age, sex, pathological grade and clinical stage.</p>
Full article ">Figure 4
<p>Evaluation of the risk model. (<b>A</b>) Uni-Cox analysis of OS-related risk score and clinical variables. (<b>B</b>) Multi-Cox analysis of OS-related risk score and clinical variables. (<b>C</b>–<b>E</b>) ROC curves for the train, test and complete sets for the 1-, 3- and 5-year OS, respectively. (<b>F</b>–<b>H</b>) ROC curves for the clinical features and risk score in these sets.</p>
Full article ">Figure 5
<p>Construction of nomogram. (<b>A</b>) Prediction of OS in GC patients using nomogram. (<b>B</b>) Calibration diagram of OS. *** <span class="html-italic">p</span> &lt; 0.001, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 6
<p>KEGG analysis by GSEA.</p>
Full article ">Figure 7
<p>The investigation of tumor immune traits and interventions. (<b>A</b>) Immune cell bubble chart in groups. (<b>B</b>) Single-sample GSEA analysis of immune cells and immunological-related pathways in the high- and low-risk groups. (<b>C</b>) Scores relating to immunity among risk groups. (<b>D</b>) Checkpoint expressions in risk groups. *** <span class="html-italic">p</span> &lt; 0.001, ** <span class="html-italic">p</span> &lt; 0.01, * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 8
<p>Consensus clustering in GC based on predictive ar-lncRNAs. (<b>A</b>) Two clusters of patients were separated. (<b>B</b>) The t-SNE of two clusters and two risk groups (C1, cluster 1; C2, cluster 2). (<b>C</b>) The PCA of clusters and risk groups (C1, cluster 1; C2, cluster 2). (<b>D</b>) Sankey diagram showing risk groups and clusters. (<b>E</b>) The comparison of OS between clusters by Kaplan–Meier curves. (<b>F</b>) Immune cell heatmap between clusters. (<b>G</b>) Immunological scores that differ between clusters (the numbers on the top of columns are <span class="html-italic">p</span>-values). (<b>H</b>) The expressions of immune checkpoints between clusters. *** <span class="html-italic">p</span> &lt; 0.001, ** <span class="html-italic">p</span> &lt; 0.01, * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">
29 pages, 11368 KiB  
Article
A New Renieramycin T Right-Half Analog as a Small Molecule Degrader of STAT3
by Preeyaphan Phookphan, Satapat Racha, Masashi Yokoya, Zin Zin Ei, Daiki Hotta, Hongbin Zou and Pithi Chanvorachote
Mar. Drugs 2024, 22(8), 370; https://doi.org/10.3390/md22080370 - 14 Aug 2024
Viewed by 1830
Abstract
Constitutive activation of STAT3 contributes to tumor development and metastasis, making it a promising target for cancer therapy. (1R,4R,5S)-10-hydroxy-9-methoxy-8,11-dimethyl-3-(naphthalen-2-ylmethyl)-1,2,3,4,5,6-hexahydro-1,5-epiminobenzo[d]azocine-4-carbonitrile, DH_31, a new derivative of the marine natural product Renieramycin T, showed potent activity against H292 and H460 cells, with IC50 values of [...] Read more.
Constitutive activation of STAT3 contributes to tumor development and metastasis, making it a promising target for cancer therapy. (1R,4R,5S)-10-hydroxy-9-methoxy-8,11-dimethyl-3-(naphthalen-2-ylmethyl)-1,2,3,4,5,6-hexahydro-1,5-epiminobenzo[d]azocine-4-carbonitrile, DH_31, a new derivative of the marine natural product Renieramycin T, showed potent activity against H292 and H460 cells, with IC50 values of 5.54 ± 1.04 µM and 2.9 ± 0.58 µM, respectively. Structure–activity relationship (SAR) analysis suggests that adding a naphthalene ring with methyl linkers to ring C and a hydroxyl group to ring E enhances the cytotoxic effect of DH_31. At 1–2.5 µM, DH_31 significantly inhibited EMT phenotypes such as migration, and sensitized cells to anoikis. Consistent with the upregulation of ZO1 and the downregulation of Snail, Slug, N-cadherin, and Vimentin at both mRNA and protein levels, in silico prediction identified STAT3 as a target, validated by protein analysis showing that DH_31 significantly decreases STAT3 levels through ubiquitin-proteasomal degradation. Immunofluorescence and Western blot analysis confirmed that DH_31 significantly decreased STAT3 and EMT markers. Additionally, molecular docking suggests a covalent interaction between the cyano group of DH_31 and Cys-468 in the DNA-binding domain of STAT3 (binding affinity = −7.630 kcal/mol), leading to destabilization thereafter. In conclusion, DH_31, a novel RT derivative, demonstrates potential as a STAT3-targeting drug that significantly contribute to understanding of the development of new targeted therapy. Full article
Show Figures

Figure 1

Figure 1
<p>Derivatives of the RT right-half analogs—DH_17, DH_20, DH_23, DH_26, DH_28, DH_30, and DH_31. (<b>A</b>) The structure of Renieramycin T, TM-(−)-18, and the core structure of the RT right-half analog with R. R represents the position of the pyridyl, thiazolyl, or naphthalenyl group in ring C of the RT right-half analog, respectively. (<b>B</b>) Structures of the present RT right-half analogs: DH_17, DH_20, DH_23, DH_26, DH_28, DH_30, and DH_31. (<b>C</b>) Step-by-step synthesis for derivatives of RT right-half analogs.</p>
Full article ">Figure 2
<p>The effect of RT right-half analogs on cytotoxicity in NSCLC and human normal lung epithelial (BEAS-2B) cell lines and apoptotic cell death in NSCLC cells. (<b>A</b>) NSCLC H292 and H460 cells were treated with derivatives of RT right-half analogs for 24 h and analyzed using MTT assay to assess cytotoxicity. (<b>B</b>) IC<sub>50</sub> values for H292 and H460 cell lines were calculated. (<b>C</b>) BEAS-2B cells were treated with DH_28, DH_30, DH_31, and TM-(−)-18 for 24 h. The cytotoxic effects were evaluated using an MTT assay, and the IC<sub>50</sub> values for BEAS-2B cells were determined. (<b>D</b>) H292 and H460 cells were seeded and treated with 0–10 μM of DH_28, DH_30, and DH_31 for 24 h. Hoechst 33342 and PI were used to stain the cell nuclei. Images were obtained under a fluorescence microscope. (<b>E</b>) The percentages of cell death were calculated based on the stained images in H292 and H460 cells. Data represent the mean ± SD (<span class="html-italic">n</span> = 3). *, **, and *** indicate a statistically significant difference between the treated and the untreated cells at <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, respectively.</p>
Full article ">Figure 3
<p>Putative analysis of NSCLC against DH_31 and the effect of DH_31 on EMT-association proteins. (<b>A</b>) Venn diagram of NSCLC and DH_31 targets and GO enrichment analysis of putative targets was performed to clarify the relevant biologic processes (<span class="html-italic">p</span> &lt; 0.01). The y-axis represents GO terms, and the x-axis indicates the number of genes enriched in that term. The color from blue to red indicates the value of <span class="html-italic">p</span>. The adjust (FDR) is becoming smaller with greater credibility and importance. (<b>B</b>) The expression levels of ZO1, ZEB1, Slug, Snail, N-cadherin, and Vimentin were visualized by fluorescence microscopy. Scale bar, 20 µm. Bar graphs show the relative levels of ZO1, ZEB1, Slug, Snail, N-cadherin, and Vimentin. (<b>C</b>) The protein expression levels of ZO1, Slug, Snail, N-cadherin, Vimentin and β–actin were evaluated by Western blot analysis. The relative protein levels were calculated by densitometry. Data represent the mean ± SD (<span class="html-italic">n</span> = 3). *, **, and *** indicate a statistically significant difference between the treated and untreated cells at <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, respectively.</p>
Full article ">Figure 4
<p>The effects of DH_31 on migration and anoikis resistance on NSCLC H460. (<b>A</b>) DH_31 decreased the migration of H460 cells. (<b>B</b>) The relative migration levels of the treated and untreated cells were determined at 24, 48, and 72 h. (<b>C</b>) DH_31 increased the sensitivity to anoikis in H460 cells. (<b>D</b>) The relative viability of cells was determined after culture under detachment conditions for 6, 12, and 24 h. Scale bar, 20 µm. Data represent the mean ± SD (<span class="html-italic">n</span> = 3). ** and *** indicate a statistically significant difference between the treated and the untreated cells at <span class="html-italic">p</span> &lt; 0.01 and <span class="html-italic">p</span> &lt; 0.001, respectively.</p>
Full article ">Figure 5
<p>STAT3 identified as a potential target of DH_31. (<b>A</b>) The top 10 targets among the 64 targets were ranked based on the number of degrees, visualized by the CytoHubba plugin. The degree values of the top 10 targets in the PPI network were ranked, with STAT3 having the highest degree. The intensity of the colors corresponded to the degree values, with purple indicating large values, pink indicating moderate values, and yellow indicating small values. (<b>B</b>) H460 cells treated with DH_31 (0–2.5 μM) for 24 h were stained with anti-STAT3 antibody (red) and examined using confocal laser scanning microscopy. Cell nuclei were stained with Hoechst 33342 (blue). Scale bar, 10 µm. Arrows denote localized STAT3 proteins. (<b>C</b>) The relative levels of STAT3 of H460 were determined by immunofluorescence analysis. (<b>D</b>) The protein expression levels of STAT3 and β–actin was evaluated by Western blot analysis. (<b>E</b>) The relative protein levels were calculated by densitometry. Data represent the mean ± SD (<span class="html-italic">n</span> = 3). * and *** indicate a statistically significant difference between the treated and untreated cells at <span class="html-italic">p</span> &lt; 0.05 and <span class="html-italic">p</span> &lt; 0.001, respectively.</p>
Full article ">Figure 6
<p>The effect of DH_31 on enhanced ubiquitin-mediated STAT3 proteasomal degradation in NSCLC H460. H460 cells were treated with DH_31 (0–2.5 μM) for 8 h. (<b>A</b>) The expression levels of STAT3 mRNA were determined by Real-time qPCR. (<b>B</b>) The ubiquitin–proteasome inhibitor MG132 reversed the inhibitory effect of DH_31 on the expression of the STAT3 protein. After treatment with or without MG132 (10 µM) for 1 h, cells were treated with DH_31 (0–2.5 µM) for 6 h. The STAT3 levels were measured using Western blot analysis and calculated by densitometry. (<b>C</b>) DH_31 induced the ubiquitin–proteasomal degradation of STAT3. After treatment with or without MG132 (10 µM) for 1 h, cells were treated with DH_31 (0 and 2.5 µM) for 6 h. The protein lysates were collected subsequent to STAT3 immunoprecipitation, and the ubiquitinated protein levels were measure by Western blot analysis. Ub-STAT3 levels were calculated by densitometry. Data represent the mean ± SD (<span class="html-italic">n</span> = 3). *, and ** indicate a statistically significant difference between the treated and untreated cells at <span class="html-italic">p</span> &lt; 0.05 and <span class="html-italic">p</span> &lt; 0.01, respectively. # and ## indicate a statistically significant difference from the cells without MG132 at <span class="html-italic">p</span> &lt; 0.05 and <span class="html-italic">p</span> &lt; 0.01, respectively.</p>
Full article ">Figure 7
<p>Domain structure of STAT3 and structure of DH_31 with in silico predicted binding configurations. (<b>A</b>) Schematic of the domain structure of STAT3 and the structure of the dimer interface of STAT3 (PDB: 1BG1) illustrating the surface locations of the DNA-binding domain (residues 321–494) (red) and the SH2 domain (residues 584–688) (green), (<b>B</b>) the binding interaction of DH_31 to the SH2 domain of STAT3, (<b>C</b>) the binding interaction of DH_31 to the DNA-binding domain, and (<b>D</b>) the binding interaction of TM-(−)-18 to the DNA-binding domain of STAT3. (<b>E</b>) The binding energy of DH_31 and TM-(−)-18 at the SH2 domain and the DNA-binding domain.</p>
Full article ">Figure 8
<p>The effect of DH_31 on the mRNA expression of EMT markers in NSCLC H460. (<b>A</b>) Schematic representation of the of STAT3 transcription factor binding sites in target genes. (<b>B</b>) The mRNA expression of <span class="html-italic">ZO1</span>, <span class="html-italic">Slug</span>, <span class="html-italic">Snail</span>, <span class="html-italic">N-cadherin</span>, and <span class="html-italic">Vimentin</span> in H460 cells treated with DH_31 (0–2.5 µM).</p>
Full article ">Scheme 1
<p>Synthesis of <b>2e</b>.</p>
Full article ">Scheme 2
<p>Synthesis of <b>2f</b>: DH_30.</p>
Full article ">Scheme 3
<p>Synthesis of <b>3a</b>: DH_17.</p>
Full article ">Scheme 4
<p>Synthesis of <b>3b</b>: DH_20.</p>
Full article ">Scheme 5
<p>Synthesis of <b>3c</b>: DH_23.</p>
Full article ">Scheme 6
<p>Synthesis of <b>3d</b>: DH_26.</p>
Full article ">Scheme 7
<p>Synthesis of <b>3e</b>: DH_28.</p>
Full article ">Scheme 8
<p>Synthesis of <b>3f</b>: DH_31.</p>
Full article ">
17 pages, 741 KiB  
Review
Decoding the Intricate Landscape of Pancreatic Cancer: Insights into Tumor Biology, Microenvironment, and Therapeutic Interventions
by Antonella Argentiero, Alessandro Andriano, Ingrid Catalina Caradonna, Giulia de Martino and Vanessa Desantis
Cancers 2024, 16(13), 2438; https://doi.org/10.3390/cancers16132438 - 2 Jul 2024
Cited by 1 | Viewed by 1508
Abstract
Pancreatic ductal adenocarcinoma (PDAC) presents significant oncological challenges due to its aggressive nature and poor prognosis. The tumor microenvironment (TME) plays a critical role in progression and treatment resistance. Non-neoplastic cells, such as cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs), contribute to tumor [...] Read more.
Pancreatic ductal adenocarcinoma (PDAC) presents significant oncological challenges due to its aggressive nature and poor prognosis. The tumor microenvironment (TME) plays a critical role in progression and treatment resistance. Non-neoplastic cells, such as cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs), contribute to tumor growth, angiogenesis, and immune evasion. Although immune cells infiltrate TME, tumor cells evade immune responses by secreting chemokines and expressing immune checkpoint inhibitors (ICIs). Vascular components, like endothelial cells and pericytes, stimulate angiogenesis to support tumor growth, while adipocytes secrete factors that promote cell growth, invasion, and treatment resistance. Additionally, perineural invasion, a characteristic feature of PDAC, contributes to local recurrence and poor prognosis. Moreover, key signaling pathways including Kirsten rat sarcoma viral oncogene (KRAS), transforming growth factor beta (TGF-β), Notch, hypoxia-inducible factor (HIF), and Wnt/β-catenin drive tumor progression and resistance. Targeting the TME is crucial for developing effective therapies, including strategies like inhibiting CAFs, modulating immune response, disrupting angiogenesis, and blocking neural cell interactions. A recent multi-omic approach has identified signature genes associated with anoikis resistance, which could serve as prognostic biomarkers and targets for personalized therapy. Full article
(This article belongs to the Section Tumor Microenvironment)
Show Figures

Figure 1

Figure 1
<p>Schematic representation of the pancreatic ductal adenocarcinoma (PDAC) tumor microenvironment (TME). In normal pancreatic tissue, stromal cells such as fibroblasts play a supportive role in tissue homeostasis and repair. However, cancer-associated fibroblasts (CAFs) are activated and secrete a variety of factors that promote tumor growth and invasion. CAFs contribute to the dense desmoplastic stroma that is a hallmark of pancreatic cancer, which can limit the delivery of chemotherapy drugs to the tumor.</p>
Full article ">
17 pages, 3771 KiB  
Article
Arsenic Nanoparticles Trigger Apoptosis via Anoikis Induction in OECM-1 Cells
by Alejandra A. Covarrubias, Mauricio Reyna-Jeldes, Seidy Pedroso-Santana, Sabrina Marín, Carolina Madero-Mendoza, Cecilia Demergasso and Claudio Coddou
Int. J. Mol. Sci. 2024, 25(12), 6723; https://doi.org/10.3390/ijms25126723 - 18 Jun 2024
Viewed by 3777
Abstract
Arsenic compounds have been used as therapeutic alternatives for several diseases including cancer. In the following work, we obtained arsenic nanoparticles (AsNPs) produced by an anaerobic bacterium from the Salar de Ascotán, in northern Chile, and evaluated their effects on the human [...] Read more.
Arsenic compounds have been used as therapeutic alternatives for several diseases including cancer. In the following work, we obtained arsenic nanoparticles (AsNPs) produced by an anaerobic bacterium from the Salar de Ascotán, in northern Chile, and evaluated their effects on the human oral squamous carcinoma cell line OECM-1. Resazurin reduction assays were carried out on these cells using 1–100 µM of AsNPs, finding a concentration-dependent reduction in cell viability that was not observed for the non-tumoral gastric mucosa-derived cell line GES-1. To establish if these effects were associated with apoptosis induction, markers like Bcl2, Bax, and cleaved caspase 3 were analyzed via Western blot, executor caspases 3/7 via luminometry, and DNA fragmentation was analyzed by TUNEL assay, using 100 µM cisplatin as a positive control. OECM-1 cells treated with AsNPs showed an induction of both extrinsic and intrinsic apoptotic pathways, which can be explained by a significant decrease in P-Akt/Akt and P-ERK/ERK relative protein ratios, and an increase in both PTEN and p53 mRNA levels and Bit-1 relative protein levels. These results suggest a prospective mechanism of action for AsNPs that involves a potential interaction with extracellular matrix (ECM) components that reduces cell attachment and subsequently triggers anoikis, an anchorage-dependent type of apoptosis. Full article
(This article belongs to the Special Issue State-of-the-Art Molecular Biology in Chile, 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>AsNP characterization. (<b>a</b>) DLS and Zeta potential analysis for three independent AsNP batches (PdI: polydispersity index). (<b>b</b>) S-TEM micrograph of AsNPs. White arrows indicate AsNPs. (<b>c</b>) Fluorescent microscopy image of agglomerated AsNPs effectively conjugated with TRITC.</p>
Full article ">Figure 2
<p>AsNP effects on OECM-1 cell viability. (<b>a</b>) Changes in cell viability, measured by resazurin reduction assay, obtained for 48 h treatments with 1–100 µM of AsNPs. (<b>b</b>) Cell viability in control cells (C) and in vehicle-treated cells (V), 0.03% chitosan for 48 h. (<b>c</b>) Loss of adherence of OECM-1 cells incubated with vehicle, 60 µM of AsNPs and 100 µM of CisP for 48 h, and stained with 0.2% methylene blue solution. In (<b>a</b>,<b>b</b>), data are expressed as mean ± SEM of 6 independent experiments.</p>
Full article ">Figure 3
<p>AsNP effects on apoptosis induction. (<b>a</b>) Bcl-2 and Bax immunodetection in OECM-1 cells incubated with 0.03% chitosan (vehicle), 60 µM of AsNPs, and 100 µM of CisP for 48 h. (<b>b</b>) Relative Bcl2/Bax protein ratio quantification. (<b>c</b>) Cleaved caspase 3 immunodetection in OECM-1 cells incubated with vehicle, 60 µM of AsNPs, and 100 µM of CisP for 48 h. (<b>d</b>) Densitometric analysis of figure (<b>c</b>). (<b>e</b>) Caspase 3/7 activities assay in OECM-1 cells incubated with 1, 30, 60, and 100 µM of AsNPs for 48 h. Data are expressed as mean ± SEM of 4 independent experiments. Parametric analysis was performed by Student’s <span class="html-italic">t</span>-test. Significance level was established at <span class="html-italic">p</span> &lt; 0.05 (*), 0.001 ≤ <span class="html-italic">p</span> &lt; 0.01 (**) and <span class="html-italic">p</span> &lt; 0.001 (***).</p>
Full article ">Figure 4
<p>AsNP activity on apoptotic DNA fragmentation. TUNEL assay in untreated control (<b>a</b>), 0.03% chitosan (vehicle, (<b>b</b>)), and 30 and 60 µM AsNP treatments for 6 h ((<b>c</b>,<b>d</b>), respectively) was performed in OECM-1 cells. Positive controls correspond to 100 µM CisP for 12 h (<b>e</b>) and DNase I for 6 h (<b>f</b>). White arrows in (<b>c</b>–<b>f</b>) represent TUNEL-positive cells examples for each treatment condition. (<b>g</b>) Percentages of TUNEL-positive cells obtained by quantifying 100 cells per condition. C = control; V = vehicle). Parametric analysis was performed via Student’s <span class="html-italic">t</span>-test. Significance level was established at 0.001 ≤ <span class="html-italic">p</span> &lt; 0.01 (**) and <span class="html-italic">p</span> &lt; 0.001 (***).</p>
Full article ">Figure 5
<p>AsNPs trigger apoptosis via <span class="html-italic">anoikis</span>. (<b>a</b>,<b>b</b>) Erk, p-Erk, Akt, and p-Akt immunodetection in OECM-1 cells incubated with 0.03% chitosan (vehicle), 60 µM of AsNPs, and 100 µM of CisP for 48 h. (<b>c</b>) Densitometric analysis of p-Akt/Akt and p-Erk/Erk relative protein ratios. (<b>d</b>) Quantification of PTEN and p53 relative expression levels in OECM-1 cells using qPCR. (<b>e</b>) Bit-1 immunodetection in OECM-1 cells incubated under the same experimental conditions. (<b>f</b>) Densitometric analysis of figure (<b>e</b>). Data are expressed as mean ± SEM of 4 independent experiments. Parametric analysis was performed by Student’s <span class="html-italic">t</span>-test. Significance level was established at <span class="html-italic">p</span> &lt; 0.05 (*) and 0.001 ≤ <span class="html-italic">p</span> &lt; 0.01 (**).</p>
Full article ">Figure 6
<p>AsNPs interact with the ECM in OECM-1 cell spheroids. (<b>a</b>) Spheroids were created using a matrix containing 90% type I collagen and 10% FITC-type I collagen. (<b>b</b>) Spheroids incubated with 60 µM of TRITC-conjugated AsNPs for 1 h. (<b>c</b>) Small spheroids under similar treatment conditions. Magnification bars: (<b>a</b>,<b>c</b>) = 10 µm; (<b>b</b>) = 20 µm.</p>
Full article ">Figure 7
<p>AsNP effects on OECM-1 cells. Arsenic nanoparticles (AsNPs) exert diverse effects in the OSCC cell line OECM-1. After AsNP treatment, an important increase in cell detachment is observed, this triggers a type of apoptosis called <span class="html-italic">anoikis</span> (red cells), which can be proven by the increases in Bit-1 relative protein levels, caspases 3/7 activity, Akt/Erk phosphorylation inhibition, and its interaction with ECM components like collagen I. In addition, AsNPs can also be internalized by OECM-1 cells, activating the intrinsic pathway of apoptosis through alterations in the Bcl2/Bax relative protein ratio. Created with BioRender.com (<a href="https://app.biorender.com/user/signin" target="_blank">https://app.biorender.com/user/signin</a>; accessed on 13 April 2024).</p>
Full article ">
13 pages, 4530 KiB  
Article
SIRT6 Inhibits Anoikis of Colorectal Cancer Cells by Down-Regulating NDRG1
by Fengying Li, Wentao Yu, Xiaoling Zhou, Jingyu Hou, Yunyi Gao, Jun Zhang and Xiangwei Gao
Int. J. Mol. Sci. 2024, 25(11), 5585; https://doi.org/10.3390/ijms25115585 - 21 May 2024
Viewed by 916
Abstract
Anoikis, a form of apoptosis resulting from the loss of cell–extracellular matrix interaction, is a significant barrier to cancer cell metastasis. However, the epigenetic regulation of this process remains to be explored. Here, we demonstrate that the histone deacetylase sirtuin 6 (SIRT6) plays [...] Read more.
Anoikis, a form of apoptosis resulting from the loss of cell–extracellular matrix interaction, is a significant barrier to cancer cell metastasis. However, the epigenetic regulation of this process remains to be explored. Here, we demonstrate that the histone deacetylase sirtuin 6 (SIRT6) plays a pivotal role in conferring anoikis resistance to colorectal cancer (CRC) cells. The protein level of SIRT6 is negatively correlated with anoikis in CRC cells. The overexpression of SIRT6 decreases while the knockdown of SIRT6 increases detachment-induced anoikis. Mechanistically, SIRT6 inhibits the transcription of N-myc downstream-regulated gene 1 (NDRG1), a negative regulator of the AKT signaling pathway. We observed the up-regulation of SIRT6 in advanced-stage CRC samples. Together, our findings unveil a novel epigenetic program regulating the anoikis of CRC cells. Full article
(This article belongs to the Special Issue The Function of Stress Proteins in Cell Death and Diseases)
Show Figures

Figure 1

Figure 1
<p><b>The protein level of SIRT6 is decreased during the anoikis of CRC cells.</b> (<b>A</b>) The protein level of SIRT6 in RKO cells cultured in ultra-low attachment plates (Detached) for the indicated time. (<b>B</b>) Immunofluorescence analysis of SIRT6 protein in RKO cells cultured in ultra-low attachment plates (Detached) for the indicated time. Pan-cytokeratin (PanCK) was used as an epithelial marker. Scale bar, 5 μm. (<b>C</b>) The mRNA level of the <span class="html-italic">SIRT6</span> gene in RKO cells cultured in ultra-low attachment plates (Detached) for the indicated time. A two-tailed <span class="html-italic">t</span>-test was used for statistical analysis. ns—not significant. (<b>D</b>) The degradation of the SIRT6 protein in RKO cells cultured in normal dishes (Attached) or ultra-low attachment dishes (Detached) for 12 h. Attached and detached cells were treated with cycloheximide (CHX, 100 μg/mL) to block protein synthesis. The remaining SIRT6 protein level was determined by immunoblotting.</p>
Full article ">Figure 2
<p><b>The protein level of SIRT6 is higher in anoikis-resistant CRC cells.</b> (<b>A</b>) The apoptosis levels of SW480 cells and SW620 cells cultured in ultra-low attachment dishes for 48 h. Cell apoptosis was monitored by annexin V-FITC/PI staining and flow cytometry. (<b>B</b>) The degradation of SIRT6 protein in SW480 cells and SW620 cells cultured in ultra-low attachment dishes for 24 h. Cells were treated with CHX (100 μg/mL) to block protein synthesis, and the remaining SIRT6 protein level was determined by immunoblotting.</p>
Full article ">Figure 3
<p><b>SIRT6 inhibits the anoikis of CRC cells.</b> (<b>A</b>) Immunoblotting analysis of SIRT6 knockdown (S6-KD) in RKO cells. (<b>B</b>) Apoptosis levels of control and SIRT6 knockdown cells cultured in normal dishes (Attached) or ultra-low attachment dishes (Detached) for 36 h. Cell apoptosis was monitored by annexin V-FITC/PI staining and flow cytometry. (<b>C</b>) Annexin-V-positive cells in (<b>B</b>) were quantified. (<b>D</b>) Immunoblotting analysis of SIRT6 overexpression (S6-OE) in RKO cells. (<b>E</b>) Apoptosis levels of control and SIRT6 overexpression cells cultured in normal dishes (Attached) or ultra-low attachment dishes (Detached) for 36 h. Cell apoptosis was monitored by annexin V-FITC/PI staining and flow cytometry. (<b>F</b>) Annexin-V-positive cells in (<b>E</b>) were quantified. All data are presented as mean ± SD. A two-tailed <span class="html-italic">t</span>-test was used for statistical analysis. *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p><b>Identification of SIRT6-regulated gene expression.</b> (<b>A</b>) Volcano plots of differentially expressed genes upon SIRT6 overexpression (S6-OE) or SIRT6 knockdown (S6-KD). (<b>B</b>) Gene ontology analysis of down-regulated genes upon SIRT6 overexpression. BP, biological function; CC, cellular compartment; MF, molecular function. (<b>C</b>) Gene ontology analysis of up-regulated genes upon SIRT6 knockdown. BP, biological function; cc, cellular compartment; MF, molecular function. (<b>D</b>) KEGG analysis of down-regulated genes upon SIRT6 overexpression and up-regulated genes upon SIRT6 knockdown.</p>
Full article ">Figure 5
<p><b>SIRT6 represses the transcription of <span class="html-italic">NDRG1</span>.</b> (<b>A</b>) Venn diagram demonstrating the overlap of up-regulated genes in SIRT6 knockdown (S6-KD) cells and down-regulated genes in SIRT6 overexpression (S6-OE) cells. (<b>B</b>) mRNA level of <span class="html-italic">NDRG1</span> gene in SIRT6 knockdown cells. (<b>C</b>) mRNA level of <span class="html-italic">NDRG1</span> gene in SIRT6 overexpression cells. (<b>D</b>) Immunoblotting analysis of NDRG1 protein levels and AKT phosphorylation levels in SIRT6 knockdown and overexpression cells. (<b>E</b>) ChIP-qPCR analysis of SIRT6 binding to <span class="html-italic">NDRG1</span> gene promoter region, with <span class="html-italic">β-actin</span> (<span class="html-italic">ACTB</span>) gene serving as the control. ChIP analysis was performed with antibodies against SIRT6 or control IgG and analyzed by qPCR. Occupancies of SIRT6 in <span class="html-italic">NDRG1</span> gene promoter region or ACTB gene were normalized to the input DNA. (<b>F</b>) Immunoblotting analysis of Ac-H3K9 level in the immunoprecipitated products by Ac-H3K9 antibody from control (Con), SIRT6 overexpression (S6-OE), and SIRT6 knockdown (S6-KD) cells. (<b>G</b>) ChIP-qPCR analysis of acetylated histone H3K9 level (Ac-H3K9) in <span class="html-italic">NDRG1</span> gene promoter region. ChIP analysis was performed with antibodies against Ac-H3K9 and analyzed by qPCR. Occupancies of Ac-H3K9 in <span class="html-italic">NDRG1</span> gene promoter region were normalized to the input DNA. All data are presented as mean ± SD. A two-tailed <span class="html-italic">t</span>-test was used for statistical analysis. * <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, ns—not significant.</p>
Full article ">Figure 6
<p><b>Down-regulation of NDRG1 contributes to SIRT6-inhibited anoikis.</b> (<b>A</b>) Immunoblotting analysis of AKT phosphorylation level in NDRG1 knockdown (NDRG1-KD) cells. (<b>B</b>) Apoptosis levels of cells cultured in normal dishes (Attached) or ultra-low attachment plates (Detached) for 36 h. Cell apoptosis was monitored by annexin V-FITC staining and flow cytometry. Annexin-V-positive cells were quantified. (<b>C</b>) Immunoblotting analysis of AKT phosphorylation levels in control cells and SIRT6 overexpression cells with or without NDRG1 expression. (<b>D</b>) Apoptosis levels of cells cultured in normal dishes (Attached) or ultra-low attachment plates (Detached) for 36 h. Cell apoptosis was monitored by annexin V-FITC staining and flow cytometry. Annexin-V-positive cells were quantified. All data are presented as mean ± SD. A two-tailed <span class="html-italic">t</span>-test was used for statistical analysis. *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 7
<p><b>SIRT6 expression is elevated in advanced-stage CRC samples.</b> (<b>A</b>) Representative immunohistochemistry (IHC) images depicting expression levels of SIRT6 protein in adjacent normal tissue, stage I CRC tissue, and stage III CRC tissue in a CRC tissue array. (<b>B</b>) IHC scoring of adjacent normal tissues and CRC tissues (left). Distribution of low (+) and high (+) levels of SIRT6 protein expression in adjacent normal tissues and CRC tissues (right). (<b>C</b>) IHC scoring of stage I–II tissues and stage III–IV tissues (left). Distribution of low (+) and high (+) levels of SIRT6 protein expression in stage I–II tissues and stage III–IV tissues (right). Statistical analysis was performed using the Chi-Square test. * <span class="html-italic">p</span> &lt; 0.05, ns—not significant.</p>
Full article ">
9 pages, 246 KiB  
Article
Major Stressful Life Events and the Risk of Pancreatic, Head and Neck Cancers: A Case–Control Study
by Arthi Sridhar, Vishaldeep Kaur Sekhon, Chandler Nguyen, Kamelah Abushalha, Amirali Tahanan, Mohammad Hossein Rahbar and Syed Hasan Jafri
Cancers 2024, 16(2), 451; https://doi.org/10.3390/cancers16020451 - 20 Jan 2024
Viewed by 1358
Abstract
Background: Major stressful life events have been shown to be associated with an increased risk of lung cancer, breast cancer and the development of various chronic illnesses. The stress response generated by our body results in a variety of physiological and metabolic changes [...] Read more.
Background: Major stressful life events have been shown to be associated with an increased risk of lung cancer, breast cancer and the development of various chronic illnesses. The stress response generated by our body results in a variety of physiological and metabolic changes which can affect the immune system and have been shown to be associated with tumor progression. In this study, we aim to determine if major stressful life events are associated with the incidence of head and neck or pancreatic cancer (HNPC). Methods: This is a matched case–control study. Cases (CAs) were HNPC patients diagnosed within the previous 12 months. Controls (COs) were patients without a prior history of malignancy. Basic demographic data information on major stressful life events was collected using the modified Holmes–Rahe stress scale. A total sample of 280 was needed (79 cases, 201 controls) to achieve at least 80% power to detect odds ratios (ORs) of 2.00 or higher at the 5% level of significance. Results: From 1 January 2018 to 31 August 2021, 280 patients were enrolled (CA = 79, CO = 201) in this study. In a multivariable logistic regression analysis after controlling for potential confounding variables (including sex, age, race, education, marital status, smoking history), there was no difference between the lifetime prevalence of major stressful event in cases and controls. However, patients with HNPC were significantly more likely to report a major stressful life event within the preceding 5 years when compared to COs (p = 0.01, OR = 2.32, 95% CI, 1.18–4.54). Conclusions: Patients with head, neck and pancreatic cancers are significantly associated with having a major stressful life event within 5 years of their diagnosis. This study highlights the potential need to recognize stressful life events as risk factors for developing malignancies. Full article
(This article belongs to the Section Cancer Informatics and Big Data)
21 pages, 1969 KiB  
Review
The Mechanisms of Regulated Cell Death: Structural and Functional Proteomic Pathways Induced or Inhibited by a Specific Protein—A Narrative Review
by Diego Fernández-Lázaro, Begoña Sanz and Jesús Seco-Calvo
Proteomes 2024, 12(1), 3; https://doi.org/10.3390/proteomes12010003 - 5 Jan 2024
Cited by 1 | Viewed by 2612
Abstract
Billions of cells die in us every hour, and our tissues do not shrink because there is a natural regulation where Cell Death (CD) is balanced with cell division. The process in which cells eliminate themselves in a controlled manner is called Programmed [...] Read more.
Billions of cells die in us every hour, and our tissues do not shrink because there is a natural regulation where Cell Death (CD) is balanced with cell division. The process in which cells eliminate themselves in a controlled manner is called Programmed Cell Death (PCD). The PCD plays an important role during embryonic development, in maintaining homeostasis of the body’s tissues, and in the elimination of damaged cells, under a wide range of physiological and developmental stimuli. A multitude of protein mediators of PCD have been identified and signals have been found to utilize common pathways elucidating the proteins involved. This narrative review focuses on caspase-dependent and caspase-independent PCD pathways. Included are studies of caspase-dependent PCD such as Anoikis, Catastrophe Mitotic, Pyroptosis, Emperitosis, Parthanatos and Cornification, and Caspase-Independent PCD as Wallerian Degeneration, Ferroptosis, Paraptosis, Entosis, Methuosis, and Extracellular Trap Abnormal Condition (ETosis), as well as neutrophil extracellular trap abnormal condition (NETosis) and Eosinophil Extracellular Trap Abnormal Condition (EETosis). Understanding PCD from those reported in this review could shed substantial light on the processes of biological homeostasis. In addition, identifying specific proteins involved in these processes is mandatory to identify molecular biomarkers, as well as therapeutic targets. This knowledge could provide the ability to modulate the PCD response and could lead to new therapeutic interventions in a wide range of diseases. Full article
Show Figures

Figure 1

Figure 1
<p>Structure of caspases. Abbreviations = CARD: Caspase Recruitment and Activation Domain; DED: Death Effector Domain; p20: large subunit (p20); p10: small subunit.</p>
Full article ">Figure 2
<p>Main pathways of initiation of apoptosis. Abbreviations = TNF: Tumor Necrosis Factor; FAS: Cell Surface Receptor that when binding to its ligand causes apoptosis. (APO-1/CD95).</p>
Full article ">Figure 3
<p>Pyroptosis.</p>
Full article ">Figure 4
<p>Ferroptosis pathways. Abbreviations = xCT: cystine-glutamate antiporter; ROS: Reactive Oxygen Species; PUFAs: polyunsaturated fatty acids; GSH: glutathione GPX4: glutathione peroxidase 4; GSSG: glutathione disulfide.</p>
Full article ">Figure 5
<p>Necroptosis.</p>
Full article ">
21 pages, 3197 KiB  
Article
Lipophilic Statins Eliminate Senescent Endothelial Cells by inducing Anoikis-Related Cell Death
by Barbora Belakova, Nicholas K. Wedige, Ezzat M. Awad, Simon Hess, André Oszwald, Marlene Fellner, Shafaat Y. Khan, Ulrike Resch, Markus Lipovac, Karel Šmejkal, Pavel Uhrin and Johannes M. Breuss
Cells 2023, 12(24), 2836; https://doi.org/10.3390/cells12242836 - 14 Dec 2023
Cited by 3 | Viewed by 2279
Abstract
Pre-clinical studies from the recent past have indicated that senescent cells can negatively affect health and contribute to premature aging. Targeted eradication of these cells has been shown to improve the health of aged experimental animals, leading to a clinical interest in finding [...] Read more.
Pre-clinical studies from the recent past have indicated that senescent cells can negatively affect health and contribute to premature aging. Targeted eradication of these cells has been shown to improve the health of aged experimental animals, leading to a clinical interest in finding compounds that selectively eliminate senescent cells while sparing non-senescent ones. In our study, we identified a senolytic capacity of statins, which are lipid-lowering drugs prescribed to patients at high risk of cardiovascular events. Using two different models of senescence in human vascular endothelial cells (HUVECs), we found that statins preferentially eliminated senescent cells, while leaving non-senescent cells unharmed. We observed that the senolytic effect of statins could be negated with the co-administration of mevalonic acid and that statins induced cell detachment leading to anoikis-like apoptosis, as evidenced by real-time visualization of caspase-3/7 activation. Our findings suggest that statins possess a senolytic property, possibly also contributing to their described beneficial cardiovascular effects. Further studies are needed to explore the potential of short-term, high-dose statin treatment as a candidate senolytic therapy. Full article
(This article belongs to the Special Issue Senescence in the Cardiovascular System)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Assessment of Ki-67 and γH2AX expression in young and senescent HUVECs (old and irradiated). (<b>a</b>) Immunofluorescent co-detection of Ki-67 (red) and γH2AX (green); nuclear counterstain with DAPI scale bar 50 µm. (<b>b</b>) Quantification of the portion of Ki-67 positive cells and of cells showing the senescence-associated dotted pattern of γH2AX. Statistical analysis was made using unpaired <span class="html-italic">t</span>-tests. Significance levels are indicated as follows: **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 2
<p>Assessment of p16 expression. (<b>a</b>) Immunofluorescent staining for p16 showing the different signal intensities of young versus senescent HUVECs (old and irradiated). P16 in red, nuclear staining in blue, and corresponding phase contrast images; scale bar 150 µm. (<b>b</b>) Differences in p16 expression were assessed as integrated cellular fluorescent signal intensities per field of view divided by the number of cell nuclei present (given in arbitrary units (a.u.)) by assessing at least five different fields of view using the Olympus software package cellSens and analyzed with unpaired <span class="html-italic">t</span>-tests using GraphPad Prism. Significance levels are indicated as follows: ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.005, ns = not significant. (<b>c</b>) Western blot analysis of lamin B1 expression in young, old, and irradiated HUVECs. The detection of β-actin was used as an internal standard.</p>
Full article ">Figure 3
<p>Elimination of senescent HUVECs and inhibition of proliferation in young HUVECs with 1 µM simvastatin. Time-response curves depict cell count changes for (<b>a</b>,<b>b</b>) senescent (replication/irradiation-induced) and (<b>c</b>) young HUVECs when exposed to varying simvastatin concentrations (ranging from 0.11 to 10 µM) over a 96-hour period. Additionally, time-response curves for (<b>d</b>,<b>e</b>) senescent and (<b>f</b>) young HUVECs treated with combinations of quercetin (10 µM) and dasatinib (0.05 µM, 0.15 µM or 0.45 µM) for 96 h are presented for comparison. Data in panels (<b>a</b>,<b>b</b>,<b>d</b>,<b>e</b>) are expressed as percentages of the untreated control (depicted as a solid gray line as 100%), while data in panels (<b>c</b>,<b>f</b>) are shown as percentages of the initial cell count (represented by the dotted line at 100%; solid gray lines represent the values for the untreated control). These results are based on three independent experiments conducted in duplicate. Statistical analysis was performed using ANOVA followed by a post hoc Dunnett’s Multiple Comparison test. Significance levels are indicated as follows: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.005, and ns = not significant.</p>
Full article ">Figure 4
<p>Elimination of propagation-induced senescent HUVECs (old) by statins and prevention thereof by mevalonic acid. (<b>a</b>–<b>d</b>) Time-response curves depict cell count changes for the effect of selected lipophilic and hydrophilic statins on the cell count of senescent HUVECs over a 96-h period: (<b>a</b>) simvastatin, (<b>b</b>) lovastatin, (<b>c</b>) atorvastatin, and (<b>d</b>) the hydrophilic pravastatin. (<b>e</b>–<b>h</b>) Concentration–response curves including EC50 values for old, irradiated, and young HUVECs show the cell counts (in % of the initial count) 96 h after exposure to different concentrations of statins. (<b>i</b>–<b>l</b>) Time–response curves for the cell count changes in old HUVECs exposed concomitantly to different concentrations of (<b>i</b>) simvastatin, (<b>j</b>) lovastatin, (<b>k</b>) atorvastatin, and (<b>l</b>) pravastatin and to 100 µM mevalonic acid. The used statin concentrations ranged from 0.11 to 10 µM and were applied for 96 h. Data in (<b>a</b>–<b>d</b>) are expressed as percentages of the untreated control (no statin, depicted as a solid gray line as 100%), in the graph legend for (<b>i</b>–<b>l</b>), 0/0 labels the untreated control (no statin and no mevalonate, black), while 0 labels the control only exposed to mevalonic acid but not to a statin (gray). These results are based on three independent experiments conducted in duplicates. Statistical analysis was performed using ANOVA followed by a post hoc Dunnett’s Multiple Comparison test. * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.005, and ns = not significant. All concentration–response curves, which were ascertained using two different lack-of-fit tests, are shown with 95% confidence intervals.</p>
Full article ">Figure 5
<p>Western blot analysis of caspase-3 and PARP-1 activation. (<b>a</b>) Young, old, and irradiated HUVECs were exposed to 0.33 µM, 0.6 µM, and 1 µM activated simvastatin (aSV) for 72 h or alternatively, to 0.2 µM staurosporine (STS) for 3.5 h. (<b>b</b>) Caspase-3 and PARP-1 activation in supernatants collected after the initial 48 h of statin treatment. Samples in both cases, were analyzed with an antibody cocktail consisting of antibodies against pro-caspase-3, activated caspase-3, cleaved PARP-1, and actin as an internal standard.</p>
Full article ">Figure 6
<p>Examples of time-lapse sequences tracking morphological changes and caspase-3/7 activation in simvastatin-treated senescent HUVECs. (<b>a</b>) After exposing cells to a complete endothelial growth medium containing 0.33 µM activated simvastatin for 24 hours, the Caspase-3/7 Green Apoptosis Assay Reagent 4440 was added, and the cells were recorded at a frequency of 10 images per hour using both bright-field and fluorescence imaging (488/510 nm excitation/emission filter). The dashed lines highlight the cell of interest. The full-length video of the presented sequence is available in the <a href="#app1-cells-12-02836" class="html-app">Supplementary Material</a> as <a href="#app1-cells-12-02836" class="html-app">Supplementary Movie S1</a>. (<b>b</b>) For the control, vehicle-treated senescent cells at the same time points.</p>
Full article ">
25 pages, 4043 KiB  
Article
Self-Renewal Inhibition in Breast Cancer Stem Cells: Moonlight Role of PEDF in Breast Cancer
by Carmen Gil-Gas, Marta Sánchez-Díez, Paloma Honrubia-Gómez, Jose Luis Sánchez-Sánchez, Carmen B. Alvarez-Simón, Sebastia Sabater, Francisco Sánchez-Sánchez and Carmen Ramírez-Castillejo
Cancers 2023, 15(22), 5422; https://doi.org/10.3390/cancers15225422 - 15 Nov 2023
Cited by 3 | Viewed by 1306
Abstract
Breast cancer is the leading cause of death among females in developed countries. Although the implementation of screening tests and the development of new therapies have increased the probability of remission, relapse rates remain high. Numerous studies have indicated the connection between cancer-initiating [...] Read more.
Breast cancer is the leading cause of death among females in developed countries. Although the implementation of screening tests and the development of new therapies have increased the probability of remission, relapse rates remain high. Numerous studies have indicated the connection between cancer-initiating cells and slow cellular cycle cells, identified by their capacity to retain long labeling (LT+). In this study, we perform new assays showing how stem cell self-renewal modulating proteins, such as PEDF, can modify the properties, percentage of biomarker-expressing cells, and carcinogenicity of cancer stem cells. The PEDF signaling pathway could be a useful tool for controlling cancer stem cells’ self-renewal and therefore control patient relapse, as PEDF enhances resistance in breast cancer patient cells’ in vitro culture. We have designed a peptide consisting of the C-terminal part of this protein, which acts by blocking endogenous PEDF in cell culture assays. We demonstrate that it is possible to interfere with the self-renewal capacity of cancer stem cells, induce anoikis in vivo, and reduce resistance against docetaxel treatment in cancer patient cells in in vitro culture. We have also demonstrated that this modified PEDF protein produces a significant decrease in the percentage of expressed cancer stem cell markers. Full article
(This article belongs to the Section Cancer Epidemiology and Prevention)
Show Figures

Figure 1

Figure 1
<p>Long-term label-retaining cells (LT+) are present in cell lines and cultures from patient cells and display cancer stem cell characteristics. (<b>A</b>) MDA-MB-231 cells were stained with DDAO and cultivated 8DIV (days in vitro) in monolayers or as mammospheres (CM-sph). (<b>B</b>) Also, the MCF7 cell line showed an LT+ population, which was higher in mammosphere assays (cytometry assay) than in monolayers. (<b>C</b>) Growing patterns of LT+ and LT− cells from patient Pa00 cells stained and grown 8DIV (400 cells/well) and then sorted according to their DDAO content. The number of living cells after 3DIV was checked by methyl purple assay. LT− cells grew similar to controls and faster than LT+ cells. The growth of the control cells has been represented with a blue horizontal line. (<b>D</b>) Docetaxel dose–response curves for LT+, LT−, and control cells. Pa00 cells were stained with DDAO and grown for 8DIV, sorted by their content of DDAO, and grown with increasing concentrations of docetaxel. LT+ cells showed greater resistance against docetaxel than LT−, and respective IC50 are marked by perpendicular lines. (<b>E</b>) 5000 Pa00 cells were injected in nude mice in each case. The tumor volumes are similar when injecting LT− and control non-separated cells but smaller when injecting LT+ cells. All tumors were palpable at the same time. LT+ tumors grew slowly compared to those in the control or LT− group. (<b>F</b>) Table of number of injected mice and tumor formation of the different cell types (<span class="html-italic">n</span> = 3 all experiment; * <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 2
<p>Pigmented Epithelium-Derived Factor (PEDF) increases the number of LT+ cells and the docetaxel resistance of breast cancer cells. (<b>A</b>) Cells treated with chronic PEDF showed a different morphology than control cells. (<b>B</b>) Quantification of morphological differences induced by PEDF treatment. Cells in the micrographs were measured via ImageJ program using the mask shown in Figure (<b>A</b>). (<b>C</b>) Growing pattern after 3DIV of PEDF treated cells and control. PEDF chronically treated cells grew slower than control. (<b>D</b>) Docetaxel dose–response curve of PEDF chronically treated cells and control. PEDF chronically treated cells were more resistant against docetaxel. (<b>E</b>) Histology of PEDF treated tumors and control tumors. Optical microscopic observation shows visible changes in cells chronically treated with PEDF, with a qualitative increase in both cytoplasmic density and the external matrix. Necrotic areas are bigger in controls compared to those with PEDF treatment. Black arrows show mask examples of areas compatible with acellular necrotic spaces. (<b>F</b>) Docetaxel dose–response curve of LT+ PEDF treated cells and LT+ untreated cells, <span class="html-italic">n</span> = 3. LT+ PEDF chronically treated cells were more resistant to docetaxel than untreated ones. (<b>G</b>) PEDF-treated cells grew more slowly than control cells, meaning that the dye-retaining population is up to three times larger than with PEDF treatment. (<b>H</b>) BCRP1 marker immunohistochemistry in control cells. (<b>I</b>) BCRP1 marker immunohistochemistry in PEDF-treated cells. (<b>J</b>) CD133 marker immunohistochemistry in control cells (<b>K</b>) CD133 marker immunohistochemistry in PEDF-treated cells. White arrows in (<b>H</b>–<b>K</b>) shown examples of the abundance of cells in different mitotic phases. (<b>L</b>–<b>O</b>) Insert at high magnification of the yellow square in (<b>H</b>–<b>K</b>). <span class="html-italic">n</span> = 3 in all experiments; * <span class="html-italic">p</span> &lt; 0.05. Bar in (<b>H</b>–<b>K</b>) is 50 µm. Bar in (<b>L</b>–<b>O</b>) is 10 µm.</p>
Full article ">Figure 3
<p>CTE-PEDF induces loss of anchorage and cell death in vivo and reduces resistance against docetaxel. (<b>A</b>) CTE-PEDF construction from 195 to 418 aa of PEDF and with glutamic acid instead of serine in position 227. (<b>B</b>) Pa00 cells were treated chronically with CTE (200 ng/µL). After a week, they showed an increase in anoikis figures. (<b>C</b>) Quantification of CTE-PEDF induced morphological changes in nuclear and cytoplasmic areas and cell distance. (<b>D</b>) Growing pattern after 3DIV of CTE-PEDF treated cells and control. Treated cells survive for less time than control cells. (<b>E</b>) Docetaxel dose–response curve of CTE-treated cells and control. CTE chronically treated cells were less resistant to docetaxel (<span class="html-italic">n</span> = 3 in all experiments; * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001). Bar 50 µm.</p>
Full article ">Figure 4
<p>Cter-PEDF and CTE-PEDF treatments decrease in vivo cancer stem cell markers. (<b>A</b>) Cytometry assay showing the smaller number of CSC in treated cells compared to control. (<b>B</b>) Quantification of positive cells in acute treatment compared to control. (<b>C</b>) Quantification of positive cells in chronic treatment compared to control (<span class="html-italic">n</span> = 3 in all experiments; *** <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 5
<p>PEDF and CTE-PEDF treatments modified in vivo cancer stem cell markers. (<b>A</b>) Examples of xenografts of the PEDF-, CTE-, and control-injected cells. (<b>B</b>) Immunofluorescence of cells positive (arrows) for CD133 and BCRP1, respectively, in xenografts of chronic treatments. Immunocytochemistry with hematoxylin is shown at 20×. Immunocytochemistry assays with CD133 and BCRP1 are shown at 40× with DAPI nuclear staining. (<b>C</b>) Differential effects on cell viability of PEDF and CtE-PEDF treatments over Pa00 tumoral cells in culture. a: Statistically significant differences between Cter and control or PEDF-treated cells, <span class="html-italic">p</span> &lt; 0.05. b: Statistically significant differences between PEDF and control or Cter-treated cells, <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 6
<p>LT+ and BCRP1+ cells used in xenograft assays showing the effect of CTE-PEDF treatments in vivo and the synergy of this effect with chemo and radiotherapy. (<b>A</b>) Cell cytometry of PEDF-treated and untreated cells. LT+ cells are more abundant after PEDF treatment. (<b>B</b>) Quantification of three independent flow cytometry experiments in A. (<b>C</b>) Xenografts resulting from cells positive for stem cell markers and slow-cycling LT+ cells are more resistant tumors when subjected to dose–response assays after dissection and placed in cell culture. (<b>D</b>) Cells expressing the cancer stem cell marker BCRP1 were injected at different cell concentrations. Control-negative cells were also injected. In all cases, an assay was performed with and without treatment with the carboxyl end of PEDF (Cter-PEDF). The xenograft tumors of the Cter-treated cells appear time-delayed (in green) with respect to their controls (in red). (<b>E</b>) Consecutive treatment with radiotherapy (8 treatments and 22 treatments at 6 Gy) downregulates stem cell cancer markers such as the p21 mRNA involved in cell cycle arrest of these cells. (<b>F</b>) The effect of CTE is also synergistic with radiotherapeutic treatments, as it decreases cancer stem cell viability significantly more with CTE treatment and radiotherapy (gray bars) than with radiotherapy alone or with a negative control peptide (CTA peptide without negative charge, white bars). a: statistically significant differences (<span class="html-italic">p</span> &lt; 0.05; compared to the non-irradiated control. b: statistically significant differences compared to 6 Gy treatment without CTE. (<span class="html-italic">p</span> &lt; 0.05; *; <span class="html-italic">p</span> &lt; 0.01; ***).</p>
Full article ">Figure 7
<p>Hypothesis of the exhaustion and depletion of tumor stem cells by blocking the PEDF self-renewal factor signaling pathway. This could result in increased proliferation of chemotherapy-responsive cells and decreased tumor resistance and relapse frequency.</p>
Full article ">
29 pages, 1744 KiB  
Review
Types of Cell Death from a Molecular Perspective
by Fatemeh Hajibabaie, Navid Abedpoor and Parisa Mohamadynejad
Biology 2023, 12(11), 1426; https://doi.org/10.3390/biology12111426 - 13 Nov 2023
Cited by 7 | Viewed by 3806
Abstract
The former conventional belief was that cell death resulted from either apoptosis or necrosis; however, in recent years, different pathways through which a cell can undergo cell death have been discovered. Various types of cell death are distinguished by specific morphological alterations in [...] Read more.
The former conventional belief was that cell death resulted from either apoptosis or necrosis; however, in recent years, different pathways through which a cell can undergo cell death have been discovered. Various types of cell death are distinguished by specific morphological alterations in the cell’s structure, coupled with numerous biological activation processes. Various diseases, such as cancers, can occur due to the accumulation of damaged cells in the body caused by the dysregulation and failure of cell death. Thus, comprehending these cell death pathways is crucial for formulating effective therapeutic strategies. We focused on providing a comprehensive overview of the existing literature pertaining to various forms of cell death, encompassing apoptosis, anoikis, pyroptosis, NETosis, ferroptosis, autophagy, entosis, methuosis, paraptosis, mitoptosis, parthanatos, necroptosis, and necrosis. Full article
Show Figures

Figure 1

Figure 1
<p>Systematic classification and analysis of cell death, encompassing morphological, biochemical, and functional aspects. Atg: autophagy-related gene.</p>
Full article ">Figure 2
<p>The molecular mechanism of apoptotic cell death. TNF: tumor necrosis factor, TNFR1: tumor necrosis factor receptor 1, FADD: FAS-associated death domain, Bid: BH3 interacting domain death agonist, Bad: BCL2-associated agonist of cell death, Bax: Bcl-2-associated X protein, Apaf1: apoptotic protease activating factor-1, IL-10: interleukin 10, Jak2: janus kinase 2, and STAT3: signal transducer and activator of transcription 3. ↑ Indicated increasing of the factor. ↓ Indicated reducing of the factor.</p>
Full article ">Figure 3
<p>Molecular mechanism of pyroptosis in the form of lytic programmed cell death is characterized by its highly inflammatory nature and is most frequently observed following intracellular pathogen infections. TLR: Toll-like receptor, TRIF: TIR-domain-containing adapter-inducing interferon-β, MYD88: myeloid differentiation primary response protein 88, TAk1: transforming growth factor (TGF)-β-activated kinase 1, TAB: TAK1-binding protein, IKK: nuclear factor kappa-B kinase, NF-κB: nuclear factor kappa B, NLRP3: nod-like receptor family pyrin domain containing 3, AIM2: absent in melanoma-2, NLRC4: nod-like receptor family CARD domain containing 4, and NALP1: nucleotide-binding oligomerization domain-like receptor family, pyrin domain-containing 1. Purple ellipse indicted a complex.</p>
Full article ">Figure 4
<p>Molecular mechanism of ferroptosis as an iron-dependent cell death. Fe<sup>3+</sup>: ferric ion, TFR1: transferrin receptor 1, HSPB1: heat shock protein beta-1, GPX: glutathione peroxidase, and GSH: glutathione. The blue ellipse is the channel.</p>
Full article ">
25 pages, 37540 KiB  
Article
Identification of a Novel Oxidative Stress- and Anoikis-Related Prognostic Signature and Its Immune Landscape Analysis in Non-Small Cell Lung Cancer
by Hanqing Zhao, Ying Huang, Guoshun Tong, Wei Wu and Yangwu Ren
Int. J. Mol. Sci. 2023, 24(22), 16188; https://doi.org/10.3390/ijms242216188 - 10 Nov 2023
Viewed by 1252
Abstract
The objective of this study was to identify a kind of prognostic signature based on oxidative stress- and anoikis-related genes (OARGs) for predicting the prognosis and immune landscape of NSCLC. Initially, We identified 47 differentially expressed OARGs that primarily regulate oxidative stress and [...] Read more.
The objective of this study was to identify a kind of prognostic signature based on oxidative stress- and anoikis-related genes (OARGs) for predicting the prognosis and immune landscape of NSCLC. Initially, We identified 47 differentially expressed OARGs that primarily regulate oxidative stress and epithelial cell infiltration through the PI3K-Akt pathway. Subsequently, 10 OARGs related to prognosis determined two potential clusters. A cluster was associated with a shorter survival level, lower immune infiltration, higher stemness index and tumor mutation burden. Next, The best risk score model constructed by prognostic OARGs was the Random Survival Forest model, and it included SLC2A1, LDHA and PLAU. The high-risk group was associated with cluster A and poor prognosis, with a higher tumor mutation burden, stemness index and proportion of M0-type macrophages, and a lower immune checkpoint expression level, immune function score and IPS score. The calibration curve and decision-making curve showed that the risk score combined with clinical pathological characteristics could be used to construct a nomogram for guiding the clinical treatment strategies. Finally, We found that all three hub genes were highly expressed in tumor tissues, and LDHA expression was mainly regulated by has-miR-338-3p, has-miR-330-5p and has-miR-34c-5p. Altogether, We constructed an OARG-related prognostic signature to reveal potential relationships between the signature and clinical characteristics, TME, stemness, tumor mutational burden, drug sensitivity and immune landscape in NSCLC patients. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

Figure 1
<p>Identification of OARGs associated with prognosis. (<b>A</b>) 185 genes at the intersection of anoikis and oxidative stress. (<b>B</b>) 47 Differentially expressed OARGs in normal and tumor tissues (red indicates high expression and blue indicates low expression in the heat map). (<b>C,D</b>) Differentially expressed genes are involved in functions and pathways. (<b>E</b>) 10 prognostic-related OARGs.</p>
Full article ">Figure 2
<p>Characteristics of OARGs associated with prognosis. (<b>A</b>) Relationships between 10 prognostic-related OARGs. (<b>B</b>) CNV of prognostic-related OARGs. Green represents a decrease in the gene copy number variation and red represents an increase. (<b>C</b>) Location of CNV in prognostic-associated OARGs on human chromosomes. Blue dots represent a higher frequency of copy number loss and red represents a higher frequency of copy number gain. (<b>D</b>) Occurrence of somatic mutations in prognostic-related OARGs. (<b>E</b>,<b>F</b>) Expression differences of prognostic-related OARGs between wild-type and mutant types.</p>
Full article ">Figure 3
<p>Consensus clustering of OARGs. (<b>A</b>) A total of 1193 NSCLC patients were divided into two clusters by consensus clustering. The light blue 1 area represents cluster A, and the dark blue 2 area is cluster B. (<b>B</b>) The consistent cumulative distribution function (CDF) plot shows that the CDF reaches its maximum value when k is set to 2 (k indicates several clusters). (<b>C</b>) PCA showed the distinction between the two clusters. (<b>D</b>) The KM curve revealed a significant difference in survival time between the two clusters (<span class="html-italic">p</span> &lt; 0.001). (<b>E</b>) The relationship between OARG clusters and clinical features and OARG expression in NSCLC patients. (Red represents expression values above the mean and is denoted as positive, while blue represents expression values below the mean and is denoted as negative. The darker the color, the more different it is from the mean).</p>
Full article ">Figure 4
<p>Analysis of related characteristics of OARG clusters. (<b>A</b>) Expression of OARGs in different clusters. (<b>B</b>) The ssGSEA analysis showed the infiltration of immune cells in different clusters. (<b>C</b>–<b>F</b>) GSVA analysis and GSEA analysis revealed pathways and functions that were enriched in the two clusters, respectively. (<b>G</b>) Tumor-mutation burden in the two clusters. (<b>H</b>) The situation of different stemness indices in the two subpopulations. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4 Cont.
<p>Analysis of related characteristics of OARG clusters. (<b>A</b>) Expression of OARGs in different clusters. (<b>B</b>) The ssGSEA analysis showed the infiltration of immune cells in different clusters. (<b>C</b>–<b>F</b>) GSVA analysis and GSEA analysis revealed pathways and functions that were enriched in the two clusters, respectively. (<b>G</b>) Tumor-mutation burden in the two clusters. (<b>H</b>) The situation of different stemness indices in the two subpopulations. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>Construction of OARG-related risk scoring model. (<b>A</b>–<b>F</b>) Lasso+Cox model. (<b>A</b>): Lasso regression of lambda graph. (<b>B</b>): Coefficient plot for Lasso regression. The numbers on the upper abscissions represent the number of nonzero coefficients in the model; The ordinate represents the magnitude of the gene coefficients; The lower abscissa represents the normalized coefficient vector; The numbers on each line represent the number of the different genes. (<b>C</b>,<b>D</b>): KM curves for the training and test sets. (<b>E</b>,<b>F</b>): ROC curves for the time dependence of the training and test sets. (<b>G</b>–<b>O</b>) Random Survival Forest model. (<b>G</b>): The model was built for the first time. (<b>H</b>): The model was constructed by a grid search to find the optimal parameters. (<b>I</b>): The optimized model was established. (<b>J</b>): Grid search after filtering variables by the minimum depth method. (<b>K</b>): The model was built after screening the variables and optimizing. (<b>L</b>,<b>M</b>): KM curves for the training and test sets. (<b>N</b>,<b>O</b>): ROC curves for the time dependence of the training and test sets. (<b>P</b>–<b>S</b>) GBM model. (<b>P</b>,<b>Q</b>): KM curves for the training and test sets. (<b>R</b>,<b>S</b>): ROC curves for the time dependence of the training and test sets.</p>
Full article ">Figure 6
<p>Establishment of nomogram for NSCLC patients. (<b>A</b>) Multivariate Cox regression analysis in NSCLC patients. (<b>B</b>) Nomogram constructed by risk score and other clinicopathological factors. (<b>C</b>) Calibration plot showed the differences between nomogram-predicted survival rates and actual survival rates. (<b>D</b>) Cumulative hazard curve showed the probability of survival over time progression based on different scores. (<b>E</b>–<b>G</b>) DCA curves of the nomogram at 1, 3 and 5 years. Decision making based on risk scores had higher clinical benefits in the first year, while both the third and fifth years had higher benefits based on the nomogram. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 7
<p>Relationship between different risk scores and OARG-related characteristics. (<b>A</b>) Differences in risk scores in subgroups A, B. (<b>B</b>) The Sankey plot shows that group A is mainly distributed in the high-risk group. (<b>C</b>) Hub genes with risk scores and clinical features of patients with NSCLC. (Red indicates high expression and blue indicates low expression). (<b>D</b>) Differences in TMB between the high- and low-risk groups. (<b>E</b>) Spearman rank correlation analysis found a positive correlation between TMB and risk scores. (<b>F</b>) Differences in stemness index between high- and low-risk groups. * <span class="html-italic">p</span> &lt; 0.05,*** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 8
<p>Relationship between risk score and immune cell infiltration and TME. (<b>A</b>) Differences in the component of different immune cells between the high- and low-risk groups. (<b>B</b>) Correlation analysis indicated that M0 macrophages and risk scores were positively correlated. (<b>C</b>) Correlation between immune cells. (<b>D</b>) Correlation between immune cells and 3 hub OARGs. (<b>E</b>) StromalScore, ImmuneScore and ESTIMATEScore in high-risk group and low-risk group. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 9
<p>Immune checkpoint expression, immune cell function score and IPS score in high- and low-risk groups. (<b>A</b>) Differences in immune checkpoint expression between high-risk and low-risk groups. (<b>B</b>) Differences in immune function between high- and low-risk groups. (<b>C</b>–<b>F</b>) Differences in IPS scores between high- and low-risk groups. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 10
<p>Correlation between OARGs involved in risk score model and tumor immune microenvironment. (<b>A</b>,<b>B</b>) Annotation of 12 major cell clusters in GSE131907 and the percentage of each cell clusters (<b>C</b>,<b>D</b>) Percentage and expression of SLC2A1. (<b>E</b>,<b>F</b>) Percentage and expression of LDHA. (<b>G</b>,<b>H</b>) Percentage and expression of PLAU.</p>
Full article ">Figure 11
<p>Competitive endogenous RNA (ceRNA) network analysis of LDHA.</p>
Full article ">Figure 12
<p>Validation of expression levels of SLC2A1, LDHA, PLAU. (<b>A</b>–<b>C</b>) Immunohistochemical results for SLC2A1, LDHA, and PLAU from HPA database. (<b>D</b>–<b>F</b>) Expression levels of SLC2A1, LDHA, and PLAU in the GSE101929 dataset. (<b>G</b>–<b>I</b>) Expression levels of SLC2A1, LDHA, and PLAU in the GSE74706 dataset.</p>
Full article ">
18 pages, 8542 KiB  
Article
Suspension-Induced Stem Cell Transition: A Non-Transgenic Method to Generate Adult Stem Cells from Mouse and Human Somatic Cells
by Behzad Yeganeh, Azadeh Yeganeh, Kyle Malone, Shawn T. Beug and Robert P. Jankov
Cells 2023, 12(20), 2508; https://doi.org/10.3390/cells12202508 - 23 Oct 2023
Viewed by 2067
Abstract
Adult stem cells (ASCs) can be cultured with difficulty from most tissues, often requiring chemical or transgenic modification to achieve adequate quantities. We show here that mouse primary fibroblasts, grown in suspension, change from the elongated and flattened morphology observed under standard adherent [...] Read more.
Adult stem cells (ASCs) can be cultured with difficulty from most tissues, often requiring chemical or transgenic modification to achieve adequate quantities. We show here that mouse primary fibroblasts, grown in suspension, change from the elongated and flattened morphology observed under standard adherent culture conditions of generating rounded cells with large nuclei and scant cytoplasm and expressing the mesenchymal stem cell (MSC) marker (Sca1; Ly6A) within 24 h. Based on this initial observation, we describe here a suspension culture method that, irrespective of the lineage used, mouse fibroblast or primary human somatic cells (fibroblasts, hepatocytes and keratinocytes), is capable of generating a high yield of cells in spheroid form which display the expression of ASC surface markers, circumventing the anoikis which often occurs at this stage. Moreover, mouse fibroblast-derived spheroids can be differentiated into adipogenic and osteogenic lineages. An analysis of single-cell RNA sequence data in mouse fibroblasts identified eight distinct cell clusters with one in particular comprising approximately 10% of the cells showing high levels of proliferative capacity expressing high levels of genes related to MSCs and self-renewal as well as the extracellular matrix (ECM). We believe the rapid, high-yield generation of proliferative, multi-potent ASC-like cells via the process we term suspension-induced stem cell transition (SIST) could have significant implications for regenerative medicine. Full article
(This article belongs to the Section Stem Cells)
Show Figures

Figure 1

Figure 1
<p>Mouse TEFs grown in suspension culture display MSC surface markers while undergoing detachment-induced apoptosis (anoikis). (<b>A</b>) Comparison of <span class="html-italic">Sca-1-</span>GFP TEFs morphology as a monolayer (top, pointed by white and black arrows) and after 30 min in suspension (bottom, pointed by white and black arrows). GFP fluorescence images are combined with their corresponding phase-contrast images. (<b>B</b>) Flow cytometric analysis of GFP expression of adherent wild-type or <span class="html-italic">Sca-1-</span>GFP TEFs cultured as a monolayer and cells cultured in suspension. (<b>C</b>) Quantitative evaluation of GFP expression of adherent <span class="html-italic">Sca-1-</span>GFP TEFs from (<b>B</b>). (<b>D</b>) Representative immunoblots of GFP protein in <span class="html-italic">Sca-1-</span>GFP TEFs grown as a monolayer and 24 h in suspension. (<b>E</b>) Densitometric analysis of GFP protein expression in monolayer <span class="html-italic">Sca-1-</span>GFP TEFs vs. suspension (<span class="html-italic">n</span> = 3). (<b>F</b>) Representative histograms for flow cytometric analysis of wild-type mouse TEFs cultured as a monolayer and 24 h in suspension. TEFs were analyzed for mMSCs surface markers, Sca-1, CD29, CD44, CD90.1, CD105, CD106 and CD45R of adherent (top) and suspension cells after 24 h (bottom). (<b>G</b>) Flow cytometry analysis for detection of Annexin V (Alexa 555) of TEFs grown for 14 h in adherent or suspension conditions. (<b>H</b>) Quantification of Annexin V-positive cells from (<b>F</b>). *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 2
<p>Mouse TEF-derived spheroids grown in SIST express mMSC markers, proliferate and exhibit self-renewal capacity. (<b>A</b>) Schematic representation of two suspension culture methods designed to promote cell–cell contact. (<b>B</b>) Representative microscope images captured from live cell imaging from <span class="html-italic">Sca-1-</span>GFP fibroblasts grown in suspension culture (left) and sphere formation (right) at 24 h. (<b>C</b>) IF staining of cleaved caspase 3 (C.CASP3) of mouse TEFs treated with 0.4 mM hydrogen peroxide as positive control (top) and mouse TEFs spheroids after 7 days (bottom). (<b>D</b>) IF staining of mouse MSCs-specific surface markers Sca-1, CD29, CD44, CD90.1, CD105, CD106 and CD45R as a negative marker of adherent cells (top) and spheroids (bottom). (<b>E</b>) Quantification of spheroid volume over a 7-day time course (<span class="html-italic">n</span> = 6 spheroids). (<b>F</b>) Schematic representation of methods used for self-renewal evaluation of mouse fibroblast-derived spheroids in this study. (<b>G</b>) Representative 96-well plate stained with crystal violet to identify wells with new spheroids. *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>Mouse TEF-derived spheroids are multipotent with no tumorigenic potential. (<b>A</b>) Seeding of a single sphere of wild-type (top) and Sca-1-GFP (bottom) fibroblasts on a coverslip after 1 and 5 days in culture. (<b>B</b>) Co-IF staining of Sca-1 (red) with another marker of MSCs CD44 (green) in a single sphere after 1 (left panel) and 5 days (right panel) in culture. Scale bars are indicated in the images. (<b>C</b>) Representative images of Oil Red O (left) and Alizarin Red S (right) staining demonstrating adipogenic and osteogenic differentiation of adherent fibroblasts (middle panel) from TEFs derived from spheroids (bottom panel) and adherent fibroblasts (left panel). Mouse liver and bone tissue were used as positive controls (top panel). (<b>D</b>) Representative hematoxylin and eosin staining of tissues dissected from injection sites in mice receiving cell-free vehicle (Vehicle), adherent monolayer fibroblasts or spheroid-derived cells (Spheroids).</p>
Full article ">Figure 4
<p>Characterization of mouse TEF-derived spheroids vs. monolayer via differential expression gene analysis. (<b>A</b>) UMAP projection of cells calculated from a principal component analysis (PCA) reduction from the Seurat integrated assay, which attempts to bring similar cells close together. Cells are colored by (<b>A</b>) cluster, identified from the integrated assay at a resolution of 0.3, or by (<b>B</b>) source library, showing the overlap of UMAP coordinates. (<b>C</b>) Violin plots of MSC surface marker genes observed in IF staining in <a href="#cells-12-02508-f002" class="html-fig">Figure 2</a>C, showing expression profiles split by source library within each cluster. (<b>D</b>) Pie chart demonstrating distribution of spheroid cells across clusters. (<b>E</b>) Violin plot visualization of expression of MSC surface markers genes identified as significantly enriched in cluster 4. (<b>F</b>) Violin plot visualization of expression of selected collagen, fibronectin and laminin genes identified as significantly enriched in cluster 4. (<b>G</b>) Violin plots of stem cell self-renewal genes identified as significantly enriched in cluster 4. (<b>H</b>) Violin plot visualization of expression of homeobox genes identified as significantly enriched in cluster 7.</p>
Full article ">Figure 5
<p>Human dermal fibroblast- and hepatocyte-derived spheroids possess SC properties. (<b>A</b>) Representative phase-contrast images of adherent monolayer human dermal fibroblasts (top) and a spheroid after 7 days in culture (bottom). (<b>B</b>) Representative histograms for flow cytometric analysis of primary HDFs cultured as a monolayer and 24 h in suspension. HDFs were analyzed for surface hMSC surface markers CD73, CD105, CD106, CD146, CD166, STRO-1 and CD45 as a negative marker of adherent (top) and suspension cells after 24 h (bottom). (<b>C</b>) IF staining of human MSC-specific surface markers CD73, CD105, CD106, CD146, CD166, STRO-1 and CD45 as a negative marker of adherent cells (top) and spheroids (bottom). (<b>D</b>) Representative phase contrast images of adherent monolayer human hepatocytes (top) and a spheroid after 7 days in culture (bottom). (<b>E</b>) Characterization of human hepatocyte-derived spheroids via IF staining using human hepatic stem cell-specific surface markers CD117, CD133, EPCAM, and AFP and ALB as a hepatocyte marker on adherent cells (top) and spheroids (bottom). Sections from human liver tissue were used as positive control (top panel). Scale bars are indicated in the images.</p>
Full article ">Figure 6
<p>Differential capacity of human hepatocyte-derived spheroids and formation of spheroids by human keratinocytes that proliferate and express keratinocyte-specific stem cell markers. (<b>A</b>) A single sphere of human hepatocytes after seeding on coverslip at day 1 (left) and day 5 (right) in culture. (<b>B</b>) Co-IF staining of CD117 (red) with AFP (green) in a single sphere after 3 days in culture. (<b>C</b>) IF staining of AFP (green) and CK19 (red, ductal cell marker) on human hepatocytes derived from spheroids after 7 days culture in transition/expansion medium. (<b>D</b>) Representative phase-contrast images of monolayer human keratinocytes (top) and a spheroid at day 7 (bottom). (<b>E</b>) IF staining of human keratinocytes CK1, CK5 and CK14 and keratinocyte stem cell markers TfR/CD71 and p63/TP73L on adherent cells and spheroids. Scale bars are indicated in the images.</p>
Full article ">
17 pages, 3539 KiB  
Article
A Novel Role of Connective Tissue Growth Factor in the Regulation of the Epithelial Phenotype
by Radhika P. Gogoi, Sandra Galoforo, Alexandra Fox, Colton Morris, Harry Ramos, Vir K. Gogoi, Hussein Chehade, Nicholas K. Adzibolosu, Chenjun Shi, Jitao Zhang, Roslyn Tedja, Robert Morris, Ayesha B. Alvero and Gil Mor
Cancers 2023, 15(19), 4834; https://doi.org/10.3390/cancers15194834 - 2 Oct 2023
Viewed by 1467
Abstract
Background: Epithelial–mesenchymal transition (EMT) is a biological process where epithelial cells lose their adhesive properties and gain invasive, metastatic, and mesenchymal properties. Maintaining the balance between the epithelial and mesenchymal stage is essential for tissue homeostasis. Many of the genes promoting mesenchymal transformation [...] Read more.
Background: Epithelial–mesenchymal transition (EMT) is a biological process where epithelial cells lose their adhesive properties and gain invasive, metastatic, and mesenchymal properties. Maintaining the balance between the epithelial and mesenchymal stage is essential for tissue homeostasis. Many of the genes promoting mesenchymal transformation have been identified; however, our understanding of the genes responsible for maintaining the epithelial phenotype is limited. Our objective was to identify the genes responsible for maintaining the epithelial phenotype and inhibiting EMT. Methods: RNA seq was performed using an vitro model of EMT. CTGF expression was determined via qPCR and Western blot analysis. The knockout of CTGF was completed using the CTGF sgRNA CRISPR/CAS9. The tumorigenic potential was determined using NCG mice. Results: The knockout of CTGF in epithelial ovarian cancer cells leads to the acquisition of functional characteristics associated with the mesenchymal phenotype such as anoikis resistance, cytoskeleton remodeling, increased cell stiffness, and the acquisition of invasion and tumorigenic capacity. Conclusions: We identified CTGF is an important regulator of the epithelial phenotype, and its loss is associated with the early cellular modifications required for EMT. We describe a novel role for CTGF, regulating cytoskeleton and the extracellular matrix interactions necessary for the conservation of epithelial structure and function. These findings provide a new window into understanding the early stages of mesenchymal transformation. Full article
(This article belongs to the Special Issue Multiple Signaling Pathways in Ovarian Cancer)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Differentially expressed pathways and biological processes in epithelial ovarian cancer cells and cells in the E/M hybrid state. (<b>A</b>). Model outlining the process of epithelial–mesenchymal plasticity in R182 ovarian cancer cells as described previously in our lab by Tedja et al. [<a href="#B15-cancers-15-04834" class="html-bibr">15</a>]. (<b>B</b>). Volcano plot of differentially expressed genes (DEGs). Blue dots represent downregulated DEGs and red dots represent upregulated DEGs. (<b>C</b>). Bar plot of top 10 differentially regulated pathways. (<b>D</b>). Chord diagram of top three differentially regulated pathways and their associated DEGs (Star points to CTGF). (<b>E</b>). Top 20 differentially regulated biological processes (arrows highlight processes associated with ECM reorganization).</p>
Full article ">Figure 2
<p>CTGF regulates epithelial and mesenchymal markers in ovarian cancer cells. (<b>A</b>). Western blot demonstrating CTGF expression in various OC cell lines. (<b>B</b>). Western blot demonstrating loss of CTGF by CRISPR KO in R182 OC cell lines. (<b>C</b>). Sanger sequencing verifying deletion of CTGF in R182 CTGF KO cell line. (<b>D</b>). Western blot evaluating expression of epithelial and mesenchymal markers in wild-type and CTGF-KO R182 cell lines. Representative figures of three independent experiments. N = 3. The uncropped blots are shown in <a href="#app1-cancers-15-04834" class="html-app">Supplementary File S1</a>.</p>
Full article ">Figure 3
<p>CTGF negatively regulates anoikis resistance. R182 WT and R182 CTG-KO cells were cultured in ultra-low attachment conditions. (<b>A</b>) Culture morphology was assessed via microscopy after 24 and 48 h. Scale bar 1000 μm. (<b>B</b>) Cell viability was quantified at designated time points using Celltiter96 assay. Experiments were performed independently and in triplicate. Data are presented as mean ± SEM and an unpaired <span class="html-italic">t</span>-test was used to calculate statistical significance. (<b>C</b>) R182 WT, R182 CTG-KO, and R182 CTG-KO cells treated with 100 ng/mL recombinant CTGF were cultured in 50% Matrigel. Culture morphology was assessed via microscopy at day 6. Scale bar 1000 μm. (<b>D</b>) Quantitation of invasion assay at 160 h. Independent experiments were performed in triplicate. Data are presented as mean ± SEM and an unpaired <span class="html-italic">t</span>-test was used to calculate statistical significance. *** <span class="html-italic">p</span> ≤ 0.001. **** <span class="html-italic">p</span> ≤ 0.0001.</p>
Full article ">Figure 4
<p>Loss of CTGF reprograms cell adhesion and ECM in OC cells. RNA sequencing was performed in R182 WT and R182 CTG_KO cells. (<b>A</b>) Volcano plot of differentially expressed genes (DEGs). Blue dots represent downregulated DEGs and red dots represent upregulated DEGs. (<b>B</b>) Bar plot of top 10 differentially regulated pathways. (<b>C</b>) Top seven differentially regulated biological processes. (<b>D</b>) Dendogram of top seven differentially regulated biological processes.</p>
Full article ">Figure 5
<p>CTGF reprograms ECM–receptor interaction. (<b>A</b>) DEGs in the ECM–receptor interaction pathway comparing R81 WT and R182 CTGF-KO cells. (<b>B</b>) Validation of identified DEGs via qPCR. Data are presented as mean <span class="underline">+</span> SEM and an unpaired <span class="html-italic">t</span>-test was used to calculate statistical significance. *** <span class="html-italic">p</span> ≤ 0.001; ** <span class="html-italic">p</span> ≤ 0.01 and * <span class="html-italic">p</span> ≤ 0.05. (<b>C</b>) LAMC2 protein expression in cell lysate of WT and CTGF KO cells (arrow). (<b>D</b>) LAMC2 IF in R182 WT, R182 CTGF-KO, and R182CTGF-KO treated with 100 ng/mL rCTGF. Scale bar 10 μm. (<b>E</b>) Secreted LAMC2 protein expression in media of R182 WT and R182 CTGF-KO cells. (<b>F</b>) Addition of conditioned media from R182-CTGF KO cells to R182 WT cells confers anoikis resistance in R182 WT cells. Anoikis resistance was measured as described in the Materials and Methods section. Briefly, R182 cells were plated in either optimum or CTGF KO media and anoikis resistance was measured up to 72 h. Three independent experiments were performed in triplicate. Data are presented as mean ± SEM and an unpaired <span class="html-italic">t</span>-test was used to calculate statistical significance. * denotes <span class="html-italic">p</span> ≤ 0.01. The uncropped blots are shown in <a href="#app1-cancers-15-04834" class="html-app">Supplementary File S1</a>.</p>
Full article ">Figure 6
<p>Loss of CTGF promotes extracellular matrix remodeling. (<b>A</b>) Proposed model of the role of CTGF in epithelial to mesenchymal cell transition in OC. (<b>B</b>) F-actin IF in R182 WT and R182 CTGF-KO cells demonstrates the presence of lamellopodia and (<b>C</b>) the reorganization of actin filaments. A representative figure of three independent experiments performed in triplicates.</p>
Full article ">Figure 7
<p>Cell stiffness of CTGF KO and WT R182 cells. (<b>A</b>) Top rows are Brillouin images. Bottom rows are co-registered bright-field images. Dashed line indicates the cell body. Scale bar: 10 µm. (<b>B</b>) Brillouin shift results. Multiple measurements were taken for each cell line; wt (n = 50); KO (n = 26); and rCTGF (n = 39). * <span class="html-italic">p</span> &lt; 1.6 × 10<sup>−5</sup>. ** <span class="html-italic">p</span> = 0.015. *** <span class="html-italic">p</span> = 0.002. (<b>C</b>) Invasion assay performed with R182 WT and R182-CTGF KO with 25% and 50% Matrigel measured at 120 h. Independent experiments were performed in quadruplicate. Data are presented as mean ± SEM and an unpaired <span class="html-italic">t</span>-test was used to calculate statistical significance. Scale bar 1000 μm. (<b>D</b>) Representative images of invasion assay with R182 WT and CTGF KO cells in 25 and 50% Matrigel.</p>
Full article ">Figure 8
<p>Tumorigenicity. (<b>A</b>) Tumor growth curves of R182 WT and R182 CTGF-KO cells in NCG mice. R182 CTGF-KO cells can form s.c. tumors while no detectable tumors were observed with R182 wt cells. (<b>B</b>) Histology of tumors formed by R182 CTGF-KO cells. Note cells invading the Matrigel (M) and the presence of neovascular process (BV). Representative figures of five independent animals. Scale bar: 100 μm.</p>
Full article ">
19 pages, 10334 KiB  
Article
The Comprehensive Analysis of m6A-Associated Anoikis Genes in Low-Grade Gliomas
by Hui Zheng, Yutong Zhao, Hai Zhou, Yuguang Tang and Zongyi Xie
Brain Sci. 2023, 13(9), 1311; https://doi.org/10.3390/brainsci13091311 - 12 Sep 2023
Cited by 2 | Viewed by 1506
Abstract
The relationship between N6-methyladenosine (m6A) regulators and anoikis and their effects on low-grade glioma (LGG) is not clear yet. The TCGA-LGG cohort, mRNAseq 325 dataset, and GSE16011 validation set were separately obtained via the Cancer Genome Atlas (TCGA), Chinese Glioma Genome Altas (CGGA), [...] Read more.
The relationship between N6-methyladenosine (m6A) regulators and anoikis and their effects on low-grade glioma (LGG) is not clear yet. The TCGA-LGG cohort, mRNAseq 325 dataset, and GSE16011 validation set were separately obtained via the Cancer Genome Atlas (TCGA), Chinese Glioma Genome Altas (CGGA), and Gene Expression Omnibus (GEO) databases. In total, 27 m6A-related genes (m6A-RGs) and 508 anoikis-related genes (ANRGs) were extracted from published articles individually. First, differentially expressed genes (DEGs) between LGG and normal samples were sifted out by differential expression analysis. DEGs were respectively intersected with m6A-RGs and ANRGs to acquire differentially expressed m6A-RGs (DE-m6A-RGs) and differentially expressed ANRGs (DE-ANRGs). A correlation analysis of DE-m6A-RGs and DE-ANRGs was performed to obtain DE-m6A-ANRGs. Next, univariate Cox and least absolute shrinkage and selection operator (LASSO) were performed on DE-m6A-ANRGs to sift out risk model genes, and a risk score was gained according to them. Then, gene set enrichment analysis (GSEA) was implemented based on risk model genes. After that, we constructed an independent prognostic model and performed immune infiltration analysis and drug sensitivity analysis. Finally, an mRNA-miRNA-lncRNA regulatory network was constructed. There were 6901 DEGs between LGG and normal samples. Six DE-m6A-RGs and 214 DE-ANRGs were gained through intersecting DEGs with m6A-RGs and ANRGs, respectively. A total of 149 DE-m6A-ANRGs were derived after correlation analysis. Four genes, namely ANXA5, KIF18A, BRCA1, and HOXA10, composed the risk model, and they were involved in apoptosis, fatty acid metabolism, and glycolysis. The age and risk scores were finally sifted out to construct an independent prognostic model. Activated CD4 T cells, gamma delta T cells, and natural killer T cells had the largest positive correlations with risk model genes, while activated B cells were significantly negatively correlated with KIF18A and BRCA1. AT.9283, EXEL.2280, Gilteritinib, and Pracinostat had the largest correlation (absolute value) with a risk score. Four risk model genes (mRNAs), 12 miRNAs, and 21 lncRNAs formed an mRNA-miRNA-lncRNA network, containing HOXA10-hsa-miR-129-5p-LINC00689 and KIF18A-hsa-miR-221-3p-DANCR. Through bioinformatics, we constructed a prognostic model of m6A-associated anoikis genes in LGG, providing new ideas for research related to the prognosis and treatment of LGG. Full article
(This article belongs to the Section Molecular and Cellular Neuroscience)
Show Figures

Figure 1

Figure 1
<p>Analysis of six differentially expressed m6A-RGs (DE-m6A-RGs) and 214 differentially expressed anoikis-related genes (DE-ANRGs) in the TCGA-LGG cohort (<b>a</b>) Volcano plot and (<b>b</b>) heatmap for differentially expressed genes (DEGs) between LGG and normal samples (<b>c</b>) Venn plot to identify six DE-m6A-RGs (<b>d</b>) Venn plot to identify 214 DE-ANRGs (<b>e</b>) Bubble chart for the Gene Ontology (GO) analysis of six DE-m6A-RGs (<b>f</b>) Bubble chart for the enriched GO terms of 214 DE-ANRGs (<b>g</b>) Bar chart for the Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis of 214 DE-ANRGs.</p>
Full article ">Figure 2
<p>Spearman correlation analysis to identify 149 DE-m6A-ANRGs.Green nodes indicate M6A genes, red nodes indicate M6A-related DE-ANRG. Red edges indicate positive correlation and blue edges indicate negative correlation.</p>
Full article ">Figure 3
<p>A prognostic model was established based on four risk model genes. (<b>a</b>) Univariate Cox regression analysis to screen 10 survival-related genes. Green (HR &lt; 1) indicates the protective factor, red (HR &gt; 1) indicates the risk factor. (<b>b</b>) A least absolute shrinkage and selection operator (LASSO) regression model was built based on four risk model genes, including Cross-validation diagram (left) and LASSO coefficients profiles (right). The two vertical dashed lines in the chart are the logλ values corresponding to λmin (the logarithm of the minimum mean square error lambda, the left dashed line) and λ1se (the logarithm of the standard error of the minimum distance lambda, the right dashed line). From left to right along the x-axis, with the increases of lambda, the compression parameter increases and the absolute value of the coefficient decreases. The number on top are the number of variables with non-zero regression coefficients in the LASSO model. Variables with non-zero coefficients are important features for our screening. A line indicates a gene. (<b>c</b>) Kaplan–Meier survival curves of the risk model in LGG patients (<span class="html-italic">p</span> &lt; 0.0001). Green indicates the low risk groups, red indicates the high risk groups. (<b>d</b>) Receiver operating characteristic (ROC) curves for the predictive accuracy of the risk model in LGG patients Different colors indicate the different followed-up years.</p>
Full article ">Figure 4
<p>Gene set enrichment analysis (GSEA) of four risk model genes (<b>a</b>) Results of GSEA for ANXA5. (<b>b</b>) Results of the GSEA of KIF18A (<b>c</b>) Results of GSEA for BRCA1. (<b>d</b>) Results of GSEA for HOXA10.</p>
Full article ">Figure 5
<p>Clinical correlation analysis of four risk model genes and the risk model (<b>a</b>) Correlation scatter plots for the relationship between four risk model genes and the risk score (<b>b</b>) Boxplot of risk scores in different clinical subtypes (<b>c</b>) Clinical stratification analysis for the risk model (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 6
<p>Construction of the nomogram and the effect of high/low-risk groups on LGG progression (<b>a</b>) Univariate and (<b>b</b>) Multivariate Cox regression analysis to screen independent prognostic factors, including age and risk score. (<b>c</b>) The nomogram was built based on the independent prognostic factors. (<b>d</b>) Calibration curves of the nomogram to predict survival at 1, 3, and 5 years (<b>e</b>) ROC curves to evaluate the predictive accuracy of the nomogram at 1, 3, and 5 years. (<b>f</b>) Gene set variation analysis (GSVA) of all genes in the high/low-risk groups.</p>
Full article ">Figure 7
<p>Immune-related analysis and drug susceptibility analysis of the risk model (<b>a</b>) Boxplot of the stromal score, immune score, and estimated score in high/low-risk groups (<b>b</b>) Histogram for the infiltration score of 28 immune cells in TCGA-LGG cohorts (<b>c</b>) Violin plot and (<b>d</b>) heatmap for the infiltration levels of 28 immune cells in high/low-risk groups (<b>e</b>) Correlation heatmap of four risk model genes and 25 significantly differentially expressed immune cells. ns indicates not significance, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 8
<p>Construction of the mRNA-miRNA-lncRNA regulatory network based on four risk model genes (<b>a</b>) Volcano plot for differentially expressed miRNAs (DE-miRNAs) in the TCGA-LGG cohort. (<b>b</b>) Volcano plot for differentially expressed lncRNAs (DE-lncRNAs) in the TCGA-LGG cohort. (<b>c</b>) Venn plot to identify 16 intersected miRNAs (<b>d</b>) Venn plot to identify 21 intersected lncRNAs (<b>e</b>) The mRNA-miRNA-lncRNA regulatory network, red represents up-regulated genes, blue represents down-regulated genes.</p>
Full article ">Figure 9
<p>Expression variation of four prognostic genes (<b>a</b>) Boxplot of ANXA5, KIF18A, BRCA1, and HOXA10 in the TCGA-LGG cohort (wilcox.test). (<b>b</b>) Boxplot of ANXA5, KIF18A, BRCA1, and HOXA10 in the GSE16011 set (wilcox.test), **** <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. (<b>c</b>) The results of immunohistochemistry (IHC) methods for the protein expression levels of risk model genes between glioma and normal tissues through the human protein atlas (HPA) database The deeper the yellow in the diagram, the higher the protein expression of the target gene.</p>
Full article ">
Back to TopTop