[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

Journals

Article Types

Countries / Regions

Search Results (41)

Search Parameters:
Keywords = TCF/LEF

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
13 pages, 2219 KiB  
Article
CRABP1 Enhances the Proliferation of the Dermal Papilla Cells of Hu Sheep through the Wnt/β-catenin Pathway
by Zahid Hussain, Tingyan Hu, Yuan Gou, Mingliang He, Xiaoyang Lv, Shanhe Wang and Wei Sun
Genes 2024, 15(10), 1291; https://doi.org/10.3390/genes15101291 - 30 Sep 2024
Viewed by 533
Abstract
Background: The homologous proteins identified as cellular retinoic acid-binding proteins I and II (CRABP-I and CRABP-II) belong to a subset of intracellular proteins characterized by their robust affinity for retinoic acid, which plays an indispensable role in the development of hair [...] Read more.
Background: The homologous proteins identified as cellular retinoic acid-binding proteins I and II (CRABP-I and CRABP-II) belong to a subset of intracellular proteins characterized by their robust affinity for retinoic acid, which plays an indispensable role in the development of hair follicle, including differentiation, proliferation, and apoptosis in keratinocytes. Previous research on Hu sheep hair follicles revealed the specific expression CRABP1 in dermal papilla cells (DPCs), suggesting that CRABP1 has a potential role in regulating the DPC population. Therefore, the main purpose of this study is to expose the performance of the CRABP1 genes in the development and proliferation of DPCs. Methods: Initially, overexpression and inhibition of CRABP1 in the DPCs were conducted through overexpression vector and siRNA. CCK-8, EDU, and RT-PCR cell cycle assays and immunostaining were performed to evaluate the proliferation and cell cycle of dermal papilla cells (DPCs). Although, the influence of CRABP1 upon β-catenin in dermal papilla cells (DPCs) was found using immunofluorescence labeling. Finally, RT-PCR was conducted to assess the impact of CRABP1 on the expression levels of CTNNB1, TCF4, and LEF1 in DPCs involved in the Wnt/β-catenin signaling pathway. Results: The results showed that CRABP1 overexpression promotes the growth rates of DPCs and significantly enhances the proportion of S-phase cells compared with the control group (p < 0.05). The results were the opposite when CRABP1 was a knockdown. In contrast, there was a significant decline in the mRNA expression levels of CTNNβ1, LEF1 (p < 0.05), and TCF4 (p < 0.01) by CRABP1 knockdown. Conclusions: This study found that CRABP1 influences the expression of important genes within the Wnt/β-catenin signaling pathway and promotes DPC proliferation. This investigation provides a theoretical framework to explain the mechanisms that control hair follicle morphogenesis and development. Full article
(This article belongs to the Special Issue Advances in Cattle, Sheep, and Goats Molecular Genetics and Breeding)
Show Figures

Figure 1

Figure 1
<p>The construction of PcDNA3.1 (+)—CRABP1. (<b>A</b>) The CRABP1 coding sequence (CDS) of Hu sheep was amplified to its full length. Lanes 1, 2, and 3 are PCR products, while lane M is the DL 5000 marker. (<b>B</b>) Double enzymes digestion verification of PcDNA3.1 (+)-CRABP1. Lanes 1–6 represent the PCR product, and lane M is the DL10,000 marker. (<b>C</b>) The mRNA expression level in the DPCs of Hu sheep after CRABP1 transfection (<span class="html-italic">n</span> = 3 samples/group). (<b>D</b>) The relative expression levels of CRABP1 mRNA were assessed after transfection with different siRNA. (<b>E</b>) Immunofluorescence identification of CRABP1 in hair follicle of Hu sheep DPCs. Scale bar, 50 µm. (**) stands for highly significant differences (<span class="html-italic">p</span> &lt; 0.01), and (***) stands for highly significant differences (<span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 2
<p>The influence of overexpressing CRABP1 on theDPCs proliferationin Hu sheep. (<b>A</b>) After overexpressing CRABP1, the mRNA levels of PCNA, CDK2, and CCND1 were measured. (<b>B</b>) CCK-8 is employed to evaluate DPC activity after the overexpression of the CRABP1 gene. (<b>C</b>) Flow cytometry is employed to analyze the cell cycle of DPC after CRABP1 overexpression. (<b>D</b>) The EDU test was applied to quantify the proliferation of DPCs in Hu sheep after CRABP1 overexpression. Scale bar, 50 µm. (<b>E</b>) The percentage of DPCs that test positive for EDU in Hu sheep. (<b>F</b>) The proportion of various phases within the cell cycle. (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01) denotes statistical significance. (ns) stands for highly significant differences (<span class="html-italic">p</span> &gt; 0.05).</p>
Full article ">Figure 3
<p>The effect of downregulation of CRABP1 on the DPCs proliferation in Hu sheep. (<b>A</b>) The comparative expression mRNA levels of PCNA, CDK2, and CCND1 after CRABP1 knockdown. (<b>B</b>) DPC activity is detected by CCK-8 after CRABP1 knockdown plasmid. (<b>C</b>) The EDU assay was used to measure the percentage of proliferating DPCs in Hu sheep after the downregulation of CRABP1. Proliferating cells are indicated by red EdUstaining, while nuclei are indicated by blue Hoechst staining. Scale bar, 50 µm. (<b>D</b>) The percentage of Hu sheep DPCs that are EdU-positive. * <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
<p>CRABP1 regulates nuclear translocation β-catenin. After CRABP1 overexpression, β-catenin was stained by immunofluorescence in DPCs. Red fluorescence showed the expression of β-catenin. Nucleus was stained with DAPI in blue. Scale bar, 50 µm.</p>
Full article ">Figure 5
<p>After CRABP1 transfection, both overexpression and knockdown of genes are observed in the Wnt/β-catenin signaling pathway. (<b>A</b>) The effect of CRABP1 overexpression on the gene expression of the Wnt/β-catenin signaling pathway. (<b>B</b>) The expression of genes in the Wnt/β-catenin signaling pathways is influenced by the downregulation of CRABP1. A “*” indicates a significant difference (<span class="html-italic">p</span> &lt; 0.05), while a “**” indicates a highly significant difference (<span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">
11 pages, 1459 KiB  
Communication
The Chronic Toxicity of Endocrine-Disrupting Chemical to Daphnia magna: A Transcriptome and Network Analysis of TNT Exposure
by Jun Lee, Hyun Woo Kim, Dong Yeop Shin, Jun Pyo Han, Yujin Jang, Ju Yeon Park, Seok-Gyu Yun, Eun-Min Cho and Young Rok Seo
Int. J. Mol. Sci. 2024, 25(18), 9895; https://doi.org/10.3390/ijms25189895 - 13 Sep 2024
Viewed by 750
Abstract
Endocrine-disrupting chemicals (EDCs) impair growth and development. While EDCs can occur naturally in aquatic ecosystems, they are continuously introduced through anthropogenic activities such as industrial effluents, pharmaceutical production, wastewater, and mining. To elucidate the chronic toxicological effects of endocrine-disrupting chemicals (EDCs) on aquatic [...] Read more.
Endocrine-disrupting chemicals (EDCs) impair growth and development. While EDCs can occur naturally in aquatic ecosystems, they are continuously introduced through anthropogenic activities such as industrial effluents, pharmaceutical production, wastewater, and mining. To elucidate the chronic toxicological effects of endocrine-disrupting chemicals (EDCs) on aquatic organisms, we collected experimental data from a standardized chronic exposure test using Daphnia magna (D. magna), individuals of which were exposed to a potential EDC, trinitrotoluene (TNT). The chronic toxicity effects of this compound were explored through differential gene expression, gene ontology, network construction, and putative adverse outcome pathway (AOP) proposition. Our findings suggest that TNT has detrimental effects on the upstream signaling of Tcf/Lef, potentially adversely impacting oocyte maturation and early development. This study employs diverse bioinformatics approaches to elucidate the gene-level toxicological effects of chronic TNT exposure on aquatic ecosystems. The results provide valuable insights into the molecular mechanisms of the adverse impacts of TNT through network construction and putative AOP proposition. Full article
Show Figures

Figure 1

Figure 1
<p>Biological networks and putative AOP for TNT chronic toxicity in <span class="html-italic">D. magna</span>. (<b>a</b>) Interactions between all DEGs; the interactions represent relationships between translated proteins of each DEG. (<b>b</b>) Biological network for DEGs associated with chronic exposure to TNT. This biological network shows that reproduction- and development-related adverse effect are strongly associated; (<b>c</b>) Endocrine-focused biological network for DEGs associated with chronic exposure to TNT. This endocrine-focused biological network demonstrates strong associations with reproduction, development, and adverse effects related to female reproduction. The red ovals represent upregulated genes of <span class="html-italic">D. magna</span>, while blue ovals indicate downregulated genes of <span class="html-italic">D. magna</span>. The yellow box and purple box indicate cellular process and phenotype respectively. The blue connecting lines indicate biological association between genes. The gray line and dashed gray line indicate gene–phenotype interaction and gene–cellular process interaction, respectively. (<b>d</b>) A putative AOP for chronic TNT exposure. This putative AOP demonstrates Ck1ε and Ck2 are differentially expressed after a putative molecular initiating event (pMIE), and the transcription of target genes which play roles in oocyte maturation and early development is regulated after Tcf/Lef regulation.</p>
Full article ">
14 pages, 2047 KiB  
Article
Lysine Demethylase KDM2A Promotes Proteasomal Degradation of TCF/LEF Transcription Factors in a Neddylation-Dependent Manner
by Tijana Šopin, František Liška, Tomáš Kučera, Dušan Cmarko and Tomáš Vacík
Cells 2023, 12(22), 2620; https://doi.org/10.3390/cells12222620 - 13 Nov 2023
Cited by 3 | Viewed by 1226
Abstract
Canonical Wnt signaling is essential for a plethora of biological processes ranging from early embryogenesis to aging. Malfunctions of this crucial signaling pathway are associated with various developmental defects and diseases, including cancer. Although TCF/LEF transcription factors (TCF/LEFs) are known to be essential [...] Read more.
Canonical Wnt signaling is essential for a plethora of biological processes ranging from early embryogenesis to aging. Malfunctions of this crucial signaling pathway are associated with various developmental defects and diseases, including cancer. Although TCF/LEF transcription factors (TCF/LEFs) are known to be essential for this pathway, the regulation of their intracellular levels is not completely understood. Here, we show that the lysine demethylase KDM2A promotes the proteasomal destabilization of TCF/LEFs independently of its demethylase domain. We found that the KDM2A-mediated destabilization of TCF/LEFs is dependent on the KDM2A zinc finger CXXC domain. Furthermore, we identified the C-terminal region of TCF7L2 and the CXXC domain of KDM2A as the domains responsible for the interaction between the two proteins. Our study is also the first to show that endogenous TCF/LEF proteins undergo KDM2A-mediated proteasomal degradation in a neddylation-dependent manner. Here, we reveal a completely new mechanism that affects canonical Wnt signaling by regulating the levels of TCF/LEF transcription factors through their KDM2A-promoted proteasomal degradation. Full article
(This article belongs to the Section Cell Signaling)
Show Figures

Figure 1

Figure 1
<p>KDM2A negatively affects canonical Wnt signaling by destabilizing TCF/LEF transcription factors. (<b>A</b>) The overexpression of KDM2A in HEK293T cells was confirmed through Western blotting. Elevated levels of KDM2A result in decreased protein levels of endogenous TCF/LEF proteins. GAPDH serves as a loading control; (<b>B</b>) the overexpression of KDM2A leads to a statistically significant decrease in TCF/LEF protein levels. The Western blot signals were quantified using Fiji software v1.54f [<a href="#B39-cells-12-02620" class="html-bibr">39</a>]. The TCF/LEF signals were normalized against those of the GAPDH loading control. The data are presented as the ratio between the normalized signal from the cells transfected with the empty pCS2 plasmid (empty) and that from the cells transfected with the pCS2-KDM2A plasmid (KDM2A); (<b>C</b>) quantitative RT-PCR confirmed that the overexpression of KDM2A does not affect the <span class="html-italic">TCF/LEF</span> mRNA levels when related to the HPRT1 mRNA levels. Similar results were obtained when the <span class="html-italic">TCF/LEF</span> mRNA levels were normalized against RPL32; (<b>D</b>) KDM2A and its domains. The JmjC demethylase domain is localized at the N-terminus (in blue), and its function is abolished by the H212A/D214A mutation (mutJmjC) [<a href="#B20-cells-12-02620" class="html-bibr">20</a>]. The zinc finger CXXC domain (in yellow) is disrupted by the K601A mutation [<a href="#B20-cells-12-02620" class="html-bibr">20</a>]; (<b>E</b>) the canonical Wnt signaling luciferase reporter TOP5 is activated by the pathway agonist BIO but repressed by elevated levels of both the wild-type KDM2A (wt) and the mutant KDM2A, whose demethylase domain was disrupted (mutJmjC). The mutant KDM2A protein whose CXXC domain was disrupted (mutCXXC) was not able to repress the activated reporter. The luciferase reporter activity is expressed as a fold change ratio between the normalized luciferase signal from the TOP5 transfected cells and that from the FOP5 transfected cells; (<b>F</b>) the overexpression of both the wild-type and the mutant demethylase domain defective KDM2A proteins results in lower TCF/LEF levels, whereas the mutant KDM2A protein with a disrupted CXXC domain failed to destabilize TCF/LEFs; (<b>G</b>) the overexpression of both the wild-type (wt) and the JmjC demethylase domain defective (mutJmjC) KDM2A protein results in a significant decrease in the TCF/LEF protein levels, whereas the CXXC mutant KDM2A protein (mutCXXC) does not affect the TCF/LEF protein levels. The blots were analyzed using the same approach as in (<b>B</b>). The statistical significance was determined by the student’s <span class="html-italic">t</span>-test as follows: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 (n = 3).</p>
Full article ">Figure 2
<p>The KDM2A-mediated destabilization of TCF/LEFs is proteasome-dependent. (<b>A</b>) TCF/LEFs and beta-catenin are stabilized when the proteasome is inhibited by MG132. The cycloheximide treatment (CHX) was used to analyze the half-life of TCF/LEFs and beta-catenin; (<b>B</b>) the KDM2A-mediated degradation of TCF/LEFs and the nuclear beta-catenin (non-phospho) are prevented by blocking the function of the proteasome with MG132. Ubiquitinated proteins cannot be degraded and accumulate in the cells treated with MG132; (<b>C</b>) blocking the proteasome with MG132 stabilizes the TCF7L2 M/S isoforms and leads to their significantly higher levels. The Western blot signals were quantified using Fiji software v1.54f [<a href="#B39-cells-12-02620" class="html-bibr">39</a>], and they were normalized against the GAPDH loading control. The data are presented as the ratio between the normalized signals from the cells treated with MG132 and those treated with DMSO; (<b>D</b>) blocking the proteasome with MG132 prevents the KDM2A-mediated degradation of TCF7LS M/S isoforms and results in their significantly higher levels in comparison to the DMSO control. Blots were quantified as in (<b>C</b>), and the data are presented as the ratio between the normalized signals from the cells transfected with the empty pCS2 plasmid (empty) and those from the cells transfected with the pCS2-KDM2A plasmid (KDM2A). The statistical significance was determined by the student’s <span class="html-italic">t</span>-test as follows: * <span class="html-italic">p</span> &lt; 0.05 (n = 3).</p>
Full article ">Figure 3
<p>The KDM2A-mediated destabilization of TCF/LEFs is neddylation dependent. (<b>A</b>) The inhibition of neddylation with the neddylation inhibitor MLN4924 stabilizes TCF/LEFs and prevents them from being destabilized by elevated levels of KDM2A. The amount of neddylated cullins, the major NEDD8 substrate in the cell [<a href="#B44-cells-12-02620" class="html-bibr">44</a>], is lower in the cells treated with MLN4924; (<b>B</b>) blocking neddylation with MLN4924 stabilizes the TCF7L2 M/S isoforms and leads to their significantly higher levels. The Western blot signals were quantified using Fiji software v1.54f [<a href="#B39-cells-12-02620" class="html-bibr">39</a>], and they were normalized against the GAPDH loading control. The data are presented as the ratio between the normalized signals from the cells treated with MLN4924 and those treated with DMSO; (<b>C</b>) blocking neddylation with MLN4924 prevents the KDM2A-mediated degradation of TCF7LS M/S isoforms and results in their significantly higher levels in comparison to the DMSO control. Blots were quantified as in (<b>B</b>), and the data are presented as the ratio between the normalized signals from the cells transfected with the empty pCS2 plasmid (empty) and those from the cells transfected with the pCS2-KDM2A plasmid (KDM2A); (<b>D</b>) the TOP5 luciferase reporter is induced by the neddylation inhibitor MLN4924. The luciferase reporter activity is expressed as a fold change ratio between the normalized luciferase signal from the TOP5 transfected cells and that from the FOP5 transfected cells; (<b>E</b>) blocking neddylation with MLN4924 results in significantly higher mRNA levels of the canonical Wnt signaling target gene <span class="html-italic">AXIN2</span> when related to HPRT1. Similar results were obtained when the <span class="html-italic">AXIN2</span> mRNA levels were normalized against RPL32; (<b>F</b>) the KDM2A-mediated degradation of TCF7L2 and LEF1 is prevented when ubiquitination is blocked with PYR-41; (<b>G</b>) the TCF7L2 M/S levels are not significantly changed after treatment with PYR-41. The Western blot signals were quantified as in (<b>B</b>) using Fiji software v1.54f [<a href="#B39-cells-12-02620" class="html-bibr">39</a>] and normalized against the LAMIN B1 loading control. The data are presented as the ratio between the normalized signals from the cells treated with PYR-41 and those treated with DMSO. (<b>H</b>) Blocking ubiquitination with PYR-41 prevents the KDM2A-promoted destabilization of the TCF7L2 M/S proteins. The signals were analyzed as in (<b>G</b>). The data are presented as the ratio between the normalized signals from the cells transfected with the empty pCS2 plasmid (empty) and those from the cells transfected with the pCS2-KDM2A plasmid (KDM2A). The statistical significance was determined by the student’s <span class="html-italic">t</span>-test as follows: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 (n = 3).</p>
Full article ">Figure 4
<p>KDM2A interacts with TCF7L2 and beta-catenin. (<b>A</b>) Both TCF7L2 isoforms (E and M/S) were co-trapped with EGFP-KDM2A. The GFP-trap confirmed the interaction between beta-catenin and KDM2A; (<b>B</b>) the endogenous KDM2A and beta-catenin were co-trapped with EGFP-TCF7L2; (<b>C</b>) the endogenous TCF7L2 was co-trapped with EGFP-beta-catenin; (<b>D</b>) TCF7L2 and its domains. The beta-catenin domain (BD, in green) lies at the N-terminus. The evolutionarily conserved domain (CD, in orange) and DNA binding domain (HMG, in blue) lie in the central region. The HMG domain and the nuclear localization domain (NLS) are missing in the TCF7L2delC mutant protein; (<b>E</b>) Myc-tagged (MT) KDM2A co-immunoprecipitated with both the full-length (TCF7L2AA1-461) and the N-terminally truncated FLAG-tagged TCF7L2 (delN = TCF7L2AA162-461), but not with the C-terminally truncated FLAG-tagged TCF7L2 (delC = TCF7L2AA1-321); (<b>F</b>) KDM2A whose CXXC domain was disrupted by the K601A mutation failed to co-immunoprecipitate with FLAG-tagged TCF7L2.</p>
Full article ">
16 pages, 2524 KiB  
Article
Transcriptional Regulation of the Human 5-HT1A Receptor Gene by Lithium: Role of Deaf1 and GSK3β
by Emerson F. Harkin, Georges Nasrallah, Brice Le François and Paul R. Albert
Int. J. Mol. Sci. 2023, 24(21), 15620; https://doi.org/10.3390/ijms242115620 - 26 Oct 2023
Cited by 1 | Viewed by 1585
Abstract
Serotonin 1A (5-HT1A) autoreceptors located on serotonin neurons inhibit their activity, and their upregulation has been implicated in depression, suicide and resistance to antidepressant treatment. Conversely, post-synaptic 5-HT1A heteroreceptors are important for antidepressant response. The transcription factor deformed epidermal autoregulatory factor 1 (Deaf1) [...] Read more.
Serotonin 1A (5-HT1A) autoreceptors located on serotonin neurons inhibit their activity, and their upregulation has been implicated in depression, suicide and resistance to antidepressant treatment. Conversely, post-synaptic 5-HT1A heteroreceptors are important for antidepressant response. The transcription factor deformed epidermal autoregulatory factor 1 (Deaf1) acts as a presynaptic repressor and postsynaptic enhancer of 5-HT1A transcription, but the mechanism is unclear. Because Deaf1 interacts with and is phosphorylated by glycogen synthase kinase 3β (GSK3β)—a constitutively active protein kinase that is inhibited by the mood stabilizer lithium at therapeutic concentrations—we investigated the role of GSK3β in Deaf1 regulation of human 5-HT1A transcription. In 5-HT1A promoter-reporter assays, human HEK293 kidney and 5-HT1A-expressing SKN-SH neuroblastoma cells, transfection of Deaf1 reduced 5-HT1A promoter activity by ~45%. To identify potential GSK3β site(s) on Deaf1, point mutations of known and predicted phosphorylation sites on Deaf1 were tested. Deaf1 repressor function was not affected by any of the mutants tested except the Y300F mutant, which augmented Deaf1 repression. Both lithium and the selective GSK3 inhibitors CHIR-99021 and AR-014418 attenuated and reversed Deaf1 repression compared to vector. This inhibition was at concentrations that maximally inhibit GSK3β activity as detected by the GSK3β-sensitive TCF/LEF reporter construct. Our results support the hypothesis that GSK3β regulates the activity of Deaf1 to repress 5-HT1A transcription and provide a potential mechanism for actions of GSK3 inhibitors on behavior. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Activity of point mutants of reported and predicted Deaf1 phosphorylation sites. (<b>A</b>) Schematic diagram of the structural domains characterized in Deaf1 and positions of reported and predicted phosphorylation sites. Domains are the Alanine-Acidic domain (AA), the SAND domain (SAND), the nuclear localization signal (NLS), the nuclear export signal (NES) and the MYND domain (MYND). Previously published phosphorylated sites are framed in red. (<b>B</b>,<b>C</b>) Deaf1 and mutant activity at the 5-HT1A promoter in HEK-293 (<b>B</b>) or SKN-SH (<b>C</b>) cells. Cells were transfected with the pGL3B reporter construct (with or without the 5-HT1A promoter), the pCDNA3 construct (with or without HisDeaf1 or mutants), and pCMV-β-gal. Results for reported sites and predicted sites are both shown. Data are shown as mean ± SEM of relative normalized luminescence (RLN), i.e., the relative luciferase activity was normalized to pGL3B-Vector. Values of all samples were compared by two-way ANOVA followed by Bonferroni-adjusted pairwise comparisons to find <span class="html-italic">p</span> values. * <span class="html-italic">p</span> &lt; 0.05 compared to HisDEAF1; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.01 compared to pCDNA3.</p>
Full article ">Figure 2
<p>Two-dimensional-gel analysis of Deaf1 and Deaf1 Y300F mutant.</p>
Full article ">Figure 3
<p>Effect of Lithium or PMA on Deaf1 activity at the 5-HT1A promoter in HEK293 cells. (<b>A</b>) 5-HT1A promoter regulation by Deaf1 in HEK293 cells. pGL3B-luciferase reporter without or with the 5-HT1A promoter (5-HT1A) was cotransfected with pcDNA3 without (Vector) or with Deaf1 coding sequence. Units are relative normalized luminescence (RNL) normalized to 5-HT1A-Vector and shown as mean ± SEM; * <span class="html-italic">p</span> &lt; 0.05 compared to Vector control. (<b>B</b>,<b>C</b>) Drug treatments: Cells were transfected with 5-HT1A-luciferase, pcDNA3 without or with HisDeaf1, and pCMV-β-gal. Data are plotted as mean ± SEM of 3 independent experiments each. (<b>B</b>) Assays with the indicated concentration (mM) of lithium (LiCl, GSK3β inhibitor) or sodium chloride (NaCl) as osmotic control; RNL was normalized to 2.5 mM NaCl vector control. (<b>C</b>) Assays with PMA (PKC activator) or vehicle (DMSO) treatment; RNL was normalized to 2.5 µL DMSO control. The RNL values of all samples were compared by two-way ANOVA followed by Bonferroni-adjusted pairwise comparisons to find <span class="html-italic">p</span> values: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001. (<b>D</b>) Deaf1-independent enhancer activity of lithium. Mouse embryo fibroblast cells from Deaf1 knockout mice were transfected with the 5-HT1A promoter construct and empty Deaf1 vector and treated with 10 mM LiCl or NaCl. ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 4
<p>Effect of GSK3β inhibitors on Deaf1 activity at the 5-HT1A promoter. (<b>A</b>) AR014418. Effect of AR on the 5-HT1A promoter in the presence and absence of overexpressed Deaf1. Significant concentration × plasmid interaction (F<sub>(1,49)</sub> = 20.2, <span class="html-italic">p</span> &lt; 0.001, R<sup>2</sup> = 0.47). (<b>B</b>) CHIR-99021 (CHIR). Effect of CHIR on the 5-HT1A promoter in the presence and absence of overexpressed Deaf1. Significant concentration × plasmid interaction (F<sub>(1,51)</sub> = 73.6, <span class="html-italic">p</span> &lt; 0.001, R<sup>2</sup> = 0.63). (<b>C</b>) SB216763 (SB). Effect of SB on the 5-HT1A promoter in the presence and absence of overexpressed Deaf1. Concentration × plasmid interaction (F<sub>(1,57)</sub> = 0.50, <span class="html-italic">p</span> = 0.48, R<sup>2</sup> = 0.40). Data are presented as mean ± SEM, <span class="html-italic">n</span> = 3–4. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 compared to paired vector; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 compared to respective vehicle control. The % repression is shown numerically above each concentration.</p>
Full article ">Figure 5
<p>Concentration-dependent inhibition of GSK3β activity on TCF-LEF promoter. Drugs were tested on cells transfected with a TCF/LEF firefly luciferase reporter and relative luciferase activity normalized to Vector control (RNL), with higher RNL representing greater inhibition. Data are plotted as mean ± SEM, <span class="html-italic">n</span> = 3. (<b>A</b>) Effect of lithium on GSK3β activity. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 compared with the lowest concentration of the same drug; <sup><span>$</span></sup> <span class="html-italic">p</span> &lt; 0.05 for lithium vs. sodium. <span class="html-italic">n</span> = 3. (<b>B</b>) Effect of CHIR on GSK3β activity. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. vehicle. (<b>C</b>) Effect of SB on GSK3β activity. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 compared with vehicle.</p>
Full article ">
22 pages, 3243 KiB  
Article
DCUN1D1 Is an Essential Regulator of Prostate Cancer Proliferation and Tumour Growth That Acts through Neddylation of Cullin 1, 3, 4A and 5 and Deregulation of Wnt/Catenin Pathway
by Akhona Vava, Juliano D. Paccez, Yihong Wang, Xuesong Gu, Manoj K. Bhasin, Michael Myers, Nelson C. Soares, Towia A. Libermann and Luiz F. Zerbini
Cells 2023, 12(15), 1973; https://doi.org/10.3390/cells12151973 - 31 Jul 2023
Cited by 5 | Viewed by 2240
Abstract
Defective in cullin neddylation 1 domain containing 1 (DCUN1D1) is an E3 ligase for the neddylation, a post-translational process similar to and occurring in parallel to ubiquitin proteasome pathway. Although established as an oncogene in a variety of squamous cell carcinomas, the precise [...] Read more.
Defective in cullin neddylation 1 domain containing 1 (DCUN1D1) is an E3 ligase for the neddylation, a post-translational process similar to and occurring in parallel to ubiquitin proteasome pathway. Although established as an oncogene in a variety of squamous cell carcinomas, the precise role of DCUN1D1 in prostate cancer (PCa) has not been previously explored thoroughly. Here, we investigated the role of DCUN1D1 in PCa and demonstrated that DCUN1D1 is upregulated in cell lines as well as human tissue samples. Inhibition of DCUN1D1 significantly reduced PCa cell proliferation and migration and remarkably inhibited xenograft formation in mice. Applying both genomics and proteomics approaches, we provide novel information about the DCUN1D1 mechanism of action. We identified CUL3, CUL4B, RBX1, CAND1 and RPS19 proteins as DCUN1D1 binding partners. Our analysis also revealed the dysregulation of genes associated with cellular growth and proliferation, developmental, cell death and cancer pathways and the WNT/β-catenin pathway as potential mechanisms. Inhibition of DCUN1D1 leads to the inactivation of β-catenin through its phosphorylation and degradation which inhibits the downstream action of β-catenin, reducing its interaction with Lef1 in the Lef1/TCF complex that regulates Wnt target gene expression. Together our data point to an essential role of the DCUN1D1 protein in PCa which can be explored for potential targeted therapy. Full article
(This article belongs to the Section Cellular Metabolism)
Show Figures

Figure 1

Figure 1
<p>DCUN1D1 is upregulated in prostate cancer cell lines and prostate cancer tissue samples. (<b>a</b>) RT-PCR analysis of DCUN1D1 in prostate cancer cell lines. Total RNA was collected from PREC (normal), DU145, DUCaP, PC3, CL1 (androgen independent) and CW22, CW19, LNCaP and VCaP (androgen dependent cells). Normalization of each sample was carried out by measuring the amount of human GAPDH cDNA. (<b>b</b>) Western blot analysis of DCUN1D1 expression in the prostate cancer cell lines. DCUN1D1 expression was probed with anti-DCUN1D1 antibody. Normalization of each sample was carried out by β-tubulin antibodies. (<b>c</b>) Immunohistochemical staining of DCUN1D1 in representative prostate cancer cells (arrowhead) of tumour tissue exhibited intense expression compared to adjacent normal prostate tissue (long arrow). On the left, staining of DCUN1D1 in normal and prostate cancer tissue (Magnification 100×). On the right, DCUN1D1 immunohistochemical staining of prostate cancer tissue (Magnification 200×). Experiments were independently repeated three times and a representative image of an independent experiment is represented.</p>
Full article ">Figure 2
<p>Blockage of DCUN1D1 expression inhibits proliferation and migration and induces apoptosis in prostate cancer cell lines. Prostate cancer DU145 and PC3 cell lines were infected with lentivirus-encoding shRNA against GFP and DCUN1D1. (<b>a</b>) Real-time PCR analysis of DCUN1D1 in prostate cancer cell lines 48 h post-infection. Total RNA was collected from DU145 and PC3 cell lines and normalized by measuring the amount of glyceraldehyde-3-phosphate dehydrogenase (GAPDH)-complementary DNA. (<b>b</b>) Western blot analysis of DCUN1D1 expression in DU145 and PC3 cell lines 48 h post-infection. DCUN1D1 expression was probed with anti-DCUN1D1 antibody and normalized by measuring the amount of glyceraldehyde-3-phosphate dehydrogenase (GAPDH). (<b>c</b>) Proliferations of DU145 and PC3 48 h post-infection. Data means ± s.d. of triplicate independent experiments. (<b>d</b>) Migrations of DU145 and PC3 cell lines were measured 48 h post-infection in transwell plates. Migrating cells were fixed, stained and 3–5 random microscopic fields were counted. Values shown are mean ± s.d. from a representative experiment. (<b>e</b>) Apoptosis analysis of DU145 cell lines post-infection. The Cell Death Elisa Plus kit was used to quantify apoptosis 24 h post-seeding of the cells. Data means ± s.d. of triplicate independent experiments. * <span class="html-italic">p</span> &lt; 0.05, two-tailed Student’s <span class="html-italic">t</span>-test. Experiments were independently repeated three times and a representative image of an independent experiment is represented.</p>
Full article ">Figure 3
<p>Inhibition of DCUN1D1 reduces tumour formation in MF1 nude mice. DU145 cells infected with Lv-shRNA DCUN1D1 (DU145 DCUN1D1-KD) or Lv-shRNA GFP (DU145-GFP) were implanted subcutaneously into MF1 nude mice. (<b>a</b>) The tumour weight was measured 60 days after implantation. Values are represented as mean ± s.d. of six individuals. (<b>b</b>) Tumour development throughout the study. Values are represented as mean ± s.d. of six individuals at each time point. * <span class="html-italic">p</span> &lt; 0.05, two-tailed Student’s <span class="html-italic">t</span>-test.</p>
Full article ">Figure 4
<p>Bioinformatics analysis of functions, pathways and proteins dysregulated following DCUN1D1 knockdown in PCa cells (<b>a</b>) Enriched functions dysregulated in DU145 cells expressing the DCUN1D1 shRNA. (<b>b</b>) Top 10 pathways dysregulated following DCUN1D1 knockdown in DU145 prostate cancer cells. The list of genes up- and downregulated in DU145 cells expressing the DCUN1D1 shRNA was imported into the Ingenuity Pathway Analysis tool and canonical pathways were determined. Output represents dysregulated functions or pathways expressed as −log (<span class="html-italic">p</span>-value). (<b>c</b>) Bar chart of Panther pathway output from DU145 cells expressing DCUN1D1 shRNA.</p>
Full article ">Figure 5
<p>Blockage of DCUN1D1 decreases global neddylation, ubiquitination and expression of neddylation components and shows preferential neddylation activity of cullin proteins in PCa. Western blot analysis of DU145 and DU145 DCUN1D1-KD cells. Protein extracts were obtained from the cells and subjected to Western blot analysis using (<b>a</b>) anti-ubiquitin, (<b>b</b>) anti-Nedd8. Blockage of DCUN1D1 decreased the expression of the neddylation pathway components including (<b>c</b>) the E1 NAE heterodimer APPB1 (top panel) and the neddylation-conjugating enzyme, UBC12 (bottom panel) and (<b>d</b>) the cullin-associated proteins RBX1 (top panel) and CAND1 (bottom panel). Inhibition of DCUN1D1 showed preferential NEDD8 modification of the cullin family of proteins. Immunoblot analysis of DU145 and DU145 DCUN1D1 knockdown cell protein extracts using (<b>e</b>) anti-cullin 1, (<b>f</b>) anti-cullin 3, (<b>g</b>) anti-cullin 4A, (<b>h</b>) anti-cullin 4B and (<b>i</b>) anti-cullin 5. The GAPDH loading control was probed using the anti-GAPDH antibody. Experiments were independently repeated three times and a representative image of an independent experiment is represented.</p>
Full article ">Figure 6
<p>Inhibition of DCUN1D1 deactivated the WNT signalling pathway. Protein extracts containing DU145-GFP and DU145 DCUN1D1-KD cell lines were analysed using Western blot analysis. Inhibition of DCUN1D1 (<b>a</b>) increased phosphorylation of β-catenin and reduced expression of total β-catenin and (<b>b</b>) increased the phosphorylated levels of Gsk-3β. The GAPDH was used as loading control. (<b>c</b>) Blockage of DCUN1D1 inhibits ubiquitination of β-catenin. DU145 DCUN1D1-KD and DU145-GFP cells were transfected with pCDNA Myc–Ub and pCDNA Flag–DCUN1D1 expression vector or parental vector and treated with proteasome inhibitor (MG132) for 4 h. Proteins were immunoprecipitated using anti-β-catenin antibody. Ubiquitinated β-catenin was detected by using anti-ubiquitin antibody. Re-expression of DCUN1D1 protein restores β-catenin expression. (<b>d</b>) Blockage of DCUN1D1 expression inhibited β-catenin/ Lef1 interaction. Proteins from DU145 and DU145 DCUN1D1-KD transfected with pCDNA Flag–DCUN1D1 cells were immunoprecipitated using anti-β-catenin antibody. The interaction between the proteins was detected by anti- Lef1 antibodies (<b>e</b>) Schematic representation of the DCUN1D1 pathway in PCa. Experiments were independently repeated three times and a representative image of an independent experiment is represented.</p>
Full article ">
25 pages, 4369 KiB  
Article
WNT3a Signaling Inhibits Aromatase Expression in Breast Adipose Fibroblasts—A Possible Mechanism Supporting the Loss of Estrogen Responsiveness of Triple-Negative Breast Cancers
by Alexander Kaiser, Gabriele Eiselt, Joachim Bechler, Otmar Huber and Martin Schmidt
Int. J. Mol. Sci. 2023, 24(5), 4654; https://doi.org/10.3390/ijms24054654 - 28 Feb 2023
Cited by 1 | Viewed by 1865
Abstract
Estrogen-dependent breast cancers rely on a constant supply of estrogens and expression of estrogen receptors. Local biosynthesis, by aromatase in breast adipose fibroblasts (BAFs), is their most important source for estrogens. Triple-negative breast cancers (TNBC) rely on other growth-promoting signals, including those from [...] Read more.
Estrogen-dependent breast cancers rely on a constant supply of estrogens and expression of estrogen receptors. Local biosynthesis, by aromatase in breast adipose fibroblasts (BAFs), is their most important source for estrogens. Triple-negative breast cancers (TNBC) rely on other growth-promoting signals, including those from the Wnt pathway. In this study, we explored the hypothesis that Wnt signaling alters the proliferation of BAFs, and is involved in regulation of aromatase expression in BAFs. Conditioned medium (CM) from TNBC cells and WNT3a consistently increased BAF growth, and reduced aromatase activity up to 90%, by suppression of the aromatase promoter I.3/II region. Database searches identified three putative Wnt-responsive elements (WREs) in the aromatase promoter I.3/II. In luciferase reporter gene assays, promoter I.3/II activity was inhibited by overexpression of full-length T-cell factor (TCF)-4 in 3T3-L1 preadipocytes, which served as a model for BAFs. Full-length lymphoid enhancer-binding factor (LEF)-1 increased the transcriptional activity. However, TCF-4 binding to WRE1 in the aromatase promoter, was lost after WNT3a stimulation in immunoprecipitation-based in vitro DNA-binding assays, and in chromatin immunoprecipitation (ChIP). In vitro DNA-binding assays, ChIP, and Western blotting revealed a WNT3a-dependent switch of nuclear LEF-1 isoforms towards a truncated variant, whereas β-catenin levels remained unchanged. This LEF-1 variant revealed dominant negative properties, and most likely recruited enzymes involved in heterochromatin formation. In addition, WNT3a induced the replacement of TCF-4 by the truncated LEF-1 variant, on WRE1 of the aromatase promoter I.3/II. The mechanism described here may be responsible for the loss of aromatase expression predominantly associated with TNBC. Tumors with (strong) expression of Wnt ligands actively suppress aromatase expression in BAFs. Consequently a reduced estrogen supply could favor the growth of estrogen-independent tumor cells, which consequently would make estrogen receptors dispensable. In summary, canonical Wnt signaling within (cancerous) breast tissue may be a major factor controlling local estrogen synthesis and action. Full article
(This article belongs to the Special Issue Prevention and Drug Treatment of Breast Cancer)
Show Figures

Figure 1

Figure 1
<p>Breast cancer cell line conditioned medium (CM) and WNT3a CM potentiate breast adipose fibroblast (BAF) proliferation. Starting two days after seeding, BAFs were incubated for 8 days with various concentrations of CM. Media were changed every other day. Vital cells were assayed with the fluorescein diacetate (FDA) assay. (<b>A</b>) BAFs were cultured in M199 medium, without or with different volume percentages of serum-free CM from MDA-MB231 cells, MCF-7 cells, or with 30% serum-free media alone. MCF-7 CM caused no significant changes in vital cells (parameter for vital cells: RFU, relative fluorescence units). MDA-MB231 CM induced a dose-dependent increase in vital cells. Numbers indicate biological replicates tested in triplicate. (<b>B</b>) BAFs were grown in the presence of 50% M199/FBS and various concentrations of serum-free CM from the indicated cell lines (the balance was filled with serum-free medium). n = 2 experiments tested in quadruplicate. Subgroup mean values were calculated from the mean values obtained for the triple-negative and receptor-positive cell lines, respectively. (<b>C</b>) WNT3a CM induced a concentration-dependent increase in vital cells, whereas 30% serum-free CM from L-M(TK-)-null cells, not expressing WNT3a, had no such effect. All values represent means ± SEM, and were normalized to 100% M199/fetal bovine serum (FBS) (*, <span class="html-italic">p</span> &lt; 0.05; **, <span class="html-italic">p</span> &lt; 0.005; versus M199/FBS or 0% CM, respectively).</p>
Full article ">Figure 2
<p>Aromatase in BAFs is inhibited by MDA-MB231 CM, canonical Wnt signaling, or active HDACs. BAFs were incubated for 24 h in serum-free medium supplemented with 10 µM forskolin for aromatase induction (grey bars), or vehicle (no forskolin, white bars). Different volume percentages of MDA-MB231 CM, WNT3a CM, or different concentrations of glycogen synthase kinase-3β (GSK-3β) inhibitors (BIO, lithium chloride), or the histone deacetylase (HDAC) inhibitor panobinostat (50 nM) were applied throughout the incubation time, as indicated. For measurement of aromatase activity, <sup>3</sup>H-labeled androstenedione was added 6 h before termination of the incubation time. Cell lysates for mRNA analyses were prepared after 24 h of treatment with forskolin, in the absence or presence of 50% (<span class="html-italic">v</span>/<span class="html-italic">v</span>) WNT3a CM. (<b>A</b>) MDA-MB231 CM inhibited aromatase activity dose-dependently. (<b>B</b>) Basal aromatase activity was not affected by WNT3a CM, however, forskolin stimulated aromatase activity was reduced by more than 90% in the presence of 50% (<span class="html-italic">v</span>/<span class="html-italic">v</span>) WNT3a CM. (<b>C</b>) This inhibitory effect of WNT3a CM was dose-dependent. (<b>D</b>) Consistently, aromatase mRNA expression was inhibited upon treatment of BAFs with WNT3a CM. (<b>E</b>,<b>F</b>) WNT3a CM similarly suppressed transcription via promoters I.3 and II, respectively, on RNA level. The effects of the GSK-3β inhibitors BIO (<b>G</b>) and lithium chloride (<b>H</b>) confirm the involvement of canonical Wnt signaling in the mechanism of WNT3a-triggered inhibition of aromatase activity. (<b>I</b>) HDAC inhibition by panobinostat markedly increased aromatase activity, and WNT3a CM significantly inhibited aromatase activity in panobinostat-treated BAFs. Data are normalized to values from cells treated with forskolin alone, and are presented as means ± SEM of 3 (in <b>A</b>,<b>C</b>,<b>G</b>–<b>I</b>) or 4 (in <b>B</b>,<b>D</b>–<b>F</b>) independent experiments. Significant differences, as compared to values for the respective controls treated with forskolin, were identified by Student’s <span class="html-italic">t</span>-test (*, <span class="html-italic">p</span> &lt; 0.05) or (in <b>B</b>,<b>D</b>–<b>F</b>) the Mann–Whitney rank sum test (*, <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 3
<p>Transcription factor binding sites and Wnt response elements (WRE) in the aromatase promoter I.3/II. MatInspector (Genomatix) database searches revealed three putative WREs in the aromatase promoter I.3/II region (GenBank Nc_000015.10 chromosome 15 reference GRCh38 primary assembly). The core binding element positions are indicated with respect to the promoter II transcriptional start site (TSS). In addition, their genomic localization is given. WRE core binding elements are highlighted in black; full-length WRE sequences are in bold type and underlined. WRE2 combined two core binding sequences in forward and reverse direction, respectively. WRE search results (lower table) were evaluated by MatInspector’s specific matrix similarity score. The higher the score, the higher the sequence identity with the ideal WRE sequence (maximum score 1). The promoter elements in boxes are derived from a review of Chen et al. [<a href="#B1-ijms-24-04654" class="html-bibr">1</a>].</p>
Full article ">Figure 4
<p>Expression and in vitro WRE-binding of Wnt signaling proteins. (<b>A</b>) 3T3-L1 preadipocytes were incubated overnight in serum-free medium with 10 µM forskolin in the absence (Wnt(−)) or in the presence of 50% WNT3a CM (Wnt(+)). For Western blotting, every lane contained 100 µg soluble nuclear extract. Membranes were (re)probed from left to right with the antibodies (ab) indicated. Comparable protein loading was tested by anti-Lamin A/C antibody. MW, molecular weight. One of two experiments is shown. (<b>B</b>–<b>D</b>) For each condition, 50 µg soluble nuclear extracts from 3T3-L1 cells were subjected to immunoprecipitation, with either anti-T-cell factor-4 (TCF-4) (L40C3) or anti- lymphoid enhancer factor-1 (LEF-1) (2D12) antibody. The precipitates were incubated with WRE-specific fluorescent double-stranded oligonucleotides (WRE1, WRE2, WRE3). Binding of labeled WRE-oligonucleotides was suppressed by an excess of the corresponding nonfluorescent competitor oligonucleotide (cWRE1, cWRE2, cWRE3). Immunoprecipitates from WNT3a-stimulated cells showed no specific binding to WRE1 or WRE2. Means ± SE; n = 3; Student’s <span class="html-italic">t</span>-test (*, <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 5
<p>LEF-1 replaces TCF-4 on WRE1 of the aromatase promoter in BAFs upon WNT3a treatment. BAFs were incubated for 24 h in serum-free medium with 10 µM forskolin ±50% WNT3a CM (indicated as Wnt(−) and Wnt(+), respectively). ChIP was performed as described in Methods. Antibodies used for immunoprecipitation were anti-TCF-4 (H-125) (TCF-4), anti-LEF-1 292 (LEF-1), or anti-β-catenin (clone 14) (β-Cat). (<b>A</b>) Two primer sets were used for subsequent PCR: primer set 1 defines an amplicon, which includes all WREs in the aromatase promoter I.3/II region, whereas primer set 2 amplifies a region containing only WRE2 and WRE3. Boxes representing amplicons: numbers in parentheses indicate position in relation to the promoter II transcriptional start site; middle line indicates positions of identified and putative cis-elements in the promoter region (for details see <a href="#ijms-24-04654-f003" class="html-fig">Figure 3</a>). (<b>B</b>) Representative polyacrylamide gels from one PCR with primer set 1. In: 1:50 diluted DNA input; H<sub>2</sub>O: water used as PCR control without template; neg. control: Protein G Sepharose alone used for immunoprecipitation (IP). For quantitation, band intensities obtained from ChIP reactions were expressed as %-values of input signal: (<b>C</b>) Means ± SEM obtained with BAFs from (n = 4) donors using primer set 1. Student’s <span class="html-italic">t</span>-test was used to identify differences between groups (horizontal bars, numbers indicate <span class="html-italic">p</span>-values). (<b>D</b>) Ratios of band intensities obtained for WNT3a-treated and untreated BAFs, using primer set 1, were calculated for each antibody target examined. No response to treatment equals a ratio of 1 (dotted line). Differences to a no-response situation were identified with the Mann–Whitney rank sum test (n = 4; *, <span class="html-italic">p</span> &lt; 0.05). (<b>E</b>) Means ± SEM obtained with BAFs from (n = 4) donors using primer set 2. (<b>F</b>) Ratios of WNT3a responses using primer set 2.</p>
Full article ">Figure 6
<p>Functional consequences of TCF-4 or LEF-1 binding to WREs in the aromatase promoter I.3/II region. 3T3-L1 preadipocytes were transfected with wildtype aromatase promoter I.3/II reporter constructs pGL3-PII-522 (PII-WT), or derived constructs with inactivating mutations in WRE1 (PII-mWRE1), WRE2 (PII-mWRE2), or WRE3 (PII-mWRE3). Alternatively, the TOPflash Wnt reporter vector was used. For co-transfection vectors containing one of the following, TCF-4 or LEF-1 constructs were used as indicated: pCMV4-FLAG containing full-length TCF-4 (TCF-4) or TCF-4 lacking the N-terminal β-catenin binding domain (ΔN-TCF-4); or pCS2+ containing full-length LEF-1 (LEF-VP16), LEF-1 lacking the N-terminal β-catenin binding domain (ΔN-LEF-VP16) or LEF-1 lacking the β-catenin binding domain, and in addition the context-dependent regulatory domain (ΔΔN-LEF-VP16), which were all fused to the VP16 transactivation domain. As controls, empty pCMV4-FLAG or pCS2+ were used (indications PII-WT or TOPflash in the panels). After 24 h, cells were stimulated in serum-free medium with 10 µM forskolin ± 50% WNT3a CM. Cells were lysed 16 h later, and luciferase activities were measured. For better comparison of effects, the results were normalized to the activities of the respective controls (PII-WT), separately for untreated and WNT3a-treated conditions, respectively, in (<b>A</b>–<b>F</b>). (<b>A</b>) Mutations in WRE1 or WRE2 led to an up to 70% increase in luciferase activities after WNT3a incubation, but tended to be inhibitory in controls. (<b>B</b>) Inhibitory effects of TCF-4 and ΔN-TCF-4. (<b>C</b>) TCF-4 effects were independent of single WRE mutations. (<b>D</b>) Differential effects of LEF-1-VP16 constructs without or with N-terminal deletions. (<b>E</b>) WRE1 mutation abolished the significant stimulatory effect of full-length LEF-1-VP16. (<b>F</b>) The inhibitory effect of ΔN-LEF-VP16 depended on an intact WRE1. (<b>G</b>) LEF-1-VP16 overexpression increased WNT3a stimulation-dependent and -independent reporter gene activities. (<b>H</b>) TCF-4 overexpression did not significantly affect luciferase activities. All data are means ± SE of at least 3 experiments. Significant differences to the corresponding controls (PII-WT or TOPflash) (*, <span class="html-italic">p</span> &lt; 0.05), or to the corresponding values without WNT3a treatment (#, <span class="html-italic">p</span> &lt; 0.05), were identified with Student’s <span class="html-italic">t</span>-test.</p>
Full article ">Figure 7
<p>Model for key events in WNT3a-initiated suppression of aromatase expression in BAFs. WNT3a binds Frizzled (FZD) [<a href="#B51-ijms-24-04654" class="html-bibr">51</a>], in association with the co-receptor low-density lipoprotein receptor-related protein 5 or 6 (LRP5/6) [<a href="#B52-ijms-24-04654" class="html-bibr">52</a>]. In consequence, the β-catenin degradation complex (axin, adenomatous polyposis coli (APC), glycogen synthase kinase 3-β (GSK-3β) and casein kinase 1 (CK1)) is inactivated. GSK-3β and CK1, together with axin and APC, translocate to dishevelled (Dvl) at the membrane, and phosphorylate (P) LRP5/6 [<a href="#B53-ijms-24-04654" class="html-bibr">53</a>], but no longer β-catenin. Only phosphorylated β-catenin is ubiquitinylated (U) by E3 ubiquitin ligase β-Trcp (β-Trcp), leading to proteasomal degradation [<a href="#B54-ijms-24-04654" class="html-bibr">54</a>]. In canonical Wnt signaling, unphosphorylated β-catenin accumulates and translocates into the nucleus. In forskolin stimulated BAFs, the Wnt-responsive elements (WREs) within the aromatase promoter I.3/II region bind full-length TCF-4 or LEF-1 proteins. WNT3a stimulation of BAFs induces a switch in WRE occupancy: increasing amounts of a nuclear short LEF-1 variant lead to its binding to WRE1. Aromatase promoter I.3/II activation and aromatase expression may be inhibited via heterochromatin formation, involving transducin-like enhancer of split (TLE) and histone deacetylases (HDACs), associated with the short LEF-1 variant. The figure is based on information from some reviews [<a href="#B6-ijms-24-04654" class="html-bibr">6</a>,<a href="#B7-ijms-24-04654" class="html-bibr">7</a>,<a href="#B8-ijms-24-04654" class="html-bibr">8</a>,<a href="#B9-ijms-24-04654" class="html-bibr">9</a>].</p>
Full article ">Figure A1
<p>Breast cancer cell line CM and WNT3a CM potentiate BAF proliferation. Microscopic images of representative wells from an experiment subsequently used for the proliferation assay (for details see legend to <a href="#ijms-24-04654-f001" class="html-fig">Figure 1</a>B,C).</p>
Full article ">Figure A2
<p><span class="html-italic">WNT1</span> and <span class="html-italic">WNT3A</span> expression in breast cancer cell lines. RNA was isolated from cells used for production of CM under FBS containing or serum-free conditions, respectively, for three days. Expression levels were quantified by qPCR and are normalized to <span class="html-italic">GAPDH</span> expression. (<b>A</b>) <span class="html-italic">WNT1</span> and (<b>B</b>) <span class="html-italic">WNT3A</span> expression in the presence of FBS. (<b>C</b>) <span class="html-italic">WNT1</span> and (<b>D</b>) <span class="html-italic">WNT3A</span> expression in the absence of serum. The boxes indicate median and range for three independent experiments, absence of box indicates lack of quantifiable expression. (<b>E</b>) <span class="html-italic">WNT1</span> and (<b>F</b>) <span class="html-italic">WNT3A</span> expression in the absence of serum were correlated with the proliferation rates of BAFs treated with 30% CM (indicated as vital cells in <a href="#ijms-24-04654-f001" class="html-fig">Figure 1</a>B).</p>
Full article ">
24 pages, 1456 KiB  
Review
Novel Insights into the Role of Kras in Myeloid Differentiation: Engaging with Wnt/β-Catenin Signaling
by Noriko Yokoyama, Hitoshi Nakayama and Kazuhisa Iwabuchi
Cells 2023, 12(2), 322; https://doi.org/10.3390/cells12020322 - 14 Jan 2023
Cited by 2 | Viewed by 2460
Abstract
Cells of the HL-60 myeloid leukemia cell line can be differentiated into neutrophil-like cells by treatment with dimethyl sulfoxide (DMSO). The molecular mechanisms involved in this differentiation process, however, remain unclear. This review focuses on the differentiation of HL-60 cells. Although the Ras [...] Read more.
Cells of the HL-60 myeloid leukemia cell line can be differentiated into neutrophil-like cells by treatment with dimethyl sulfoxide (DMSO). The molecular mechanisms involved in this differentiation process, however, remain unclear. This review focuses on the differentiation of HL-60 cells. Although the Ras proteins, a group of small GTP-binding proteins, are ubiquitously expressed and highly homologous, each has specific molecular functions. Kras was shown to be essential for normal mouse development, whereas Hras and Nras are not. Kras knockout mice develop profound hematopoietic defects, indicating that Kras is required for hematopoiesis in adults. The Wnt/β-catenin signaling pathway plays a crucial role in regulating the homeostasis of hematopoietic cells. The protein β-catenin is a key player in the Wnt/β-catenin signaling pathway. A great deal of evidence shows that the Wnt/β-catenin signaling pathway is deregulated in malignant tumors, including hematological malignancies. Wild-type Kras acts as a tumor suppressor during DMSO-induced differentiation of HL-60 cells. Upon DMSO treatment, Kras translocates to the plasma membrane, and its activity is enhanced. Inhibition of Kras attenuates CD11b expression. DMSO also elevates levels of GSK3β phosphorylation, resulting in the release of unphosphorylated β-catenin from the β-catenin destruction complex and its accumulation in the cytoplasm. The accumulated β-catenin subsequently translocates into the nucleus. Inhibition of Kras attenuates Lef/Tcf-sensitive transcription activity. Thus, upon treatment of HL-60 cells with DMSO, wild-type Kras reacts with the Wnt/β-catenin pathway, thereby regulating the granulocytic differentiation of HL-60 cells. Wild-type Kras and the Wnt/β-catenin signaling pathway are activated sequentially, increasing the levels of expression of C/EBPα, C/EBPε, and granulocyte colony-stimulating factor (G-CSF) receptor. Full article
(This article belongs to the Special Issue Wnt Signaling in Development and Aging)
Show Figures

Figure 1

Figure 1
<p>Wnt/β-catenin signaling pathway. The (<b>left side</b>) shows signaling in the absence of Wnt. In the absence of Wnt signals, β-catenin is degraded by a complex of proteins comprising APC, Axin, CK1, GSK3β, β-catenin, and β-TrCP (the E3-ubiquitin ligase). The rectangle shows the β-catenin destruction complex. Tyrosine phosphorylated GSK3β is active. Sequential phosphorylation of β-catenin by CK1 and GSK3β triggers its ubiquitination by the β-TrCP complex. Ubiquitylated and phosphorylated β-catenin is degraded by the proteasome. PP2A dephosphorylates β-catenin and inhibits its degradation. Red “P” shows tyrosine phosphorylation. The (<b>right side</b>) shows signaling in the presence of Wnt. Wnt ligand binds to its cognate receptors, FZ and Lrp5/6, forming a multiprotein complex known as the signalosome. Lrp5/6 receptors are phosphorylated by CK1α and GSK3β. The cytoplasmic protein Dishevelled (Dvl) is recruited to the plasma membrane and interacts with the FZ receptor and other Dvls. Interactions of Axin with phosphorylated Lrp5/6 and Dvl polymer inactivate the destruction complex. Consequently, β-catenin is stabilized and translocated into the nucleus. β-catenin associates with the Lef/Tcf transcription factor and activates the transcription of target genes. Black “P” shows serine/threonine phosphorylation. The model presented is a modified version and part of a previous model (FASEB Bioadvances, 2019; 00:1–15).</p>
Full article ">Figure 2
<p>Non-canonical Wnt signaling pathways. <b>Left panel</b>: Wnt/PCP pathway; the binding of Wnt to FZ receptor activates the recruitment of Dvl and interacts with Daam1, thereby stimulating a cascade involving the small Rho GTPase family members Rho and Rac, as well as JNK. The PCP pathway regulates actin polymerization and cytoskeletal modifications. <b>Right panel</b>: Wnt/Ca<sup>2+</sup> signaling pathway; upon Wnt binding to an FZ receptor, Dvl and G-protein are recruited to the receptor. PLC activation results in an elevation of IP3, which triggers Ca<sup>2+</sup> release from the ER. Elevated Ca<sup>2+</sup> level in the cytoplasm stimulates PKC, CaMKII, and Calcineurin, activating the transcriptional regulator NFAT. Elevated cytoplasmic Ca<sup>2+</sup> also activates TAK1 and the Nemo-like kinase (NLK) signaling pathway through CaMKII. NLK phosphorylates Tcf/Lef, inhibiting the formation of the β-catenin-Tcf/Lef complex.</p>
Full article ">Figure 3
<p>Association of WT-Kras with Wnt/β-catenin signaling networks in the DMSO-induced differentiation of HL-60 cells. DMSO triggers Kras activation through the recruitment of Kras to the plasma membrane (a,b). AKT phosphorylates GSK3β and inactivates GSK3β (c,d). Accumulated unphosphorylated β-catenin in the cytoplasm translocates to the nucleus (e,f), where it binds to Lef/Tcf and stimulates the transcription of target genes, such as those encoding CEBPα, CEBPɛ, and G-CSF receptor (g). In the absence of DMSO, β-catenin is phosphorylated by a destruction complex consisting of core proteins, including Axin, APC, CK1, β-catenin, GSK3β, and E3-ubiquitin ligase β-TrCP. Degradation of phosphorylated β-catenin follows its ubiquitination by proteasomes (h). Black “P” shows serine/threonine phosphorylation. Red “P” shows tyrosine phosphorylation. The model presented slightly modified a previous model (FASEB Bioadvances, 2019; 00:1-15).</p>
Full article ">
16 pages, 2526 KiB  
Article
Immobilization of Wnt Fragment Peptides on Magnetic Nanoparticles or Synthetic Surfaces Regulate Wnt Signaling Kinetics
by Bin Hu, Michael Rotherham, Neil Farrow, Paul Roach, Jon Dobson and Alicia J. El Haj
Int. J. Mol. Sci. 2022, 23(17), 10164; https://doi.org/10.3390/ijms231710164 - 5 Sep 2022
Cited by 5 | Viewed by 1976
Abstract
Wnt signaling plays an important role in embryogenesis and adult stem cell homeostasis. Its diminished activation is implicated in osteoporosis and degenerative neural diseases. However, systematic administration of Wnt-signaling agonists carries risk, as aberrantly activated Wnt/β-catenin signaling is linked to cancer. Therefore, technologies [...] Read more.
Wnt signaling plays an important role in embryogenesis and adult stem cell homeostasis. Its diminished activation is implicated in osteoporosis and degenerative neural diseases. However, systematic administration of Wnt-signaling agonists carries risk, as aberrantly activated Wnt/β-catenin signaling is linked to cancer. Therefore, technologies for local modulation and control of Wnt signaling targeted to specific sites of disease or degeneration have potential therapeutic value in the treatment of degenerative diseases. We reported a facile approach to locally activate the canonical Wnt signaling cascade using nanomagnetic actuation or ligand immobilized platforms. Using a human embryonic kidney (HEK293) Luc-TCF/LEF reporter cell line, we demonstrated that targeting the cell membrane Wnt receptor, Frizzled 2, with peptide-tagged magnetic nanoparticles (MNPs) triggered canonical Wnt signaling transduction when exposed to a high-gradient, time-varying magnetic field, and the induced TCF/LEF signal transduction was shown to be avidity-dependent. We also demonstrated that the peptide retained signaling activity after functionalization onto glass surfaces, providing a versatile platform for drug discovery or recreation of the cell niche. In conclusion, these results showed that peptide-mediated Wnt signaling kinetics depended not only on ligand concentration but also on the presentation method of the ligand, which may be further modulated by magnetic actuation. This has important implications when designing future therapeutic platforms involving Wnt mimetics. Full article
(This article belongs to the Special Issue Bioactive Nanoparticles)
Show Figures

Figure 1

Figure 1
<p>Frizzled 2 is expressed by HEK293 reporter cells. (<b>A</b>) Reverse transcription-PCR confirmed Frizzled 2 expression at transcriptional level, <span class="html-italic">n</span> = 3 (1–3 = sample number, NTC = no template control). (<b>B</b>) Immunocytochemistry confirmed Frizzled 2 expression at translational level, representative images of <span class="html-italic">n</span> = 3; Frizzled 2 is shown in green (<b>top left</b>), and cell nuclei are shown in blue by DAPI stain (<b>top right</b>), merge channel is shown below, scale bar represents 50 μm.</p>
Full article ">Figure 2
<p>Effect of peptides and two agonists on the activation of canonical Wnt signaling transduction. (<b>A</b>,<b>B</b>) Sequence of UM206_L, UM206_C. (<b>C</b>,<b>D</b>) TCF/LEF signaling transduction induced by indicated peptide concentration for UM206_L (<b>C</b>), UM206_C (<b>D</b>) 3 h (■) and 24 h (<span style="color:red">●</span>) post-treatment in HEK293 Luc-TCF/LEF cells (<span class="html-italic">n</span> = 3). (<b>E</b>,<b>F</b>) The stimulatory effect of 500 ng/mL rmWnt3a (<b>E</b>) and 30 mM LiCl (<b>F</b>) treatment on TCF/LEF transduction at the indicated time points (<span class="html-italic">n</span> = 4). In (<b>C</b>–<b>F</b>), luminescence was presented as fold change with respect to non-treated control (NTC), which was set to 1. The data are expressed as mean ± SEM obtained from two independent experiments (* <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 3
<p>Schematics of the nanomagnetic actuation technique using a magnetic force bioreactor (MFB). MNPs are first functionalized with antibodies or peptides and then incubated with cells. After removing unbound MNPs, cells are exposed to a high-gradient, time-varying magnetic field generated by vertically oscillating NdFeB magnet arrays mounted on stepper motors under computer control [<a href="#B17-ijms-23-10164" class="html-bibr">17</a>].</p>
Full article ">Figure 4
<p>Dose response of UM206 functionalized MNPs on TCF/LEF signaling induction in HEK293 Luc-TCF/LEF cells. (<b>A</b>) Left and right: the starting ratio of peptide-to-MNP is 10 μg-per-mg MNP and 1 μg-per-mg MNP, respectively. (<b>B</b>) Effects of control MNPs without conjugation. (<b>C</b>) Viability of HEK293 cells in response to various treatments and (<b>D</b>) to indicated concentration of peptides. “Loading” indicates MFB exposure, and “Static” indicates no exposure to MFBs. In (<b>A</b>,<b>B</b>), luminescence was defined as fold change with respect to the non-treated control (NTC) group. For (<b>A</b>–<b>D</b>), data are presented as mean ± SEM, <span class="html-italic">n</span> = 4 and shown here is a representative set of results of two independent experiments (* <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 5
<p>Immobilized UM206 peptide retained mild signaling activity on glass substrate. (<b>A</b>) Schematic of UM206 immobilization process onto amine-functionalized glass coverslips using oxygen plasma activation, APTES treatment, then ligand conjugation. (<b>B</b>) Fluorescent images of uncoated glass (<b>left</b>) and glass substrate coated with antibody-ATTO-488 (<b>right</b>) confirmed protein immobilization on glass surface. Bar represents 50 µm, images representative of <span class="html-italic">n</span> = 6. (<b>C</b>) HEK293 Luc-TCF/LEF reporter was mildly activated by both immobilized UM206_L and UM206_C to a comparable level as immobilized Wnt3a after 3 h (<b>C</b>(<b>i</b>)) but remained at similar levels after 24 h (<b>C</b>(<b>ii</b>)). Data are presented as mean fold change ± SEM, <span class="html-italic">n</span> = 6. Shown here are representative results of two independent experiments (* <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001). (<b>D</b>) Immunofluorescent images of active β-catenin in response to UM206 functionalized surfaces. A base level of activated β-catenin was observed in the control (BSA) coated group. An increase in activated nuclear β-catenin was observed in response to surfaces coated with UM206_C, UM206_L, or with Wnt3a positive control after 3 h. Representative images of <span class="html-italic">n</span> = 3 shown. Scale bar represents 50 μm.</p>
Full article ">Figure 5 Cont.
<p>Immobilized UM206 peptide retained mild signaling activity on glass substrate. (<b>A</b>) Schematic of UM206 immobilization process onto amine-functionalized glass coverslips using oxygen plasma activation, APTES treatment, then ligand conjugation. (<b>B</b>) Fluorescent images of uncoated glass (<b>left</b>) and glass substrate coated with antibody-ATTO-488 (<b>right</b>) confirmed protein immobilization on glass surface. Bar represents 50 µm, images representative of <span class="html-italic">n</span> = 6. (<b>C</b>) HEK293 Luc-TCF/LEF reporter was mildly activated by both immobilized UM206_L and UM206_C to a comparable level as immobilized Wnt3a after 3 h (<b>C</b>(<b>i</b>)) but remained at similar levels after 24 h (<b>C</b>(<b>ii</b>)). Data are presented as mean fold change ± SEM, <span class="html-italic">n</span> = 6. Shown here are representative results of two independent experiments (* <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001). (<b>D</b>) Immunofluorescent images of active β-catenin in response to UM206 functionalized surfaces. A base level of activated β-catenin was observed in the control (BSA) coated group. An increase in activated nuclear β-catenin was observed in response to surfaces coated with UM206_C, UM206_L, or with Wnt3a positive control after 3 h. Representative images of <span class="html-italic">n</span> = 3 shown. Scale bar represents 50 μm.</p>
Full article ">
24 pages, 1025 KiB  
Review
Countering Triple Negative Breast Cancer via Impeding Wnt/β-Catenin Signaling, a Phytotherapeutic Approach
by Laleh Arzi, Homa Mollaei and Reyhane Hoshyar
Plants 2022, 11(17), 2191; https://doi.org/10.3390/plants11172191 - 24 Aug 2022
Cited by 9 | Viewed by 2893
Abstract
Triple negative breast cancer (TNBC) is characterized as a heterogeneous disease with severe malignancy and high mortality. Aberrant Wnt/β-catenin signaling is responsible for self-renewal and mammosphere generation, metastasis and resistance to apoptosis and chemotherapy in TNBC. Nonetheless, in the absence of a targeted [...] Read more.
Triple negative breast cancer (TNBC) is characterized as a heterogeneous disease with severe malignancy and high mortality. Aberrant Wnt/β-catenin signaling is responsible for self-renewal and mammosphere generation, metastasis and resistance to apoptosis and chemotherapy in TNBC. Nonetheless, in the absence of a targeted therapy, chemotherapy is regarded as the exclusive treatment strategy for the treatment of TNBC. This review aims to provide an unprecedented overview of the plants and herbal derivatives which repress the progression of TNBC through prohibiting the Wnt/β-catenin pathway. Herbal medicine extracts and bioactive compounds (alkaloids, retinoids. flavonoids, terpenes, carotenoids and lignans) alone, in combination with each other and/or with chemotherapy agents could interrupt the various steps of Wnt/β-catenin signaling, i.e., WNT, FZD, LRP, GSK3β, Dsh, APC, β-catenin and TCF/LEF. These phytotherapy agents diminish proliferation, metastasis, breast cancer stem cell self-renewal and induce apoptosis in cell and animal models of TNBC through the down-expression of the downstream target genes of Wnt signaling. Some of the herbal derivatives simultaneously impede Wnt/β-catenin signaling and other overactive pathways in triple negative breast cancer, including: mTORC1; ER stress and SATB1 signaling. The herbal remedies and their bioactive ingredients perform essential roles in the treatment of the very fatal TNBC via repression of Wnt/β-catenin signaling. Full article
(This article belongs to the Special Issue Plant Therapeutics)
Show Figures

Figure 1

Figure 1
<p>Chemical structure of natural derivatives combating TNBC through interfering with the Wnt/β-catenin pathway (Pubchem).</p>
Full article ">Figure 1 Cont.
<p>Chemical structure of natural derivatives combating TNBC through interfering with the Wnt/β-catenin pathway (Pubchem).</p>
Full article ">Figure 1 Cont.
<p>Chemical structure of natural derivatives combating TNBC through interfering with the Wnt/β-catenin pathway (Pubchem).</p>
Full article ">
15 pages, 2325 KiB  
Article
Identification and Functional Analysis of a Novel CTNNB1 Mutation in Pediatric Medulloblastoma
by Lide Alaña, Caroline E. Nunes-Xavier, Laura Zaldumbide, Idoia Martin-Guerrero, Lorena Mosteiro, Piedad Alba-Pavón, Olatz Villate, Susana García-Obregón, Hermenegildo González-García, Raquel Herraiz, Itziar Astigarraga, Rafael Pulido and Miguel García-Ariza
Cancers 2022, 14(2), 421; https://doi.org/10.3390/cancers14020421 - 14 Jan 2022
Cited by 1 | Viewed by 3161
Abstract
Medulloblastoma is the primary malignant tumor of the Central Nervous System (CNS) most common in pediatrics. We present here, the histological, molecular, and functional analysis of a cohort of 88 pediatric medulloblastoma tumor samples. The WNT-activated subgroup comprised 10% of our cohort, and [...] Read more.
Medulloblastoma is the primary malignant tumor of the Central Nervous System (CNS) most common in pediatrics. We present here, the histological, molecular, and functional analysis of a cohort of 88 pediatric medulloblastoma tumor samples. The WNT-activated subgroup comprised 10% of our cohort, and all WNT-activated patients had exon 3 CTNNB1 mutations and were immunostained for nuclear β-catenin. One novel heterozygous CTNNB1 mutation was found, which resulted in the deletion of β-catenin Ser37 residue (ΔS37). The ΔS37 β-catenin variant ectopically expressed in U2OS human osteosarcoma cells displayed higher protein expression levels than wild-type β-catenin, and functional analysis disclosed gain-of-function properties in terms of elevated TCF/LEF transcriptional activity in cells. Our results suggest that the stabilization and nuclear accumulation of ΔS37 β-catenin contributed to early medulloblastoma tumorigenesis. Full article
(This article belongs to the Section Pediatric Oncology)
Show Figures

Figure 1

Figure 1
<p>Molecular groups of pediatric medulloblastomas. (<b>A</b>): Non-WNT/SHH group are more than 60 cases of the cohort (62/88), Group SHH <span class="html-italic">TP53</span>wt are 15 cases (15/88), Group WNT-activated 9 cases (9/88) and Group SHH <span class="html-italic">TP53</span>mut, 2 cases (2/88). (<b>B</b>): The four groups showed differences in histological distributions.</p>
Full article ">Figure 2
<p>DNA sequencing of <span class="html-italic">CTNNB</span><span class="html-italic">1</span> exon 3 from a tumor specimen. Sanger DNA sequencing showed the deletion c.109-111del, resulting in deletion of a TCT codon in the tumor sample (p.Ser37del).</p>
Full article ">Figure 3
<p><span class="html-italic">CTNNB1</span> mutation distribution in medulloblastoma and functional characterization of the β-catenin ΔS37 variant. (<b>A</b>): The top line shows β-catenin amino acid composition between residues 25–49. Arrows indicate β-catenin residues mutated in our cohort of 88 pediatric medulloblastoma. Residues in red are regulatory phosphorylated residues. The bottom line shows the novel p.Ser37del β-catenin variant (ΔS37) found. Amino acids are denoted using the one-letter code. (<b>B</b>): The plot showing the identity and number of <span class="html-italic">CTNNB1</span> mutations found in pediatric medulloblastoma. Data are from Saint Jude pecan database.</p>
Full article ">Figure 4
<p>Hematoxylin-eosin staining of tumor specimen. Histological sections display tumoral (<b>A</b>): Hematoxylin-eosin stain, 40× magnification and no reticulin net enhancement (<b>B</b>): Reticulin stain, 100× magnification. No anaplastic or large cell changes are noticeable. (<b>C</b>): Hematoxylin-eosin, 200× magnification and (<b>D</b>): Hematoxylin-eosin, 400× magnification.</p>
Full article ">Figure 5
<p>Immunostaining of tumor specimen. The immunoprofile shows patchy nuclear staining for β-catenin (<b>A</b>): 400× magnification, diffuse nuclear stain for YAP1 (<b>B</b>): 100× magnification and OTX2 (<b>C</b>): 200× magnification, and negative staining for GAB1 (<b>D</b>): 200× magnification and p53 (<b>E</b>): 200× magnification.</p>
Full article ">Figure 6
<p>Functional characterization of the novel β-catenin variant c.109-111del (p.Ser37del). (<b>A</b>): Top panel. Immunoblot of endogenous β-catenin (φ), and recombinant β-catenin wild-type (WT) and S33C and ΔS37 variants. U2OS cells were transfected with empty vector (φ) or with plasmids containing the indicated β-catenin variants. Cell lysates were resolved on 4–10% SDS-PAGE under reducing conditions followed by immunoblot using anti-β-catenin antibody. Anti-α-tubulin was used to monitor protein loading. Bottom panel. Plot showing β-catenin/α-tubulin ratio, in arbitrary units (AU), from quantified immunoblot bands from two independent experiments ± SD. (<b>B</b>): Transcriptional activity of β-catenin variants. SEAP-normalized TCF/LEF-driven luciferase activity of β-catenin from U2OS transfected cells, as described in top panel. Luminescence is shown in arbitrary units (AU), from three independent experiments. Statistically significant results (<span class="html-italic">p</span> ˂ 0.05) are marked with *.</p>
Full article ">
13 pages, 1818 KiB  
Article
Tectoridin Stimulates the Activity of Human Dermal Papilla Cells and Promotes Hair Shaft Elongation in Mouse Vibrissae Hair Follicle Culture
by Gary Ka-Wing Yuen, Bryan Siu-Yin Ho, Lish Sheng-Ying Lin, Tina Ting-Xia Dong and Karl Wah-Keung Tsim
Molecules 2022, 27(2), 400; https://doi.org/10.3390/molecules27020400 - 8 Jan 2022
Cited by 9 | Viewed by 3691
Abstract
To search hair growth-promoting herbal extract, a screening platform of having HEK293T fibroblast being transfected with pTOPFLASH DNA construct was developed over a thousand of herbal extracts and phytochemicals were screened. One of the hits was ethanolic extract of Rhizoma Belamcandae, the rhizome [...] Read more.
To search hair growth-promoting herbal extract, a screening platform of having HEK293T fibroblast being transfected with pTOPFLASH DNA construct was developed over a thousand of herbal extracts and phytochemicals were screened. One of the hits was ethanolic extract of Rhizoma Belamcandae, the rhizome of Belamcanda chinensis (L.) DC. Tectoridin, an isoflavone from Rhizoma Belamcandae, was shown to be responsible for this activation of promoter construct, inducing the transcription of pTOPFLASH in the transfected fibroblasts in a dose-dependent manner. The blockage by DKK-1 suggested the action of tectoridin could be mediated by the Wnt receptor. The hair growth-promoting effects of tectoridin were illustrated in human follicular dermal papilla cells and mouse vibrissae organ cultures. In tectoridin-treated dermal papilla cultures, an activation of Wnt signaling was demonstrated by various indicative markers, including TCF/LEF1 transcriptional activity, nuclear translocation of β-catenin, expressions level of mRNAs encoding axin-related protein, (AXIN2), β-catenin, lymphoid enhancer-binding factor-1 (LEF-1), insulin-like growth factor 1 (IGF-1) and alkaline phosphatase (ALP). In addition, an increase of hair shaft elongation was observed in cultured mouse vibrissae upon the treatment of tectoridin. Tectoridin, as well as the herbal extract of Rhizoma Belamcandae, possesses hair promoting activity, which deserves further development. Full article
Show Figures

Figure 1

Figure 1
<p>Chemical fingerprints of extracts deriving from Rhizoma Belamcandae and its role in activating Wnt/β-catenin signaling. (<b>A</b>): Different extracts of Rhizoma Belamcandae (10 mg/mL, injection volume = 10 μL) were subjected to HPLC-DAD analysis. The peaks were revealed at 266 nm. Tectoridin at 20 mg/L was used as the chemical marker here. Representative figures are shown, <span class="html-italic">n</span> = 4. (<b>B</b>): HEK293T cells were transfected with pTOPFLASH DNA construct (insert) for 4 h. The cultures were treated with different extracts for another 24 h. The cell lysate was subjected for luciferase assays. Data are normalized and expressed as the fold (x basal) of control (control group was treated with 0.02% DMSO), in mean ± SEM, <span class="html-italic">n</span> = 4, each with duplicate.</p>
Full article ">Figure 2
<p>Tectoridin activates Wnt/β-catenin signaling. (<b>A</b>) HEK293T cells were transfected with pTOPFLASH DNA construct for 4 h. The cultures were treated with different doses of tectoridin for another 24 h. The cell lysate was used for luciferase assays. (<b>B</b>) HEK293T cells were transfected with as in (<b>A</b>). Then, the cultures were pre-treated with or without DKK-1 (200 ng/mL) for 1 h before the treatment of tectoridin (20 μM), or Wnt3a (200 ng/mL) for 24 h. (<b>C</b>) HEK293T cells were transfected with a DNA construct containing β-catenin tagged with green fluorescent protein (GFP; see insert), followed by treatment of tectoridin (50 µM) or valproic acid (VPA; an activator of Wnt receptor; 5 mM) for 24 h. Data are normalized and expressed as the % of increase, or total, in comparison to control (control group was treated with 0.02% DMSO), in mean ± SEM, <span class="html-italic">n</span> = 4, each with duplicate samples. * <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 3
<p>Tectoridin increases expression of downstream target genes of Wnt/β-catenin signaling. In cultured DPC, different doses of tectoridin (3, 10, 20 and 50 µM, as indicated), or Wnt3a (200 ng/mL), were applied for 48 h. The isolated total RNA from the culture was subjected to RT-PCR analyses of AXIN2, LEF-1, β-catenin, IGF-1, and ALP. Data are normalized and expressed as the fold (x basal) of control (control group was treated with 0.02% DMSO), in mean ± SEM, <span class="html-italic">n</span> = 5, each with duplicate samples. * <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
<p>Tectoridin promotes hair shaft elongation. Individual anagen vibrissae hair follicles were isolated from the upper lip pad of 4-week-old C57BL/6 male mice and cultured in William’s medium E in 5% CO<sub>2</sub> at 37 °C. The cultures were treated with different doses of tectoridin, or WAY316606 (2 μM), as indicated for 3 days. Hair shaft elongation was measured from bottom of hair follicle to the epidermis (upper panel). The measurement of hair length from the hair follicles was performed (low panel). Data are normalized and expressed as the % of increase in comparison to control (control group was treated with 0.02% DMSO), in mean ± SEM, <span class="html-italic">n</span> = 4. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>Proposed action of tectoridin in Wnt/β -catenin signaling.</p>
Full article ">
15 pages, 2983 KiB  
Article
Metformin Dysregulates the Unfolded Protein Response and the WNT/β-Catenin Pathway in Endometrial Cancer Cells through an AMPK-Independent Mechanism
by Domenico Conza, Paola Mirra, Gaetano Calì, Luigi Insabato, Francesca Fiory, Francesco Beguinot and Luca Ulianich
Cells 2021, 10(5), 1067; https://doi.org/10.3390/cells10051067 - 30 Apr 2021
Cited by 16 | Viewed by 3524
Abstract
Multiple lines of evidence suggest that metformin, an antidiabetic drug, exerts anti-tumorigenic effects in different types of cancer. Metformin has been reported to affect cancer cells’ metabolism and proliferation mainly through the activation of AMP-activated protein kinase (AMPK). Here, we show that metformin [...] Read more.
Multiple lines of evidence suggest that metformin, an antidiabetic drug, exerts anti-tumorigenic effects in different types of cancer. Metformin has been reported to affect cancer cells’ metabolism and proliferation mainly through the activation of AMP-activated protein kinase (AMPK). Here, we show that metformin inhibits, indeed, endometrial cancer cells’ growth and induces apoptosis. More importantly, we report that metformin affects two important pro-survival pathways, such as the Unfolded Protein Response (UPR), following endoplasmic reticulum stress, and the WNT/β-catenin pathway. GRP78, a key protein in the pro-survival arm of the UPR, was indeed downregulated, while GADD153/CHOP, a transcription factor that mediates the pro-apoptotic response of the UPR, was upregulated at both the mRNA and protein level. Furthermore, metformin dramatically inhibited β-catenin mRNA and protein expression. This was paralleled by a reduction in β-catenin transcriptional activity, since metformin inhibited the activity of a TCF/LEF-luciferase promoter. Intriguingly, compound C, a well-known inhibitor of AMPK, was unable to prevent all these effects, suggesting that metformin might inhibit endometrial cancer cells’ growth and survival through the modulation of specific branches of the UPR and the inhibition of the Wnt/β-catenin pathway in an AMPK-independent manner. Our findings may provide new insights on the mechanisms of action of metformin and refine the use of this drug in the treatment of endometrial cancer. Full article
(This article belongs to the Special Issue Unfolded Protein Response in Inflammation and Cancer)
Show Figures

Figure 1

Figure 1
<p>Metformin inhibits cell growth and viability in endometrial cancer cells. (<b>A</b>) Ishikawa, HEC1B, or AN3CA cells were seeded at a density of 5 × 10<sup>3</sup> cells in a 96-well plate. After 16 h, cells were treated or not with 1, 5, or 10 mM metformin. Cell viability was measured after 48 h using the MTT assay. Values represent the mean absorbance at 570 nm ± SD of triplicates of three independent experiments. * indicates a <span class="html-italic">p</span>-value &lt; 0.05; *** indicates a <span class="html-italic">p</span>-value &lt; 0.001. (<b>B</b>) Ishikawa, HEC1B, or AN3CA cells were seeded onto six-well plates at a density of 1 × 10<sup>3</sup> cells per well. After cell attachment, 5 mM metformin was added or not to the wells for 4 h. The cells were then cultured with fresh medium. After 2 weeks, the resultant colonies were fixed with 4% paraformaldehyde and stained with hematoxylin. The colonies were then photographed, counted under a microscope, and colony efficiency formation was calculated. ** indicates a <span class="html-italic">p</span>-value &lt; 0.01; *** indicates a <span class="html-italic">p</span>-value &lt; 0.001. (<b>C</b>) Ishikawa, HEC1B, or AN3CA cells (1 × 10<sup>6</sup> per well) were seeded in six-well plates and allowed to form a cell monolayer for 24 h. Cell layers were wounded with a micropipette tip and then incubated in fresh culture medium containing or not 5 mM metformin for 48 h. Cell migration toward the wounded area was observed, photographed, and measured. Experiments were performed three times in triplicate. Graphs show the percentage of wound healing rate. ** indicates a <span class="html-italic">p</span>-value &lt; 0.01. (<b>D</b>) Ishikawa, HEC1B, or AN3CA cells were treated or not with 1, 5, or 10 mM metformin for 48 h. Total cellular proteins were extracted and Western blot experiments were performed with antibodies against PARP and β-actin.</p>
Full article ">Figure 2
<p>Inhibition of AMPK by CC does not alter metformin effects on endometrial cancer cells. (<b>A</b>) Ishikawa, HEC1B, or AN3CA cells were treated or not for 24 h with 5 mM metformin or 10 µM CC or pretreated for 1 h with 10 µM CC followed by treatment with 5 mM metformin. Total cellular proteins were extracted and Western blot experiments were performed, as described in the <a href="#sec2dot3-cells-10-01067" class="html-sec">Section 2.3</a>, using antibodies against p-AMPK (left panels) or p-S6 kinase (right panels). Data represent the mean ± SD of three independent experiments. * <span class="html-italic">p</span> &lt; 0.1; ** <span class="html-italic">p</span> &lt; 0.05; *** <span class="html-italic">p</span> &lt; 0.01. (<b>B</b>) Ishikawa, HEC1B, or AN3CA cells were seeded at a density of 5 × 10<sup>3</sup> cells in a 96-well plate. After 16 h, cells were treated or not with increasing concentration of metformin in the presence or absence of 1 h pretreatment with 10 µM CC. Cell viability was measured after 48 h using the MTT assay. Values represent the mean absorbance at 570 nm ±SD of triplicates of three independent experiments. * indicates a <span class="html-italic">p</span>-value &lt; 0.05; ** indicates a <span class="html-italic">p</span>-value &lt; 0.01. (<b>C</b>) Ishikawa, HEC1B, or AN3CA cells were treated or not for 48 h with 5 mM metformin or 10 µM CC or pretreated for 1 h with 10 µM CC followed by treatment with 5 mM metformin. Total cellular proteins were extracted and Western blot experiments were performed, as described in the <a href="#sec2dot3-cells-10-01067" class="html-sec">Section 2.3</a>, with antibodies against PARP or β-actin (loading control).</p>
Full article ">Figure 3
<p>Metformin modulates the mRNA expression of UPR genes in an AMPK-independent manner in endometrial cancer cells. Ishikawa, HEC1B, or AN3CA cells were treated or not for 24 h with 5 mM metformin in the presence or absence of 1 h pretreatment with 10 µM CC. Total RNA was extracted and real-time RT-PCR experiments were performed using oligonucleotides specific for GRP78, ATF6, ATF4, CHOP, and GAPDH as described in the <a href="#sec2dot4-cells-10-01067" class="html-sec">Section 2.4</a>. Values shown represent the mean (± s.d.) of triplicate samples of three independent experiments. * <span class="html-italic">p</span> &lt; 0.1; ** <span class="html-italic">p</span> &lt; 0.05; *** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 4
<p>Metformin modulates the expression/phosphorylation of UPR proteins and AKT in an AMPK-independent manner in endometrial cancer cells. Ishikawa (<b>A</b>), HEC1B (<b>B</b>), or AN3CA (<b>C</b>) cells were treated or not for 24 h with 5 mM metformin in the presence or absence of 1 h pretreatment with 10 µM CC. Total cellular proteins were extracted and Western blot experiments were performed with antibodies against GRP78, ATF6, ATF4, p-eIF2α, <span class="html-italic">p</span>-AKT, or β-actin (loading control), as described in the <a href="#sec2dot3-cells-10-01067" class="html-sec">Section 2.3</a>. Values shown represent the mean (± s.d.) of three independent experiments. * <span class="html-italic">p</span> &lt; 0.1; ** <span class="html-italic">p</span> &lt; 0.05; *** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 5
<p>Metformin inhibits β-catenin expression independently from AMPK activation in endometrial cancer cells. (<b>A</b>) Ishikawa, HEC1B, or AN3CA cells were treated or not for 24 h with 5 mM metformin. Total RNA was extracted and real-time RT-PCR experiments were performed using oligonucleotides specific to β-catenin and GAPDH as described in the <a href="#sec2dot4-cells-10-01067" class="html-sec">Section 2.4</a>. Values shown represent the mean (± s.d.) of triplicate samples of three independent experiments. ** <span class="html-italic">p</span> &lt; 0.05. (<b>B</b>) Ishikawa, HEC1B, or AN3CA cells were treated or not for 24 h with 5 mM metformin in the presence or absence of 1 h pretreatment with 10 µM CC. Total cellular proteins were extracted and Western blot experiments were performed with antibodies against β-catenin or β-actin (loading control), as described in the <a href="#sec2dot3-cells-10-01067" class="html-sec">Section 2.3</a>. Values shown represent the mean (± s.d.) of three independent experiments. * <span class="html-italic">p</span> &lt; 0.1; ** <span class="html-italic">p</span> &lt; 0.05. (<b>C</b>) Ishikawa and AN3CA cells were grown on glass coverslips for 48 h, then were treated or not for 24 h with 5 mM metformin. Cells were fixed in 4% paraformaldehyde in PBS for 20 min, washed twice in 50 mm NH4Cl in PBS, and permeabilized for 5 min in 0.1% Triton X-100 in PBS. Cells were double-stained with anti-β-catenin antibodies and HOECHST 33258 (Nuclei). Bars, 10 μm. (<b>D</b>) Ishikawa and AN3CA cells were plated in six-well plates to approximately 80% confluence 24 h before transfection. Cells were then transfected with 1.0 μg of the reporter vector BAT-LUX TCF/LEF and 50 ng of pRL-TK vector with Lipofectamine 3000. After 24 h, transfection medium was replaced with fresh medium and cells were treated or not for 24 h with 5 mM metformin, 10 µM CC, or pretreated for 1 h with 10 µM CC followed by treatment with 5 mM metformin. Firefly and renilla activities were determined in cell lysates using the Dual-Luciferase Reporter Assay System and a luminometer. Results were expressed as the ratio of firefly to renilla activity. Values shown represent the mean (± s.d.) of triplicate samples of three independent experiments. * <span class="html-italic">p</span> &lt; 0.1.</p>
Full article ">Figure 6
<p>Metformin inhibits GSK3β phosphorylation in endometrial cancer cells. Ishikawa or AN3CA cells were treated or not for 24 h with 5 mM metformin or 10 µM CC or pretreated for 1 h with 10 µM CC followed by treatment with 5 mM metformin. Total cellular proteins were extracted and Western blot experiments were performed with antibodies against p-GSK3β (Ser9) or vinculin (loading control), as described in the <a href="#sec2dot3-cells-10-01067" class="html-sec">Section 2.3</a>. Values shown represent the mean (± s.d.) of three independent experiments. ** <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 7
<p>Scheme summarizing the observed mechanisms affected by metformin in endometrial cancer cells. Besides AMPK activation, metformin modulates the UPR by activating the PERK/ATF4/CHOP axis and inhibiting the ATF6/GRP78 axis. Furthermore, metformin inhibits the Wnt/β-catenin signaling pathway by reducing β-catenin expression.</p>
Full article ">
15 pages, 1809 KiB  
Article
Zinc Modulates Several Transcription-Factor Regulated Pathways in Mouse Skeletal Muscle Cells
by Parisa Vahidi Ferdowsi, Rachel Ng, John Adulcikas, Sukhwinder Singh Sohal and Stephen Myers
Molecules 2020, 25(21), 5098; https://doi.org/10.3390/molecules25215098 - 3 Nov 2020
Cited by 14 | Viewed by 2912
Abstract
Zinc is an essential metal ion involved in many biological processes. Studies have shown that zinc can activate several molecules in the insulin signalling pathway and the concomitant uptake of glucose in skeletal muscle cells. However, there is limited information on other potential [...] Read more.
Zinc is an essential metal ion involved in many biological processes. Studies have shown that zinc can activate several molecules in the insulin signalling pathway and the concomitant uptake of glucose in skeletal muscle cells. However, there is limited information on other potential pathways that zinc can activate in skeletal muscle. Accordingly, this study aimed to identify other zinc-activating pathways in skeletal muscle cells to further delineate the role of this metal ion in cellular processes. Mouse C2C12 skeletal muscle cells were treated with insulin (10 nM), zinc (20 µM), and the zinc chelator TPEN (various concentrations) over 60 min. Western blots were performed for the zinc-activation of pAkt, pErk, and pCreb. A Cignal 45-Reporter Array that targets 45 signalling pathways was utilised to test the ability of zinc to activate pathways that have not yet been described. Zinc and insulin activated pAkt over 60 min as expected. Moreover, the treatment of C2C12 skeletal muscle cells with TPEN reduced the ability of zinc to activate pAkt and pErk. Zinc also activated several associated novel transcription factor pathways including Nrf1/Nrf2, ATF6, CREB, EGR1, STAT1, AP-1, PPAR, and TCF/LEF, and pCREB protein over 120 min of zinc treatment. These studies have shown that zinc’s activity extends beyond that of insulin signalling and plays a role in modulating novel transcription factor activated pathways. Further studies to determine the exact role of zinc in the activation of transcription factor pathways will provide novel insights into this metal ion actions. Full article
Show Figures

Figure 1

Figure 1
<p>MTT assay of C2C12 cells following treatment with 10 nM of insulin, 20 µM of zinc, 10 µM of NaPy, and 100 µM of TPEN for 0, 15, 30, 60, and 120 min.</p>
Full article ">Figure 2
<p>Analysis of pAkt in C2C12 mouse skeletal muscle cells treated with control (DMSO), zinc, NaPy, and insulin over 60 min. Time is shown from 0, 15, 30, and 60 min and total Akt was used as an internal loading control. The levels of pAkt were normalised to total Akt.</p>
Full article ">Figure 3
<p>Western blot result of pAkt and pErk in C2C12 cells treated with increasing concentrations of TPEN. (<b>a</b>,<b>c</b>). Cells were treated with increasing concentrations of TPEN (0, 10, 20, 40, 100 µM) for 60 min followed by 30 min of 20 µM zinc and 10 µM NaPy treatment before harvesting total protein. Phosphorylated Akt and Erk were immunoprobed by Western blotting. Total Akt and total Erk were used as internal controls. Three independent Western blots on three independent treatments were performed. (<b>b</b>,<b>d</b>). Densitometry graphs for pAkt and pErk, respectively, from three independent data Western blots ** = <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 4
<p>Western blot result of pAkt and pErk in C2C12 cells treated with increasing concentrations of TPEN. (<b>a</b>,<b>c</b>). Cells were treated with increasing concentrations of TPEN (0, 10, 20, 40, 100 µM) for 60 min followed by 30 min of 10 nM insulin treatment before harvesting total protein. Phosphorylated Akt and Erk were immunoprobed by Western blotting. Total Akt and Total Erk were used as internal controls. Three independent Western blots on three independent treatments were performed. (<b>b</b>,<b>d</b>). Densitometry graphs for pAkt and pErk, respectively, from three independent Western blots * = <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 5
<p>Western blot result of pAkt and pErk expression in C2C12 cells treated with increasing concentrations of TPEN. (<b>a</b>,<b>c</b>). Cells were treated with increasing concentrations of TPEN (0, 10, 20, 40, 100 µM) for 60 min followed by 30 min of 20 µM zinc,10 µM NaPy and 10nM insulin treatment before harvesting total protein. Phosphorylated Akt and Erk were immunoprobed by Western blotting. Total Akt and Total Erk were used as internal controls. Three independent Western blots on three independent treatments were performed. (<b>b</b>,<b>d</b>). Densitometry graphs for pAkt and pErk, respectively, from three independent data Western blots * = <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 6
<p>Western blot and densitometry of pCREB expression in C2C12 cells treated with zinc and insulin over 120 min. (<b>a</b>). Western blot of pCREB and total CREB treated with 20 µM zinc or 10 nM of insulin over 15, 30, 60, and 120 min. (<b>b</b>). Densitometry plot of pCreb/Creb from the Western blot data (<b>a</b>) from three independent experiments, ** = <span class="html-italic">p</span> &lt; 0.01. Control = no treatment. Zn + NaPy = zinc plus sodium pyrithione.</p>
Full article ">
20 pages, 3303 KiB  
Communication
Metformin Reduces NGF-Induced Tumour Promoter Effects in Epithelial Ovarian Cancer Cells
by Maritza P. Garrido, Renato Salvatierra, Manuel Valenzuela-Valderrama, Christopher Vallejos, Nicole Bruneau, Andrea Hernández, Margarita Vega, Alberto Selman, Andrew F. G. Quest and Carmen Romero
Pharmaceuticals 2020, 13(10), 315; https://doi.org/10.3390/ph13100315 - 16 Oct 2020
Cited by 12 | Viewed by 3871
Abstract
Epithelial ovarian cancer (EOC) is a lethal gynaecological neoplasm characterized by rapid growth and angiogenesis. Nerve growth factor (NGF) and its high affinity receptor tropomyosin receptor kinase A (TRKA) contribute to EOC progression by increasing the expression of c-MYC, survivin and vascular endothelial [...] Read more.
Epithelial ovarian cancer (EOC) is a lethal gynaecological neoplasm characterized by rapid growth and angiogenesis. Nerve growth factor (NGF) and its high affinity receptor tropomyosin receptor kinase A (TRKA) contribute to EOC progression by increasing the expression of c-MYC, survivin and vascular endothelial growth factor (VEGF) along with a decrease in microRNAs (miR) 23b and 145. We previously reported that metformin prevents NGF-induced proliferation and angiogenic potential of EOC cells. In this study, we sought to obtain a better understanding of the mechanism(s) by which metformin blocks these NGF-induced effects in EOC cells. Human ovarian surface epithelial (HOSE) and EOC (A2780/SKOV3) cells were stimulated with NGF and/or metformin to assess the expression of c-MYC, β-catenin, survivin and VEGF and the abundance of the tumor suppressor miRs 23b and 145. Metformin decreased the NGF-induced transcriptional activity of MYC and β-catenin/T-cell factor/lymphoid enhancer-binding factor (TCF-Lef), as well as the expression of c-MYC, survivin and VEGF in EOC cells, while it increased miR-23b and miR-145 levels. The preliminary analysis of ovarian biopsies from women users or non-users of metformin was consistent with these in vitro results. Our observations shed light on the mechanisms by which metformin may suppress tumour growth in EOC and suggest that metformin should be considered as a possible complementary therapy in EOC treatment. Full article
(This article belongs to the Special Issue Metformin: Mechanism and Application 2022)
Show Figures

Figure 1

Figure 1
<p>Metformin blocks the nerve growth factor (NGF)-mediated effects on c-MYC in ovarian cells. Ovarian cells were treated with metformin 10 mM for 48 h and/or NGF 100 ng/mL or 150 ng/mL (A2780/human ovarian surface epithelial HOSE cells and SKOV3 cells, respectively) for 24 h or the last 2 h. (<b>A</b>) Representative Images of c-MYC immunodetection in HOSE cells with semi-quantification analysis. Bar = 100 µm. Lower right inserts: 400× magnification. Upper right insert: negative control (cells without primary antibody). <span class="html-italic">N</span> = 4 independent experiments (8 images were evaluated per experiment). (<b>B</b>,<b>C</b>) Western blots of c-MYC in A2780 and SKOV3 cells. (<b>D</b>,<b>E</b>) Gen-reporter assays to evaluate MYC transcriptional activity in the epithelial ovarian cancer (EOC) cells A2780 and SKOV3. <span class="html-italic">N</span> = 4 independent experiments. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001. Statistical analysis: Kruskal–Wallis test and Dunn’s post-test. B: basal condition (without stimuli), N: NGF, M: metformin treatment. Results are expressed as the mean ± standard error of the mean (SEM).</p>
Full article ">Figure 2
<p>Metformin decreases the NGF-induced β-catenin/TCF-Lef transcriptional activity in EOC cells. Ovarian cells were treated with metformin 10 mM for 48 h and/or NGF 100 ng/mL or 150 ng/mL (A2780/HOSE cells and SKOV3 cells, respectively) for 24 h or the last 2 h. (<b>A</b>–<b>C</b>) Western blots of β-catenin in HOSE, A2780 and SKOV3 cells after the respective treatments. (<b>D</b>,<b>E</b>) Gene reporter assays to evaluate β-catenin/TCF-Lef transcriptional activity in the EOC cells A2780 and SKOV3. <span class="html-italic">N</span> = 4 independent experiments. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01. Statistical analysis: Kruskal–Wallis test and Dunn’s post-test. <sup>†</sup> <span class="html-italic">p</span> &lt; 0.05 as indicated according Mann–Whitney test. B: basal condition (without stimuli), N: NGF, M: metformin treatment. Results are expressed as the mean ± standard error of the mean (SEM).</p>
Full article ">Figure 3
<p>Metformin decreases the NGF-dependent increase in survivin expression in ovarian cells. Ovarian cells were treated with metformin 10 mM for 48 h and/or NGF 100 ng/mL or 150 ng/mL (A2780/HOSE cells and SKOV3 cells, respectively) for 24 h or the last 2 h. Then, survivin mRNA was detected in the ovarian cell lines HOSE, A2780 and SKOV3 by RT-PCR and protein levels were determined by Western blotting. (<b>A</b>–<b>C</b>) mRNA levels of survivin in the ovarian cell lines following metformin and NGF treatment. (<b>D</b>–<b>F</b>) Western blots of survivin in ovarian cell lines. <span class="html-italic">N</span> = 4 experiments per condition. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01 as indicated or with respect to basal condition. Statistical analysis: Kruskal–Wallis and Dunn´s post-test. <sup>†</sup> <span class="html-italic">p</span> &lt; 0.05 with respect to basal condition or where indicated using the Mann–Whitney test. B: basal condition (without stimuli), N: NGF, M: metformin treatment. Results are expressed as the mean ± standard error of the mean (SEM).</p>
Full article ">Figure 4
<p>Metformin prevents the NGF induced VEGF expression and angiogenic properties of ovarian cells. Ovarian cells were treated with metformin 10 mM for 48 h and/or NGF 100 ng/mL or 150 ng/mL (A2780/HOSE cells and SKOV3 cells, respectively) for 24 h or the last 2 h, and then processed to detect VEGF expression. (<b>A</b>,<b>C</b>,<b>E</b>) Representative agarose gels with PCR products of different VEGF transcripts (VEGF 121, VEGF 165 and VEGF 189 amino acids) in ovarian cells. Semi-quantification of <span class="html-italic">N</span> = 4 or more independent experiments. (<b>B</b>,<b>D</b>,<b>F</b>) VEGF detection in the culture supernatants of ovarian cells by enzyme-linked immunosorbent assay (ELISA). <span class="html-italic">N</span> = 4 or more independent experiments induplicate. (<b>G</b>,<b>H</b>) Angiogenic score calculated as indicated in the methodology section with conditioned medium from A2780 or SKOV3 cells, respectively. Bar = 50 µm. <span class="html-italic">N</span> = 4 or more independent experiments (4–8 images were evaluated per experiment). * <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 as indicated or with respect to basal conditions. Statistical analysis: Kruskal–Wallis and Dunn´s post-test. <sup>†</sup> <span class="html-italic">p</span> &lt; 0.05 and <sup>††</sup> <span class="html-italic">p</span> &lt; 0.01 with respect to basal conditions or as indicated using the Mann–Whitney test. B: basal condition (without stimuli), N: NGF, M: metformin treatment. Results are expressed as the mean ± standard error of the mean (SEM).</p>
Full article ">Figure 5
<p>Metformin increases the expression of miR-145 and miR-23b in ovarian cell lines. Ovarian cells were treated with metformin (10 mM for 48 h) and NGF (100 or 150 ng/mL, last 3 h) to detect miR expression. miR-23b and miR-145 were quantified in HOSE, A2780 or SKOV3 cells by qPCR. <span class="html-italic">N</span> = 4 or more independent experiments in duplicate. * <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 as indicated or with respect to basal conditions. Statistical analysis: Kruskal–Wallis and Dunn´s post-test. <sup>†</sup> <span class="html-italic">p</span> &lt; 0.05 and <sup>††</sup> <span class="html-italic">p</span> &lt; 0.01 with respect to basal conditions or where indicated using the Mann–Whitney test. B: basal condition (without stimuli), N: NGF, M: metformin treatment. Results are expressed as the mean ± standard error of the mean (SEM).</p>
Full article ">Figure 6
<p>Metformin intake decreases the presence of β-catenin, c-MYC and survivin in epithelial ovarian tumours. Sequential paraffin-embedded samples from female users (M+) and non-users (M−) of metformin were used to detect oncoproteins by immunohistochemistry: β-catenin (<b>A</b>) c-MYC (<b>B</b>), or survivin (<b>C</b>) levels. <span class="html-italic">N</span> = 4 independent experiments (6–8 images were evaluated per experiment). Bar = 40 µm. * <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 according to the Mann–Whitney test. Results are expressed as the mean ± standard error of the mean (SEM).</p>
Full article ">Figure 7
<p>Proposed mechanisms by which metformin prevents NGF–mediated effects in EOC cells. (<b>A</b>) Metformin blocks the NGF-induced c-MYC, survivin and VEGF protein levels, decreasing the proliferative and angiogenic potential of EOC cells. NGF and VEGF are depicted as acting directly on endothelial cells, while c-MYC and survivin changes contribute to EOC cell proliferation and VEGF expression. (<b>B</b>) Metformin increases the expression of oncosuppressor miR-145 and miR-23b, which may be responsible for the observed decreases in c-MYC, survivin and VEGF levels in EOC cells. Filled arrows mean stimulation, thick perpendicular bars mean inhibition and dotted lines mean indirect association.</p>
Full article ">
16 pages, 2622 KiB  
Article
TRAP1 Regulates Wnt/β-Catenin Pathway through LRP5/6 Receptors Expression Modulation
by Giacomo Lettini, Valentina Condelli, Michele Pietrafesa, Fabiana Crispo, Pietro Zoppoli, Francesca Maddalena, Ilaria Laurenzana, Alessandro Sgambato, Franca Esposito and Matteo Landriscina
Int. J. Mol. Sci. 2020, 21(20), 7526; https://doi.org/10.3390/ijms21207526 - 13 Oct 2020
Cited by 9 | Viewed by 3073
Abstract
Wnt/β-Catenin signaling is involved in embryonic development, regeneration, and cellular differentiation and is responsible for cancer stemness maintenance. The HSP90 molecular chaperone TRAP1 is upregulated in 60–70% of human colorectal carcinomas (CRCs) and favors stem cells maintenance, modulating the Wnt/β-Catenin pathway and preventing [...] Read more.
Wnt/β-Catenin signaling is involved in embryonic development, regeneration, and cellular differentiation and is responsible for cancer stemness maintenance. The HSP90 molecular chaperone TRAP1 is upregulated in 60–70% of human colorectal carcinomas (CRCs) and favors stem cells maintenance, modulating the Wnt/β-Catenin pathway and preventing β-Catenin phosphorylation/degradation. The role of TRAP1 in the regulation of Wnt/β-Catenin signaling was further investigated in human CRC cell lines, patient-derived spheroids, and CRC specimens. TRAP1 relevance in the activation of Wnt/β-Catenin signaling was highlighted by a TCF/LEF Cignal Reporter Assay in Wnt-off HEK293T and CRC HCT116 cell lines. Of note, this regulation occurs through the modulation of Wnt ligand receptors LRP5 and LRP6 that are both downregulated in TRAP1-silenced cell lines. However, while LRP5 mRNA is significantly downregulated upon TRAP1 silencing, LRP6 mRNA is unchanged, suggesting independent mechanisms of regulation by TRAP1. Indeed, LRP5 is regulated upon promoter methylation in CRC cell lines and human CRCs, whereas LRP6 is controlled at post-translational level by protein ubiquitination/degradation. Consistently, human CRCs with high TRAP1 expression are characterized by the co-upregulation of active β-Catenin, LRP5 and LRP6. Altogether, these data suggest that Wnt/β-Catenin signaling is modulated at multiple levels by TRAP1. Full article
(This article belongs to the Special Issue The Wnt Signaling Pathway in Cancer)
Show Figures

Figure 1

Figure 1
<p>TRAP1 modulates the activity of Wnt/β-Catenin pathway. (<b>A</b>,<b>C</b>) The activity of Wnt/β-Catenin signaling was evaluated in control (siNeg) and TRAP1-silenced (siTRAP1) HEK293T (A) and HCT116 (C) cells, upon transfection with a Cignal TCF/LEF Reporter and subsequent stimulation with 150 ng/µL Wnt3A recombinant protein for 6 h. Statistical significance respect to the respective control: * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; n/s, not significant. Data represent means + S.D. MFI, Mean Fluorescence Intensity. (<b>B</b>,<b>D</b>) Total lysates from control and TRAP1-silenced HEK293T (B) and HCT116 (D) cells, treated as reported in panels (<b>A</b>,<b>C</b>), were separated by SDS-PAGE and immunoblotted with indicated antibodies.</p>
Full article ">Figure 2
<p>TRAP1 enhances the sphere-forming ability of colon carcinoma and cancer stem cells. (<b>A</b>,<b>B</b>) Representative images (<b>A</b>) and quantitative representation (<b>B</b>) of the sphere-forming ability in control (siNeg) and TRAP1-silenced (siTRAP1) HCT116, SA41 and SA54 cells. Statistical significance respect to the respective siNeg control: * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01. Data represent means + S.D. (<b>A</b>) Insert: TRAP1 immunoblot analysis in siNeg and siTRAP1 HCT116, SA41 and SA54 cells. (<b>C</b>) Relative CD44 MFI levels in control (siNeg) and TRAP1-silenced (siTRAP1) SA41 and SA54 cells evaluated by CD44 immunostaining and subsequent flow cytometry. Statistical significance respect to the respective siNeg control: * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; n/s. Data represent means + S.D. MFI, Mean Fluorescence Intensity.</p>
Full article ">Figure 3
<p>TRAP1 silencing results in the downregulation of LRP5/6 receptors and Wnt/β-Catenin-related proteins. (<b>A</b>,<b>B</b>) Total cell lysates collected from control (siNeg) and TRAP1-silenced (siTRAP1) HCT116 and HEK293T cells (<b>A</b>), and SA54, SA41, and SA21 patient-derived spheroids (<b>B</b>) were separated by SDS-PAGE and immunoblotted with the indicated antibodies. (<b>C</b>) Real-time PCR gene expression analysis of LRP5 and LRP6 in TRAP1-silenced HCT116 and SA54 cells compared to cells transfected with negative siRNA. Statistical significance respect to siNeg control: * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; n/s, not significant. Data represent means + S.D.</p>
Full article ">Figure 4
<p>TRAP1 regulates LRP6 expression through a quality control by the regulation of its ubiquitination/degradation. (<b>A</b>) Total lysates from HCT116, SA41, SA54, and SA21 cells, transfected with control (siNeg) or TRAP1/TBP7 siRNAs, were separated by SDS-PAGE and immunoblotted with indicated antibodies. (<b>B</b>) Anti-TRAP1 immunoprecipitates were obtained from HCT116 cells, separated by SDS-PAGE and immunoblotted with anti-LRP6 and anti-TRAP1 antibodies. IgG Neg, total cellular extracts incubated with nonspecific antibody. Input: Total cell lysates from HCT116 cells were separated by SDS-PAGE and immunoblotted with the indicated antibodies. (<b>C</b>) Anti-LRP6 (upper panel) and anti-ubiquitin (lower panel) immunoprecipitates were obtained from HCT116 cells transfected with control (siNeg) or TRAP1 siRNAs, separated by SDS-PAGE and immunoblotted with anti-ubiquitin (upper panel) and anti-LRP6 (lower panel) antibodies. Cells were incubated with 10 mmol/L MG132 for 2 h before cell lysis. Densitometric band intensities represent the ratios between ubiquitinated and total LRP6 bands in IPs. IgG Neg, total cellular extracts incubated with nonspecific antibody. Input: Total cell lysates from HCT116 cells transfected with control or TRAP1 siRNA were separated by SDS-PAGE and immunoblotted with the indicated antibodies.</p>
Full article ">Figure 5
<p>TRAP1 regulates LRP5 expression through DNA methylation mechanisms. (<b>A</b>) Scatter plot of 361 TCGA colorectal adenocarcinomas. Promoter methylation is reported on the <span class="html-italic">x</span>-axis, mRNA expression on the <span class="html-italic">y</span>-axis. Solid line shows the linear regression. Rho is the Spearman’s rank correlation and the <span class="html-italic">p</span>-value is the result of the correlation test. (<b>B</b>) Methylation-specific PCR representing the level of LRP5 promoter methylation in control (siNeg) TRAP1-silenced (siTRAP1) HCT116 and SA54 cells. PCR analysis of a LRP5 un-methylated region is reported as internal control.</p>
Full article ">Figure 6
<p>TRAP1 is co-expressed with active β-Catenin and LRP5/6 in human CRCs. (<b>A</b>) Total cell lysates from eight representative human CRCs (T) and the respective peritumoral non-infiltrated mucosas (M) were separated by SDS-PAGE and immunoblotted with the indicated antibodies. (<b>B</b>,<b>C</b>) Dot plots representing the correlation between active β-Catenin and LRP5/LRP6 (<b>B</b>) and between TRAP1 and active β-Catenin/LRP5/LRP6 (<b>C</b>) in our series of 41 colorectal carcinomas. (<b>D</b>) Real Time PCR analysis of LRP5 mRNA expression in two representative cases of human CRCs, represented as time increase compared to the respective non-infiltrated normal mucosa. Statistical significance respect to the respective control: ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001. (<b>E</b>) Methylation specific PCR representing the level of LRP5 promoter methylation in two representative cases of human CRCs.</p>
Full article ">
Back to TopTop