[go: up one dir, main page]

 
 

Wnt Signaling in Development, Regeneration and Cancer

A special issue of Genes (ISSN 2073-4425). This special issue belongs to the section "Human Genomics and Genetic Diseases".

Deadline for manuscript submissions: closed (30 June 2020) | Viewed by 42827

Special Issue Editors


E-Mail Website
Guest Editor
Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA
Interests: Wnt Signaling; intestinal epithelium; tissue homeostasis; kinase signaling; chemical biology; GI cancer

E-Mail Website
Guest Editor
Department of Cell Biology & Physiology, Department of Otolaryngology, Washington University in St. Louis,St. Louis, MO 63130, USA
Interests: Wnt signaling; nrf2 signaling; cancer; pProteomics; ubiquitin ligases; functional genomics

Special Issue Information

Dear Colleagues,

We write to invite you to contribute to a Special Issue in Genes that will be focused on Wnt signaling in development, regeneration, and cancer. As an evolutionarily conserved signaling network of embryonic patterning, Wnt signaling plays a major role in the maintenance, renewal, and differentiation of a number of stem cells and progenitor cell lineages in the adult multicellular organism. Thus, modulating this pathway has enormous potential in regenerative medicine and in the treatment of major human diseases, such as cancer. Major questions that are currently being investigated include the role of Wnt signaling in regulating stem cell self-renewal, proliferation, and differentiation as well as the characterization of cells in the niche that produce and receive the Wnt signal. In addition, the detailed mechanisms and dynamics by which the Wnt pathway is controlled and can crosstalk with other pathways are poorly defined. Moreover, the involvement of Wnt signaling in drug sensitivity or resistance remains unclear.

This Special Issue will highlight reviews, new methods, and original articles that advance our understanding of the role of the Wnt pathway in development, regeneration, and disease. We welcome contributions in the areas of Wnt signaling broadly defined. Topics of interest include but are not limited to the role of Wnt signaling in maintenance, proliferation, and differentiation of stem cells of various organ lineages; the role of Wnt signaling in embryonic stem cells and cancer stem cells; and the role of Wnt signaling in controlling and regulating stem cell niches. We also welcome studies that highlight new technologies, new modes of regulation, and/or novel components of the Wnt signaling pathway that reveal underlying biochemical mechanisms that may ultimately control cell behavior. We encourage studies that explore the effect of Wnt signaling on therapeutic response. Finally, we welcome studies that make use of traditional as well as nontraditional model organisms that may provide insight into the evolutionarily conserved role of Wnt signaling across phyla. We look forward to your contributions.

Prof. Curtis Thorne
Prof. Michael Ben Major
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Genes is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2600 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • Wnt Signaling
  • Stem Cells
  • Regenerative medicine
  • Drug resistance
  • Cancer
  • Development

Benefits of Publishing in a Special Issue

  • Ease of navigation: Grouping papers by topic helps scholars navigate broad scope journals more efficiently.
  • Greater discoverability: Special Issues support the reach and impact of scientific research. Articles in Special Issues are more discoverable and cited more frequently.
  • Expansion of research network: Special Issues facilitate connections among authors, fostering scientific collaborations.
  • External promotion: Articles in Special Issues are often promoted through the journal's social media, increasing their visibility.
  • e-Book format: Special Issues with more than 10 articles can be published as dedicated e-books, ensuring wide and rapid dissemination.

Further information on MDPI's Special Issue polices can be found here.

Published Papers (8 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

24 pages, 1451 KiB  
Article
Diverse LEF/TCF Expression in Human Colorectal Cancer Correlates with Altered Wnt-Regulated Transcriptome in a Meta-Analysis of Patient Biopsies
by Claus-Dieter Mayer, Soizick Magon de La Giclais, Fozan Alsehly and Stefan Hoppler
Genes 2020, 11(5), 538; https://doi.org/10.3390/genes11050538 - 11 May 2020
Cited by 13 | Viewed by 3818
Abstract
Aberrantly activated Wnt signaling causes cellular transformation that can lead to human colorectal cancer. Wnt signaling is mediated by Lymphoid Enhancer Factor/T-Cell Factor (LEF/TCF) DNA-binding factors. Here we investigate whether altered LEF/TCF expression is conserved in human colorectal tumor sample and [...] Read more.
Aberrantly activated Wnt signaling causes cellular transformation that can lead to human colorectal cancer. Wnt signaling is mediated by Lymphoid Enhancer Factor/T-Cell Factor (LEF/TCF) DNA-binding factors. Here we investigate whether altered LEF/TCF expression is conserved in human colorectal tumor sample and may potentially be correlated with indicators of cancer progression. We carried out a meta-analysis of carefully selected publicly available gene expression data sets with paired tumor biopsy and adjacent matched normal tissues from colorectal cancer patients. Our meta-analysis confirms that among the four human LEF/TCF genes, LEF1 and TCF7 are preferentially expressed in tumor biopsies, while TCF7L2 and TCF7L1 in normal control tissue. We also confirm positive correlation of LEF1 and TCF7 expression with hallmarks of active Wnt signaling (i.e., AXIN2 and LGR5). We are able to correlate differential LEF/TCF gene expression with distinct transcriptomes associated with cell adhesion, extracellular matrix organization, and Wnt receptor feedback regulation. We demonstrate here in human colorectal tumor sample correlation of altered LEF/TCF gene expression with quantitatively and qualitatively different transcriptomes, suggesting LEF/TCF-specific transcriptional regulation of Wnt target genes relevant for cancer progression and survival. This bioinformatics analysis provides a foundation for future more detailed, functional, and molecular analyses aimed at dissecting such functional differences. Full article
(This article belongs to the Special Issue Wnt Signaling in Development, Regeneration and Cancer)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Forest plots of gene expression of all four LEF/TCF genes (<b>A</b>): <span class="html-italic">TCF7</span>, (<b>B</b>): <span class="html-italic">LEF1</span>, (<b>C</b>): <span class="html-italic">TCF7L1</span>, (<b>D</b>): <span class="html-italic">TCF7L2</span>, and (<b>E</b>): <span class="html-italic">AXIN2</span>, (<b>F</b>): <span class="html-italic">DICKKOPF-1</span> (<span class="html-italic">DKK1</span>), (<b>G</b>): <span class="html-italic">FZD7</span>, and (<b>H</b>): <span class="html-italic">LGR5</span>, from six selected studies. Columns from left to right indicate: accession no. of study (with GSE20842 [<a href="#B27-genes-11-00538" class="html-bibr">27</a>] separated between kras-positive and kras-mutant samples); number of patients in individual studies; horizontal segments indicate the standardized mean difference between tumor and normal, and their confidence interval, with the size of the square dot being proportional with the weight of the study in the meta-analysis using a ‘random effects’ model. The corresponding values are written in the column on the right: weight of the individual study in percent as part of the meta-analysis; standardized mean difference; and in square brackets confidence interval. The red polygon in the bottom of each plot shows the summary estimate based on the random-effect model. Values to the left of the midline indicated higher expression in the control relative to the tumor sample, e.g., see <span class="html-italic">AXIN2</span> and <span class="html-italic">LGR5</span>. Individual studies with small sample size (i.e., few patients) as expected often have larger confidence intervals (therefore less reliability, e.g., see <span class="html-italic">TCF7</span> and <span class="html-italic">LEF1</span> data for GSE46622 study), but in the meta-analysis (in red) much tighter confidence intervals (therefore higher reliability). Note that, among the four LEF/TCF genes, <span class="html-italic">TCF7</span>, <span class="html-italic">LEF1</span>, are expressed higher, while <span class="html-italic">TCF7L1</span>, <span class="html-italic">TCF7L2</span> lower in tumor tissue.</p>
Full article ">Figure 2
<p>Correlation plot matrix of relative gene expression between eight selected genes in six selected studies (<b>A</b>–<b>N</b>), with normal control (<b>A</b>, <b>C</b>, <b>E</b>, <b>G</b>, <b>I</b>, <b>K</b>, <b>M</b>) separated from tumor sample (<b>B</b>, <b>D</b>, <b>F</b>, <b>H</b>, <b>J</b>, <b>L</b>, <b>N</b>), and additionally for the GSE20842 [<a href="#B27-genes-11-00538" class="html-bibr">27</a>] between kras-mutant (“mut”) samples (<b>C</b>, <b>D</b>) and kras-positive (“wild” as in wildtype) samples (<b>E</b>, <b>F</b>). Blue dots indicate positive and red dots negative correlation. The size of the circle and the intensity of the color is proportional to the correlation coefficient; therefore, as an internal control, expected diagonal series of large blue dots where expression of genes is compared to the expression of the same gene). Missing values in GSE46622 [<a href="#B30-genes-11-00538" class="html-bibr">30</a>] is due to low value data for <span class="html-italic">LEF1</span> in this study. Note positive correlation between <span class="html-italic">AXIN2</span> expression and <span class="html-italic">LGR5</span>, <span class="html-italic">TCF7</span> and <span class="html-italic">LEF1</span> expression, yet negative correlation with <span class="html-italic">TCF7L1</span> expression, while <span class="html-italic">TCF7L1</span> and <span class="html-italic">FZD7</span> expression are positively correlated, though clearly much more so in normal control tissue than in tumor. In contrast, the correlation between <span class="html-italic">AXIN2</span> and <span class="html-italic">TCF7</span> expression is clearly more robust in tumor compared to normal tissue. Interestingly, the unearthed correlation between <span class="html-italic">TCF7L1</span> and <span class="html-italic">FZD7</span> expression appears to be dependent on wild-type <span class="html-italic">kRAS</span> in the tumor (compare <b>D</b> with <b>F</b>, yet not in normal control <b>C</b>).</p>
Full article ">Figure 3
<p>Correlation of transcript expression between eight selected genes (<span class="html-italic">TCF7</span>, <span class="html-italic">LEF1</span>, <span class="html-italic">TCF7L1</span>, <span class="html-italic">TCF7L2</span>, <span class="html-italic">AXIN2</span>, <span class="html-italic">DKK1</span>, <span class="html-italic">FZD7</span> and <span class="html-italic">LGR5</span>) in normal control tissue (<b>A</b>), in tumor tissue (<b>B</b>), and when analyzed combined in normal and tumor tissue (<b>C</b>) (negative numbers and graded red highlighting indicates negative correlation; positive numbers and graded green highlighting positive correlation). (<b>D</b>) Mean difference between normal and tumor tissue of correlation of transcript expression between eight selected genes (red highlighting with positive numbers indicates reduced negative correlation in tumor tissue; green highlighting with negative numbers indicates reduced, with positive numbers increased, positive correlation in tumor tissue; yellow highlighting with positive numbers indicates a switch from negative to positive, and with negative numbers to negative, correlation in tumor tissue. Note generally reduced correlations in tumor tissue; particularly note, reduced positive correlation between <span class="html-italic">TCF7</span> and <span class="html-italic">LEF1</span>, between <span class="html-italic">TCF7</span> and <span class="html-italic">LGR5</span>, and between <span class="html-italic">TCF7L1</span> and <span class="html-italic">FZD7</span> expression; and reduced negative correlation between <span class="html-italic">AXIN2</span> and <span class="html-italic">TCF7L1</span> expression. However, also note exceptional increased positive correlation between <span class="html-italic">AXIN2</span> and <span class="html-italic">TCF7</span> in tumor tissue.</p>
Full article ">
15 pages, 2602 KiB  
Communication
Vulpinic Acid Controls Stem Cell Fate toward Osteogenesis and Adipogenesis
by Sang Ah Yi, Ki Hong Nam, Sil Kim, Hae Min So, Rhim Ryoo, Jeung-Whan Han, Ki Hyun Kim and Jaecheol Lee
Genes 2020, 11(1), 18; https://doi.org/10.3390/genes11010018 - 23 Dec 2019
Cited by 7 | Viewed by 3460
Abstract
Vulpinic acid, a naturally occurring methyl ester of pulvinic acid, has been reported to exert anti-fungal, anti-cancer, and anti-oxidative effects. However, its metabolic action has not been implicated yet. Here, we show that vulpinic acid derived from a mushroom, Pulveroboletus ravenelii controls the [...] Read more.
Vulpinic acid, a naturally occurring methyl ester of pulvinic acid, has been reported to exert anti-fungal, anti-cancer, and anti-oxidative effects. However, its metabolic action has not been implicated yet. Here, we show that vulpinic acid derived from a mushroom, Pulveroboletus ravenelii controls the cell fate of mesenchymal stem cells and preadipocytes by inducing the acetylation of histone H3 and α-tubulin, respectively. The treatment of 10T1/2 mesenchymal stem cells with vulpinic acid increased the expression of Wnt6, Wnt10a, and Wnt10b, which led to osteogenesis inhibiting the adipogenic lineage commitment, through the upregulation of H3 acetylation. By contrast, treatment with vulpinic acid promoted the terminal differentiation of 3T3-L1 preadipocytes into mature adipocytes. In this process, the increase in acetylated tubulin was accompanied, while acetylated H3 was not altered. As excessive generation of adipocytes occurs, the accumulation of lipid drops was not concentrated, but dispersed into a number of adipocytes. Consistently, the expressions of lipolytic genes were upregulated and inflammatory factors were downregulated in adipocytes exposed to vulpinic acid during adipogenesis. These findings reveal the multiple actions of vulpinic acid in two stages of differentiation, promoting the osteogenesis of mesenchymal stem cells and decreasing hypertrophic adipocytes, which can provide experimental evidence for the novel metabolic advantages of vulpinic acid. Full article
(This article belongs to the Special Issue Wnt Signaling in Development, Regeneration and Cancer)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Vulpinic acid (VA) modulates acetylation of histone H3 and α-tubulin in 10T1/2 and 3T3-L1 cells. (<b>A</b>) Microscopic images (100X) of 10T1/2 and 3T3-L1 cells treated with vulpinic acid for 24 h at the indicated concentrations. (<b>B</b>) Immunoblot analysis of 10T1/2 cells treated with vulpinic acid for 24 h at the indicated concentrations. (<b>C</b>) Immunoblot analysis of 3T3-L1 cells treated with vulpinic acid for 24 h at the indicated concentrations. Data represent means ± SEM (standard effort of the mean) for n = 3. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 2
<p>Vulpinic acid promotes the expression of Wnt genes via H3 acetylation in 10T1/2 MSCs. (<b>A</b>) Schematic representation of Wnt signaling-mediated cell fate determination of mesenchymal stem cells (MSCs). (<b>B</b>) The mRNA levels of Wnt6, Wnt10a, and Wnt10b genes in 10T1/2 cells treated with vulpinic acid (10 or 40 μM) for 24 h. (<b>C</b>) 10T1/2 cells were treated with or without vulpinic acid (40 μM) for 24 h. ChIP assay was performed with IgG and acetylated H3 antibodies followed by real time PCR with primers for promoter regions of Wnt6, Wnt10a, and Wnt10b genes. Data represent means ± SEM for n = 3. * <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>Vulpinic acid induces osteogenesis via H3 acetylation in 10T1/2 MSCs. (<b>A</b>) Schematic representation of BMPs-mediated osteogenic or adipogenic commitment of MSCs. (<b>B</b>) Immunoblot analysis of 10T1/2 cells treated with vulpinic acid (10 or 40 μM) for 24 h in the presence of BMP2 or BMP4. (<b>C</b>) 10T1/2 cells were treated with or without vulpinic acid (40 μM) for 24 h in the presence of BMP2 or BMP4. ChIP assay was performed with IgG and acetylated H3 antibodies followed by real time PCR with primers for promoter region of Runx2 gene. (<b>D</b>) The mRNA levels of the <span class="html-italic">Bmp2</span>, <span class="html-italic">Ocn</span>, and <span class="html-italic">Sp7</span> genes in 10T1/2 cells treated with vulpinic acid (10 or 40 μM) for 24 h. (<b>E</b>) The mRNA levels of Cebpa and PPARγ genes in 10T1/2 cells treated with vulpinic acid (10 or 40 μM) for 24 h in the presence of BMP4. Data represent means ± SEM for n = 3. * <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>Vulpinic acid promotes adipogenesis from 3T3-L1 preadipocytes. (<b>A</b>) Schematic representation of adipogenesis process. 3T3-L1 cells were incubated with an adipogenic medium in the absence or presence of vulpinic acid. (<b>B</b>) The mRNA levels of Adipsin, Fabp4, and PPARγ genes in 3T3-L1 adipocytes incubated with vulpinic acid (10 or 40 μM) during adipogenesis. (<b>C</b>) The mRNA levels of Adipoq and Leptin genes in 3T3-L1 adipocytes incubated with vulpinic acid (10 or 40 μM) during adipogenesis. (<b>D</b>) Immunoblot analysis of 3T3-L1 adipocytes incubated with vulpinic acid (10 or 40 μM) during adipogenesis. Data represent means ± SEM for n = 3. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>Vulpinic acid reduces hypertrophic adipocytes by promoting hyperplasia. (<b>A</b>) Oil-red-O staining of 3T3-L1 adipocytes incubated with vulpinic acid (10 or 40 μM) during adipogenesis. Scale bar = 200 μm (40X). (<b>B</b>) The mRNA levels of ATGL and MCAD genes in 3T3-L1 adipocytes incubated with vulpinic acid (10 or 40 μM) during adipogenesis. (<b>C</b>) The mRNA levels of IL6 and TNFα genes in 3T3-L1 adipocytes incubated with vulpinic acid (10 or 40 μM) during adipogenesis. Data represent means ± SEM for n = 3. * <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 6
<p>Molecular model underlying the mechanism of actions of vulpinic acid. During the early stage of mesenchymal stem cells, vulpinic acid promotes osteogenic commitment while preventing adipogenic commitment via H3 acetylation-mediated gene modulations. During terminal differentiation to mature adipocytes from preadipocytes, vulpinic acid enhances de novo generation of adipocytes via acetylation of α-tubulin, reducing hypertrophic adipocytes.</p>
Full article ">
13 pages, 1303 KiB  
Article
Wnt-11 Expression Promotes Invasiveness and Correlates with Survival in Human Pancreatic Ductal Adeno Carcinoma
by Dafydd A. Dart, Damla E Arisan, Sioned Owen, Chunyi Hao, Wen G. Jiang and Pinar Uysal-Onganer
Genes 2019, 10(11), 921; https://doi.org/10.3390/genes10110921 - 11 Nov 2019
Cited by 11 | Viewed by 3355
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest forms of cancer, proving difficult to manage clinically. Wnt-11, a developmentally regulated gene producing a secreted protein, has been associated with various carcinomas but has not previously been studied in PDAC. The present study [...] Read more.
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest forms of cancer, proving difficult to manage clinically. Wnt-11, a developmentally regulated gene producing a secreted protein, has been associated with various carcinomas but has not previously been studied in PDAC. The present study aimed to elucidate these aspects first in vitro and then in a clinical setting in vivo. Molecular analyses of Wnt-11 expression as well as other biomarkers involved qRT-PCR, RNA-seq and siRNA. Proliferation was measured by MTT; invasiveness was quantified by Boyden chamber (Matrigel) assay. Wnt-11 mRNA was present in three different human PDAC cell lines. Wnt-11 loss affected epithelial-mesenchymal transition and expression of neuronal and stemness biomarkers associated with metastasis. Indeed, silencing Wnt-11 in Panc-1 cells significantly inhibited their Matrigel invasiveness without affecting their proliferative activity. Consistently with the in vitro data, human biopsies of PDAC showed significantly higher Wnt-11 mRNA levels compared with matched adjacent tissues. Expression was significantly upregulated during PDAC progression (TNM stage I to II) and maintained (TNM stages III and IV). Wnt-11 is expressed in PDAC in vitro and in vivo and plays a significant role in the pathophysiology of the disease; this evidence leads to the conclusion that Wnt-11 could serve as a novel, functional biomarker PDAC. Full article
(This article belongs to the Special Issue Wnt Signaling in Development, Regeneration and Cancer)
Show Figures

Figure 1

Figure 1
<p>Wnt-11 mRNA relative expression profile in Bx-PC3, Panc-1 and MiaPaca-2 cells, and Wnt-11 siRNA on Panc-1 cells. (<b>A</b>) Real-time PCR result of Wnt-11 relative mRNA expression. Data are plotted as fold-differences relative to the Bx-PC3 cell Wnt-11 mRNA expression level after normalising with housekeeping gene (RPII) level. Panc-1 cells express the highest amount of Wnt-11 mRNA (folds more than Bx–PC3; <span class="html-italic">n</span> = 6; <span class="html-italic">p</span> &lt; 0.01) among three pancreatic cancer cell lines and Wnt-11 siRNA reduces the expression 4.52-fold (light grey bar; <span class="html-italic">n</span> = 6; <span class="html-italic">p</span> = 0.011). All data were analysed as means ± standard errors. Statistical significance was determined using Student’s <span class="html-italic">t</span>-test or ANOVA with Newman–Keuls post-hoc analysis were used, as appropriate. (<b>B</b>,<b>C</b>) Immunostaining for Wnt-11 (green) and To-Pro (blue) in Panc-1 cells transfected with Wnt-11siRNA. (<span class="html-italic">B</span>) Control. (<span class="html-italic">C</span>) Wnt-11 siRNA for 48 hours. Scale bar, 20 µm (applicable to both panels).</p>
Full article ">Figure 2
<p>Wnt-11 promotes epithelial-mesenchymal transition (EMT) and neuronal differentiation in Panc-1 cells line. (<b>A</b>) qRT-PCR for NSE, NeuroD, Hes6 and Nanog relative mRNA expressions after transfection of Panc-1 cells either with scrambled or Wnt-11 siRNA after 48 hours. Wnt-11 siRNA. (<b>B</b>) qRT-PCR for EMT markers; Ecad, Snail, Vim and Twist relative mRNA expressions after transfection of Panc-1 cells either with scrambled or Wnt-11 siRNA after 48 hours. NSE, NeuroD, Hes6 and Nanog mRNA expressions and they were reduced after silencing Wnt-11 (53%, 89%, 51.5% and 94% respectively; <span class="html-italic">n</span> = 6; <span class="html-italic">p</span> &lt; 0.05 for all). Wnt-11 siRNA increased Ecad mRNA expression 5.29-fold in Panc-1 cells (<span class="html-italic">n</span> = 6; <span class="html-italic">p</span> = 0.02). Other EMT markers, such as Snail, Vim and Twist expressed in Panc-1 and MiaPaca-2 cells and this expression was reduced by Wnt-11 siRNA in Panc-1 cells (Fig 2B; min 70% reduction for all, <span class="html-italic">n</span> = 6; <span class="html-italic">p</span> &lt; 0.05). Statistical significance was determined using Student’s <span class="html-italic">t</span>-test and ANOVA were used.</p>
Full article ">Figure 3
<p>Functional evidence for Wnt-11-induced reduction of Matrigel invasion on Panc-1 cell lines. Panc-1 cells were transfected for 72 hours with control or Wnt-11 siRNAs, plated on Matrigel coated transwell filters and the extent of invasion determined after 16 hours. (<b>A</b>) Wnt-11 siRNA decreased invasion by 23% (<span class="html-italic">n</span> = 3; <span class="html-italic">p</span> = 0.02). The results are plotted as Invasion Index (InvI, %), which is the percentage of invaded cells compared to the total number of cells seeded. (<b>B</b>) The total cell number/proliferation did not change during the course of the experiment (<span class="html-italic">n</span> = 3; <span class="html-italic">p</span> &gt; 0.05). All data were analysed as means ± standard errors.</p>
Full article ">Figure 4
<p>Clinical evidence that Wnt-11 was significantly upregulated in a cohort of pancreatic tumour samples. (<b>A</b>) In total, 111 pairs of tumour and adjacent control tissues were studied and Wnt-11 expression (relative) appeared to be increased in the tumour tissue compared to adjacent normal tissues (<span class="html-italic">p</span> = 0.02). (<b>B</b>) A high level of Wnt-11 expression (relative) was found in patients classified according to T staging (<span class="html-italic">p</span> &lt; 0.05 for T1 vs T2 or T3 or T4; T2 vs T3 or T3 vs T4 <span class="html-italic">p</span> &gt; 0.05). All data were analysed as means ± standard errors.</p>
Full article ">Figure 5
<p>A survival curve gained from Human Protein Atlas shows that a high expression of Wnt-11 reduces cumulative survival of PDAC. (<span class="html-italic">p</span> = 0.02). A low expression (<span class="html-italic">n</span> = 121)<b>.</b> High expression (<span class="html-italic">n</span> = 55). 5-year survival high 26%; 5-year survival low 31%. [<a href="#B34-genes-10-00921" class="html-bibr">34</a>]. Kaplan–Meier survival estimators examined the prognosis of each group of patients, and the survival outcomes of the two groups were compared. Bylog ranktests. <a href="https://www.proteinatlas.org/ENSG00000085741WNT11/pathology/pancreatic" target="_blank">https://www.proteinatlas.org/ENSG00000085741WNT11/pathology/pancreatic</a> + cancer # Location. Accessed on 22 October 2019.</p>
Full article ">

Review

Jump to: Research

20 pages, 2325 KiB  
Review
The Canonical Wnt Pathway as a Key Regulator in Liver Development, Differentiation and Homeostatic Renewal
by Sebastian L. Wild, Aya Elghajiji, Carmen Grimaldos Rodriguez, Stephen D. Weston, Zoë D. Burke and David Tosh
Genes 2020, 11(10), 1163; https://doi.org/10.3390/genes11101163 - 30 Sep 2020
Cited by 17 | Viewed by 8301
Abstract
The canonical Wnt (Wnt/β-catenin) signalling pathway is highly conserved and plays a critical role in regulating cellular processes both during development and in adult tissue homeostasis. The Wnt/β-catenin signalling pathway is vital for correct body patterning and is involved in fate specification of [...] Read more.
The canonical Wnt (Wnt/β-catenin) signalling pathway is highly conserved and plays a critical role in regulating cellular processes both during development and in adult tissue homeostasis. The Wnt/β-catenin signalling pathway is vital for correct body patterning and is involved in fate specification of the gut tube, the primitive precursor of liver. In adults, the Wnt/β-catenin pathway is increasingly recognised as an important regulator of metabolic zonation, homeostatic renewal and regeneration in response to injury throughout the liver. Herein, we review recent developments relating to the key role of the pathway in the patterning and fate specification of the liver, in the directed differentiation of pluripotent stem cells into hepatocytes and in governing proliferation and zonation in the adult liver. We pay particular attention to recent contributions to the controversy surrounding homeostatic renewal and proliferation in response to injury. Furthermore, we discuss how crosstalk between the Wnt/β-catenin and Hedgehog (Hh) and hypoxia inducible factor (HIF) pathways works to maintain liver homeostasis. Advancing our understanding of this pathway will benefit our ability to model disease, screen drugs and generate tissue and organ replacements for regenerative medicine. Full article
(This article belongs to the Special Issue Wnt Signaling in Development, Regeneration and Cancer)
Show Figures

Figure 1

Figure 1
<p>Summary of Wnt/β-catenin, Hedgehog and hypoxia inducible factor (HIF) pathways and their downstream mediators in the liver. (<b>A</b>) Active Wnt/β-catenin signalling. Wnt2 and Wnt9b secreted from central vein endothelial cell complex with LRP5/6 and FZD receptors. The β-catenin destruction complex is prevented from assembling allowing cytoplasmic accumulation of β-catenin which in turn permits β-catenin’s translocation to the nucleus. β-catenin then complexes with co-factors including CREB binding protein (CBP), TCF/LEF, B-cell lymphoma 9 (BCL-9) and pygopus protein (Pygo) to promote transcription of genes under β-catenin transcriptional control including c-Myc, CyclinD1, GS, aryl hydrocarbon receptor (AHR) and constitutive androstane receptor (CAR). (<b>B</b>) Active HIF1 signalling. Constitutively expressed HIF1α subunits avoid ubiquitin ligase, HIF prolyl hydroxylase (PHD) and VHL (von Hippel Lindau) facilitated proteasomal degradation when PHD is inactivated by low pO<sub>2</sub>. HIF1α subunits are then free to translocate to the nucleus where they dimerise with aryl hydrocarbon receptor nuclear translocator (ARNT) beta subunits to form HIF1 which is then able to bind to HIFresponse elements (HREs) and affect transcription of genes including vascular endothelial growth factor (VEGF), plasminogen activator inhibitor 1 (PAI1), P4HA2, HO1, erythropoietin, ceruloplasmin glucokinase and bone morphogenic protein (BMP). (<b>C</b>) Active Hh signalling. Hh ligands bind and inhibit their cognate receptor patched (PTCH). PTCH is no longer able to inhibit the G-protein coupled receptor smoothened (SMO), and SMO is phosphorylated by CKI and GPRK2 causing translocation from intracellular endosomes to the plasma membrane of primary cilia. Gli proteins (Gli1, Gli2, Gli3) can then escape association with suppressor of fused homolog (SUFU) and phosphorylation by GSK3β, protein kinase A (PKA), CKI and resulting in proteasomal degradation. It is then free to translocate to the nucleus to effect transcription of Hh target genes including VEGF, angiopoietin 1 and 2; Snail, Nanog, SOX2 and 9; Twist2, α-SMA and vimentin.</p>
Full article ">Figure 2
<p>Structural and functional zonation in the adult liver. (<b>A</b>) The liver receives oxygenated blood from the heart via the hepatic artery (red; 25%) and deoxygenated blood via the portal vein (blue; 75%). Blood drains via the inferior vena cava (purple). Bile drains from the liver and gallbladder via the common hepatic duct (green). (<b>B</b>) Liver lobules are roughly hexagonal. The portal triads comprise the portal vein, hepatic artery and bile duct and are found at each vertex surrounding a central vein. (<b>C</b>) The acinus is divided into 3 zones: periportal, intermediate and perivenous. APC and carbamoyl phosphate synthase 1 (CPS1) are expressed throughout the periportal and intermediate zones in a mutually exclusive relationship with glutamine synthetase (GS) which is expressed only in the last 1–2 layers of hepatocytes in the perivenous zone as result of the transcriptional activity of nuclear β-catenin. Wnt/β-catenin signalling is, therefore, largely restricted to the perivenous zone. Oxygen (O<sub>2</sub>) concentration (pO<sub>2</sub>) decreases as blood moves through the acinus and is lowest in the perivenous zone. HIF activity is highest in the perivenous region. Hh ligands are secreted by periportal endothelial cells. (<b>D</b>) The acinus comprises a sinusoid connecting the hepatic artery and portal vein with the central vein. Oxygenated blood from the hepatic artery mixes with deoxygenated blood from the portal vein as it flows towards the central vein. Stellate cells and Kupffer cells are present throughout the endothelial sinusoid. Bile secreted from the apical membrane of hepatocytes drains into the canal of Herring which empties into the bile duct. The basolateral membranes of hepatocytes line the sinusoid and project into the space of Disse where proteins and metabolites are exchanged. Stellate cells are found in the space of Disse, a cavity which separates hepatocytes from the sinusoidal endothelial cells. Kupffer cells, the resident macrophage cell, are present within the endothelial sinusoid.</p>
Full article ">Figure 3
<p>Role of Wnt/β-catenin signalling in gut tube patterning. After gastrulation, the definitive endoderm (DE) forms into the primitive gut tube along the anterior–posterior axis. This gives rise to the foregut, midgut and hindgut domains. Wnt/β-catenin signalling acts by inducing midgut and hindgut development in the posterior axis. Foregut formation occurs when Wnt antagonists secreted from the anterior endoderm suppress Wnt/β-catenin signalling, allowing for subsequent liver development.</p>
Full article ">
18 pages, 1322 KiB  
Review
Wnt/β-catenin Signaling in Tissue Self-Organization
by Kelvin W. Pond, Konstantin Doubrovinski and Curtis A. Thorne
Genes 2020, 11(8), 939; https://doi.org/10.3390/genes11080939 - 14 Aug 2020
Cited by 23 | Viewed by 5849
Abstract
Across metazoans, animal body structures and tissues exist in robust patterns that arise seemingly out of stochasticity of a few early cells in the embryo. These patterns ensure proper tissue form and function during early embryogenesis, development, homeostasis, and regeneration. Fundamental questions are [...] Read more.
Across metazoans, animal body structures and tissues exist in robust patterns that arise seemingly out of stochasticity of a few early cells in the embryo. These patterns ensure proper tissue form and function during early embryogenesis, development, homeostasis, and regeneration. Fundamental questions are how these patterns are generated and maintained during tissue homeostasis and regeneration. Though fascinating scientists for generations, these ideas remain poorly understood. Today, it is apparent that the Wnt/β-catenin pathway plays a central role in tissue patterning. Wnt proteins are small diffusible morphogens which are essential for cell type specification and patterning of tissues. In this review, we highlight several mechanisms described where the spatial properties of Wnt/β-catenin signaling are controlled, allowing them to work in combination with other diffusible molecules to control tissue patterning. We discuss examples of this self-patterning behavior during development and adult tissues’ maintenance. The combination of new physiological culture systems, mathematical approaches, and synthetic biology will continue to fuel discoveries about how tissues are patterned. These insights are critical for understanding the intricate interplay of core patterning signals and how they become disrupted in disease. Full article
(This article belongs to the Special Issue Wnt Signaling in Development, Regeneration and Cancer)
Show Figures

Figure 1

Figure 1
<p>General model for the Wnt/β-catenin signaling pathway and its regulation by the destruction complex. The destruction complex: Axin, Adenomatous polyposis coli (APC), casein kinase 1 α (CK1α), glycogen synthase kinase 3 (GSK3), protein phosphatase 2A (PP2a), and the ubiquitin ligase βTrCP. Membrane receptors: Frizzled (FZD) and low-density lipoprotein receptor-related protein 6 (Lrp6). Dishevelled (Dvl) T-cell factor (TCF).</p>
Full article ">Figure 2
<p>Wnt/β-catenin signaling in tissue self-organization. Two main categories are shown, Embryogenesis/Development (top) and Regeneration/Homeostasis (bottom). The categories were partitioned due to reversibility. Development of the embryo and body axis as well as appendage development are irreversible and temporally regulated patterning processes. Uniquely, regeneration and homeostasis are ongoing processes that an organism must call upon when needed. These processes have a greater need to be autonomous and could not withstand removal of a morphogen source unless there was a mechanism in place for sensing and replacing a missing niche.</p>
Full article ">Figure 3
<p>Examples of Wnt/β-catenin-dependent Reaction-Diffusion (RD) mechanisms implicated in tissue patterning. (<b>A</b>) Over time, a short-range activator and a long-range inhibitor interact to form local morphogen concentrations in patterns. (<b>B</b>) Basic model for the RD activator/inhibitor pair. (<b>C</b>) Examples of Wnt-driven patterning driven by RD mechanisms. Although the morphogen pairs/triplets and their mechanisms of control varies, the basic activator/inhibitor concepts are maintained and predicted by mathematical modeling. Patterning images C1–C4 were adapted from References [<a href="#B37-genes-11-00939" class="html-bibr">37</a>,<a href="#B46-genes-11-00939" class="html-bibr">46</a>,<a href="#B47-genes-11-00939" class="html-bibr">47</a>,<a href="#B48-genes-11-00939" class="html-bibr">48</a>].</p>
Full article ">
11 pages, 1677 KiB  
Review
Nuclear Regulation of Wnt/β-Catenin Signaling: It’s a Complex Situation
by Christin C. Anthony, David J. Robbins, Yashi Ahmed and Ethan Lee
Genes 2020, 11(8), 886; https://doi.org/10.3390/genes11080886 - 4 Aug 2020
Cited by 79 | Viewed by 7827
Abstract
Wnt signaling is an evolutionarily conserved metazoan cell communication pathway required for proper animal development. Of the myriad of signaling events that have been ascribed to cellular activation by Wnt ligands, the canonical Wnt/β-catenin pathway has been the most studied and best understood. [...] Read more.
Wnt signaling is an evolutionarily conserved metazoan cell communication pathway required for proper animal development. Of the myriad of signaling events that have been ascribed to cellular activation by Wnt ligands, the canonical Wnt/β-catenin pathway has been the most studied and best understood. Misregulation of Wnt/β-catenin signaling has been implicated in developmental defects in the embryo and major diseases in the adult. Despite the latter, no drugs that inhibit the Wnt/β-catenin pathway have been approved by the FDA. In this review, we explore the least understood step in the Wnt/β-catenin pathway—nuclear regulation of Wnt target gene transcription. We initially describe our current understanding of the importation of β-catenin into the nucleus. We then focus on the mechanism of action of the major nuclear proteins implicated in driving gene transcription. Finally, we explore the concept of a nuclear Wnt enhanceosome and propose a modified model that describes the necessary components for the transcription of Wnt target genes. Full article
(This article belongs to the Special Issue Wnt Signaling in Development, Regeneration and Cancer)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Model of the Wnt pathway. (<b>a</b>)—ln the absence of Wnt ligand, the β-catenin β destruction complex maintains low cytoplasmic levels of β-catenin. (<b>b</b>)—In the presence of Wnt ligand, the signalosome is assembled and the β-catenin degradation is disrupted. Entry of β-catenin into the nucleus promotes the formation of the enhanceosome to drive the transcription of Wnt target genes. See text for more details.</p>
Full article ">Figure 2
<p>Cartoon of β-catenin and its nuclear interacting partners.</p>
Full article ">Figure 3
<p>Proposed modified model of a Wnt enhanceosome with a β-catenin-TCF/LEF core complex and other requisite components that are context dependent. We propose that at least four additional components are required to form a functional enhanceosome: (1) a chromatin remodeling complex (e.g., SET-1, a histone methyltransferase, and CBP/p300, a histone acetyltransferase) that promotes gene transcription; (2) a bridging factor to link enhancer regions to the WRE and which may coordinate context-dependent factors (e.g., the ChILS complex and BCL9); (3) a mediator recruiter (e.g., Pygo and β-catenin); (4) an E3 ubiquitin ligase that promotes Gro/TLE dissociation from TCF/LEF (e.g., XIAP or Hyd/UBR5).</p>
Full article ">
25 pages, 1238 KiB  
Review
Glia and Neural Stem and Progenitor Cells of the Healthy and Ischemic Brain: The Workplace for the Wnt Signaling Pathway
by Tomas Knotek, Lucie Janeckova, Jan Kriska, Vladimir Korinek and Miroslava Anderova
Genes 2020, 11(7), 804; https://doi.org/10.3390/genes11070804 - 16 Jul 2020
Cited by 17 | Viewed by 4619
Abstract
Wnt signaling plays an important role in the self-renewal, fate-commitment and survival of the neural stem/progenitor cells (NS/PCs) of the adult central nervous system (CNS). Ischemic stroke impairs the proper functioning of the CNS and, therefore, active Wnt signaling may prevent, ameliorate, or [...] Read more.
Wnt signaling plays an important role in the self-renewal, fate-commitment and survival of the neural stem/progenitor cells (NS/PCs) of the adult central nervous system (CNS). Ischemic stroke impairs the proper functioning of the CNS and, therefore, active Wnt signaling may prevent, ameliorate, or even reverse the negative effects of ischemic brain injury. In this review, we provide the current knowledge of Wnt signaling in the adult CNS, its status in diverse cell types, and the Wnt pathway’s impact on the properties of NS/PCs and glial cells in the context of ischemic injury. Finally, we summarize promising strategies that might be considered for stroke therapy, and we outline possible future directions of the field. Full article
(This article belongs to the Special Issue Wnt Signaling in Development, Regeneration and Cancer)
Show Figures

Figure 1

Figure 1
<p>Overview of the canonical and non-canonical Wnt signaling pathways. (<b>A</b>) β-catenin-dependent signaling is activated by binding of a canonical Wnt ligand to the frizzled (FZD) receptor and the low-density lipoprotein receptor-related protein 5/6 (LRP5/6) co-receptor. Subsequently, β-catenin is released from the destruction complex and translocated to the nucleus, where it forms a complex with the T-cell factor/lymphoid enhancer-binding factor (TCF/LEF) proteins in order to activate transcription of the Wnt-responsive genes; (<b>B</b>) Activation of the non-canonical—β-catenin-independent—Wnt signaling branches influences cell properties and results in transcription of alternative Wnt target genes. In addition, in some cells non-canonical signaling inhibits β-catenin-mediated transcription. A more detailed description of the pathways is given in the text. Abbreviations: AP1, activator protein 1; APC, adenomatous polyposis coli; AXIN, axis inhibition; β-CAT, β-catenin; β-TrCP, β-transducin repeats-containing protein; C-JUN, transcription factor C-JUN; Ca<sup>2+</sup>, calcium; CaMKII, Ca<sup>2+</sup>/calmodulin-dependent protein kinase II; CDC42, GTPase CDC42; CK1, casein kinase 1; DAAM1, DVL-associated activator of morphogenesis 1; DAG, diacylglycerol; DKK, dickkopf; DVL, dishevelled; ER, endoplasmic reticulum; GSK3β, glycogen synthase kinase 3β; IP3, inositol trisphosphate; JNK, c-Jun N-terminal kinase; NFAT, nuclear factor of activated T-cells; NLK, nemo-like kinase; P, phosphorylation; PCP, planar cell polarity; PKC, protein kinase C; PLC, phospholipase C; RAC1, Rac family small GTPase 1; RhoA, Ras homolog family member A; ROCK, Rho-associated kinase; ROR, receptor tyrosine kinase ROR; RYK, receptor tyrosine kinase RYK; sFRP, secreted FZD-related proteins; TLE, transducin-like enhancer of split; Ub, ubiquitination; WIF, Wnt inhibitory factor.</p>
Full article ">Figure 2
<p>Neurogenic niches of the adult mammalian brain. Schematic diagrams illustrating the main cellular components within the neurogenic niche in the (<b>A</b>) subventricular zone (SVZ) of the lateral ventricles (LVs) and (<b>B</b>) dentate gyrus (DG) of the hippocampus (HIP) containing the neurogenic niche in the subgranular zone (SGZ), and representative immunofluorescence images of the corresponding regions showing the main markers of cell types indicated in the illustrated schemes. The neuronal lineage in both compartments is represented by neural stem cells (NSCs), neural progenitor cells (NPCs) and migrating neuroblasts. In addition, neuronal maturation is shown in the granule cell layer (GCL) adjacent to the SGZ. Glial cells, represented by astrocytes and neuron-glial antigen 2 (NG2)-positive glia in both regions, and by ependymal cells constituting the ventricular wall (VW) of the LVs, release Wnt ligands (in red color) in order to activate quiescent NSCs and mediate their proliferation/differentiation. Glial fibrillary acidic protein (GFAP) represents NSCs and a subpopulation of astrocytes; proliferating cell nuclear antigen (PCNA) marks proliferating progenitors; doublecortin (DCX)-positive cells identify both neuroblasts and immature neurons; NG2 is present in NG2 glia; 4′,6-diamidino-2-phenylindole (DAPI) stains cell nuclei. Note that the magnified field and DG histology in panel B are rotated approximately 90° counterclockwise compared to the scheme of the frontal section. Scale bar: 15 µm. Abbreviations: CC, corpus callosum; CTX, cortex; STR, striatum.</p>
Full article ">
19 pages, 1114 KiB  
Review
The Role of Wnt Signalling in Chronic Kidney Disease (CKD)
by Soniya A. Malik, Kavindiya Modarage and Paraskevi Goggolidou
Genes 2020, 11(5), 496; https://doi.org/10.3390/genes11050496 - 30 Apr 2020
Cited by 23 | Viewed by 4985
Abstract
Chronic kidney disease (CKD) encompasses a group of diverse diseases that are associated with accumulating kidney damage and a decline in glomerular filtration rate (GFR). These conditions can be of an acquired or genetic nature and, in many cases, interactions between genetics and [...] Read more.
Chronic kidney disease (CKD) encompasses a group of diverse diseases that are associated with accumulating kidney damage and a decline in glomerular filtration rate (GFR). These conditions can be of an acquired or genetic nature and, in many cases, interactions between genetics and the environment also play a role in disease manifestation and severity. In this review, we focus on genetically inherited chronic kidney diseases and dissect the links between canonical and non-canonical Wnt signalling, and this umbrella of conditions that result in kidney damage. Most of the current evidence on the role of Wnt signalling in CKD is gathered from studies in polycystic kidney disease (PKD) and nephronophthisis (NPHP) and reveals the involvement of β-catenin. Nevertheless, recent findings have also linked planar cell polarity (PCP) signalling to CKD, with further studies being required to fully understand the links and molecular mechanisms. Full article
(This article belongs to the Special Issue Wnt Signaling in Development, Regeneration and Cancer)
Show Figures

Figure 1

Figure 1
<p>Wnt component expression in normal, PKD and NPHP kidneys. The listed Wnt components (black) are expressed in all three normal, PKD and NPHP kidneys, with differential expression in PKD indicated in orange and differential expression in NPHP shown in purple.</p>
Full article ">Figure 2
<p>An illustration of the potential mechanisms linking Wnt signaling, PKD and NPHP. The associations highlight the relationship of PKD1 and PKD2 with canonical Wnt components, while Fibrocystin and Inversin are linked to PCP signaling.</p>
Full article ">
Back to TopTop