Open Access

Wnt6 influences the viability of mouse embryonic palatal mesenchymal cells via the β‑catenin pathway

  • Authors:
    • Zheng Jiang
    • Lin Pan
    • Xiaoling Chen
    • Zhiqun Chen
    • Dongwei Xu
  • View Affiliations

  • Published online on: October 2, 2017     https://doi.org/10.3892/etm.2017.5240
  • Pages: 5339-5344
  • Copyright: © Jiang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The embryological stages of palatal shelf elongation and elevation, mainly induced by the proliferation and extracellular matrix secretion of embryonic palatal mesenchymal (MEPM) cells, are essential for normal palatal development. Wingless‑related MMTV integration site gene family (Wnt) signaling pathways serve key roles in craniofacial development and palate formation. Recent studies have indicated that Wnt6 participates in embryonic development of the palate, though its exact role in palate development remains unclear. In the present study, to investigate the role of Wnt6 during the stages of palatal shelves elongation and elevation, mouse MEPM cells were cultured from dissected palatal shelves at embryonic day 13.5. Results of an MTT assay and flow cytometric analysis demonstrated that treatment with recombinant Wnt6 increased the viability of MEPM cells (P<0.01) and the proportion of cells in the S and G2/M phases (P<0.01). Meanwhile, Wnt6 activated the β‑catenin signaling pathway as indicated by the dual luciferase assay result, and blockade of the WNT/β‑catenin pathway reduced the cytoactivity of Wnt6 in MEPM cells (P<0.01). Collectively, these findings indicate that Wnt6 promotes the vitality of MEPM cells by increasing the S + G2/M‑phase cell population, potentially through activation of the β‑catenin pathway during palatal shelf elongation and elevation.

Introduction

Cleft palate is among the most common congenital birth defects in humans (13), and is caused by a failure in secondary palate formation (4,5). From embryonic days (E) 12.5 to E15.5, the secondary palate is initiated to extend from the maxillary processes to form the palatal shelves (6). At approximately E13.5, the bilateral palatal shelves elongate and elevate to form the midline edge seam (7), while the size of the palatal shelves is determined by the proliferation and apoptosis of mouse embryonic palatal mesenchymal (MEPM) cells (8). During this period, disturbances in the normal viability or extracellular matrix secretion of MEPM cells can cause cleft palate (9). These two processes are controlled by complex signaling cascades that are induced by a number of growth factors, including members of the wingless-related MMTV integration site gene family (Wnt) (8,10,11).

The Wnt signaling glycoproteins serve critical roles in cell viability, differentiation, migration, polarity, and death, and thereby regulate embryonic development (1218). On the basis of their downstream signal transduction, Wnts have been divided into ‘β-catenin-dependent’ and ‘β-catenin-independent’ signaling proteins. β-catenin-dependent signaling is defined by the stabilization of β-catenin, which enters the nucleus and forms a complex with lymphoid-enhancer-binding factor/T cell factor (TCF) DNA-binding proteins to activate transcription of Wnt target genes (1216). Meanwhile, β-catenin-independent signaling performed without β-catenin includes a number of less well characterized intracellular pathways (1820). In previous studies, the Wnt signaling pathways have been suggested to serve important roles in craniofacial development (21,22) and palate formation (8,21,2325).

Wnt family member 6 (Wnt6), as a member of the Wnt signaling family, has been reported to be essential for the viability of macrophages (26), myoblasts (27) and stromal cells (28), and to be expressed in mouse embryonic palatal tissue from E12.5 to E14.5 (10). Furthermore, the WNT6-WNT10A cluster has been identified to exhibit linkage and disequilibrium in cleft palate (29). These previous data indicate that Wnt6 participates in embryonic development of the palate. However, the exact role of Wnt6 in palate development remains unclear.

The purpose of the present study was to investigate the effect of Wnt6 in MEPM cells using the MTT assay, flow cytometry, western blot analysis and reporter gene assay. The results suggest that Wnt6 may regulate the viability of palatal mesenchymal cells through the β-catenin pathway.

Materials and methods

Ethics statement

This study was carried out in strict accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals of the National Institutes of Health (Bethesda, MD, USA) (6). The protocol was approved by the Committee for the Ethics of Animal Experiments of Xiamen University, Xiamen, China (permit no. SCXK2013-0001). All surgical procedures were performed under urethane (Thermo Fisher Scientific, Inc., Waltham, MA, USA) anesthesia (1.0 g/kg via intraperitoneal injection), and all efforts were made to minimize suffering.

MEPM cell culture

A total of 60 female and 20 male wild-type CD1 mice (Charles River Laboratories, Inc., Wilmington, MA, USA) of the same strain were housed at an ambient temperature of 22°C with 12-h light/dark cycle and had access to food and water ad libitum. Mature male and female mice (6 weeks old, weighing 17–23 g) were mated overnight, and the presence of a vaginal plug the following morning was designated as E0.5. Primary MEPM cell cultures were established from secondary palatal tissue microdissected from E13.5 mouse embryos (11). The dissected palate shelves were treated with 1 U/ml dispase II (Roche Diagnostics, Indianapolis, IN, USA) at 37°C for 15 min (8,30). Following removal of the desquamated epithelia, the tissue was digested with 0.25% trypsin/0.05% EDTA, and then the dissociated cells were cultured for 48 h in Dulbecco's modified Eagle's medium/nutrient mixture F-12 (DMEM/F12; Hyclone; GE Healthcare Life Sciences, Logan, UT, USA) with 10% fetal bovine serum (FBS; Hyclone; GE Healthcare Life Sciences) and 1% penicillin/streptomycin at 37°C in a humidified atmosphere of 5% CO2 in air. Cells were passaged and seeded at 4×105 cells/25 cm2 in DMEM/F12 without FBS and incubated at 37°C for 40 min; in such conditions, the epithelial cells adhered more slowly than the MEPM cells. After culturing for 40 min at 37°C, the medium without FBS was replaced with fresh DMEM/F12 including FBS and antibiotics to acquire MEPM cells of high purity. Cells at passage 3 cultured at 37°C for 8 h were used for subsequent assays. Experimental cell groups were established, in which cells were always incubated with additional 100 ng/ml recombinant Wnt6 (Abgent, Inc., San Diego, CA, USA) with or without additional 100 ng/ml recombinant dickkopf WNT signaling pathway inhibitor 1 (DKK1; R&D Systems, Inc., Minneapolis, MN, USA) in DMEM/F12 containing 10% FBS and 1% penicillin/streptomycin at 37°C. In the control cell group, MEPM cells were only cultured with 10% FBS and 1% penicillin/streptomycin in DMEM/F12.

MTT assay

The MEPM cells were transferred to 24-well plates (Corning Incorporated, Corning, NY, USA) at a density of 1.6×104 cells/well and cultured at 37°C in DMEM/F12 with 10% FBS for 1–4 days. MTT solution (5 mg/ml) was added to each well and the plates were incubated at 37°C for 4 h. Subsequently, the supernatant was removed and dimethyl sulfoxide 0.5 mg/ml was added to each well to dissolve the formazan crystals. The optical density at 490 nm was quantified using the PowerWave XS2 package at daily intervals over the 4-day assay period (BioTek Instruments, Inc., Winooski, VT, USA). Three independent replicates were performed for each cell group.

Flow cytometry

Following treatment with or without Wnt6 or Wnt6 + DKK1 for 48 h as stated above, the MEPM cells were harvested and fixed in cold 75% ethanol at −4°C overnight, cells were pelleted and washed twice in 1× PBS at 1,000 × g for 5 min at 4°C. Subsequently, cells were incubated with 50 µg/ml propidium iodide and 5 µg/ml RNAase at room temperature for 30 min. Following supplementation with 50 mg/l propidium iodide and incubation in the dark for 1 h at room temperature, cell cycle analysis was performed using a BD FACS Calibur flow cytometer (BD Biosciences, Franklin Lakes, NJ, USA) and CellQuest Pro 5.1 software (BD Biosciences) with a minimum of 20,000 events for each cell group.

Reporter gene assay

After 48 h of treatment with or without 100 ng/ml recombinant Wnt6 as stated above, the MEPM cells were seeded at 1.6×105 cells/well onto 12-well plates, and transient transfections for 5 min at room temperature were performed using Lipofectamine 2,000 (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA). Cells were transfected with 1.6 µg of TCF luciferase reporter constructs (Promega Corporation, Madison, WI, USA) (TOPflash or FOPflash, respectively) and 0.08 µg of Renilla reniformis gene vectors (Promega Corporation) for normalization. The TOPflash TCF reporter plasmid contains two sets of three copies of the binding site upstream of the thymidine kinase minimal promoter and luciferase open reading frame, while FOPflash contains mutated TCF binding sites and was used as a negative control (3134). A subset of the cells lacking Wnt6 treatment were also cotransfected with β-catenin pcDNA (1.6 µg; Biocytogen LLC, Beijing, China) as a positive control. Following 48 h of incubation, the luciferase assay was performed using a Dual Luciferase Assay System kit (Promega Corporation) according to the manufacturer's protocol. Relative luciferase activity was reported as the ratio of firefly/Renilla luciferase activity.

Western blot analysis

Total protein was isolated from cultured MEPM cells after 48 h of treatment with or without Wnt6 ± DKK1 using RIPA lysis and extraction buffer (Beyotime Institute of Biotechnology, Haimen, China). Protein quantification was performed with a Bio-Rad DC Protein Assay kit (Bio-Rad Laboratories, Inc., Hercules, CA, USA). The intracellular protein expression levels of β-catenin and β-actin and the Wnt6 protein levels in the cell culture supernatant were assessed. To obtain Wnt6 protein from the supernatant, conditioned culture media were collected and concentrated with Amicon® Ultra-4 Centrifugal Filter Units (10,000 NMWL; EMD Millipore, Billerica, MA, USA), then extracted with a Protein Extraction kit II (Applygen Technologies, Inc., Beijing, China), as described previously (22). Equal amounts of protein (60 µg per lane) were separated on 10% SDS-polyacrylamide gels and transferred onto polyvinylidenedifluoride membranes (Roche Diagnostics). The membrane was blocked in a 6% non-fat milk solution in Tris-buffered saline with 0.5% Tween-20 (TBST) (Roche Diagnostics) at room temperature for 1 h, and then incubated with rabbit anti-Wnt6 monoclonal antibody (Abcam, Cambridge, UK; cat. no. ab154144; dilution 1:200), rabbit anti-β-catenin monoclonal antibody (Abcam; cat. no. ab32572; dilution 1:500) or rabbit anti-β-actin monoclonal antibody (Wuhan Antgene Biotechnology Co., Ltd, Wuhan, China; cat. no. ANT009; dilution 1:800) overnight at 4°C. After rinsing with TBST for three times, the goat anti-rabbit HRP-conjugated secondary antibody (Wuhan Antgene Biotechnology Co., Ltd., cat. no. ANT020; dilution 1:5,000) was applied to the membranes at room temperature for 1 h. The blot was visualized using SuperSignal West Pico Chemiluminesent Substrate (Thermo Fisher Scientific, Inc.) and the protein bands were analyzed with ImageJ 1.48 software (National Institutes of Health).

Statistical analysis

All quantitative data were presented as the mean ± standard deviation. Statistical analysis of differences was performed with SPSS 19.0 software (SPSS, Inc., Chicago, IL, USA) P<0.05 was considered to indicate statistical significance. The significance of data was determined by one-way analysis of variance followed by a Bonferroni post-hoc test.

Results

Wnt6 promotes the viability of MEPM cells

The expression of Wnt6 was identified in primary (control) MEPM cells at E13.5 (Fig. 1). To investigate the effect of Wnt6 on MEPM cell viability, recombinant Wnt6 was added to the culture medium, and cell viability was assessed with an MTT assay. It was observed that the viability of MEPM cells treated with Wnt6 was significantly higher than that of control cells between 2 and 4 days after treatment (P<0.01; Fig. 2).

In addition, the results of flow cytometric analysis indicated that the proportion of primary MEPM cells in S + G2/M phases (29.91±0.72%) was significantly increased by treatment with Wnt6 (50.93±0.89%; P<0.01; Fig. 3). These findings suggested that Wnt6 promoted the viability of MEPM cells and increased the population of S + G2/M-phase cells.

Wnt6 activates the β-catenin/TCF signaling pathway

A TOP flash luciferase reporter assay was performed to investigate whether Wnt6 activates Wnt/β-catenin signaling in MEPM cells. Cells treated with Wnt6 or pcDNA-β-catenin exhibited significantly higher baselines of β-catenin/TCF-mediated transcriptional activity when compared with the control cells (P<0.01). In addition, the TOPflash reporter activity did not differ significantly between the Wnt6 and β-catenin groups (FOPflash data not shown) (Fig. 4). Furthermore, the total amount of β-catenin was markedly increased in the Wnt6-treated cells when compared with the control cells (Fig. 1).

Blockade of the WNT/β-catenin pathway reduces the cytoactivity of Wnt6

DKK1 binds to low density lipoprotein receptor-related protein 5/6 and is considered to be a general canonical Wnt signaling inhibitor (35). In the present study, DKK1 was used to confirm the effect of Wnt6⁄β-catenin signaling on MEPM cell viability. Cells treated with Wnt6 were incubated with or without DKK1. As depicted in Fig. 1, the additional treatment with DKK1 suppressed the expression of β-catenin. In addition, the results in Fig. 2 demonstrated that DKK1 significantly reduced the Wnt6-mediated increase in cell viability from day 2 post-treatment (P<0.01). Furthermore, when the Wnt6/β-catenin pathway was blocked, the viability of MEPM cells was significantly lower than that of control cells (P<0.01). A notable decrease in the S + G2/M cell proportion was also observed in the DKK1 + Wnt6 group (20.43±0.59%; Fig. 3) when compared with the control and Wnt6 groups (P<0.01), and cell cycle arrest in G0/G1 phase was indicated in the DKK1 + Wnt6 group.

Discussion

Embryonic development of the palate is a temporospatially coordinated process. The secondary palate is initiated to extend from the maxillary processes to form the palatal shelves between E12.5 and E15.5 (6). At approximately E13.5, the bilateral palatal shelves, composed of a core of mesenchyme cells, elongate and elevate toward each other (7). In the present study, to investigate the role of Wnt6 during the stages of palatal shelf elongation and elevation, MEPM cells were cultured from palatal shelves dissected from mouse fetuses at E13.5. The results of MTT assay and flow cytometry analysis indicated that the viability and S + G2/M population of the MEPM cells was increased by Wnt6. Meanwhile, a dual luciferase assay and analysis with DKK1 identified a pivotal role of β-catenin signaling in the development of MEPM cells. Collectively, the results indicated that Wnt6 promoted the viability of the MEPM cells by increasing the population of S + G2/M cells. The involvement of Wnt6 in the β-catenin pathway during palatal shelf elongation and elevation was also implicated.

Former studies have already demonstrated that Wnt6 is a necessary cytoactive factor in macrophages (26), myoblasts (27) and stromal cells (28). Wnt6 has also been reported to be expressed in the mouse embryonic palatal tissue from E12.5 to E14.5 (10), and the WNT6-WNT10A cluster exhibits linkage and disequilibrium in cleft palate (29). In the present study, the effect of Wnt6 on MEPM cell viability during embryonic palate development was elucidated in vitro. Further studies into the effects of Wnt6 knockout on palatogenesis in mice should now be performed to identify potentially associated malformations and regulatory mechanisms.

Numerous different factors are involved in the complex transduction of Wnt signals, though particular cells only respond to specific Wnt factors (36). It has been reported that Wnt6 activates β-catenin signaling in F9 embryonal carcinoma cells and serves an important role in endoderm specification (37,38) and formation (39). Furthermore, Wnt6 restricted heart muscle development in embryonic mesenchymal cells through the β-catenin pathway (40), and the osteoblastogenesis of mesenchymal stem cells was regulated by Wnt6⁄β-catenin signaling (41). In the current study, Wnt6 was indicated to stimulate the transcriptional activity of β-catenin, suggesting that it may activate Wnt/β-catenin signaling to enhance the viability of MEPM cells. Treatment with a recombinant version of the Wnt signaling antagonist DKK1 was also performed to verify the relationship between Wnt6 and β-catenin signaling. The results demonstrated that the increased viability of Wnt6-treated MEPM cells was inhibited by DKK1. This data confirmed the stimulatory effect of Wnt6 on the viability of MEPM cells. However, as DKK1 is not a specific inhibitor of Wnt6, further studies are required to confirm the exact signal transduction mechanism of the Wnt6/β-catenin pathway in MEPM cells.

In addition, the present study identified that compromised Wnt6 activity and β-catenin signaling in MEPM cells reduced the cytoactivity of Wnt6 and caused cell cycle arrest in G0/G1 phase when compared with control cells. Thus, conditional inhibition of the β-catenin pathway may disturb the normal cytoactivity of MEPM cells, which may ultimately lead to the development of cleft palate.

It is established that reduced Wnt/β-catenin signaling is involved in the development of cleft palate (21,2325). Furthermore, the Wnt/β-catenin signaling pathway has previously been associated with alterations in the viability and apoptosis of MEPM cells (8). These previous findings and the present data suggest that Wnt/β-catenin pathway is among the most important signaling cascades for palatal development. However, the exact mechanism regarding the involvement of β-catenin in MEPM cell activity requires further investigation.

In conclusion, to the best of our knowledge, the present results are the first to indicate that Wnt6/β-catenin signaling is involved in the viability of MEPM cells during the stages of palatal shelf elongation and elevation in vitro, thus suggesting that Wnt6/β-catenin signaling is involved in the process of palatogenesis.

Acknowledgements

The present study was supported by the Science and Technology Program of Xiamen City, China (grant no. 3502Z20154074).

References

1 

Mossey PA, Little J, Munger RG, Dixon MJ and Shaw WC: Cleft lip and palate. Lancet. 374:1773–1785. 2009. View Article : Google Scholar : PubMed/NCBI

2 

Fraser FC: The genetics of cleft lip and cleft palate. Am J Hum Genet. 22:336–352. 1970.PubMed/NCBI

3 

Schutte BC and Murray JC: The many faces and factors of orofacial clefts. Hum Mol Genet. 8:1853–1859. 1999. View Article : Google Scholar : PubMed/NCBI

4 

Meng T, Shi JY, Wu M, Wang Y, Li L, Liu Y, Zheng Q, Huang L and Shi B: Overexpression of mouse TTF-2 gene causes cleft palate. J Cell Mol Med. 16:2362–2368. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Ferguson MW: Palate development. Development. 103 Suppl:S41–S60. 1988.

6 

Wu C, Endo M, Yang BH, Radecki MA, Davis PF, Zoltick PW, Spivak RM, Flake AW, Kirschner RE and Nah HD: Intra-amniotic transient transduction of the periderm with a viral vector encoding TGFβ3 prevents cleft palate in Tgfβ3(−/-) mouse embryos. Mol Ther. 21:8–17. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Meng L, Bian Z, Torensma R and Von den Hoff JW: Biological mechanisms in palatogenesis and cleft palate. J Dent Res. 88:22–33. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Feng C, Xu Z, Li Z, Zhang D, Liu Q and Lu L: Down-regulation of Wnt10a by RNA interference inhibits proliferation and promotes apoptosis in mouse embryonic palatal mesenchymal cells through Wnt/β-catenin signaling pathway. J Physiol Biochem. 69:855–863. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Li L, Shi JY, Zhu GQ and Shi B: MiR-17-92 cluster regulates cell proliferation and collagen synthesis by targeting TGFB pathway in mouse palatal mesenchymal cells. J Cell Biochem. 113:1235–1244. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Warner DR, Smith HS, Webb CL, Greene RM and Pisano MM: Expression of Wnts in the developing murine secondary palate. Int J Dev Biol. 53:1105–1112. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Warner DR, Mukhopadhyay P, Brock GN, Pihur V, Pisano MM and Greene RM: TGFβ-1 and Wnt-3a interact to induce unique gene expression profiles in murine embryonic palate mesenchymal cells. Reprod Toxicol. 31:128–133. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Miller JR: The Wnts. Genome Biol. 3:REVIEWS30012002.PubMed/NCBI

13 

Lee HY, Kléber M, Hari L, Brault V, Suter U, Taketo MM, Kemler R and Sommer L: Instructive role of Wnt/beta-catenin of in sensory fate specification in neural crest stem cells. Science. 303:1020–1023. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Miki T, Yasuda SY and Kahn M: Wnt/β-catenin signaling in embryonic stem cell self-renewal and somatic cell reprogramming. Stem Cell Rev. 7:836–846. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Kühl SJ and Kühl M: On the role of Wnt/β-catenin signaling in stem cells. Biochim Biophys Acta. 1830:2297–2306. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Reya T and Clevers H: Wnt signalling in stem cells and cancer. Nature. 434:843–850. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Staal FJ and Luis TC: Wnt signaling in hematopoiesis: Crucial factors for self-renewal, proliferation and cell fate decisions. J Cell Biochem. 109:844–849. 2010.PubMed/NCBI

18 

Nusse R: Wnt signaling and stem cell control. Cell Res. 18:523–527. 2008. View Article : Google Scholar : PubMed/NCBI

19 

Moon RT, Bowerman B, Boutros M and Perrimon N: The promise and perils of Wnt signaling through beta-catenin. Science. 296:1644–1646. 2002. View Article : Google Scholar : PubMed/NCBI

20 

Angers S and Moon RT: Proximal events in Wnt signal transduction. Nat Rev Mol Cell Biol. 10:468–477. 2009.PubMed/NCBI

21 

Jin YR, Han XH, Taketo MM and Yoon JK: Wnt9b-dependent FGF signaling is crucial for outgrowth of the nasal and maxillary processes during upper jaw and lip development. Development. 139:1821–1830. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Jiang Z, Von den Hoff JW, Torensma R, Meng L and Bian Z: Wnt16 is involved in intramembranous ossification and suppresses osteoblast differentiation through the Wnt/β-catenin pathway. J Cell Physiol. 229:384–392. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Hu X, Gao J, Liao Y, Tang S and Lu F: Retinoic acid alters the proliferation and survival of the epithelium and mesenchyme and suppresses Wnt/β-catenin signaling in developing cleft palate. Cell Death Dis. 4:e8982013. View Article : Google Scholar : PubMed/NCBI

24 

Hu X, Gao JH, Liao YJ, Tang SJ and Lu F: Dexamethasone alters epithelium proliferation and survival and suppresses Wnt/β-catenin signaling in developing cleft palate. Food Chem Toxicol. 56:67–74. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Mostowska A, Hozyasz KK, Wójcicki P, Lasota A, Dunin-Wilczyńska I and Jagodziński PP: Association of DVL2 and AXIN2 gene polymorphisms with cleft lip with or without cleft palate in a Polish population. Birth Defects Res A Clin Mol Teratol. 94:943–950. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Schaale K, Brandenburg J, Kispert A, Leitges M, Ehlers S and Reiling N: Wnt6 is expressed in granulomatous lesions of Mycobacterium tuberculosis-infected mice and is involved in macrophage differentiation and proliferation. J Immunol. 191:5182–5195. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Hitchins L, Fletcher F, Allen S and Dhoot GK: Role of Sulf1A in Wnt1- and Wnt6-induced growth regulation and myoblast hyper-elongation. FEBS Open Bio. 3:30–34. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Wang Q, Lu J, Zhang S, Wang S, Wang W, Wang B, Wang F, Chen Q, Duan E, Leitges M, et al: Wnt6 is essential for stromal cell proliferation during decidualization in mice. Biol Reprod. 88:52013. View Article : Google Scholar : PubMed/NCBI

29 

Beaty TH, Hetmanski JB, Fallin MD, Park JW, Sull JW, McIntosh I, Liang KY, Vanderkolk CA, Redett RJ, Boyadjiev SA, et al: Analysis of candidate genes on chromosome 2 in oral cleft case-parent trios from three populations. Hum Genet. 120:501–518. 2006. View Article : Google Scholar : PubMed/NCBI

30 

Matsumura K, Taketomi T, Yoshizaki K, Arai S, Sanui T, Yoshiga D, Yoshimura A and Nakamura S: Sprouty2 controls proliferation of palate mesenchymal cells via fibroblast growth factor signaling. Biochem Biophys Res Commun. 404:1076–1082. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Thompson MD, Dar MJ and Monga SP: Pegylated interferon alpha targets Wnt signaling by inducing nuclear export of β-catenin. J Hepatol. 54:506–512. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Kang YJ, Park HJ, Chung HJ, Min HY, Park EJ, Lee MA, Shin Y and Lee SK: Wnt/β-catenin signaling mediates the antitumor activity of magnolol in colorectal cancer cells. Mol Pharmacol. 82:168–177. 2012. View Article : Google Scholar : PubMed/NCBI

33 

An JH, Yang JY, Ahn BY, Cho SW, Jung JY, Cho HY, Cho YM, Kim SW, Park KS, Kim SY, et al: Enhanced mitochondrial biogenesis contributes to Wnt induced osteoblastic differentiation of C3H10T1/2 cells. Bone. 47:140–150. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Kim DY, Park YG, Quan HY, Kim SJ, Jung MS and Chung SH: Ginsenoside Rd stimulates the differentiation and mineralization of osteoblastic MC3T3-E1 cells by activating AMP-activated protein kinase via the BMP-2 signaling pathway. Fitoterapia. 83:215–222. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Mao B, Wu W, Li Y, Hoppe D, Stannek P, Glinka A and Niehrs C: LDL-receptor-related protein 6 is a receptor for Dickkopf proteins. Nature. 411:321–325. 2001. View Article : Google Scholar : PubMed/NCBI

36 

Gordon MD and Nusse R: Wnt signaling: Multiple pathways, multiple receptors, and multiple transcription factors. J Biol Chem. 281:22429–22433. 2006. View Article : Google Scholar : PubMed/NCBI

37 

Lhomond G, McClay DR, Gache C and Croce JC: Frizzled1/2/7 signaling directs β-catenin nuclearisation and initiates endoderm specification in macromeres during sea urchin embryogenesis. Development. 139:816–825. 2012. View Article : Google Scholar : PubMed/NCBI

38 

Krawetz R and Kelly GM: Wnt6 induces the specification and epithelialization of F9 embryonal carcinoma cells to primitive endoderm. Cell Signal. 20:506–517. 2008. View Article : Google Scholar : PubMed/NCBI

39 

Hwang JT and Kelly GM: GATA6 and FOXA2 regulate Wnt6 expression during extraembryonic endoderm formation. Stem Cells Dev. 21:3220–3232. 2012. View Article : Google Scholar : PubMed/NCBI

40 

Lavery DL, Martin J, Turnbull YD and Hoppler S: Wnt6 signaling regulates heart muscle development during organogenesis. Dev Biol. 323:177–188. 2008. View Article : Google Scholar : PubMed/NCBI

41 

Cawthorn WP, Bree AJ, Yao Y, Du B, Hemati N, Martinez-Santibañez G and MacDougald OA: Wnt6, Wnt10a and Wnt10b inhibit adipogenesis and stimulate osteoblastogenesis through a β-catenin-dependent mechanism. Bone. 50:477–489. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2017
Volume 14 Issue 6

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Jiang Z, Pan L, Chen X, Chen Z and Xu D: Wnt6 influences the viability of mouse embryonic palatal mesenchymal cells via the β‑catenin pathway. Exp Ther Med 14: 5339-5344, 2017
APA
Jiang, Z., Pan, L., Chen, X., Chen, Z., & Xu, D. (2017). Wnt6 influences the viability of mouse embryonic palatal mesenchymal cells via the β‑catenin pathway. Experimental and Therapeutic Medicine, 14, 5339-5344. https://doi.org/10.3892/etm.2017.5240
MLA
Jiang, Z., Pan, L., Chen, X., Chen, Z., Xu, D."Wnt6 influences the viability of mouse embryonic palatal mesenchymal cells via the β‑catenin pathway". Experimental and Therapeutic Medicine 14.6 (2017): 5339-5344.
Chicago
Jiang, Z., Pan, L., Chen, X., Chen, Z., Xu, D."Wnt6 influences the viability of mouse embryonic palatal mesenchymal cells via the β‑catenin pathway". Experimental and Therapeutic Medicine 14, no. 6 (2017): 5339-5344. https://doi.org/10.3892/etm.2017.5240