scholarly journals Differentiation-Inducing Factor-1 Alters Canonical Wnt Signaling and Suppresses Alkaline Phosphatase Expression in Osteoblast-Like Cell Lines

2006 ◽  
Vol 21 (8) ◽  
pp. 1307-1316 ◽  
Author(s):  
Etsuko Matsuzaki ◽  
Fumi Takahashi-Yanaga ◽  
Yoshikazu Miwa ◽  
Masato Hirata ◽  
Yutaka Watanabe ◽  
...  
Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2742-2742 ◽  
Author(s):  
Nicola Giuliani ◽  
Simona Colla ◽  
Paola Storti ◽  
Gaetano Donofrio ◽  
Marina Bolzoni ◽  
...  

Abstract Osteoblast suppression is the hallmark of Multiple Myeloma (MM) osteolytic bone lesions mainly due to the capacity of MM cells to inhibit the osteogenic differentiation of bone marrow (BM) mesenchymal cells (MSC). Many evidences suggest that Wnt signaling is critically involved in the regulation of osteoblast formation. Recently, in murine osteoprogenitor cells and in MM mouse models it has been shown that activation of canonical Wnt pathway stimulate osteoblast formation and blunts MM-induced bone destruction. In this study we have investigated whether modulation of both canonical and noncanonical Wnt signaling pathway may affect osteogenic differentiation of human MSC and counterbalance the suppressive effect of MM cells. First we checked the potential expression of Wnt activators and inhibitors by human MSC and osteoprogenitor cells (PreOB) by gene arrays. We found that both cells expressed the activator of non-canonical Wnt pathways Wnt5a but lack of express the main activators of canonical Wnt signaling as Wnt1, Wnt3a and Wnt8. The presence of the Wnt5a receptor FZD2 and FZD5 was also detected in both cells as well as that of FZD3, FZD6 and FDZ7 and the Wnt canonical co-receptors LRP5 and LRP6. On the other hand we found that both inhibitors of canonical and non-canonical Wnt pathways DKK-1 and sFRP-1 were expressed by MSC. Secondly, activation of either canonical or non-canonical Wnt signaling pathway by Wnt3a and Wnt5a treatment respectively was performed in human MSC to evaluate the effect on osteogenic differentiation and the expression of osteoblast related markers (Collagen I, Osteocalcin and Alkaline Phosphatase). We found that Wnt5a treatment but not Wnt3a significantly increased the early osteogenic differentiation and the expression of alkaline phosphatase in MSC. Consistently in a co-culture system with MM cells Wnt5a treatment blunted, at least in part, the inhibitory effect of MM cells on alkaline phosphatase expression by MSC and PreOB. To go further inside, we evaluated in both primary human BM MSC and the human MSC cell line HS-5 the effect of either the activation of non-canonical Wnt signaling by Wnt5a overexpression using a lentivirus vector or the Wnt5a suppression using siRNA. Wnt5a over-expression in MSC induced the activation of Wnt/Ca++ non-canonical pathway as demonstrated by the increase of Wnt5a secretion and phospho-PKC expression detected by westernblot analysis. Consequently to non-canonical Wnt signal activation we found a significant increase of alkaline phosphatase expression by MSC cells as well as of their osteogenic differentiation. Interestingly, analyzing the gene expression profile by microarray, we found that Wnt5a overexpression in MSC also affects the expression of chemokines, inflammatory cytokines and pro-angiogenic molecules. In conclusion our data indicate that activation of non-canonical Wnt signal pathway may represent a potential target in MM microenvironment to counterbalance the inhibitory effect of MM cells on osteogenic differentiation of human MSC.


2011 ◽  
Vol 45 (5) ◽  
pp. 816-822 ◽  
Author(s):  
K. V. Kulikova ◽  
A. V. Posvyatenko ◽  
N. V. Gnuchev ◽  
G. P. Georgiev ◽  
A. V. Kibardin ◽  
...  

2019 ◽  
Vol 17 (1) ◽  
Author(s):  
Disharee Nath ◽  
Xiang Li ◽  
Claudia Mondragon ◽  
Dawn Post ◽  
Ming Chen ◽  
...  

Abstract Background Prostate cancer development involves various mechanisms, which are poorly understood but pointing to epithelial mesenchymal transition (EMT) as the key mechanism in progression to metastatic disease. ABI1, a member of WAVE complex and actin cytoskeleton regulator and adaptor protein, acts as tumor suppressor in prostate cancer but the role of ABI1 in EMT is not clear. Methods To investigate the molecular mechanism by which loss of ABI1 contributes to tumor progression, we disrupted the ABI1 gene in the benign prostate epithelial RWPE-1 cell line and determined its phenotype. Levels of ABI1 expression in prostate organoid tumor cell lines was evaluated by Western blotting and RNA sequencing. ABI1 expression and its association with prostate tumor grade was evaluated in a TMA cohort of 505 patients and metastatic cell lines. Results Low ABI1 expression is associated with biochemical recurrence, metastasis and death (p = 0.038). Moreover, ABI1 expression was significantly decreased in Gleason pattern 5 vs. pattern 4 (p = 0.0025) and 3 (p = 0.0012), indicating an association between low ABI1 expression and highly invasive prostate tumors. Disruption of ABI1 gene in RWPE-1 cell line resulted in gain of an invasive phenotype, which was characterized by a loss of cell-cell adhesion markers and increased migratory ability of RWPE-1 spheroids. Through RNA sequencing and protein expression analysis, we discovered that ABI1 loss leads to activation of non-canonical WNT signaling and EMT pathways, which are rescued by re-expression of ABI1. Furthermore, an increase in STAT3 phosphorylation upon ABI1 inactivation and the evidence of a high-affinity interaction between the FYN SH2 domain and ABI1 pY421 support a model in which ABI1 acts as a gatekeeper of non-canonical WNT-EMT pathway activation downstream of the FZD2 receptor. Conclusions ABI1 controls prostate tumor progression and epithelial plasticity through regulation of EMT-WNT pathway. Here we discovered that ABI1 inhibits EMT through suppressing FYN-STAT3 activation downstream from non-canonical WNT signaling thus providing a novel mechanism of prostate tumor suppression.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3433-3433
Author(s):  
Francesca Morandi ◽  
Sara Tagliaferri ◽  
Sabrina Bonomini ◽  
Mirca Lazzaretti ◽  
Luca Ferrari ◽  
...  

Abstract Osteoblast impairment occurs within myeloma (MM) cell infiltration into the bone marrow (BM). Wnt signaling is involved in the regulation of osteoblast formation. Canonical Wnt signaling pathway is activated by Wnt 1/3a that induce the activation of GSK3/Axin complex leading to the stabilization and nuclear translocation of beta-catenin that in turn activates the transcription system Lef1/TCF. Recently it has been reported that MM cells produce the Wnt inhibitors DKK-1 demonstrating a correlation between its expression and the presence of bone lesions in MM patients. However the effect of MM cells on Wnt signaling cascade in osteoblasts and osteoblast progenitors has not been investigated. To clarify this issue, first we checked DKK-1 production by human myeloma cell lines (HMCLs), purified CD138+ MM cells and BM plasma of MM patients by PCR and ELISA. Following we performed a co-culture system with HMCLs or CD138+ MM cells and either human osteoblast line (HOBIT) and with BM osteoprogenitor cells (PreOB) obtained after differentiation from mesenchymal cells or murine osteoprogenitor cell lines C2C12 and MC3T3. Both DKK-1 positive HMCLs (XG-1 and JJN3) and negative ones (RPMI-8226, OPM-2) have been used in co-culture as well as DKK-1 positive and negative purified CD138+ MM cells. Similarly we tested the effect of BM plasma of MM patients positive and negative for DKK-1 production on both human and murine cells. Wnt signaling in osteoblasts and osteoblast progenitors was evaluated either at mRNA level by specific human and murine Wnt Array kits and by quantitative PCR or at protein one by Western blot analysis for GSK3b/Axin and LEF-1/TCF expression. We evaluated active de-phosphorylated beta-catenin and inactive phosphorilated one by westernblot and by ELISA in cytosolic and nuclear extracts. DKK-1 median levels detected in the conditioned media of XG-1 and JJN3, MM cells and in BM plasma of DKK-1 positve MM patients were 0.60 ng/mL and 0.38 and 8.84 (range: 1.55–91) ng/mL respectively. Any significant inhibitory effect on WNT signaling and active beta-catenin expression and levels was not observed in HOBIT and human PreOB after co-culture with both HMCLs and MM cells or BM plasma independently to DKK-1 expression. On the contrary DKK-1 positive MM cells or BM plasma suppressed active beta-catenin expression in murine osteoprogenitor cell lines in presence of BMP-2. Consistently Wnt3a stimulation as well as anti-DKK-1 abs. did not restore the inhibitory effects on osteoblast formation and differentiation induced by MM cells in human PreOB. Consistently any significant difference was not detected on beta-catenin expression by stromal/osteoblastic cells on bone biopsies by immunohistochemistry between osteolytic (n°=10) and non-osteolytic (N°=10) MM patients. The different behavior between human and murine osteoblastic cells was further investigated. We found that both cells expressed significant levels of active beta-catenin however DKK-1 suppressed active nuclear and cytosol beta-catenin at concentration of 20–30 ng/mL in C2C12 and MC3T3 whereas only DKK-1 concentrations higher to 500 ng/mL are able to inhibited beta-catenin in HOBIT and human PreOB as well as osteoblast formation and differentiation in human BM cultures. In conclusion our data indicate that MM cells block canonical Wnt signaling in murine osteoblastic cells but not in human osteoblasts and osteoblast progenitors. Beta-catenin independent mechanisms could be involved in DKK-1 mediated bone destruction in MM patients.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2398-2398
Author(s):  
Elena K Siapati ◽  
Magda Papadaki ◽  
Zoi Kozaou ◽  
Erasmia Rouka ◽  
Evridiki Michali ◽  
...  

Abstract Abstract 2398 Poster Board II-375 B-catenin is the central effector molecule of the canonical wnt signaling pathway which governs cell fate and differentiation during embryogenesis as well as self-renewal of hematopoietic stem cells. Deregulation of the pathway has been observed in various malignancies including myeloid leukemias where over-expression of β-catenin is an independent adverse prognostic factor. In the present study we examined the functional outcome of stable β-catenin down-regulation through lentivirus-mediated expression of short hairpin RNA (shRNA). Reduction of the β-catenin levels in AML cell lines and patient samples diminished their in vitro proliferation ability without significantly affecting cell viability. In order to study the role of β-catenin in vivo, we transplanted leukemic cell lines with control or reduced levels of β-catenin in NOD/SCID animals and analyzed the engraftment levels in the bone marrow. We observed that while the immediate homing of the cells was not affected by the β-catenin levels, the bone marrow engraftment was directly dependent on its levels. Subsequent examination of bone marrow sections revealed that the reduced engraftment was partly due to the inability of the cells with lower β-catenin levels to dock to the endosteal niches, a finding that was confirmed in competitive repopulation assays with untransduced cells. When we examined the expression levels of adhesion molecules and integrins in engrafted cells in vivo, we observed a significant down-regulation of CD44 expression, a molecule that participates in the interaction of HSCs with the niche. Gene expression analysis of the components of the wnt signaling pathway showed that the pathway is subject to tight transcriptional regulation with minor expression deviations. We did, however, observe an up-regulation in components that participate in the non-canonical wnt signaling pathways such as the WNT5B ligand. Ongoing experiments in normal cord blood CD34+ cells will determine the in vivo role of β-catenin signaling in normal hematopoietic progenitors. In conclusion, our study showed that β-catenin comprises an integral part in the development and progression of AML in vivo, indicating that manipulation of the wnt pathway may hold a therapeutic potential in the management of AML. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1851-1851
Author(s):  
Ya-Wei Qiang ◽  
Bo Hu ◽  
Yu Chen ◽  
Wei Qiang ◽  
Christoph Heuck ◽  
...  

Abstract Abstract 1851 Background: The proteasome inhibitor Bortezomib (Bz) shows significant activity in Multiple Myeloma (MM) by acting on MM cell directly as well as by augmenting bone formation in vitro and in vivo. Its effect on the bone could be traced to promoting differentiation of mesenchymal stem cells into osteoblast cells by regulating BMP2 and canonical Wnt signaling. However, the molecular mechanism mediating the direct anti-MM activity of Bz remains to be fully understood. Initially the rationale for the use of Bz in MM was inhibition of NF-kB signaling, yet subsequent studies showed that Bz actually induces activation of this pathway. In this study, we examined whether Bz regulates the activity of canonical Wnt signaling pathway in MM and whether the growth-inhibition effect of Bz was associated with activation of this pathway by using multiple MM cell lines including EJM, H929, INA6, KMS28BM, JJN3, L363, OPM1, OPM2, RPMI8226, UTMC, XG2 and XG6 as well as primary plasma cells (PC) from six patients with newly diagnosed MM. Methods/Results: Immunoblotting demonstrated that Bz induces stabilization of b-catenin protein in three MM cell lines (H929, OPM2 and UTMC) in a time- and dose-dependent manner. These changes were not seen when the same cell lysate were immunoblotted for other catenin family members, a-catenin and g-catenin. Increased levels of b-catenin protein response to Bz treatment were observed in other 9 MM cell lines (EJM, INA6, KMS28BM, JJN3, L363, OPM1, RPMI8226, XG2 and XG6) and in the 6 CD138+ sorted bone marrow PC from patients with MM. To determine if Bz regulation of b-catenin level is a specific effect of the inhibition of 26S proteasome subunit we treated the same MM cell lines with another proteasome inhibitor, MG132. Similar results were observed in response to MG132 for all four MM cell lines, suggesting the effect of Bz on b-catenin protein is 26S proteosome inhibitor specific. Increases in b-catenin protein levels in MM cells were not due to increased Ctnnb1/CTNNB (b-catenin) gene transcription as b-catenin mRNA did not change in these cells treated with Bz. These results indicate that proteasome inhibition increases b-catenin is independent of transcriptional upregulation. To determine whether Bz induces the nuclear localization and transcriptional activity of b-catenin, cells were incubated with Bz for 6 hours and then fractionated to separate the nuclear and cytoplasmic fractions. Treatment with Bz resulted an increase in nuclear b-catenin as well as b-catenin in cytoplasm in four cell lines including H929, INA6, OPM1 and MM144. Increase in cytoplasmic and nuclear b-catenin was further confirmed by immunofluorescence with antibodies specific for active form of b-catenin. To determine whether Bz affects b-catenin-mediated transcriptional activity, we used a TCF/LEF luciferase reporter construct cloned in lentiviral vector. OPM2 cells were infected with lentiviral particle containing the TCF reporter or containing empty vector and were then treated with serial concentrations of Bz. The luciferase activity exhibited a dose-dependent response to Bz analogous to the stabilization of b-catenin. Similar results were observed in 7 out of 8 MM cell lines compared with untreated control. Stimulation of TCF transcriptional activity by Bz was independent of modifiers of extracellular Wnt ligands, such as Frizzled receptors, LRP5/6 co-receptors and sFRPs or the activation of intracellular GSK3b. Conclusion: These results indicate that Bz augments activation of canonical Wnt signaling by preventing b-catenin protein from proteosome-mediated degradation in MM cells. Concentrations of Bz for stimulating TCF transcriptional activity are comparable to those being used to induce inhibition of MM proliferation. Experiments modulating cytoplasmic as well as the nuclear players and interactions of the Wnt-pathway are ongoing to determine if Bz mediated activation of b-catenin signaling is responsible for its direct anti-MM effect. Disclosures: Barlogie: Celgene, Genzyme, Novartis, Millennium: Consultancy, Honoraria, Patents & Royalties. Shaughnessy:Myeloma Health, Celgene, Genzyme, Novartis: Consultancy, Employment, Equity Ownership, Honoraria, Patents & Royalties.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2951-2951
Author(s):  
Eileen R Grigson ◽  
Alexandra Pisklakova ◽  
Brittany Weissman ◽  
Yulia Nefedova

Abstract Abstract 2951 Multiple Myeloma (MM) is characterized by clonal expansion of malignant plasma cells, which preferentially reside in the bone marrow (BM). It has been previously shown that canonical Wnt signaling plays a critical role in proliferation and differentiation in MM. ICG-001 is a selective Wnt/ß-catenin signaling inhibitor that binds to the amino terminus of CREB, thus preventing ß-catenin/TCF signaling in the nucleus. Here we investigated whether the pharmacological inhibition of canonical Wnt signaling with ICG-001 could affect viability of MM cells and produce anti-myeloma effect. C-myc and cyclin D1 are known to be downstream target genes of canonical Wnt signaling. MM cells lines treated with ICG-001 revealed down regulation of c-myc and cyclin D1 as compared to control. To investigate the varying sensitivity of cell lines to ICG-001, four cells lines were used: RPMI-8226, NCI-H929, U266, and MM1S. MM cells lines were treated with ICG-001 (1uM – 40uM) and data was collected using MTT assay or flow cytometry (Annexin binding assay). ICG-001 significantly reduced viability in all four cell lines due to induction of apoptosis, with RPMI-8226 showing the greatest sensitivity. We performed cleaved-caspase assays and demonstrated that apoptosis was caspase-3 dependent. We have also evaluated the effect of ICG-001 in BM samples from patients with MM. BM cells were treated with ICG-001 for 24 hours and the level of cleaved-caspase-3 was evaluated by flow cytometry. MM cells were defined as double positive CD138+ and Lambda/Kappa+. Our results demonstrated that MM cells were sensitive to ICG-001, while this drug was not able to induce cell death in non-tumor BM cells. To test the effect of ICG-001 in vivo we injected SCID/Beige mice with 10 × 105 8226 cells in 100 μL phosphate-buffered saline (PBS). After 3 weeks when a tumor was measurable, the mice were split into two groups and were treated with either ICG-001 (100 mg/kg) or vehicle control (PBS) daily, for 14 days. Tumor size was consistently monitored during treatment and 3 weeks after treatment. We observed that ICG-001 produced a significant antitumor effect as compared to control group. These data indicate targeting Wnt/ß-catenin Pathway with ICG-001 could be therapeutically beneficial to patients with MM. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3010-3010
Author(s):  
Raphael Koch ◽  
Martin Demant ◽  
Thiha Aung ◽  
Annemarie Guentsch ◽  
Nina Diering ◽  
...  

Abstract Introduction Patients with aggressive B-cell lymphoma are treated in curative intention. However, some patients experience fatal relapse, originating from refractory lymphoma cells with the capacity for clonogenic regrowth. We here addressed repopulation capacity of lymphoma cell subpopulations and the mechanisms regulating the populational composition in the growing tumor. Material & Methods We identified side population (SP) cells in diffuse large B-cell lymphoma cell lines and patient samples with the DNA-binding dye Hoechst33342, analyzed clonogenicity in vitro and in vivo and screened for differentially expressed genes and DNA-methylation patterns. A GFP-containing lentiviral vector construct was used to keep track of side population cells cultured among mixed cultures of SP and nonSP cells. Manipulation of canonical wnt-signaling was performed by lentiviral sh-RNA constructs as well as pharmacological tankyrase-inhibition by XAV-939. In vitro data were supported by in vivo experiments using a chorioallantoic membrane-assay. Results Colony assays and suspension cultures of sorted SP and nonSP cells revealed restriction of clonogenic potential to the SP cell population as well as resurgence of nonSP cells from purified SP cell progenitors, while mixed culture assays using a GFP-vector construct tracing the SP vs. nonSP-population revealed homeostasis between the two populations, showing both SP and nonSP cells contributing to either cell compartment. SP cells show enhanced canonical wnt-signaling and increased exosomal secretion of wnt3a. Suppression of canonical wnt-signaling resulted in reduced clonogenicity. Exosome stimulation of DLBCL cell lines resulted in increased clonogenicity, stabilization of beta catenin and enhanced TOP/FOP activity. Conclusion Here we show that tumor cells reversibly switch between states of autonomous and non-autonomous clonogenicity, and that such transitions are regulated by exosome-mediated wnt signaling. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 741-741
Author(s):  
Marina Bolzoni ◽  
Simona Colla ◽  
Paola Storti ◽  
Gaetano Donofrio ◽  
Manuela Abeltino ◽  
...  

Abstract Abstract 741 Osteogenic differentiation of human mesenchymal stem cells (hMSC) is typically impaired in multiple myeloma (MM) patients leading to osteoblast deficiency. Canonical Wnt signal pathway is critical in the regulation of bone formation process and its activation in osteoblastic cells improves bone mass reducing the development of osteolytic lesions in MM mouse model. Together to canonical Wnt signaling, a non-canonical Wnt pathway, independently to β-catenin activation, has been identified. Non-canonical Wnt signaling is transduced through FZD receptor and Ror2 co-receptor to several cascades either disheveled pathways involving Rho family small GTPase and JNK or Ca++ dependent pathways involving the nuclear factor of activated T cells (NFAT). Interestingly, recent evidences suggest that non-canonical Wnt pathway activation by Wnt5a or Wnt4, rather than canonical one by Wnt3a, stimulates the osteogenic properties of hMSC through Ror2 activation. The effect of MM cells on non-canonical Wnt pathway as well as the role of the activation of this pathway in hMSC on the osteogenic differentiation impairment induced by MM cells are not known and have been investigated in the present study. First we checked the expression of non-canonical Wnt related molecules by bone marrow (BM) hMSC and osteoprogenitor cells (PreOB) at the first passage by oligonucleotide arrays. We found that both cells expressed the activator of non-canonical Wnt pathways Wnt5a but lack of express the main activators of canonical Wnt signaling as Wnt1, Wnt3a and Wnt8. The presence of the Wnt5a receptor FZD2 and FZD5 was also detected in both cells as well as of Ror2. Interestingly we found that osteogenic differentiation of hMSC towards preOB significantly increased Ror2 but not Wnt5a expression. Secondly, we performed a series of co-culture between PreOB and MM cells using either the human myeloma cell lines (JJN3, XG-1, XG-6, KMS12, KMS27) or purified CD138+ cells obtained from MM patients finding that MM cells inhibit Ror2 protein expression by PreOB and consistently the activity of NFATc1 at nuclear level. Following activation of non-canonical Wnt signaling pathway either by Wnt5a treatment or by the induction of both Wnt5a and Ror2 overexpression by lentivirus vectors have been performed in primary hMSC obtained from MM patients. The efficiency of Wnt5a and Ror2 trasduction was checked by GFP expression using flow cytometry whereas the efficacy was evaluated by the level of Wnt5a and Ror2 mRNA and protein expression as well as by the intracytoplasmatic increase of Ca++ influx, phospho-PKC expression and NFATc1 activity. We found that Wnt5a treatment as well as Wnt5a or Ror2 overexpression significantly increased osteogenic differentiation and the expression of alkaline phosphatase in hMSC. Consistently, in the co-culture system with MM cells, Wnt5a and Ror2 overexpression by hMSC blunted the inhibitory effect of MM cells on alkaline phosphatase expression and osteogenic differentiation. Finally, these observations were further confirmed showing that Wnt5a or Ror2 silencing in PreOB by siRNA or shRNA trasfection, respectively inhibited the expression of osteogenic markers alkaline phosphatase, osteocalcin and collagen I. In conclusion our data indicate that activation of non-canonical Wnt5a/Ror2/Ca++ signal pathway in hMSC increases osteogenic differentiation and counterbalance the inhibitory effect of MM cell suggesting that this pathway could represent a potential target in MM microenvironment. Disclosures: No relevant conflicts of interest to declare.


2008 ◽  
Vol 87 (2) ◽  
pp. 126-130 ◽  
Author(s):  
E.L. Scheller ◽  
J. Chang ◽  
C.Y. Wang

Dental pulp stem cells (DPSCs) are a unique precursor population isolated from postnatal human dental pulp and have the ability to regenerate a reparative dentin-like complex. Canonical Wnt signaling plays a critical role in tooth development and stem cell self-renewal through β-catenin. In this study, the regulation of odontoblast-like differentiation of DPSCs by canonical Wnt signaling was examined. DPSCs were stably transduced with canonical Wnt-1 or the active form of β-catenin, with retrovirus-mediated infection. Northern blot analysis found that Wnt-1 strongly induced the expression of matricellular protein osteopontin, and modestly enhanced the expression of type I collagen in DPSCs. Unexpectedly, Wnt-1 inhibited alkaline phosphatase (ALP) activity and the formation of mineralized nodules in DPSCs. Moreover, over-expression of β-catenin was also sufficient to suppress the differentiation and mineralization of DPSCs. In conclusion, our results suggest that canonical Wnt signaling negatively regulates the odontoblast-like differentiation of DPSCs. Abbreviations used: DPSC, dental pulp stem cell; ALP, alkaline phosphatase; BSP, bone sialoprotein; MSC, mesenchymal stem cell; β-GP, β-glycerophosphate; APC, adenomatous polyposis coli; GSK-3β, glycogen synthase kinase-3β; LRP, LDL receptor-related protein; Tcf, T-cell factor; LEF, lymphoid enhancer factor; FCS, fetal calf serum; AA, L-ascorbic acid 2-phosphate; α-MEM, α-modified Eagle’s medium; PBS, phosphate-buffered saline; HA, hemagglutinin; ON, osteonectin; OPN, osteopontin.


Sign in / Sign up

Export Citation Format

Share Document