• Title/Summary/Keyword: Wnt3a

Search Result 125, Processing Time 0.025 seconds

Differential Wnt11 Expression Related to Wnt5a in High- and Low-grade Serous Ovarian Cancer: Implications for Migration, Adhesion and Survival

  • Jannesari-Ladani, Farnaz;Hossein, Ghamartaj;Izadi-Mood, Narges
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.15 no.3
    • /
    • pp.1489-1495
    • /
    • 2014
  • Wnt is a powerful signaling pathway that plays a crucial role in cell fate determination, survival, proliferation and motility during development, in adult tissues and cancer. The aims of the present study were three fold: i) to assess Wnt11 immunoexpression and its possible relationship with Wnt5a in high- and low-grade human serous ovarian cancer (HGSC and LGSC) specimens; ii) to assess Wnt11 expression levels in Wnt5a overexpressing SKOV-3 cells; iii) to reveal the role of Wnt11 in viability, adhesion, migration and invasion of SKOV-3 cells using recombinant human Wnt11 (rhWnt11). Immunohistochemistry revealed a significant difference in Wnt11 expression between HGSC and LGSC groups (p=0.001). Moreover, a positive correlation was observed between Wnt5a and Wnt11 expression in the HGSC (r=0.713, p=0.001), but not the LGSC group. The expression of Wnt11 was decreased by 35% in Wnt5a overexpressing cells (SKOV-3/Wnt5a) compared to mock controls. Similarly Wnt11 expression levels were decreased by 47% in the presence of exogenous Wnt5a compared to untreated cells. In the presence of rhWnt11, 31% increased cell viability (p<0.001) and 21% increased cell adhesion to matrigel (p<0.01) were observed compared to control. Cell migration was increased by 1.6-fold with rhWnt11 as revealed by transwell migration assay (p<0.001). However, 45% decreased cell invasion was observed in the presence of rhWnt11 compared to control (p<0.01). Our results may suggest that differential Wnt11 immunoexpression in HGSC compared to LGSC could play important roles in serous ovarian cancer progression and may be modulated by Wnt5a expression levels.

Wnt5a attenuates the pathogenic effects of the Wnt/β-catenin pathway in human retinal pigment epithelial cells via down-regulating β-catenin and Snail

  • Kim, Joo-Hyun;Park, Seoyoung;Chung, Hyewon;Oh, Sangtaek
    • BMB Reports
    • /
    • v.48 no.9
    • /
    • pp.525-530
    • /
    • 2015
  • Activation of the Wnt/β-catenin pathway plays a pathogenic role in age-related macular degeneration (AMD) and is thus a potential target for the development of therapeutics for this disease. Here, we demonstrated that Wnt5a antagonized β-catenin response transcription (CRT) induced with Wnt3a by promoting β-catenin phosphorylation at Ser33/Ser37/Thr41 and its subsequent degradation in human retinal pigment epithelial (RPE) cells. Wnt5a decreased the levels of vascular endothelial growth factor (VEGF), tumor necrosis factor-α(TNF-α), and nuclear factor-κB (NF-κB), which was up-regulated by Wnt3a. Furthermore, Wnt5a increased E-cadherin expression and decreased cell migration by down-regulating Snail expression, thereby abrogating the Wnt3a-induced epithelial-mesenchymal transition (EMT) in human RPE cells. Our findings suggest that Wnt5a suppresses the pathogenic effects of canonical Wnt signaling in human RPE cells by promoting β-catenin phosphorylation and degradation. Therefore, Wnt5a has significant therapeutic potential for the treatment of AMD. [BMB Reports 2015; 48(9): 525-530]

Silybin Synergizes with Wnt3a in Activation of the Wnt/${\beta}$-catenin Signaling Pathway through Stabilization of Intracellular ${\beta}$-Catenin Protein (Silybin에 의한 Wnt/${\beta}$-catenin 신호전달체계의 활성화)

  • Kim, Tae-Yeoun;Oh, Sang-Taek
    • Microbiology and Biotechnology Letters
    • /
    • v.40 no.1
    • /
    • pp.50-56
    • /
    • 2012
  • The Wnt/${\beta}$-catenin signaling pathway regulates diverse developmental processes and adult tissue homeostasis. Inappropriate regulation of this pathway has been associated with human diseases, such as cancers, osteoporosis, and Alzheimer's disease. Using a cell-based chemical screening with natural compounds, we discovered silybin, a plant flavonoid isolated from the Silybum marianum, which activated the Wnt/${\beta}$-catenin signaling pathway in a synergy with Wnt3a-conditioned medium (Wnt3a-CM). In the presence of Wnt3a-CM, silybin up-regulated ${\beta}$-catenin response transcription (CRT) in HEK293-FL reporter cells and 3T3-L1 preadipocytes through stabilization of intracellular ${\beta}$-catenin protein. Silybin and Wnt3a-CM synergistically reduced expression of important adipocyte marker genes including peroxisome-proliferator-activated $receptor{\gamma}$ ($PPAR{\gamma}$) and CAATT enhancer-binding protein ${\alpha}$ (C/$EBP{\alpha}$) in 3T3-L1 preadipocytes, accompanied by the activation of Wnt/${\beta}$-catenin signaling pathway. Taken together, our findings indicate that silybin is a small-molecule synergist of the Wnt/${\beta}$-catenin signaling pathway and can be used as a controllable reagent for investigating biological processes that involve the Wnt/${\beta}$-catenin signaling pathway.

Optimal Ratio of Wnt3a Expression in Human Mesenchymal Stem Cells Promotes Axonal Regeneration in Spinal Cord Injured Rat Model

  • Yoon, Hyung Ho;Lee, Hyang Ju;Min, Joongkee;Kim, Jeong Hoon;Park, Jin Hoon;Kim, Ji Hyun;Kim, Seong Who;Lee, Heuiran;Jeon, Sang Ryong
    • Journal of Korean Neurosurgical Society
    • /
    • v.64 no.5
    • /
    • pp.705-715
    • /
    • 2021
  • Objective : Through our previous clinical trials, the demonstrated therapeutic effects of MSC in chronic spinal cord injury (SCI) were found to be not sufficient. Therefore, the need to develop stem cell agent with enhanced efficacy is increased. We transplanted enhanced Wnt3-asecreting human mesenchymal stem cells (hMSC) into injured spines at 6 weeks after SCI to improve axonal regeneration in a rat model of chronic SCI. We hypothesized that enhanced Wnt3a protein expression could augment neuro-regeneration after SCI. Methods : Thirty-six Sprague-Dawley rats were injured using an Infinite Horizon (IH) impactor at the T9-10 vertebrae and separated into five groups : 1) phosphate-buffered saline injection (injury only group, n=7); 2) hMSC transplantation (MSC, n=7); 3) hMSC transfected with pLenti vector (without Wnt3a gene) transplantation (pLenti-MSC, n=7); 4) hMSC transfected with Wnt3a gene transplantation (Wnt3a-MSC, n=7); and 5) hMSC transfected with enhanced Wnt3a gene (1.7 fold Wnt3a mRNA expression) transplantation (1.7 Wnt3a-MSC, n=8). Six weeks after SCI, each 5×105 cells/15 µL at 2 points were injected using stereotactic and microsyringe pump. To evaluate functional recovery from SCI, rats underwent Basso-Beattie-Bresnahan (BBB) locomotor test on the first, second, and third days post-injury and then weekly for 14 weeks. Axonal regeneration was assessed using growth-associated protein 43 (GAP43), microtubule-associated protein 2 (MAP2), and neurofilament (NF) immunostaining. Results : Fourteen weeks after injury (8 weeks after transplantation), BBB score of the 1.7 Wnt3a-MSC group (15.0±0.28) was significantly higher than that of the injury only (10.0±0.48), MSC (12.57±0.48), pLenti-MSC (12.42±0.48), and Wnt3a-MSC (13.71±0.61) groups (p<0.05). Immunostaining revealed increased expression of axonal regeneration markers GAP43, MAP2, and NF in the Wnt3a-MSC and 1.7 Wnt3a-MSC groups. Conclusion : Our results showed that enhanced gene expression of Wnt3a in hMSC can potentiate axonal regeneration and improve functional recovery in a rat model of chronic SCI.

Opisthorchis viverrini Infection Activates the PI3K/AKT/PTEN and Wnt/β-catenin Signaling Pathways in a Cholangiocarcinogenesis Model

  • Yothaisong, Supak;Thanee, Malinee;Namwat, Nisana;Yongvanit, Puangrat;Boonmars, Thidarut;Puapairoj, Anucha;Loilome, Watcharin
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.15 no.23
    • /
    • pp.10463-10468
    • /
    • 2015
  • Opisthorchis viverrini (Ov) infection is the major etiological factor for cholangiocarcinoma (CCA), especially in northeast Thailand. We have previously reported significant involvement of PI3K/AKT/PTEN and $Wnt/{\beta}$-catenin in human CCA tissues. The present study, therefore, examined the expression and activation of PI3K/AKT/PTEN and $Wnt/{\beta}$-catenin signaling components during Ov-induced cholangiocarcinogenesis in a hamster animal model. Hamsters were divided into two groups; non-treated and Ov plus NDMA treated. The results of immunohistochemical staining showed an upregulation of PI3K/AKT signaling as determined by elevated expression of the $p85{\alpha}$-regulatory and $p110{\alpha}$-catalytic subunits of PI3K as well as increased expression and activation of AKT during cholangiocarcinogenesis. Interestingly, the staining intensity of activated AKT (p-AKT) increased in the apical regions of the bile ducts and strong staining was detected where the liver fluke resides. Moreover, PTEN, a negative regulator of PI3K/AKT, was suppressed by decreased expression and increased phosphorylation during cholangiocarcinogenesis. We also detected upregulation of $Wnt/{\beta}$-catenin signaling as determined by increased positive staining of Wnt3, Wnt3a, Wnt5a, Wnt7b and ${\beta}$-catenin, corresponded with the period of cholangiocarcinogenesis. Furthermore, nuclear staining of ${\beta}$-catenin was observed in CCA tissues. Our results suggest the liver fluke infection causes chronic inflammatory conditions which lead to upregulation of the PI3K/AKT and $Wnt/{\beta}$-catenin signaling pathways which may drive CCA carcinogenesis. These results provide useful information for drug development, prevention and treatment of CCA.

Suppression of the Wnt/${\beta}$-catenin Pathway by Bryostatin-1 (Bryostatin-1에 의한 Wnt/${\beta}$-Catenin 신호전달체계 저해효과)

  • Park, Seoyoung;Oh, Sangtaek
    • Microbiology and Biotechnology Letters
    • /
    • v.42 no.1
    • /
    • pp.89-92
    • /
    • 2014
  • The Wnt/${\beta}$-catenin pathway plays important roles in a variety of biological processes, such as cell proliferation, differentiation, and organ development. Here, we used a cell-based reporter assay to identify bryostatin-1, a natural macrocyclic lactone, as an inhibitor of the Wnt/${\beta}$-catenin pathway. Bryostatin-1 suppressed ${\beta}$-catenin response transcription (CRT), which was activated by a Wnt3a-conditioned medium (Wnt3a-CM), through a decrease in the intracellular ${\beta}$-catenin protein levels, without affecting its mRNA level. In addition, pharmacological inhibition of proteasome abrogated bryostatin-1-mediated down-regulation of the ${\beta}$-catenin protein level. Our findings suggest that bryostatin-1 attenuates the Wnt/${\beta}$-catenin pathway through the promotion of proteasomal degradation of ${\beta}$-catenin.

Current Status of Research in Wnt Signal Transduction (Wnt 신호 전달 연구의 최신 지견)

  • Kim, Wan-Tae;Cha, Bok-Sik;Jho, Eek-Hoon
    • Development and Reproduction
    • /
    • v.11 no.3
    • /
    • pp.141-153
    • /
    • 2007
  • The Wnt signaling pathway regulates cell proliferation and differentiation during development of multicellular organisms and plays pivotal roles in the maintenance of homeostasis in adult tissues. Therefore misregulation of Wnt signaling could be a pathogenesis of diverse human diseases such as cancers. Recently, the list of diseases that may be linked to the misregulation of Wnt signaling has exploded and more people are getting interested in the way of controlling Wnt signaling. There are a lot of review papers, however, since most of them have focused on specific issues for experts in Wnt signaling it may be difficult for new comers to understand the overall background and current status of Wnt signaling. In this review, we present data and interpretations for the overall processes of Wnt signal transduction to understand the past and current status of Wnt signaling.

  • PDF

The Effects of Wnt Signaling on Neural Crest Lineage Segregation and Specification (Wnt signaling이 neural crest lineage segregation과 specification에 미치는 영향)

  • Song, Jin-Su;Jin, Eun-Jung
    • Journal of Life Science
    • /
    • v.19 no.10
    • /
    • pp.1346-1351
    • /
    • 2009
  • Recent evidence has shown that many pluripotetic neural crest cells are fate-restricted and that different fate-restricted crest cells emigrate from the neural tube at different times. Jin et al. (2001) identified the expression patterns of Wnts and its antagonists at the time that neural crest cells were being specified and suggested that Wnt signaling was involved in the segregation/differentiation of neural crest cells in the trunk in vitro. In this study, we evaluated the effects of Wnt signaling in avian neural crest lineage segregation. To accomplish this, Wnt signaling was disturbed at the time of neural crest segregation and differentiation by grafting Wnt-3a expressing cells and conducting dominant negative glycogen synthase kinase (dnGSK) electroporation. Stimulation of Wnt signaling induced neural crest lineage segregation and melanoblast specification, and increased the expression levels of genes known to be involved in neural crest development such as cadherin 7 and Slug, which suggests that they are involved in Wnt-induced neural crest lineage differentiation into melanoblasts.

RNF152 negatively regulates Wnt/β-catenin signaling in Xenopus embryos

  • Yoon, Gang-Ho;Kim, Kyuhee;Park, Dong-Seok;Choi, Sun-Cheol
    • BMB Reports
    • /
    • v.55 no.5
    • /
    • pp.232-237
    • /
    • 2022
  • The Wnt/β-catenin signaling plays crucial roles in early development, tissue homeostasis, stem cells, and cancers. Here, we show that RNF152, an E3 ligase localized to lysosomes, acts as a negative regulator of the Wnt/β-catenin pathway during Xenopus early embryogenesis. Overexpression of wild-type (WT) RNF152 inhibited XWnt8-induced stabilization of β-catenin, ectopic expression of target genes, and activity of a Wnt-responsive promoter. Likewise, an E3 ligase-defective RNF152 had repressive effects on the Wnt-dependent gene responses but not its truncation mutant lacking the transmembrane domain. Conversely, knockdown of RNF152 further enhanced the transcriptional responses induced by XWnt8. RNF152 morphants exhibited defects in craniofacial structures and pigmentation. In line with this, the gain-of-RNF152 function interfered with the expression of neural crest (NC) markers, whereas its depletion up-regulated NC formation in the early embryo. Mechanistically, RNF152 inhibits the polymerization of Dishevelled, which is key to Wnt signaling, in an E3 ligase-independent manner. Together, these results suggest that RNF152 controls negatively Wnt/β-catenin signaling to fine-tune its activity for NC formation in Xenopus embryo.

Cyclic tensile stress inhibits Wnt/${\beta}$-catenin signaling in human periodontal ligament cells

  • Kim, Ji-Young;Yang, Daum;Kim, Ha-Neui;Jung, Kyoung-Suk;Chang, Young-Il;Lee, Zang-Hee
    • International Journal of Oral Biology
    • /
    • v.34 no.2
    • /
    • pp.53-59
    • /
    • 2009
  • Periodontal ligament (PDL) tissue is a connective tissue that is interposed between the roots of the teeth and the inner wall of the alveolar bone socket. PDL is always exposed to physiologic mechanical force such as masticatory force and PDL cells play important roles during orthodontic tooth movement by synthesizing and secreting different mediators involved in bone remodeling. The Wnt/${\beta}$-catenin signaling pathway was recently shown to play a significant role in the control of bone formation. In the present study, we applied cyclic tensile stress of 20% elongation to cultured human PDL cells and assessed its impact after six days upon components of the Wnt/${\beta}$-catenin signaling pathway. RTPCR analysis showed that Wnt1a, Wnt3a, Wnt10b and the Wnt receptor LRP5 were down-regulated, whereas the Wnt inhibitor DKK1 was up-regulated in response to these stress conditions. In contrast, little change was detected in the mRNA expression of Wnt5a, Wnt7b, Fz1, and LRP6. By western blotting we found decreased expression of the ${\beta}$-catenin and p-GSK-3${\beta}$ proteins. Our results thus show that mechanical stress suppresses the canonical Wnt/${\beta}$-catenin signaling pathway in PDL cells.