• Title/Summary/Keyword: Wnt/${\beta}$-catenin pathway

Search Result 93, Processing Time 0.024 seconds

Increased Expression of MET and RON Receptor Tyrosine Kinases in Canine Cutaneous Melanotic Tumor (개 피부 흑색종의 MET/RON Receptor Tyrosine Kinases 발현 평가)

  • Han, Jae-Ik;Kim, Dae-Yong;Na, Ki-Jeong
    • Journal of Veterinary Clinics
    • /
    • v.26 no.5
    • /
    • pp.429-432
    • /
    • 2009
  • Aberrant translocation of ${\beta}$-catenin can be induced by the dissociation of cadherin-catenin complex, which is mediated by the activation of receptor tyrosine kinases (RTKs). We examined the expression levels of MET/RON RTKs in tissue samples of canine cutaneous melanotic tumor. The activation of MET/RON RTKs was observed in 28% of the examined samples. Our results indicate the possibility that the activated MET/RON RTKs are implicated in the dissociation of cadherin-catenin complex in canine cutaneous melanotic tumor.

Effect and mechanism of docosahexaenoic acid on the proliferation of dermal papilla cells (Docosahexaenoic acid의 모유두세포 증식 효능 및 기전)

  • Ko, Jiyeon;Oh, Il-Joong;Kang, Jung-Il;Choi, Youn Kyung;Yoon, Hoon-Seok;Yoo, Eun-Sook;Ko, Chang-Ik;Ahn, Yong-Seok
    • Journal of Medicine and Life Science
    • /
    • v.16 no.3
    • /
    • pp.84-89
    • /
    • 2019
  • Docosahexaenoic acid (DHA), a principal of mackerel-derived fermented fish oil, increases the proliferation of dermal papilla cells (DPCs) via the upregulation of cell cycle-associated proteins such as cyclin D1 and cdc2 p34, and might promote hair-growth. However, the intracellular mechanisms that underlie the action of DHA in the proliferation of DPCs have not been investigated fully. In this study, we addressed the action mechanisms of DHA to trigger the activation of anagen in DPCs. DHA activated β-catenin signaling by the increased phosphorylation at serine 552 and serine 675 as well as the translocation and accumulation of activated β-catenin into the nucleus. In the other hand, DHA inhibited canonical TGF-β/Smad signaling by the decreased phosphorylation of Smad2/3. Taken together, the results indicate that DHA might stimulate anagen signaling via the activation of Wnt/β-catenin pathway, while the inactivation of canonical TGF-β signaling pathway in DPCs.

The Mechanism of Whole Plant Extract of Viola verecunda on the Proliferation of Dermal Papilla Cells (콩제비꽃 전초 추출물의 모유두세포 증식 기전)

  • Kang, Jung-Il;Seo, Min Jeong;Choi, Youn Kyung;Shin, Su Young;Hwang, Yong;Goh, Jae duk;Yoo, Eun-Sook;Kim, Sang-Cheol;Kang, Hee-Kyoung
    • Korean Journal of Pharmacognosy
    • /
    • v.52 no.1
    • /
    • pp.34-40
    • /
    • 2021
  • Proliferation and maintain of dermal papilla during progression of hair-cycle are crucial to the duration of anagen and regulated by diverse signaling pathway such as PI3K/Akt/Wnt/β-catenin pathway. In this study, we investigated the effects and mechanisms of Viola verecunda on dermal papilla cells. Treatment of dermal papilla cells with whole plant extract of V. verecunda resulted in cell proliferation, which was accompanied by up-regulation of cyclin D1, phospho (ser780)-pRB and cdc2 p34, and down-regulation of p27kip1. V. verecunda extract also promoted the levels of phospho (ser473)-Akt and phospho (ser780)-pRB in a time-dependent manner. Inhibition of PI3K/Akt by Wortmannin suppressed progression of cell-cycle, thereby attenuated the increases in proliferation of dermal papilla cells by V. verecunda extract. We further investigated Wnt/β-catenin pathway with respect to the effects of V. verecunda extract on the proliferation of dermal papilla cells. Treatment with V. verecunda extract results in up-regulation of Wnt/β-catenin proteins such as phospho (ser9)-GSKβ, phospho (ser552)-β-catenin and phospho (ser675)-β-catenin. In addition, Wortmannin abrogated V. verecunda extract mediated up-regulation of cdc2 p34 and down-regulation of p27kip1. These finding reveal that the proliferative effect of V. verecunda mediated by alteration of cell-cycle via activating PI3K/Akt/Wnt pathway in dermal papilla cells.

Ten-eleven translocation 1 mediating DNA demethylation regulates the proliferation of chicken primordial germ cells through the activation of Wnt4/β-catenin signaling pathway

  • Yinglin Lu;Ming Li;Heng Cao;Jing Zhou;Fan Li;Debing Yu;Minli Yu
    • Animal Bioscience
    • /
    • v.37 no.3
    • /
    • pp.471-480
    • /
    • 2024
  • Objective: The objective of this study was to investigate the regulation relationship of Ten-eleven translocation 1 (Tet1) in DNA demethylation and the proliferation of primordial germ cells (PGCs) in chickens. Methods: siRNA targeting Tet1 was used to transiently knockdown the expression of Tet1 in chicken PGCs, and the genomic DNA methylation status was measured. The proliferation of chicken PGCs was detected by flow cytometry analysis and cell counting kit-8 assay when activation or inhibition of Wnt4/β-catenin signaling pathway. And the level of DNA methylation and hisotne methylation was also tested. Results: Results revealed that knockdown of Tet1 inhibited the proliferation of chicken PGCs and downregulated the mRNA expression of Cyclin D1 and cyclin-dependent kinase 6 (CDK6), as well as pluripotency-associated genes (Nanog, PouV, and Sox2). Flow cytometry analysis confirmed that the population of PGCs in Tet1 knockdown group displayed a significant decrease in the proportion of S and G2 phase cells, which meant that there were less PGCs entered the mitosis process than that of control. Furthermore, Tet1 knockdown delayed the entrance to G1/S phase and this inhibition was rescued by treated with BIO. Consistent with these findings, Wnt/β-catenin signaling was inactivated in Tet1 knockdown PGCs, leading to aberrant proliferation. Further analysis showed that the methylation of the whole genome increased significantly after Tet1 downregulation, while hydroxyl-methylation obviously declined. Meanwhile, the level of H3K27me3 was upregulated and H3K9me2 was downregulated in Tet1 knockdown PGCs, which was achieved by regulating Wnt/β-catenin signaling pathway. Conclusion: These results suggested that the self-renewal of chicken PGCs and the maintenance of their characteristics were regulated by Tet1 mediating DNA demethylation through the activation of Wnt4/β-catenin signaling pathway.

Parkin-induced Decrease of ${\beta}$-catenin is Mediated by Protein Kinase C in TNF-${\alpha}$-treated HeLa Cells

  • Lee, Min Ho;Jung, Byung Chul;Kim, Sung Hoon;Lee, Juyeon;Jung, Dongju;Cho, Jang-Eun;Rhee, Ki-Jong;Kim, Yoon Suk
    • Biomedical Science Letters
    • /
    • v.19 no.2
    • /
    • pp.83-89
    • /
    • 2013
  • Parkin is a protein known to have tumor suppressive functions. In a previous study, we determined that Parkin expression restores susceptibility to TNF-${\alpha}$-induced death in HeLa cells. ${\beta}$-catenin is a key protein in the Wnt signaling pathway and excessive activation of the ${\beta}$-catenin pathway can promote cancer development. In this study, we found that ${\beta}$-catenin levels decreased dramatically in Parkin over-expressing HeLa cells treated with TNF-${\alpha}$. We used chemical inhibitors of cell signaling pathways to identify the signaling molecules involved in ${\beta}$-catenin down-regulation. Our results indicate that the PKC inhibitor (RO-31-7549) blocked parkin-induced down-regulation of ${\beta}$-catenin. We also show that Parkin-induced decrease in cell viability in TNF-${\alpha}$-treated HeLa cells is alleviated upon treatment with a PKC inhibitor. Taken together, these results suggest the possibility that ${\beta}$-catenin reduction may be associated with Parkin-induced decrease of cell viability in TNF-${\alpha}$ treated HeLa cells.

Regulatory Effect of Cannabidiol (CBD) on Decreased β-Catenin Expression in Alopecia Models by Testosterone and PMA Treatment in Dermal Papilla Cells

  • Park, Yoon-Jong;Ryu, Jae-Min;Na, Han-Heom;Jung, Hyun-Suk;Kim, Bokhye;Park, Jin-Sung;Ahn, Byung-Soo;Kim, Keun-Cheol
    • Journal of Pharmacopuncture
    • /
    • v.24 no.2
    • /
    • pp.68-75
    • /
    • 2021
  • Objectives: The hair follicle is composed of more than 20 kinds of cells, and mesoderm derived dermal papilla cells and keratinocytes cooperatively contribute hair growth via Wnt/β-catenin signaling pathway. We are to investigate β-catenin expression and regulatory mechanism by CBD in alopecia hair tissues and dermal papilla cells. Methods: We performed structural and anatomical analyses on alopecia patients derived hair tissues using microscopes. Pharmacological effect of CBD was evaluated by β-catenin expression using RT-PCR and immunostaining experiment. Results: Morphological deformation and loss of cell numbers in hair shaft were observed in alopecia hair tissues. IHC experiment showed that loss of β-catenin expression was shown in inner shaft of the alopecia hair tissues, indicating that β-catenin expression is a key regulatory function during alopecia progression. Consistently, β-catenin expression was decreased in testosterone or PMA treated dermal papilla cells, suggesting that those treatments are referred as a model on molecular mechanism of alopecia using dermal papilla cells. RT-PCR and immunostaining experiments showed that β-catenin expression was decreased in RNA level, as well as decreased β-catenin protein might be resulted from ubiquitination. However, CBD treatment has no changes in gene expression including β-catenin, but the decreased β-catenin expression by testosterone or PMA was restored by CBD pretreatment, suggesting that potential regulatory effect on alopecia induction of testosterone and PMA. Conclusion: CBD might have a modulating function on alopecia caused by hormonal or excess of signaling pathway, and be a promising application for on alopecia treatment.

Dishevelling Wnt and Hippo

  • Kim, Nam Hee;Lee, Yoonmi;Yook, Jong In
    • BMB Reports
    • /
    • v.51 no.9
    • /
    • pp.425-426
    • /
    • 2018
  • As highly conserved signaling cascades of multicellular organisms, Wnt and Hippo pathways control a wide range of cellular activities, including cell adhesion, fate determination, cell cycle, motility, polarity, and metabolism. Dysregulation of those pathways are implicated in many human diseases, including cancer. Similarly to ${\beta}-catenin$ in the Wnt pathway, the YAP transcription co-activator is a major player in Hippo. Although the intracellular dynamics of YAP are well-known to largely depend on phosphorylation by LATS and AMPK kinases, the molecular effector of YAP cytosolic translocation remains unidentified. Recently, we reported that the Dishevelled (DVL), a key scaffolding protein between canonical and non-canonical Wnt pathway, is responsible for nuclear export of phosphorylated YAP. The DVL is also required for YAP intracellular trafficking induced by E-cadherin, ${\alpha}-catenin$, or metabolic stress. Note that the p53/LATS2 and LKB1/AMPK tumor suppressor axes, commonly inactivated in human cancer, govern the reciprocal inhibition between DVL and YAP. Conversely, loss of the tumor suppressor allows co-activation of YAP and Wnt independent of epithelial polarity or contact inhibition in human cancer. These observations provide novel mechanistic insight into (1) a tight molecular connection merging the Wnt and Hippo pathways, and (2) the importance of tumor suppressor contexts with respect to controlled proliferation and epithelial polarity regulated by cell adhesion.

Secondary Structure, 1H, 13C and 15N Resonance Assignments and Molecular Interactions of the Dishevelled DIX Domain

  • Capelluto, Daniel G.S.;Overduin, Michael
    • BMB Reports
    • /
    • v.38 no.2
    • /
    • pp.243-247
    • /
    • 2005
  • Dishevelled (Dvl) is a positive regulator of the canonical Wnt signaling pathway, which regulates the levels of $\beta$-catenin. The $\beta$-catenin oncoprotein depends upon the association of Dvl and Axin proteins through their DIX domains, and its accumulation directs the expression of specific developmental-related genes at the nucleus. Here, the $^1H$, $^{13}C$, and $^{15}N$ resonances of the human Dishevelled 2 DIX domain are assigned using heteronuclear nuclear magnetic resonance (NMR) spectroscopy. In addition, helical and extended elements are identified based on the NMR data. The results establish a structural context for characterizing the actin and phospholipid interactions and binding sites of this novel domain, and provide insights into its role in protein localization to stress fibers and cytoplasmic vesicles during Wnt signaling.

A small molecule approach to degrade RAS with EGFR repression is a potential therapy for KRAS mutation-driven colorectal cancer resistance to cetuximab

  • Lee, Sang-Kyu;Cho, Yong-Hee;Cha, Pu-Hyeon;Yoon, Jeong-Soo;Ro, Eun Ji;Jeong, Woo-Jeong;Park, Jieun;Kim, Hyuntae;Kim, Tae Il;Min, Do Sik;Han, Gyoonhee;Choi, Kang-Yell
    • Experimental and Molecular Medicine
    • /
    • v.50 no.11
    • /
    • pp.12.1-12.12
    • /
    • 2018
  • Drugs targeting the epidermal growth factor receptor (EGFR), such as cetuximab and panitumumab, have been prescribed for metastatic colorectal cancer (CRC), but patients harboring KRAS mutations are insensitive to them and do not have an alternative drug to overcome the problem. The levels of ${\beta}$-catenin, EGFR, and RAS, especially mutant KRAS, are increased in CRC patient tissues due to mutations of adenomatous polyposis coli (APC), which occur in 90% of human CRCs. The increases in these proteins by APC loss synergistically promote tumorigenesis. Therefore, we tested KYA1797K, a recently identified small molecule that degrades both ${\beta}$-catenin and Ras via $GSK3{\beta}$ activation, and its capability to suppress the cetuximab resistance of KRAS-mutated CRC cells. KYA1797K suppressed the growth of tumor xenografts induced by CRC cells as well as tumor organoids derived from CRC patients having both APC and KRAS mutations. Lowering the levels of both ${\beta}$-catenin and RAS as well as EGFR via targeting the $Wnt/{\beta}$-catenin pathway is a therapeutic strategy for controlling CRC and other types of cancer with aberrantly activated the $Wnt/{\beta}$-catenin and EGFR-RAS pathways, including those with resistance to EGFR-targeting drugs attributed to KRAS mutations.

Role of Wnt signaling in fracture healing

  • Xu, Huiyun;Duan, Jing;Ning, Dandan;Li, Jingbao;Liu, Ruofei;Yang, Ruixin;Jiang, Jean X.;Shang, Peng
    • BMB Reports
    • /
    • v.47 no.12
    • /
    • pp.666-672
    • /
    • 2014
  • The Wnt signaling pathway is well known to play major roles in skeletal development and homeostasis. In certain aspects, fracture repair mimics the process of bone embryonic development. Thus, the importance of Wnt signaling in fracture healing has become more apparent in recent years. Here, we summarize recent research progress in the area, which may be conducive to the development of Wnt-based therapeutic strategies for bone repair.