• Title/Summary/Keyword: Wnt signaling pathway

Search Result 135, Processing Time 0.02 seconds

Therapeutic Potentiality of Celtis choseniana Nakai on Androgenic Alopecia through Repression of Androgen Action and Modulation of Wnt/β-catenin Signaling

  • Hui-Ju Lee;Geum-Lan Hong;Kyung-Hyun Kim;Yae-Ji Kim;Tae-Won Kim;Ju-Young Jung
    • Natural Product Sciences
    • /
    • v.29 no.1
    • /
    • pp.31-37
    • /
    • 2023
  • In this study, we investigated the efficacy of Celtis choseniana Nakai (C. choseniana) as complementary herbal medicine to ameliorate androgenic alopecia (AGA). The effects of C. choseniana on AGA were evaluated using testosterone propionate-induced AGA mouse model and dihydrotestosterone-treated human hair follicle dermal papilla cells. In vivo, C. choseniana treatment deactivated androgen signaling by reducing the concentration of serum dihydrotestosterone level and expressions of 5α-reductase 2 and androgen receptor. Next, C. choseniana treatment increased the hair regrowth rate. Histological studies demonstrated that C. choseniana induced the anagen phase in testosterone propionate-induced AGA mouse model. Cellular proliferation was promoted by C. choseniana treatment via increasing the expression of proliferation factors, such as proliferating cell nuclear antigen and cyclin D1. Furthermore, C. choseniana treatment increased the expression of proteins related to the Wnt/β-catenin signaling pathway. In addition, dickkopf-1, a Wnt inhibitor, was downregulated with C. choseniana treatment. Likewise, C. choseniana treatment promoted cellular proliferation in vitro. This study demonstrated the inhibitory effect of C. choseniana on androgen-induced AGA. Moreover, C. choseniana induced activation of Wnt/β-catenin signaling, resulting in prolonged anagen and cellular proliferation. Therefore, we suggest that C. choseniana can be used as a therapeutic agent to alleviate AGA.

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.

Hair-growth Promoting Effect of Grateloupia elliptica Via the Activation of Wnt Pathway (참도박의 Wnt 경로 활성화를 통한 모발성장 효과)

  • Kang, Jung-Il;Kim, Sang-Cheol;Jeon, You-Jin;Koh, Young-Sang;Yoo, Eun-Sook;Kang, Hee-Kyoung
    • Korean Journal of Pharmacognosy
    • /
    • v.47 no.2
    • /
    • pp.143-149
    • /
    • 2016
  • Grateloupia elliptica has been reported to have the proliferation effect of dermal papilla cells (DPCs), which play important roles in the regulation of hair cycle. In the present study, we examined in vitro and in vivo hair growth-promoting effect of Grateloupia elliptica. When isolated rat vibrissa follicles were treated with extract of G. elliptica, the hair-fiber lengths of the vibrissa follicles significantly increased. Furthermore, the G. elliptica extract accelerated the telogen-angen transition in C57BL/6 mice. To investigate the molecular mechanisms of the G. elliptica extract on the proliferation of DPCs, we examined the activation of $wnt/{\beta}$-catenin signaling which is known to regulate hair follicle development, differentiation and hair growth. The G. elliptica extract activated $wnt/{\beta}$-catenin signaling via the increase of ${\beta}$-catenin and phospho-$GSK3{\beta}$. In addition, the G. elliptica extract increased the level of cyclin E and CDK2, while the level of $p27^{kip1}$ was decreased. These results suggest that the the G. elliptica extract may induce hair growth by proliferation of DPCs via cell-cycle progression and the activation of $Wnt/{\beta}$-catenin signaling.

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.

CKD-581 Downregulates Wnt/β-Catenin Pathway by DACT3 Induction in Hematologic Malignancy

  • Kim, Soo Jin;Kim, Suntae;Choi, Yong June;Kim, U Ji;Kang, Keon Wook
    • Biomolecules & Therapeutics
    • /
    • v.30 no.5
    • /
    • pp.435-446
    • /
    • 2022
  • The present study evaluated the anti-cancer activity of histone deacetylase (HDAC)-inhibiting CKD-581 in multiple myeloma (MM) and its pharmacological mechanisms. CKD-581 potently inhibited a broad spectrum of HDAC isozymes. It concentration-dependently inhibited proliferation of hematologic cancer cells including MM (MM.1S and RPMI8226) and T cell lymphoma (HH and MJ). It increased the expression of the dishevelled binding antagonist of β-catenin 3 (DACT3) in T cell lymphoma and MM cells, and decreased the expression of c-Myc and β-catenin in MM cells. Additionally, it enhanced phosphorylated p53, p21, cleaved caspase-3 and the subG1 population, and reversely, downregulated cyclin D1, CDK4 and the anti-apoptotic BCL-2 family. Finally, administration of CKD-581 exerted a significant anti-cancer activity in MM.1S-implanted xenografts. Overall, CKD-581 shows anticancer activity via inhibition of the Wnt/β-catenin signaling pathway in hematologic malignancies. This finding is evidence of the therapeutic potential and rationale of CKD-581 for treatment of MM.

Mechanism of Chemoprevention against Colon Cancer Cells Using Combined Gelam Honey and Ginger Extract via mTOR and Wnt/β-catenin Pathways

  • Wee, Lee Heng;Morad, Noor Azian;Aan, Goon Jo;Makpol, Suzana;Ngah, Wan Zurinah Wan;Yusof, Yasmin Anum Mohd
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.16 no.15
    • /
    • pp.6549-6556
    • /
    • 2015
  • The PI3K-Akt-mTOR, $Wnt/{\beta}$-catenin and apoptosis signaling pathways have been shown to be involved in genesis of colorectal cancer (CRC). The aim of this study was to elucidate whether combination of Gelam honey and ginger might have chemopreventive properties in HT29 colon cancer cells by modulating the mTOR, $Wnt/{\beta}$-catenin and apoptosis signaling pathways. Treatment with Gelam honey and ginger reduced the viability of the HT29 cells dose dependently with $IC_{50}$ values of 88 mg/ml and 2.15 mg/ml respectively, their while the combined treatment of 2 mg/ml of ginger with 31 mg/ml of Gelam honey inhibited growth of most HT29 cells. Gelam honey, ginger and combination induced apoptosis in a dose dependent manner with the combined treatment exhibiting the highest apoptosis rate. The combined treatment downregulated the gene expressions of Akt, mTOR, Raptor, Rictor, ${\beta}$-catenin, $Gsk3{\beta}$, Tcf4 and cyclin D1 while cytochrome C and caspase 3 genes were shown to be upregulated. In conclusion, the combination of Gelam honey and ginger may serve as a potential therapy in the treatment of colorectal cancer through inhibiton of mTOR, $Wnt/{\beta}$ catenin signaling pathways and induction of apoptosis pathway.

Wnt/β-Catenin Signaling Pathway Is Necessary for the Specification but Not the Maintenance of the Mouse Retinal Pigment Epithelium

  • Jong-Myeong Kim;Kwang Wook Min;You-Joung Kim;Ron Smits;Konrad Basler;Jin Woo Kim
    • Molecules and Cells
    • /
    • v.46 no.7
    • /
    • pp.441-450
    • /
    • 2023
  • β-Catenin (Ctnnb1) has been shown to play critical roles in the development and maintenance of epithelial cells, including the retinal pigment epithelium (RPE). Ctnnb1 is not only a component of intercellular junctions in the epithelium, it also functions as a transcriptional regulator in the Wnt signaling pathway. To identify which of its functional modalities is critically involved in mouse RPE development and maintenance, we varied Ctnnb1 gene content and activity in mouse RPE lineage cells and tested their impacts on mouse eye development. We found that a Ctnnb1 double mutant (Ctnnb1dm), which exhibits impaired transcriptional activity, could not replace Ctnnb1 in the RPE, whereas Ctnnb1Y654E, which has reduced affinity for the junctions, could do so. Expression of the constitutively active Ctnnb1∆ex3 mutant also suppressed the development of RPE, instead facilitating a ciliary cell fate. However, the post-mitotic or mature RPE was insensitive to the loss, inactivation, or constitutive activation of Ctnnb1. Collectively, our results suggest that Ctnnb1 should be maintained within an optimal range to specify RPE through transcriptional regulation of Wnt target genes in the optic neuroepithelium.

Positional Cloning of Novel Genes in Zebrafish Developmental Mutants

  • Kim, Cheol-Hee
    • Proceedings of the Korean Society of Developmental Biology Conference
    • /
    • 2003.10a
    • /
    • pp.24-25
    • /
    • 2003
  • The zebrafish (Danio rerio) is now the pre-eminent vertebrate model system for clarification of the roles of specific genes and signaling pathways in development. I will talk about positional cloning of two developmental mutants in zebrafish. The first mutant is headless: The vertebrate organizer can induce a complete body axis when transplanted to the ventral side of a host embryo by virtue of its distinct head and trunk inducing properties. Wingless/Wntantagonists secreted by the organizer have been identified as head inducers. Their ectopic expression can promote head formation, whereas ectopic activation of Wnt signalling during early gastrulation blocks head formation. These observations suggest that the ability of head inducers to inhibit Wntsignalling during formation of anterior structures is what distinguishes them from trunk inducers that permit the operation of posteriorizing Wnt signals. I describe the zebrafish headless (hdl) mutant and show that its severe head defects are due to a mutation in T-cell factor-3 (Tcf3), a member of the Tcf/Lef family. Loss of Tcf3 function in the hdl mutant reveals that hdl represses Wnt target genes. I provide genetic evidence that a component of the Wntsignalling pathway is essential in vertebrate head formation and patterning. Second mutant is mind bomb: Lateral inhibition, mediated by Notch signaling, leads to the selection of cells that are permitted to become neurons within domains defined by proneuralgene expression. Reduced lateral inhibition in zebrafish mib mutant embryos permits too many neural progenitors to differentiate as neurons. Positional cloning of mib revealed that it is a gene in the Notch pathway that encodes a RING ubiquitin ligase. Mib interacts with the intracellular domain of Delta to promote its ubiquitylation and internalization. Cell transplantation studies suggest that mib function is essential in the signaling cell for efficient activation of Notch in neighboring cells. (중략)

  • PDF

Zebrafish as a Tool for Function Genomics (제브라피쉬를 이용한 새로운 유전자의 발굴 및 기능분석)

  • Kim Hyun Taek;Kim Cheol Hee
    • Development and Reproduction
    • /
    • v.7 no.2
    • /
    • pp.69-80
    • /
    • 2003
  • The zebrafish(Danio rerio) is a pre-eminent vertebrate model system for clarification of the roles of specific genes and signaling pathways in development. We show examples of positional cloning in two developmental mutants in zebrafish. headless: The severe head defects in headless(hdl) mutants are due to a mutation in T-cell factor-3(Tcf3). Loss of Tcf3 function in the hdl mutant reveals that Hdl represses Wnt target genes. The results provide genetic evidence that a component of the Wnt signaling pathway is essential in vertebrate head formation and patterning. mind bomb: Reduced lateral inhibition in mind bomb(mib) mutants permits too many neural precursors to differentiate as neurons. Positional cloning of mib revealed that it is a gene in the Notch pathway that encodes a ubiquitin E3 ligase. Mib interacts with the intracellular domain of Delta to promote its internalization. The results suggest a model for Notch activation where the Delta-Notch interaction is followed by endocytosis of Delta and transendocytosis of the Notch extracellular domain by the signaling cell.

  • PDF

Molecular Mechanism of Runx2-Dependent Bone Development

  • Komori, Toshihisa
    • Molecules and Cells
    • /
    • v.43 no.2
    • /
    • pp.168-175
    • /
    • 2020
  • Runx2 is an essential transcription factor for skeletal development. It is expressed in multipotent mesenchymal cells, osteoblast-lineage cells, and chondrocytes. Runx2 plays a major role in chondrocyte maturation, and Runx3 is partly involved. Runx2 regulates chondrocyte proliferation by directly regulating Ihh expression. It also determines whether chondrocytes become those that form transient cartilage or permanent cartilage, and functions in the pathogenesis of osteoarthritis. Runx2 is essential for osteoblast differentiation and is required for the proliferation of osteoprogenitors. Ihh is required for Runx2 expression in osteoprogenitors, and hedgehog signaling and Runx2 induce the differentiation of osteoprogenitors to preosteoblasts in endochondral bone. Runx2 induces Sp7 expression, and Runx2, Sp7, and canonical Wnt signaling are required for the differentiation of preosteoblasts to immature osteoblasts. It also induces the proliferation of osteoprogenitors by directly regulating the expression of Fgfr2 and Fgfr3. Furthermore, Runx2 induces the proliferation of mesenchymal cells and their commitment into osteoblast-lineage cells through the induction of hedgehog (Gli1, Ptch1, Ihh), Fgf (Fgfr2, Fgfr3), Wnt (Tcf7, Wnt10b), and Pthlh (Pth1r) signaling pathway gene expression in calvaria, and more than a half-dosage of Runx2 is required for their expression. This is a major cause of cleidocranial dysplasia, which is caused by heterozygous mutation of RUNX2. Cbfb, which is a co-transcription factor that forms a heterodimer with Runx2, enhances DNA binding of Runx2 and stabilizes Runx2 protein by inhibiting its ubiquitination. Thus, Runx2/Cbfb regulates the proliferation and differentiation of chondrocytes and osteoblast-lineage cells by activating multiple signaling pathways and via their reciprocal regulation.