• Title/Summary/Keyword: Wnt proteins

Search Result 37, Processing Time 0.024 seconds

Static magnetic fields promote osteoblastic/cementoblastic differentiation in osteoblasts, cementoblasts, and periodontal ligament cells

  • Kim, Eun-Cheol;Park, Jaesuh;Kwon, Il Keun;Lee, Suk-Won;Park, Su-Jung;Ahn, Su-Jin
    • Journal of Periodontal and Implant Science
    • /
    • v.47 no.5
    • /
    • pp.273-291
    • /
    • 2017
  • Purpose: Although static magnetic fields (SMFs) have been used in dental prostheses and osseointegrated implants, their biological effects on osteoblastic and cementoblastic differentiation in cells involved in periodontal regeneration remain unknown. This study was undertaken to investigate the effects of SMFs (15 mT) on the osteoblastic and cementoblastic differentiation of human osteoblasts, periodontal ligament cells (PDLCs), and cementoblasts, and to explore the possible mechanisms underlying these effects. Methods: Differentiation was evaluated by measuring alkaline phosphatase (ALP) activity, mineralized nodule formation based on Alizarin red staining, calcium content, and the expression of marker mRNAs assessed by reverse transcription polymerase chain reaction (RT-PCR). Signaling pathways were analyzed by western blotting and immunocytochemistry. Results: The activities of the early marker ALP and the late markers matrix mineralization and calcium content, as well as osteoblast- and cementoblast-specific gene expression in osteoblasts, PDLCs, and cementoblasts were enhanced. SMFs upregulated the expression of Wnt proteins, and increased the phosphorylation of glycogen synthase $kinase-3{\beta}$ ($GSK-3{\beta}$) and total ${\beta}-catenin$ protein expression. Furthermore, p38 and c-Jun N-terminal kinase (JNK) mitogen-activated protein kinase (MAPK), and nuclear $factor-{\kappa}B$ ($NF-{\kappa}B$) pathways were activated. Conclusions: SMF treatment enhanced osteoblastic and/or cementoblastic differentiation in osteoblasts, cementoblasts, and PDLCs. These findings provide a molecular basis for the beneficial osteogenic and/or cementogenic effect of SMFs, which could have potential in stimulating bone or cementum formation during bone regeneration and in patients with periodontal disease.

Zic3z Defines the Dorsal and Vegetal Neuroectoderm in the Zebrafish Embryonic Development

  • Lee, Kyu-Sun;Huh, Tae-Lin;Lee, Chang-Joong;Rhee, Myung-Chull
    • Animal cells and systems
    • /
    • v.12 no.1
    • /
    • pp.23-33
    • /
    • 2008
  • The Zic family is a group of genes encoding zinc finger proteins that are highly expressed in the mammalian cerebellum. Zic genes are the vertebrate homologue of Drosophila pair-rule gene, odd-paired(opa), which plays important roles in the parasegmental subdivision as well as in the visceral mesoderm development of Drosophila embryos. Recent studies on human, mouse, frog, fish and ascidian Zic homologues support that Zic genes are involved in a variety of developmental processes, including neurogenesis, myogenesis, skeletal patterning, and left-right axis establishment. In an effort to explore possible functions of Zic proteins during vertebrate embryogenesis, we initially examined more detailed expression pattern of zebrafish homologue of zic3(zic3z). zic3z transcripts are detected in the neuroectoderm, neural plate, dorsal neural tube, and brain regions including eye field during early embryonic development. Marker DNA studies found that zic3z transcription is modulated by BMP, Wnt, and Nodal signals particularly in the dorsal and vegetal neuroectoderm at gastrula. Interfering with zic3z translation with zic3z-specific morpholino causes abnormal brain formation and expansion of the optic stalk cells. Retinal ganglion cells(RGCs) undergo abnormal neuronal differentiation. These findings suggest that zic3z defines the dorsal and vegetal neuroectoderm to specify brain formation and retinal neurogenesis during early embryonic development.

Targeting of COX-2 Expression by Recombinant Adenovirus shRNA Attenuates the Malignant Biological Behavior of Breast Cancer Cells

  • Tu, Bo;Ma, Ting-Ting;Peng, Xiao-Qiong;Wang, Qin;Yang, Hong;Huang, Xiao-Ling
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.15 no.20
    • /
    • pp.8829-8836
    • /
    • 2014
  • Background: Cyclooxygenase-2 (COX-2), considered to have tumor-promoting potential, is highly expressed in a variety of tumors, including breast cancer. Since the functions and action mechanisms of COX-2 in breast cancer have not been fully elucidated, in the present study, the effects of target inhibiting COX-2 with recombinant adenovirus Ad-COX-2-shRNA on malignant biological behavior were investigated in representative cell lines. Materials and Methods: Breast cancer MDA-MB-231 and MCF-7 cells were transfected with Ad-COX-2-shRNA and COX-2 expression was tested by RT-PCR and Western blotting. Changes in proliferation, apoptosis and invasion of breast cancer cells were detected with various assays including MTT, colony forming, flowcytometry and Transwell invasion tests. The expression of related proteins involved in the cell cycle, apoptosis, invasion and signaling pathways was assessed by Western blotting. Results: COX-2 expression was significantly reduced in both breast cancer cell lines infected with Ad-COX-2-shRNA, with obvious inhibition of proliferation, colony forming rate, G2/M phase passage and invasion, as well as induction of apoptosis, in MDA-MB-231 and MCF-7 cells, respectively. At the same time, proteins related to the cell cycle, anti-apoptosis and invasion were significantly downregulated. In addition, c-myc expression and phosphorylation activation of Wnt/${\beta}$-catenin and p38MAPK pathways were reduced by the Ad-COX-2-shRNA. Conclusions: COX-2 expression is associated with proliferation, apoptosis and invasion of breast cancer cells, and its mechanisms of action involve regulating expression of c-myc through the p38MAPK and Wnt/${\beta}$-catenin pathways.

Vanillic Acid Stimulates Anagen Signaling via the PI3K/Akt/β-Catenin Pathway in Dermal Papilla Cells

  • Kang, Jung-Il;Choi, Youn Kyung;Koh, Young-Sang;Hyun, Jin-Won;Kang, Ji-Hoon;Lee, Kwang Sik;Lee, Chun Mong;Yoo, Eun-Sook;Kang, Hee-Kyoung
    • Biomolecules & Therapeutics
    • /
    • v.28 no.4
    • /
    • pp.354-360
    • /
    • 2020
  • The hair cycle (anagen, catagen, and telogen) is regulated by the interaction between mesenchymal cells and epithelial cells in the hair follicles. The proliferation of dermal papilla cells (DPCs), mesenchymal-derived fibroblasts, has emerged as a target for the regulation of the hair cycle. Here, we show that vanillic acid, a phenolic acid from wheat bran, promotes the proliferation of DPCs via a PI3K/Akt/Wnt/β-catenin dependent mechanism. Vanillic acid promoted the proliferation of DPCs, accompanied by increased levels of cell-cycle proteins cyclin D1, CDK6, and Cdc2 p34. Vanillic acid also increased the levels of phospho(ser473)-Akt, phospho(ser780)-pRB, and phospho(thr37/46)-4EBP1 in a time-dependent manner. Wortmannin, an inhibitor of the PI3K/Akt pathway, attenuated the vanillic acid-mediated proliferation of DPCs. Vanillic acid-induced progression of the cell-cycle was also suppressed by wortmannin. Moreover, vanillic acid increased the levels of Wnt/β-catenin proteins, such as phospho(ser9)-glycogen synthase kinase-3β, phospho(ser552)-β-catenin, and phospho(ser675)-β-catenin. We found that vanillic acid increased the levels of cyclin D1 and Cox-2, which are target genes of β-catenin, and these changes were inhibited by wortmannin. To investigate whether vanillic acid affects the downregulation of β-catenin by dihydrotestosterone (DHT), implicated in the development of androgenetic alopecia, DPCs were stimulated with DHT in the presence and absence of vanillic acid for 24 h. Western blotting and confocal microscopy analyses showed that the decreased level of β-catenin after the incubation with DHT was reversed by vanillic acid. These results suggest that vanillic acid could stimulate anagen and alleviate hair loss by activating the PI3K/Akt and Wnt/β-catenin pathways in DPCs.

Transcriptional Regulation and Apoptosis Induction by Tcf/$\beta$-Catenin Complex in Various T-Cells

  • Jeong, Sunjoo;Lee, Seung-Yeon;Lee, Sun-Hee
    • Animal cells and systems
    • /
    • v.4 no.4
    • /
    • pp.389-394
    • /
    • 2000
  • The Tcf-1 (1-cell factor-1) protein binds to the T-cell specific enhancer sequences and plays an architectural role in the assembly of transcriptional machinery. One of the Tcf family proteins, Tcf-4, was found to be an important regulator for colon cancer development where it activates specific genes upon binding to $\beta$-catenin following Wnt signaling. We were interested in the transcriptional regulatory activities of Tcf-1 and Tcf-4 proteins in T-cells and colon cancer cells. Transactivation assay was developed using a reporter plasmid containing luciferase gene under the control of Tcf responsive elements. Luciferase activity was determined following co-transfection of the reporter along with Tcf-1 and/or $\beta$-catenin expressing plasmids. Transcription was significantly induced by $\beta$-catenin expression in all cells. Tcf-1 by itself did not induce transcription in the mature T-cell lines, but overexpressed Tcf-1 greatly activated transcription in the immature T-cell line. In addition, transfected $\beta$-catenin induced apoptosis, but co-transfected Tcf-1 suppressed apoptosis in HEK293 cells. These results suggest that Tcf-1 and $\beta$-catenin differently regulate transcription and apoptosis.

  • PDF

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.

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.

The Promoting Effect of Rumex japonicas Houttuyn ethanol extract on Hair Growth

  • Jeong, Jang-won;Kang, Kyung-Hwa;Cho, Sung-Woo
    • The Journal of Korean Medicine
    • /
    • v.41 no.4
    • /
    • pp.27-40
    • /
    • 2020
  • Objectives: This study was conducted to evaluate the hair growth-promoting effects by Rumex japonicas Houttuyn ethanol extract (RJHEE) in C57BL/6N mice and HaCaT cells. Methods: The hair growth effect was examined by topical application of RJHEE on the shaved dorsal skin of C57BL/6 mice. Six-week old mice were depilated and separated in 4 groups; CON (vehicle treatment), MXD (2% Minoxidil), and RJHEE (2% and 4%). The treatments were applied daily for 17 days. The hair growth was determined photographically and the hair density, thickness and length were identified by Folliscope. In dorsal skin tissue, the expression of hair growth-related protein was analyzed by Western blotting. In HaCaT cells, the cell proliferation and the protection against H2O2-induced cell damage by RJHEE were analyzed. Results: Our results indicate that RJHEE promote the hair growth, hair density, thickness and length. RHE activate the Wnt/𝛽-catenin signaling and induced the expression of cell survival-related proteins, such as pERK/ERK and Bcl-2/Bax. In HaCaT, RJHEE accelerated the cell proliferation and protected the H2O2-induced cell damage. Conclusions: Our results strongly suggest that RJHEE promotes hair growth by regulating the activation of Wnt/𝛽-catenin signaling and cell survival signaling and protects oxidative stress-induced hair damage. Therefore, RJHEE has a hair growth activity and can be useful for the treatment of alopecia.

Roles of Signaling Pathways in the Epithelial-Mesenchymal Transition in Cancer

  • Liu, Xia;Yun, Fen;Shi, Lin;Li, Zhe-Hai;Luo, Nian-Rong;Jia, Yong-Feng
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.16 no.15
    • /
    • pp.6201-6206
    • /
    • 2015
  • The epithelial-mesenchymal transition (EMT) is a cellular process though which an epithelial phenotype can be converted into a phenotype of mesenchymal cells. Under physiological conditions EMT is important for embryogenesis, organ development, wound repair and tissue remodeling. However, EMT may also be activated under pathologic conditions, especially in carcinogenesis and metastatic progression. Major signaling pathways involved in EMT include transforming growth factor ${\beta}(TGF-{\beta})$, Wnt, Notch, Hedgehog and other signaling pathways. These pathways are related to several transcription factors, including Twist, Smads and zinc finger proteins snail and slug. These interact with each other to provide crosstalk between the relevant signaling pathways. This review lays emphasis on studying the relationship between EMT and signaling pathways in carcinogenesis and metastatic progression.

DNA binding partners of YAP/TAZ

  • Kim, Min-Kyu;Jang, Ju-Won;Bae, Suk-Chul
    • BMB Reports
    • /
    • v.51 no.3
    • /
    • pp.126-133
    • /
    • 2018
  • Hippo signaling plays critical roles in regulation of tissue homeostasis, organ size, and tumorigenesis by inhibiting YES-associated protein (YAP) and PDZ-binding protein TAZ through MST1/2 and LATS1/2 pathway. It is also engaged in cross-talk with various other signaling pathways, including WNT, BMPs, Notch, GPCRs, and Hedgehog to further modulate activities of YAP/TAZ. Because YAP and TAZ are transcriptional coactivators that lack DNA-binding activity, both proteins must interact with DNA-binding transcription factors to regulate target gene's expression. To activate target genes involved in cell proliferation, TEAD family members are major DNA-binding partners of YAP/TAZ. Accordingly, YAP/TAZ were originally classified as oncogenes. However, YAP might also play tumor-suppressing role. For example, YAP can bind to DNA-binding tumor suppressors including RUNXs and p73. Thus, YAP might act either as an oncogene or tumor suppressor depending on its binding partners. Here, we summarize roles of YAP depending on its DNA-binding partners and discuss context-dependent functions of YAP/TAZ.