• Title/Summary/Keyword: Luciferase activity

Search Result 360, Processing Time 0.026 seconds

Tumor Necrosis Factor ${\alpha}$ up-regulates the Expression of beta2 Adrenergic Receptor via NF-${\kappa}B$-dependent Pathway in Osteoblasts

  • Baek, Kyunghwa;Kang, Jiho;Hwang, Hyo Rin;Baek, Jeong-Hwa
    • International Journal of Oral Biology
    • /
    • v.38 no.3
    • /
    • pp.121-126
    • /
    • 2013
  • Tumor necrosis factor alpha ($TNF{\alpha}$) is a multifunctional inflammatory cytokine that regulates various cellular and biological processes. Increased levels of $TNF{\alpha}$ have been implicated in a number of human diseases including diabetes and arthritis. Sympathetic nervous system stimulation via the beta2-adrenergic receptor (${\beta}2AR$) in osteoblasts suppresses osteogenic activity. We previously reported that $TNF{\alpha}$ upregulates ${\beta}2AR$ expression in murine osteoblastic cells and that this modulation is associated with $TNF{\alpha}$ inhibition of osteoblast differentiation. In our present study, we explored whether $TNF{\alpha}$ induces ${\beta}2AR$ expression in human osteoblasts and then identified the downstream signaling pathway. Our results indicated that ${\beta}2AR$ expression was increased in Saos-2 and C2C12 cells by $TNF{\alpha}$ treatment, and that this increase was blocked by the inhibition of NF-${\kappa}B$ activation. Chromatin immunoprecipitation and luciferase reporter assay results indicated that NF-${\kappa}B$ directly binds to its cognate elements on the ${\beta}2AR$ promoter and thereby stimulates ${\beta}2AR$ expression. These findings suggest that the activation of $TNF{\alpha}$ signaling in osteoblastic cells leads to an upregulation of ${\beta}2AR$ and also that $TNF{\alpha}$ induces ${\beta}2AR$ expression in an NF-${\kappa}B$-dependent manner.

Inhibition of hypoxia-induced cyclooxygenase-2 by Korean Red Ginseng is dependent on peroxisome proliferator-activated receptor gamma

  • Song, Heewon;Lee, Young Joo
    • Journal of Ginseng Research
    • /
    • v.41 no.3
    • /
    • pp.240-246
    • /
    • 2017
  • Background: Korean Red Ginseng (KRG) is a traditional herbal medicine made by steaming and drying fresh ginseng. It strengthens the endocrine and immune systems to ameliorate various inflammatory responses. The cyclooxygenase-2 (COX-2)/prostaglandin E2 pathway has important implications for inflammation responses and tumorigenesis. Peroxisome proliferator-activated receptor gamma ($PPAR{\gamma}$) is a transcription factor that regulates not only adipogenesis and lipid homeostasis, but also angiogenesis and inflammatory responses. Methods: The effects of the KRG on inhibition of hypoxia-induced COX-2 via $PPAR{\gamma}$ in A549 cells were determined by luciferase assay, Western blot, and/or quantitative reverse transcription-polymerase chain reaction (qRT-PCR). The antimigration and invasive effects of KRG were evaluated on A549 cells using migration and matrigel invasion assays. Results and conclusion: We previously reported that hypoxia-induced COX-2 protein and mRNA levels were suppressed by KRG. This study examines the possibility of $PPAR{\gamma}$ as a cellular target of KRG for the suppression of hypoxia-induced COX-2. $PPAR{\gamma}$ protein levels and $PPAR{\gamma}$-responsive element (PPRE)-driven reporter activities were increased by KRG. Reduction of hypoxia-induced COX-2 by KRG was abolished by the $PPAR{\gamma}$ inhibitor GW9662. In addition, the inhibition of $PPAR{\gamma}$ abolished the effect of KRG on hypoxia-induced cell migration and invasion. Discussion: Our results show that KRG inhibition of hypoxia-induced COX-2 expression and cell invasion is dependent on $PPAR{\gamma}$ activation, supporting the therapeutic potential for suppression of inflammation under hypoxia. Further studies are required to demonstrate whether KRG activates directly $PPAR{\gamma}$ and to identify the constituents responsible for this activity.

DNase I Hypersensitive Site 8 as an Enhancer is Mapped in the Upstream Vicinity of the Crlz1 Promoter (Crlz1 promoter의 상위에 인접한 DNase I hypersensitive site 8의 enhancer 기능)

  • Choi, Seong-Young;Kang, Chang-Joong
    • Journal of Life Science
    • /
    • v.22 no.9
    • /
    • pp.1201-1206
    • /
    • 2012
  • The Crlz1 gene is known to be expressed specifically in the pre-B cell stage during B-cell development. With regards to the specific expression of the pre-B cell stage of the Crlz1 gene, we have previously identified three pre-B cell-specific DNase I hypersensitive sites (HSS), which are named HSS8, 9, and 10, in the upstream vicinity of this gene. In this paper, we report that HSS8 increases further the strong Crlz1 promoter activity driven by HSS9/10 and, therefore, acts as its enhancer. Furthermore, HSS8 has been finely mapped between -1055 and -1159 from the transcription start site of the Crlz1 gene.

miR-30a Regulates the Expression of CAGE and p53 and Regulates the Response to Anti-Cancer Drugs

  • Park, Deokbum;Kim, Hyuna;Kim, Youngmi;Jeoung, Dooil
    • Molecules and Cells
    • /
    • v.39 no.4
    • /
    • pp.299-309
    • /
    • 2016
  • We have previously reported the role of miR-217 in anti-cancer drug-resistance. miRNA array and miRNA hybridization analysis predicted miR-30a-3p as a target of miR-217. miR-30a-3p and miR-217 formed a negative feedback loop and regulated the expression of each other. Ago1 immunoprecipitation and co-localization analysis revealed a possible interaction between miR-30a-3p and miR-217. miR-30a-3p conferred resistance to anti-cancer drugs and enhanced the invasion, migration, angiogenic, tumorigenic, and metastatic potential of cancer cells in CAGE-dependent manner. CAGE increased the expression of miR-30a-3p by binding to the promoter sequences of miR-30a-3p, suggesting a positive feedback loop between CAGE and miR-30a-3p. miR-30a-3p decreased the expression of p53, which showed the binding to the promoter sequences of miR-30a-3p and CAGE in anti-cancer drug-sensitive cancer cells. Luciferase activity assays showed that p53 serves as a target of miR-30a. Thus, the miR-30a-3p-CAGE-p53 feedback loop serves as a target for overcoming resistance to anti-cancer drugs.

Effects of Medicinal herb Extracts and their Components on Steatogenic Hepatotoxicity in Sk-hep1 Cells

  • Choi, You-Jin;Yoon, Yu-Jin;Choi, Ho-Sung;Park, So-Ra;Oh, Se-Hee;Jeong, Se-Mi;Suh, Hyo-Ryung;Lee, Byung-Hoon
    • Toxicological Research
    • /
    • v.27 no.4
    • /
    • pp.211-216
    • /
    • 2011
  • Herbal medicines are widely used in many countries for the treatment of many diseases. Although the use of herb extracts as alternative medicine is growing, their toxicological properties have not been thoroughly investigated. In this study, we have investigated the effects of water and ethanol extracts of 18 herbs on the hepatic lipid metabolism and steatogenic hepatotoxicity. Ethanol extracts of Cirsium japonicum, Carthamus tinctorius, Rehmanniae glutinosa (preparata), Polygala tenuifolia, Foeniculum vulgare, Polygonum multiflorum, and Acorus gramineus and water extracts of Polygonum multiflorum and Rehmanniae glutinosa induced lipid accumulation in Sk-hep1 human hepatoma cells as determined by Nile red staining. These extracts increased the luciferase activity of sterol regulatory element (SRE) and decreased that of peroxisome proliferator response element (PPRE), indicating the possibilities of enhanced fatty acid synthesis and decreased fatty acid oxidation. To identify the components responsible for the fat accumulation, we tested 50 chemicals isolated from the nine herbs. Apigenin, luteolin, pectolinarin and lupeol from Cirsium japonicum, 8-methoxypsoralen and umbelliferone from Foeniculum vulgare and pomonic acid and jiocerebroside from Rehmanniae glutinosa significantly increased the accumulation of lipid droplets. These results suggest that ethanol extracts of Cirsium japonicum, Carthamus tinctorius, Rehmanniae glutinosa (preparata), Polygala tenuifolia, Foeniculum vulgare, Polygonum multiflorum, and Acorus gramineus and water extracts of Polygonum multiflorum and Rehmanniae glutinosa can cause fatty liver disease by decreasing ${\beta}$-oxidation of fatty acid and increasing lipogenesis.

FXRα Down-Regulates LXRα Signaling at the CETP Promoter via a Common Element

  • Park, Sung-Soo;Choi, Hojung;Kim, Seung-Jin;Kim, Ok Jin;Chae, Kwon-Seok;Kim, Eungseok
    • Molecules and Cells
    • /
    • v.26 no.4
    • /
    • pp.409-414
    • /
    • 2008
  • The cholesteryl ester transfer protein (CETP), a key player in cholesterol metabolism, has been shown to promote the transfer of triglycerides from very low density lipoprotein (VLDL) and low density lipoprotein (LDL) to high density lipoprotein (HDL) in exchange for cholesterol ester. Here we demonstrate that farnesoid X receptor ${\alpha}$ ($FXR{\alpha}$; NR1H4) down-regulates CETP expression in HepG2 cells. A $FXR{\alpha}$ ligand, chenodeoxycholic acid (CDCA), suppressed basal mRNA levels of the CETP gene in HepG2 cells in a dose-dependent manner. Using gel shift and chromatin immunoprecipitation (ChIP) assays, we found that $FXR{\alpha}$ could bind to the liver X receptor ${\alpha}$ ( $LXR{\alpha}$; NR1H3) binding site (LXRE; DR4RE) located within the CETP 5' promoter region. $FXR{\alpha}$ suppressed $LXR{\alpha}$-induced DR4RE-luciferase activity and this effect was mediated by a binding competition between $FXR{\alpha}$ and $LXR{\alpha}$ for DR4RE. Furthermore, the addition of CDCA together with a $LXR{\alpha}$ ligand, GW3965, to HepG2 cells was shown to substantially decrease mRNA levels of hepatic CETP gene, which is typically induced by GW3965. Together, our data demonstrate that $FXR{\alpha}$ down-regulates CETP gene expression via binding to the DR4RE sequence within the CETP 5' promoter and this $FXR{\alpha}$ binding is essential for $FXR{\alpha}$ inhibition of $LXR{\alpha}$-induced CETP expression.

PRR11 and SKA2 gene pair is overexpressed and regulated by p53 in breast cancer

  • Wang, Yitao;Zhang, Chunxue;Mai, Li;Niu, Yulong;Wang, Yingxiong;Bu, Youquan
    • BMB Reports
    • /
    • v.52 no.2
    • /
    • pp.157-162
    • /
    • 2019
  • Our previous study found that two novel cancer-related genes, PRR11 and SKA2, constituted a classic gene pair that was regulated by p53 and NF-Y in lung cancer. However, their role and regulatory mechanism in breast cancer remain elusive. In this study, we found that the expression levels of PRR11 and SKA2 were upregulated and have a negative prognotic value in breast cancer. Loss-of-function experiments showed that RNAi-mediated knockdown of PRR11 and/or SKA2 inhibited proliferation, migration, and invasion of breast cancer cells. Mechanistic experiments revealed that knockdown of PRR11 and/or SKA2 caused dysregulation of several downstream genes, including CDK6, TPM3, and USP12, etc. Luciferase reporter assays demonstrated that wild type p53 significantly repressed the PRR11-SKA2 bidirectional promoter activity, but not NF-Y. Interestingly, NF-Y was only essential for and correlated with the expression of PRR11, but not SKA2. Consistently, adriamycin-induced (ADR) activation of endogenous p53 also caused significant repression of the PRR11 and SKA2 gene pair expression. Notably, breast cancer patients with lower expression levels of either PRR11 or SKA2, along with wild type p53, exhibited better disease-free survival compared to others with p53 mutations and/or higher expression levels of either PRR11 or SKA2. Collectively, our study indicates that the PRR11 and SKA2 transcription unit might be an oncogenic contributor and might serve as a novel diagnostic and therapeutic target in breast cancer.

Flavonoid Silibinin Increases Hair-Inductive Property Via Akt and Wnt/β-Catenin Signaling Activation in 3-Dimensional-Spheroid Cultured Human Dermal Papilla Cells

  • Cheon, Hye In;Bae, Seunghee;Ahn, Kyu Joong
    • Journal of Microbiology and Biotechnology
    • /
    • v.29 no.2
    • /
    • pp.321-329
    • /
    • 2019
  • Hair loss, also known as alopecia, is a common dermatological condition of psychosocial significance; development of therapeutic candidates for the treatment of this condition is, hence, important. Silibinin, a secondary metabolite from Silybum marianum, is an effective antioxidant that also prevents various cutaneous problems. In this study, we have investigated the effect of silibinin on hair induction using three-dimensional (3D) cultured, human dermal papilla (DP) spheroids. Silibinin was found to significantly increase viability through AKT serine/threonine kinase (AKT) activation in 3D DP spheroids. This was correlated with an increase in the diameter of the 3D DP spheroids. The activation of the wingless and INT-1 (Wnt)/${\beta}$-catenin signaling pathway, which is associated with hair growth induction in the DP, was evaluated using the T cell-specific transcription factor and lymphoid enhancer-binding factor (TCF/LEF) transcription factor reporter assay; results indicated significantly increased luciferase activity. In addition, we were able to demonstrate increased expression of the target genes, WNT5a and LEF1, using quantitative real-time PCR assay. Lastly, significantly elevated expression of signature genes associated with hair induction was demonstrated in the 3D DP spheroids treated with silibinin. These results suggest that silibinin promotes proliferation and hair induction through the AKT and Wnt/${\beta}$-catenin signaling pathways in 3D DP spheroids. Silibinin can be a potential candidate to promote hair proliferation.

miR-101-3p/Rap1b signal pathway plays a key role in osteoclast differentiation after treatment with bisphosphonates

  • Li, Jie;Li, You;Wang, Shengjie;Che, Hui;Wu, Jun;Ren, Yongxin
    • BMB Reports
    • /
    • v.52 no.9
    • /
    • pp.572-576
    • /
    • 2019
  • Bisphosphonates are the mainstay of therapy worldwide for osteoporosis. However, bisphosphonates also have limitations. The objective of this study was to determine the role of miR-101-3p/Rap1b signal pathway in osteoclast differentiation after treatment with bisphosphonates. Our results revealed that miR-101-3p was an important regulator in bisphosphonates treated-osteoclasts. When miR-101-3p was down-regulated in bone marrow-derived macrophage-like cells (BMMs), the development of mature osteoclasts was promoted, and vice versa. However, alendronate decreased multinucleated cell number regardless of whether miR-101-3p was knocked down or over-expressed. TRAP activity assay confirmed the above results. Luciferase assay indicated that miR-101-3p was a negative regulator of Rap1b. Western blot analysis revealed that protein expression level of Rap1b in BMMs transfected with OV-miR-101-3p was lower than that in BMMs transfected with an empty vector. Rap1b overexpression increased TRAP-positive multinucleated cells, while Rap1b inhibition decreased the cell numbers. In vivo data showed that miR-101-3p inhibited osteoclast differentiation in ovariectomized mice while overexpressed of Rap1b blocked the differentiation. Taken together, our data demonstrate that miR-101-3p/Rap1b signal pathway plays a key role in osteoclast differentiation after treatment with bisphosphonates.

Ets-1 enhances tumor migration through regulation of CCR7 expression

  • Fang, Li-Wen;Kao, Ying-Hsien;Chuang, Ya-Ting;Huang, Huey-Lan;Tai, Tzong-Shyuan
    • BMB Reports
    • /
    • v.52 no.9
    • /
    • pp.548-553
    • /
    • 2019
  • Ets-1 is a prototype of the ETS protein family. Members of the ETS protein family contain a unique ETS domain. Ets-1 is associated with cancer progression and metastasis in many types of cancer. Many studies have shown a link between elevated expression of Ets-1 in cancer biopsies and poor survival. CCR7 is a chemokine that binds to specific ligand CCL21/CCL19. CCR7 expression is associated with tumor metastasis and infiltration into lymph nodes. The objective of this study was to test whether Ets-1 could regulate CCR7 expression and enhance tumor metastasis. Our data showed that CCR7 expression was downregulated in Ets-1-deficient T cells upon T-cell stimulation. Overexpression of Ets-1 increased CCR7 expression in breast cancer cell lines. In contrast, knockdown of Ets-1 reduced CCR7 expression. Ets-1 could directly bind to CCR7 promoter and mediate CCR7 expression in luciferase reporter assays and chromatin immunoprecipitation assays. Transactivation activity of Ets-1 was independent of the Pointed domain of Ets-1. Ets-1 could also enhance $NF-{\kappa}B$ and CBP transactivation of CCR7 promoter. Our results also showed that Ets-1 could modulate cancer cell transmigration by altering CCR7 expression in transwell assay and wound healing assay. Taken together, our data suggest that Ets-1 can enhance CCR7 expression and contribute to tumor cell migration.