• Title/Summary/Keyword: the transcription factors

Search Result 1,098, Processing Time 0.028 seconds

Histone H3 Lysine Methylation in Adipogenesis (Adipogenesis에서 히스톤 H3 lysine methylation)

  • Jang, Younghoon
    • Journal of Life Science
    • /
    • v.30 no.8
    • /
    • pp.713-721
    • /
    • 2020
  • Adipogenesis as a model system is needed to understand the molecular mechanisms of human adipocyte biology and the pathogenesis of obesity, diabetes, and other metabolic syndromes. Many relevant studies have been conducted with a focus on gene expression regulation and intracellular signaling relating to Peroxisome proliferator-activated receptor gamma (PPARγ) and CCAAT/enhancer-binding protein alpha (C/EBPα), which are master adipogenic transcription factors. However, epigenome regulation of adipogenesis by epigenomic modifiers or histone mutations is not fully understood. Histone methylation is one of the major epigenetic modifications on gene expression in mammals, and histone H3 lysine methylation (H3Kme) in particular implicates cell differentiation during various tissue and organ development. During adipogenesis, cell type-specific enhancers are marked by histone H3K4me1 with the active enhancer mark H3K27ac. Mixed-lineage leukemia 4 (MLL4) is a major H3K4 mono-methyltransferase on the adipogenic enhancers of PPARγ and C/EBPα loci. Thus, MLL4 is an important epigenomic modifier for adipogenesis. The repressive mark H3K27me3 is mediated by the enzymatic subunit Enhancer zeste homolog 2 (EZH2) of the polycomb repressive complex 2. EZH2-mediated H3K27 tri-methylation on the Wnt gene increases adipogenesis because WNT signaling is a negative regulator of adipogenesis. This review summarizes current knowledge about the epigenomic regulation of adipogenesis by histone H3 lysine methylation which fundamentally regulates gene expression.

HOXA9 is Underexpressed in Cervical Cancer Cells and its Restoration Decreases Proliferation, Migration and Expression of Epithelial-to-Mesenchymal Transition Genes

  • Alvarado-Ruiz, Liliana;Martinez-Silva, Maria Guadalupe;Torres-Reyes, Luis Alberto;Pina-Sanchez, Patricia;Ortiz-Lazareno, Pablo;Bravo-Cuellar, Alejandro;Aguilar-Lemarroy, Adriana;Jave-Suarez, Luis Felipe
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.17 no.3
    • /
    • pp.1037-1047
    • /
    • 2016
  • HOX transcription factors are evolutionarily conserved in many different species and are involved in important cellular processes such as morphogenesis, differentiation, and proliferation. They have also recently been implicated in carcinogenesis, but their precise role in cancer, especially in cervical cancer (CC), remains unclear. In this work, using microarray assays followed by the quantitative polymerase chain reaction (qPCR), we found that the expression of 25 HOX genes was downregulated in CC derived cell lines compared with non-tumorigenic keratinocytes. In particular, the expression of HOXA9 was observed as down-modulated in CC-derived cell lines. The expression of HOXA9 has not been previously reported in CC, or in normal keratinocytes of the cervix. We found that normal CC from women without cervical lesions express HOXA9; in contrast, CC cell lines and samples of biopsies from women with CC showed significantly diminished HOXA9 expression. Furthermore, we found that methylation at the first exon of HOXA9 could play an important role in modulating the expression of this gene. Exogenous restoration of HOXA9 expression in CC cell lines decreased cell proliferation and migration, and induced an epithelial-like phenotype. Interestingly, the silencing of human papilloma virus (HPV) E6 and E7 oncogenes induced expression of HOXA9. In conclusion, controlling HOXA9 expression appears to be a necessary step during CC development. Further studies are needed to delineate the role of HOXA9 during malignant progression and to afford more insights into the relationship between downmodulation of HOXA9 and viral HPV oncoprotein expression during cercical cancer development.

CCNG2 Suppressor Biological Effects on Thyroid Cancer Cell through Promotion of CDK2 Degradation

  • Li, Wei-Juan;Liu, Ge-Ling;Yu, Fang;Xiang, Xiu-Xiu;Lu, Yi-Fang;Xiao, Hong-Zhen;Shi, Yan-Ping
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.14 no.10
    • /
    • pp.6165-6171
    • /
    • 2013
  • This study aimed to analyze the expression and clinical significance of cyclin G2 (CCNG2) in thyroid carcinoma and the biological effects of CCNG2 overexpression in a cell line. Immunohistochemistry and Western blotting were used to analyze CCNG2 protein expression in 63 cases of thyroid cancer and normal tissues to allow the relationship with clinical factors to be assessed. CCNG2 lentiviral and empty vectors were transfected into the thyroid cancer K1 cell line. Reverse transcription-polymerase chain reaction (RT-PCR) and Western blotting were applied to detect the mRNA and protein levels of CCNG2. MTT assay and cell cycle were also conducted to assess the influence of up-regulated expression of CCNG2 on K1 cell biology. The level of CCNG2 protein expression was found to be significantly lower in thyroid cancer tissue than normal tissues (P<0.05). Western blot: The relative amount of CCNG2 protein in thyroid cancer tissue was respectively found to be significantly lower than in normal tissues (P<0.05), correlating with lymph node metastasis, clinic stage and histological grade (P<0.05), but not gender, age or tumor size (P>0.05). Loss of CCNG2 expression correlated significantly with poor overall survival time on Kaplan-Meier analysis (P<0.05). The results for biological functions showed that K1 cell transfected CCNG2 had a lower survival fraction, a greater percentage in the G0/G1 phases, and lower cyclin-dependent kinase 2 (CDK2) protein expression compared with K1 cells non-transfected with CCNG2 (P<0.05). CCNG2 expression decreased in thyroid cancer and correlated significantly lymph node metastasis, clinic stage, histological grade and poor overall survival, suggesting that CCNG2 may play important roles as a negative regulator in thyroid cancer K1 cells by promoting degradation of CDK2.

Overexpression of an oligopeptide transporter gene enhances heat tolerance in transgenic rice (Oligopeptide transporter 관여 유전자 도입 형질전환벼의 고온스트레스 내성 증진)

  • Jeong, Eun-Ju;Song, Jae-Young;Yu, Dal-A;Kim, Me-Sun;Jung, Yu-Jin;Kang, Kwon Kyoo;Park, Soo-Chul;Cho, Yong-Gu
    • Journal of Plant Biotechnology
    • /
    • v.44 no.3
    • /
    • pp.296-302
    • /
    • 2017
  • Rice (Oryza sativa) cultivars show an impairment of growth and development in response to abiotic stresses such as drought, salinity, heat and cold at the early seedling stage. The tolerance to heat stress in plants has been genetically modulated by the overexpression of heat shock transcription factor genes or proteins. In addition to a high temperature-tolerance that has also been altered by elevating levels of osmolytes, increasing levels of cell detoxification enzymes and through altering membrane fluidity. To examine the heat tolerance in transgenic rice plants, three OsOPT10 overexpressing lines were characterized through a physiological analysis, which examined factors such as the electrolyte leakage (EL), soluble sugar and proline contents. We further functionally characterized the OsOPT10 gene and found that heat induced the expression of OsOPT10 and P5CS gene related proline biosynthesis. It has been suggested that the expression of OsOPT10 led to elevated heat tolerance in transgenic lines.

APEX-1 Regulates Cell Proliferation through GDNF/GFRα1 Signaling (APEX-1은 GDNF/GFRα1 시그널을 통해 세포증식을 조절한다)

  • Kim, Hong-Beum;Hariharasudhan, Gurusamy;Youn, Cha-Kyung
    • Journal of Life Science
    • /
    • v.23 no.10
    • /
    • pp.1183-1191
    • /
    • 2013
  • Human apurinic/apyrimidinic endonuclease (APEX-1) is a multifunctional protein that is capable of repairing abasic sites and single-strand breaks in damaged DNA. In addition, it serves as a redox-modifying factor for a number of transcription factors. Identifying the transcriptional targets of APEX-1 is essential for understanding how it affects various cellular outcomes. Expression array analysis was used to identify glial cell-derived neurotropic factor receptor ${\alpha}1$ ($GFR{\alpha}1$), which is an encoding receptor for the glial cell-derived neurotropic factor (GDNF) family, the expression of which is induced by APEX-1. A target of GDNF/$GFR{\alpha}$ signaling, c-Src (Tyr418) was strongly phosphorylated by GNDF in the APEX-1 expressing cells. Moreover, GDNF initiated cell proliferation, measured by counting the number of cells, in the APEX-1 expressing cells. Importantly, the down-regulation of APEX-1 by siRNA caused a marked reduction in the $GFR{\alpha}1$ expression level, and it reduced the ability of GDNF to phosphorylate c-Src (Tyr418) and stimulate cell proliferation. These results demonstrate an association between APEX-1 and GDNF/$GFR{\alpha}$ signaling and suggest a potential molecular mechanism for the involvement of APEX-1 in cell survival and proliferation.

G1 Arrest of U937 Human Monocytic Leukemia Cells by Sodium Butyrate, an HDAC Inhibitor, Via Induction of Cdk Inhibitors and Down-regulation of pRB Phosphorylation (Cdk inhibitors의 발현 증가 및 pRB 인산화 저해에 의한 HDAC inhibitor인 sodium butyrate에 의한 인체백혈병세포의 G1 arrest유발)

  • Choi, Yung-Hyun
    • Journal of Life Science
    • /
    • v.19 no.7
    • /
    • pp.871-877
    • /
    • 2009
  • We investigated the effects of sodium butyrate, a histone deacetylase inhibitor, on the cell cycle progression in human monocytic leukemia U937 cells. Exposure of U937 cells to sodium butyrate resulted in growth inhibition, G1 arrest of the cell cycle and induction of apoptosis in a dose-dependent manner as measured by MTT assay and flow cytometry analysis. The increase in G1 arrest was associated with the down-regulation in cyclin D1, E, A, cyclin-dependent kinase (Cdk) 4 and 6 expression, and up-regulation of Cdk inhibitors such as p21 and p27. Sodium butyrate treatment also inhibited the phosphorylation of retinoblastoma protein (pRB) and p130, however, the levels of transcription factors E2F-1 and E2F-4 were not markedly modulated. Furthermore, the down-regulation of phosphorylation of pRB and p130 by this compound was associated with enhanced binding of pRB and E2F-1, as well as p130 and E2F-4, respectively. Overall, the present results demonstrate a combined mechanism involving the inhibition of pRBjp130 phosphorylation and induction of Cdk inhibitors as targets for sodium butyrate that may explain some of its anti-cancer effects in U937 cells.

Melittin inhibits cell migration and invasion via blocking of the epithelial-mesenchymal transition (EMT) in lung cancer cells (EMT 억제를 통한 멜리틴의 폐암세포 이동 및 침투 억제 효과)

  • Cho, Hyun-Ji;Jeong, Yun-Jeong;Kim, Mun-Hyeon;Chung, Il-Kyung;Kang, Dong Wook;Chang, Young-Chae
    • Korean Journal of Food Science and Technology
    • /
    • v.50 no.1
    • /
    • pp.105-110
    • /
    • 2018
  • Melittin is the main component of apitoxin (bee venom) that has been reported to have anti-inflammatory and anti-cancer effects. Herein, we demonstrated that inhibition of epithelial-mesenchymal transition (EMT) by melittin causes suppression of cancer cell migration and invasion. Melittin significantly suppressed the epidermal growth factor (EGF)-induced cell migration and invasion in lung cancer cells. Moreover, melittin up-regulated the expression of epithelial marker protein, E-cadherin, and down-regulated the expression of EMT related proteins, vimentin and fibronectin. Mechanistic studies revealed that melittin markedly suppressed the expression of EMT mediated transcription factors, ZEB2, Slug, and Snail. The EGF-induced phosphorylation of AKT, mTOR, P70S6K, and 4EBP1 was also inhibited by melittin, but not that of ERK and JNK. Therefore, the inhibitory effect of melittin on migration and invasion of lung cancer cells may be associated with the inhibition of EMT via blocking of the AKT-mTOR-P70S6K-4EBP1 pathway.

Isolation and characterization of Auxin/indole-3-acetic acid 1 (Aux/IAA1) gene from poplar (Populus alba × P. glandulosa) (현사시나무에서 Auxin/indole-3-acetic acid 1 (Aux/IAA1) 유전자 분리 및 발현 특성 구명)

  • Bae, Eun-Kyung;Choi, Young-Im;Lee, Hyoshin;Choi, Ji Won
    • Journal of Plant Biotechnology
    • /
    • v.46 no.3
    • /
    • pp.180-188
    • /
    • 2019
  • Auxin plays a crucial regulatory role in plant growth and development processes. Three major classes of auxin-responsive transcription factors controlled by the Auxin/indole-3-acetic acid (Aux/IAA), Gretchen Hagen 3 (GH3), and small auxin up RNA (SAUR) genes regulate auxin signaling. Aux/IAA, in particular, encodes short-lived nuclear proteins that accumulate rapidly in response to auxin signaling. In this study, we isolated a PagAux/IAA1 gene from poplar (Populus alba ${\times}$ P. glandulosa) and investigated its expression characteristics. The PagAux/IAA1 cDNA codes for putative 200 amino acids polypeptide containing four conserved domains and two nuclear localization signals (NLSs). Utilizing Southern blot analysis, we confirmed that a single copy of the PagAux/IAA1 gene was present in the poplar genome. The expression of this gene is specific to leaves and flowers of the poplar. PagAux/IAA1 expressed in the early exponential growth phase of cell-cultured in suspension. PagAux/IAA1 expression level reduced in drought and salt stress conditions, and the presence of plant hormones such as abscisic acid. However, expression enhanced in cold stress, cambial cell division, and presence of plant hormones such as gibberellic acid and jasmonic acid. Thus, these results suggest that PagAux/IAA1 participates in cold stress response as well as developmental processes in the poplar.

Conservative Genes among 1,309 Species of Prokaryotes (원핵생물 1,309종의 보존적 유전자)

  • Lee, Dong-Geun
    • Journal of Life Science
    • /
    • v.32 no.6
    • /
    • pp.463-467
    • /
    • 2022
  • As a result of applying the COG (Cluster of Orthologous Groups of Protein) algorithm to 1,309 species to confirm the conserved genes of prokaryotes, ribosomal protein S11 (COG0100) was identified. The numbers of conservative genes were 2, 5, 5, and 6 in 1,308, 1,307, 1,306, and 1,305 species, respectively. Twenty-nine genes were conserved in over 1,302 species, and they encoded 23 ribosomal proteins, 3 tRNA synthetases, 2 translation factors, and 1 RNA polymerase subunit. Most of them were related to protein production, suggesting the importance of protein expression in prokaryotes. The highest conservative COG was COG0048 (ribosomal protein S12) among the 29 COGs. The 29 conserved genes usually have one protein for each prokaryote. COG0090 (ribosomal protein L2) had not only the lowest conservation value but also the largest standard deviation of phylogenetic distance value. As COG0090 is not only a member of the ribosome, but also a regulator of replication and transcription, it could be inferred that prokaryotes have large variations in COG0090 to survive in various environments. This study could provide data necessary for basic science, tumor control, and development of antibacterial agents.

Involvement of Multiple Signaling Molecules in Peptidoglycan-induced Expression of Interleukin-1α in THP-1 Monocytes/Macrophages (THP-1 단핵구의 펩티도글리칸 유래 인터루킨-1 알파 발현에서 TLR2, PI3K/Akt/mTOR, MAPKs의 역할)

  • Heo, Weon;Son, Yonghae;Cho, Hyok-rae;Kim, Koanhoi
    • Journal of Life Science
    • /
    • v.32 no.6
    • /
    • pp.421-429
    • /
    • 2022
  • The expression of interleukin-1α (IL-1α) is elevated in monocytic cells, such as monocytes and macro-phages, within atherosclerotic arteries, yet the cellular molecules involved in cytokine upregulation remain unclear. Because peptidoglycan (PG), a major component of gram-positive bacterial cell walls, is detected within the inflammatory cell-rich regions of atheromatous plaques, it was investigated if PG contributes to IL-1α expression in monocytes/macrophages. Exposure of THP-1 monocytic cells to PG resulted in elevated levels of IL-1α gene transcripts and increased secretion of IL-1α protein. The transcription and secretion of IL-1α were abrogated by OxPAPC, an inhibitor of TLR2/4, but not by polymyxin B that inhibits lipopolysaccharide-induced TLR4 activation. To understand the molecular mechanisms of the inflammatory responses due to bacterial pathogen-associated molecular patterns (PAMPs) in diseased arteries, we attempted to determine the cellular factors involved in the PG-induced upregulation of IL-1α expression. Pharmacological inhibition of cell signaling pathways with LY294002 (a PI3K inhibitor), Akti IV (an inhibitor of Akt activation), rapamycin (an mTOR inhibitor), U0126 (a MEK inhibitor), SB202190 (a p38 MAPK inhibitor), SP6001250 (a JNK inhibitor), and DPI (a NOX inhibitor) also significantly attenuated the PG-mediated expression of IL-1α. These results suggest that PG induces the monocytic or macrophagic expression of IL-1α, thereby contributing to vascular inflammation, via multiple signaling molecules, including TLR2, PI3K/Akt/mTOR, and MAPKs.