• Title/Summary/Keyword: PPAR

Search Result 618, Processing Time 0.029 seconds

Repression of $PPAR{\gamma}$ Activity on Adipogenesis by $17{\beta}$-estradiol in Differentiated 3T3-L1 Cell

  • Yoon, Mi-Chung;Jeong, Sun-Hyo
    • Biomedical Science Letters
    • /
    • v.15 no.3
    • /
    • pp.179-185
    • /
    • 2009
  • In our previous report, we showed that $PPAR{\gamma}$ does not influence adipogenesis in females with functioning ovaries, indicating that $PPAR{\gamma}$ activity on adipogenesis is associated with sex-related factors. Among the sex-related factors, estrogen has been recognized as a major factor in inhibiting adiposgenesis in females. Thus, we hypothensized that $17{\beta}$-estradiol (E) inhibits 3T3-L1 cell adipogenesis by preventing $PPAR{\gamma}$ activity. E decreased triglyceirde accumulation in differentiated 3T3-L1 cells compared with control group. E also decreased the expression of $PPAR{\gamma}$ mRNA as well as $PPAR{\gamma}$ dependent adipocyte-specific genes, such as adipocyte fatty acid binding protein and tumor necrosis factor $\alpha$. In addition, E not only decreased luciferase reporter activity by $PPAR{\gamma}$, but also transfection of estrogen receptor $\alpha$ ($ER{\alpha}$) or $ER{\beta}$ led to decreases in $PPAR{\gamma}$ reporter gene activation. Moreover, E-activated ERs significantly decreased the luciferase reporter gene activation induced by $PPAR{\gamma}$ transfection, suggesting that estrogen-activated ERs inhibit $PPAR{\gamma}$-dependent transactivation. Accordingly, our results demonstrate that E inhibits the action of $PPAR{\gamma}$ on adipogenesis through E activated ER, providing evidence that lack of estrogen may potentiate $PPAR{\gamma}$ action on adipogenesis.

  • PDF

PPARγ Physiology and Pathology in Gastrointestinal Epithelial Cells

  • Thompson, E. Aubrey
    • Molecules and Cells
    • /
    • v.24 no.2
    • /
    • pp.167-176
    • /
    • 2007
  • Peroxisome proliferator-activated receptor-gamma ($PPAR{\gamma}$) is expressed at very high levels in the gastrointestinal epithelium. Many of the functions of $PPAR{\gamma}$ in gastrointestinal epithelial cells have been elucidated in recent years, and a pattern is emerging which suggests that this receptor plays an important role in gastrointestinal physiology. There is also strong evidence that $PPAR{\gamma}$ is a colon cancer suppressor in pre-clinical rodent models of sporadic colon cancer, and there is considerable interest in exploitation of $PPAR{\gamma}$ agonists as prophylactic or chemopreventive agents in colon cancer. Studies in mice and in human colon cancer cell lines suggest several mechanisms that might account for the tumor suppressive effects of $PPAR{\gamma}$ agonists, although it is not in all cases clear whether these effects are altogether mediated by $PPAR{\gamma}$. Conversely, several reports suggest that $PPAR{\gamma}$ agonists may promote colon cancer under certain circumstances. This possibility warrants considerable attention since several million individuals with type II diabetes are currently taking $PPAR{\gamma}$ agonists. This review will focus on recent data related to four critical questions: what is the physiological function of $PPAR{\gamma}$ in gastrointestinal epithelial cells; how does $PPAR{\gamma}$ suppress colon carcinogenesis; is $PPAR{\gamma}$ a tumor promoter; and what is the future of $PPAR{\gamma}$ in colon cancer prevention?

Peroxisome proliferator-activated receptor $\alpha$(PPAR$\alpha$) and its clinical significance

  • 윤미정
    • The Zoological Society Korea : Newsletter
    • /
    • v.18 no.2
    • /
    • pp.6-11
    • /
    • 2001
  • Peroxisome proliferator-activated receptor $\alpha$ (PPAR$\alpha$)에 대한 본격적인 연구는 고지혈증 치료제인 fibrate류의 약물들이 PPAR$\alpha$ activator로 작용한다는 사실이 밝혀짐으로써 크게 증대되었다. PPAR$\alpha$는 fibrate를 포함한 다양한 종류의 peroxisome proiferator (PP)에 의해 활성화되는데 이들을 쥐에 단기간 투여할 경우 간의 peroxisome수와 지 방산 산화효소의 유전자발현이 증가되고 장기간 투여 할 경우 간암을 발생시키지만, fibrate류의 약물들을 고지혈증 환자에게 투여 할 경우 간암을 발생시키지 않으므로써 PP에 대한 반응성에 있어서 species difference를 나타낸다 PPAR$\alpha$는 핵에 존재하는 orphan receptor로서 PP에 의해 활성화되어 9-cis-retinoic acid receptor(RXR)와 heterodimer를 이룬 후 target gene들의 upstream에 있는 peroxisome proliferator response element (PPRE)에 결합하여 target gene들의 발현을 조절한다. 지금까지 연구된 PPAR$\alpha$의 target gene들은 모두 lipid와 lipoprotein 대사를 조절하는 것으로 알려져 있으며, 이러 한 결과들을 기초로 lipid 대사 및 energy balance와 관련된 질병들 - 동맥경화증, 관상동맥질환, 비만, 제 2형 당뇨병 등에서 PPAR$\alpha$의 역할이 집중적으로 연구되고 있다. PPAR$\alpha$가 활성화되면 lipoprotein lipase와 HDL이 증가되고 apo C-III가 감소됨으로써 동맥경화증에 대한 예방적 기능을 나타내고, 몸무게를 감소시킴으로써 비만을 방지할 수 있으며, 인슐린 감수성을 증가시켜 제 2형 당뇨병의 치료효과를 가지는 것으로 보인다. 그러나 PPAR$\alpha$-null mouse에서는 이러한 효과들이 나타나지 않는 것으로 보아 이들 질병에서 PPAR$\alpha$가 중요한 역할을 하는 것으로 생각된다.

  • PDF

Cyclooxygenase-2 Inhibitor Parecoxib Was Disclosed as a PPAR-γ Agonist by In Silico and In Vitro Assay

  • Xiao, Bin;Li, Dan-dan;Wang, Ying;Kim, Eun La;Zhao, Na;Jin, Shang-Wu;Bai, Dong-Hao;Sun, Li-Dong;Jung, Jee H.
    • Biomolecules & Therapeutics
    • /
    • v.29 no.5
    • /
    • pp.519-526
    • /
    • 2021
  • In a search for effective PPAR-γ agonists, 110 clinical drugs were screened via molecular docking, and 9 drugs, including parecoxib, were selected for subsequent biological evaluation. Molecular docking of parecoxib to the ligand-binding domain of PPAR-γ showed high binding affinity and relevant binding conformation compared with the PPAR-γ ligand/antidiabetic drug rosiglitazone. Per the docking result, parecoxib showed the best PPAR-γ transactivation in Ac2F rat liver cells. Further docking simulation and a luciferase assay suggested parecoxib would be a selective (and partial) PPAR-γ agonist. PPAR-γ activation by parecoxib induced adipocyte differentiation in 3T3-L1 murine preadipocytes. Parecoxib promoted adipogenesis in a dose-dependent manner and enhanced the expression of adipogenesis transcription factors PPAR-γ, C/EBPα, and C/EBPβ. These data indicated that parecoxib might be utilized as a partial PPAR-γ agonist for drug repositioning study.

ANTI-INFLAMMATORY EFFECTS OF PPARγ ON HUMAN DENTAL PULP CELLS (치수세포에서 PPARγ의 항 염증작용에 관한 연구)

  • Kim, Jeong-Hee
    • Restorative Dentistry and Endodontics
    • /
    • v.31 no.3
    • /
    • pp.203-214
    • /
    • 2006
  • Dental pulp is a loose, mesenchymal tissue almost entirely enclosed in the dentin. It consists of cells, ground substance, and neural and vascular supplies. Damage to the dental pulp by mechanical, chemical, thermal, and microbial irritants can provoke various types of inflammatory response. Pulpal inflammation leads to the tissue degradation, which is mediated in part by Matrix metalloproteinase leads to accelerate extracellular matrix degradation with pathological pathway We have now investigated the induction of MMPs and inflammatory cytokines by Lipopolysaccharide (LPS) control of inflammatory mediators by peroxisome proliferator-activated receptors (PPARs). Human dental pulp cells exposed to various concentrations of LPS ($1-10{\mu}g/ml$) revealed elevated levels of MMP-2 and MMP-9 at 24 hrs of culture. LPS also stimulated the production of ICAM-1, VCAM-1, $IL-1{\beta},\;and\;TNF-{\alpha}$. Adenovirus $PPAR{\gamma}\;(Ad/PPAR{\gamma})\;and\;PPAR{\gamma}$ agonist rosiglitazone reduced the synthesis of MMPs, adhesion molecules and pro-inflammatory cytokines. The inhibitory effect of $Ad/PPAR{\gamma}$ was higher than that of $PPAR{\gamma}$ agonist. These result offer new insights in regard to the anti-inflammatory potential of $PPAR{\gamma}$ in human dental pulp cell.

Protein Arginine Methyltransferase 5 (PRMT5) Regulates Adipogenesis of 3T3L-1 Cells (단백질 아르기닌 메틸전이효소 5(PRMT5)에 의한 3T3L-1 세포의 지방세포 분화 조절)

  • Jang, Min Jung;Yang, Ji Hye;Kim, Eun-Joo
    • Journal of Life Science
    • /
    • v.28 no.7
    • /
    • pp.765-771
    • /
    • 2018
  • Peroxisome proliferator-activated receptor gamma ($PPAR{\gamma}$) is a key transcription factor that regulates adipogenesis, and epigenetic control of $PPAR{\gamma}$ is of great interest in obesity-inhibition research. Our previous study showed that CACUL1 (CDK2-associated cullin domain 1) acts as a corepressor that inhibits $PPAR{\gamma}$ transcriptional activity and adipocyte differentiation. Here, we investigated the roles of protein arginine methyltransferase 5 (PRMT5), a novel binding partner of CACUL1, in regulating $PPAR{\gamma}$. The interaction between PRMT5 and CACUL1 was shown by immunoprecipitation assay in vivo and GST pulldown assay in vitro. As shown by luciferase reporter assay, PRMT5 and CACUL1 cooperated to inhibit the transcriptional activity of $PPAR{\gamma}$. The suppressive role of PRMT5 in adipogenesis was examined by Oil Red O staining using 3T3-L1 cells, which stably overexpress or deplete PRMT5. Overexpression of PRMT5 suppresses $PPAR{\gamma}$-mediated adipogenesis, whereas PRMT5 knockdown increases lipid accumulation in 3T3-L1 cells. Consistently, PRMT5 attenuates the expression of Lpl and aP2, the target genes of $PPAR{\gamma}$, as demonstrated by RT-qPCR analysis. Overall, these results suggest that PRMT5 interacts with CACUL1 to impair the transcriptional activity of $PPAR{\gamma}$, leading to the inhibition of adipocyte differentiation. Therefore, the regulation of PRMT5 enzymatic activity may provide a clue to develop an anti-obesity drug.

Molecular Cloning and mRNA Expression of the Bovine Peroxisome Proliperator Receptor Gamma(PPARγ) (한우 PPARγ 유전자의 동정과 mRNA의 발현)

  • Jeoung, Y.H.;Lee, S.M.;Park, H.Y.;Yoon, D.H.;Choi, J.G.;Moon, S.J.;Kang, M.J.
    • Journal of Animal Science and Technology
    • /
    • v.46 no.1
    • /
    • pp.23-30
    • /
    • 2004
  • The peroxisome proliferator-activated receptor $\gamma$(PPAR$\gamma$), a member of the steroid/thyroid nuclear hormone receptor suferfamily of ligand-activated transcription factor, is an important regulator of adipocyte gene expression and differentiation. In this studies, we report the identification, characterization, and expression of a Hanwoo PPAR$\gamma$ gene. The PPAR$\gamma$ cDNA sequence of the Hanwoo show strong conservation with the corresponding sequences reported in other species except of three amino acid sequences. The distribution of PPAR$\gamma$ mRNA in various tissues of Korean cattle aged 12 months were investigated using Northern Blot analysis. The highest expression was detected in adipose tissue, more lower expression was detected in colon, small intestine, kidney, lung, while expression was not detected in brain, heart. PPAR$\gamma$ expression was higher in adipose tissue of Korean cattle when aged 30 months than aged 12 months. These results indicated PPAR$\gamma$, regulator adipocyte gene expression and differentiation, related on adipose differentiation in Korean native cattle(HANWOO).

Ciglitazone, in Combination with All trans Retinoic Acid, Synergistically Induces PTEN Expression in HL-60 Cells (백혈병세포에서 PTEN 발현에 대한 Ciglitazone과 retinoic Acid의 항진 작용)

  • Lee Seung-Ho;Park Chul-Hong;Kim Byeong-Su
    • Journal of Food Hygiene and Safety
    • /
    • v.21 no.3
    • /
    • pp.171-180
    • /
    • 2006
  • Peroxisome proliferatorr-activated receptor-gamma $(PPAR{\gamma})$ must form a heterodimer with the retinoid-X receptor (RXR) to bind DNA, and its transcriptional activity is thought to be maximized by ligands specific for either receptor. Activated $(PPAR{\gamma})$ and $(PPAR{\gamma})$ ligands may influence tumor growth through regulation of the tumor suppressor PTEN. Our aim in this study was to determine whether co-stimulation with the $(PPAR{\gamma})$ ligand, ciglitazone, and RXR ligand can synergistically upregulate PTEN in human acute promyelocytic leukemia (APL) cells and consequently potentate the inhibition of cell growth and cell cycle progression of these cells. Human leukemia cell line, HL-60 cells were exposed to all-trans-retinol and ciglutazone. The PTEN expression was measured as the level of PTEN mRNA expression by RT-PCR and as the level of PTEN expression by western blot analysis. Cell cycle analysis was carried out by a propidium iodide (PI) staining method and analyzed with a FACScan. The $(PPAR{\gamma})$ ligand, ciglitazone, and the RXR ligand, retinoic acid, upregulated PTEN expression by HL-60 cells in time- and dose-dependent manners, respectively. This was significantly enhanced by a combination of both ciglitazone and retinoic acid. Moreover, these compounds synergistically induced arrests of both cell growth and the $G_l$ phase of the cell cycle. Thus, the activation of the $(PPAR{\gamma})$:RXR heterodimer may represent a regulatory pathway for human leukemia cells and there may be important roles for $(PPAR{\gamma})$ and RXR ligands in prophylactic and therapeutic approaches fur controlling leukemia through the upregulation of PTEN.

Ginsenoside Rf inhibits cyclooxygenase-2 induction via peroxisome proliferator-activated receptor gamma in A549 cells

  • Song, Heewon;Park, Joonwoo;Choi, KeunOh;Lee, Jeonggeun;Chen, Jie;Park, Hyun-Ju;Yu, Byeung-Il;Iida, Mitsuru;Rhyu, Mee-Ra;Lee, YoungJoo
    • Journal of Ginseng Research
    • /
    • v.43 no.2
    • /
    • pp.319-325
    • /
    • 2019
  • Background: Ginsenoside Rf is a ginseng saponin found only in Panax ginseng that affects lipid metabolism. It also has neuroprotective and antiinflammatory properties. We previously showed that Korean Red Ginseng (KRG) inhibited the expression of cyclooxygenase-2 (COX-2) by hypoxia via peroxisome proliferator-activated receptor gamma ($PPAR{\gamma}$). The aim of the current study was to evaluate the possibility of ginsenoside Rf as an active ingredient of KRG in the inhibition of hypoxia-induced COX-2 via $PPAR{\gamma}$. Methods: The effects of ginsenoside Rf on the upregulation of COX-2 by hypoxia and its antimigration effects were evaluated in A549 cells. Docking of ginsenoside Rf was performed with the $PPAR{\gamma}$ structure using Surflex-Dock in Sybyl-X 2.1.1. Results: $PPAR{\gamma}$ protein levels and peroxisome proliferator response element promoter activities were promoted by ginsenoside Rf. Inhibition of COX-2 expression by ginsenoside Rf was blocked by the $PPAR{\gamma}-specific$ inhibitor, T0070907. The $PPAR{\gamma}$ inhibitor also blocked the ability of ginsenoside Rf to suppress cell migration under hypoxia. The docking simulation results indicate that ginsenoside Rf binds to the active site of $PPAR{\gamma}$. Conclusions: Our results demonstrate that ginsenoside Rf inhibits hypoxia induced-COX-2 expression and cellular migration, which are dependent on $PPAR{\gamma}$ activation. These results suggest that ginsenoside Rf has an antiinflammatory effect under hypoxic conditions. Moreover, docking analysis of ginsenoside Rf into the active site of $PPAR{\gamma}$ suggests that the compound binds to $PPAR{\gamma}$ in a position similar to that of known agonists.

A Molecular Study of Sopungsungi-won(Shufengshunqiyuan) about Regulation of PPARs in Mouse NMu2Li Liver Cells and C2C12 Skeletal Muscle Myogenic Progenital Cells (소풍순기원(疏風順氣元)이 mouse의 NMu2Li 간세포와 C2C12 골격근세포에서 PPARs 조절의 분자기전에 미치는 영향)

  • Oh, Young-Jin;Shin, Soon-Shik;Yoon, Mi-Chung;Kim, Bo-Kyung
    • Journal of Oriental Neuropsychiatry
    • /
    • v.20 no.1
    • /
    • pp.147-164
    • /
    • 2009
  • Objectives : We investigated the effects of Sopungsungi-won(Shu!engshunqiyuan) (SSEx1, SSEx2) to treat the metabolic syndrome by the molecular mechanism of regulation of PPAR and modulation of mitochondrial MCAD, VLCAD mRNA expression. Methods : Mouse NMu2Li liver cells and C2C12 skeletal muscle myogenic progenital cells were transiently transfected with expression plasmids for PPAR(PPAR${\alpha}$, PPAR${\delta}$), a luciferase reporter gene construct containing 3 copies of the PPRE from the rat acyl-CoA oxidase gene and ${\beta}$-galactosidase gene. Cells were treated with several concentrated kinds of SSEx1, SSEx2 at the initial time of culture and analyzed PPAR${\alpha}$, PPAR${\delta}$ reporter gene activity using spectrophotometer (405 nm). Total RNA was extracted from SSEx1, SSEx2 and measured mRNA levels of mitochondrial MCAD, VLCAD. Representative RT-PCR bands are shown. Results : 1. SSEx1 increased the expression of PPAR${\alpha}$ reporter gene activities at 0.1 ${\mu}$g/ml (p${\mu}$g/ml (p<0.05), SSEx2 at 0.1 ${\mu}$g/ml (p${\mu}$g/ml (p<0.05) significantly in NMu2Li liver cell lines. 2. SSEx1 increased the expression of PPAR${\alpha}$ reporter gene activities at 1 ${\mu}$g/ml (p${\mu}$g/ml (p${\alpha}$ reporter gene activities in C2C12 skeletal muscle cells. 4. SSEx1 increased the modulation of mitochondrial MCAD mRNA expression (p<0.05) significantly in NMu2Li liver cell lines. 5. SSEx1, SSEx2 both increased the modulation of mitochondrial MCAD mRNA expression (p<0.05) significantly in C2C12 skeletal muscle cells. Conclusions : These results show the SSEx1, SSEx2 can be used as therapeutic agent for metabolic syndrome and it's molecular mechanisms of PPAR more contribute to the activation of PPAR${\alpha}$ then PPAR${\delta}$ reporter gene activities and it's total RNA more contribute to the modulation of mitochondrial MCAD then VLCAD mRNA expression.

  • PDF