• 제목/요약/키워드: cellular signaling pathway

검색결과 434건 처리시간 0.026초

Arsenite Acutely Decreases Nitric Oxide Production via the ROS-Protein Phosphatase 1-Endothelial Nitric Oxide Synthase-Thr497 Signaling Cascade

  • Seo, Jungwon;Lee, Jee Young;Sung, Min-Sun;Byun, Catherine Jeonghae;Cho, Du-Hyong;Lee, Hyeon-Ju;Park, Jung-Hyun;Cho, Ho-Seong;Cho, Sung-Jin;Jo, Inho
    • Biomolecules & Therapeutics
    • /
    • 제22권6호
    • /
    • pp.510-518
    • /
    • 2014
  • Chronic (>24 h) exposure of arsenite, an environmental toxicant, has shown the decreased nitric oxide (NO) production in endothelial cells (EC) by decreasing endothelial NO synthase (eNOS) expression and/or its phosphorylation at serine 1179 ($eNOS-Ser^{1179}$ in bovine sequence), which is associated with increased risk of vascular diseases. Here, we investigated the acute (<24 h) effect of arsenite on NO production using bovine aortic EC (BAEC). Arsenite acutely increased the phosphorylation of $eNOS-Thr^{497}$, but not of $eNOS-Ser^{116}$ or $eNOS-Ser^{1179}$, which was accompanied by decreased NO production. The level of eNOS expression was unaltered under this condition. Treatment with arsenite also induced reactive oxygen species (ROS) production, and pretreatment with a ROS scavenger N-acetyl-L-cysteine (NAC) completely reversed the observed effect of arsenite on $eNOS-Thr^{497}$ phosphorylation. Although protein kinase C (PKC) and protein phosphatase 1 (PP1) were reported to be involved in $eNOS-Thr^{497}$ phosphorylation, treatment with PKC inhibitor, Ro318425, and overexpression of various PKC isoforms did not affect the arsenite-stimulated $eNOS-Thr^{497}$ phosphorylation. In contrast, treatment with PP1 inhibitor, calyculin A, mimicked the observed effect of arsenite on $eNOS-Thr^{497}$ phosphorylation. Lastly, we found decreased cellular PP1 activity in arsenite-treated cells, which was reversed by NAC. Overall, our study demonstrates firstly that arsenite acutely decreases NO production at least in part by increasing $eNOS-Thr^{497}$ phosphorylation via ROS-PP1 signaling pathway, which provide the molecular mechanism underlying arsenite-induced increase in vascular disease.

Transcriptional Upregulation of Plasminogen Activator Inhibitor-1 in Rat Primary Astrocytes by a Proteasomal Inhibitor MG132

  • Cho, Kyu Suk;Kwon, Kyoung Ja;Jeon, Se Jin;Joo, So Hyun;Kim, Ki Chan;Cheong, Jae Hoon;Bahn, Geon Ho;Kim, Hahn Young;Han, Seol Heui;Shin, Chan Young;Yang, Sung-Il
    • Biomolecules & Therapeutics
    • /
    • 제21권2호
    • /
    • pp.107-113
    • /
    • 2013
  • Plasminogen activator inhibitor-1 (PAI-1) is a member of serine protease inhibitor family, which regulates the activity of tissue plasminogen activator (tPA). In CNS, tPA/PAI-1 activity is involved in the regulation of a variety of cellular processes such as neuronal development, synaptic plasticity and cell survival. To gain a more insights into the regulatory mechanism modulating tPA/PAI-1 activity in brain, we investigated the effects of proteasome inhibitors on tPA/PAI-1 expression and activity in rat primary astrocytes, the major cell type expressing both tPA and PAI-1. We found that submicromolar concentration of MG132, a cell permeable peptide-aldehyde inhibitor of ubiquitin proteasome pathway selectively upregulates PAI-1 expression. Upregulation of PAI-1 mRNA as well as increased PAI-1 promoter reporter activity suggested that MG132 transcriptionally increased PAI-1 expression. The induction of PAI-1 downregulated tPA activity in rat primary astrocytes. Another proteasome inhibitor lactacystin similarly increased the expression of PAI-1 in rat primary astrocytes. MG132 activated MAPK pathways as well as PI3K/Akt pathways. Inhibitors of these signaling pathways reduced MG132-mediated upregulation of PAI-1 in varying degrees and most prominent effects were observed with SB203580, a p38 MAPK pathway inhibitor. The regulation of tPA/PAI-1 activity by proteasome inhibitor in rat primary astrocytes may underlie the observed CNS effects of MG132 such as neuroprotection.

스타틴 그리고 배아줄기세포에서의 작용 (Statins and Their Effects on Embryonic Stem Cells)

  • 이미희;한용만;조이숙
    • 한국발생생물학회지:발생과생식
    • /
    • 제11권2호
    • /
    • pp.59-66
    • /
    • 2007
  • 배아줄기세포를 이용한 치료법 개발을 위해서는 배아줄기세포의 자가재생산 및 분화과정을 조절하는 분자적 기전을 이해하는 것이 매우 중요하다. 지질합성경로(Mevalonate pathway)에 작용하는 HMG-CoA 환원효소(Hydroxymethylglutaryl-coenzyme A reductase)의 억제제인 스타틴은 콜레스테롤 저하제로 잘 알려져 있으며, 콜레스테롤 이외에 단백질 isoprenylation의 기질로 작용하는 아이소프레노이드(Isoprenoids)(Farnesyl pyrophosphate(FPP), Geranylgeranyl pyrophosphate(GGPP))의 생성을 억제하는 효능을 가지고 있다. 스타틴에 의해 매개되는 표적단백질의 isoprenylation 억제는 다양한 세포내 신호전달과정에 영향을 미치게 되며, 결과적으로 세포기능을 조절하는데 핵심적인 역할을 하게 된다. 스타틴이 첨가된 배양배지에서 배양된 배아줄기세포는 자가재생산능이 억제되고 분화가 촉진되는데, 특히 지방/골세포 직계열로의 분화가 촉진된다. 배아줄기세포에서의 스타틴의 효과 및 작용기전에 대한 이해가 아직은 미비한 수준이나, 최근 우리 연구팀에서는 스타틴이 콜레스테롤 작용과는 무관하게 RhoA G-단백질의 세포내 분포 및 활성을 억제함으로써 배아 줄기세포의 자가재생산능을 억제하고 있음을 규명하였다. 스타틴 다면효과와 그 작용에 대한 이해는 배아줄기세포의 미분화 및 분화상태를 조절하는데 관여하는 분자적 조절기전을 이해하는데 중요한 모델이 될 수 있을 것으로 추정된다.

  • PDF

Gene Expression Profiling of Acetaminophen Induced Hepatotoxicity in Mice

  • Suh, Soo-Kyung;Jung, Ki-Kyung;Jeong, Youn-Kyoung;Kim, Hyun-Ju;Lee, Woo-Sun;Koo, Ye-Mo;Kim, Tae-Gyun;Kang, Jin-Seok;Kim, Joo-Hwan;Lee, Eun-Mi;Park, Sue-Nie;Kim, Seung-Hee;Jung, Hai-Kwan
    • Molecular & Cellular Toxicology
    • /
    • 제2권4호
    • /
    • pp.236-243
    • /
    • 2006
  • Microarray analysis of gene expression has become a powerful approach for exploring the biological effects of drugs, particularly at the stage of toxicology and safety assessment. Acetaminophen (APAP) has been known to induce necrosis in liver, but the molecular mechanism involved has not been fully understood. In this study, we investigated gene expression changes of APAP using microarray technology. APAP was orally administered with a single dose of 50 mg/kg or 500 mg/kg into ICR mice and the animals were sacrificed at 6, 24 and 72 h of APAP administration. Serum biochemical markers for liver toxicity were measured to estimate the maximal toxic time and hepatic gene expression was assessed using high-density oligonucleotide microarrays capable of determining the expression profile of >30,000 well-substantiated mouse genes. Significant alterations in gene expression were noted in the liver of APAP-administered mice. The most notable changes in APAP-administered mice were the expression of genes involved in apoptosis, cell cycle, and calcium signaling pathway, cystein metabolism, glutatione metabolism, and MAPK pathway. The majority of the genes upregulated included insulin-like growth factor binding protein 1, heme oxygenase 1, metallothionein 1, S100 calcium binding protein, caspase 4, and P21. The upregulation of apoptosis and cell cycle-related genes were paralleled to response to APAP. Most of the affected gene expressions were returned to control levels after 72 hr. In conclusion, we identified potential hepatotoxicity makers, and these expressions profiling lead to a better understanding of the molecular basis of APAP-induced hapatotoxicity.

대식세포에서 Nrf2/HO-1경로를 통한 청심연자음의 항산화효과 (Anti-oxidative Effect of Chungsimyeonja-um (CSYJE) via Nrf2/HO-1 Pathway Activity in Lipopolysaccharide (LPS) Induced RAW 264.7 Macrophages)

  • 전선홍;오솔라;김소정;전보희;성진영;김용민
    • 대한화장품학회지
    • /
    • 제46권3호
    • /
    • pp.253-263
    • /
    • 2020
  • 활성산소종(reactive oxygen species, ROS)은 우리 몸의 항상성 유지에 있어 중요한 역할을 한다. 그러나 과도한 ROS의 생성은 단백질, 지질, 핵산과 같은 세포 구성성분을 손상시키고 피부노화를 촉진시킨다. 이에 본 연구에서는 과도한 산화 스트레스를 예방하기 위해 Chungsimyeonja-um (CSYJE)의 항산화 효과를 확인하였다. 먼저 DPPH 및 ABTS assay를 실시하여 CSYJE의 항산화 효과를 확인한 결과 농도 의존적으로 radical 소거 활성을 확인하였다. 세포생존율 확인을 위해 MTT assay를 실시한 결과 1,000 ㎍/mL 농도에서 세포 독성이 없음을 확인하였다. 항산화 관련 단백질인 nuclear-E2-related factor 2 (Nrf2), Heme oxygenase-1 (HO-1)의 발현 수준을 확인하기 위해 western blotting을 실시한 결과 농도 의존적으로 발현이 증가하는 것을 확인하였다. 세포 내 ROS유발 물질인 lipopolysaccharide (LPS)로 ROS를 유도한 후, ROS생성 억제효과를 확인하기 위해 DCF-DA 염색법을 실시한 결과 농도 의존적으로 ROS 생성 억제효과를 확인하였으며 ROS의 생성으로 인한 염증성 사이토카인과 염증인자의 mRNA발현 수준을 확인하기 위해 real-time RT-PCR을 실시한 결과 농도 의존적으로 염증성 사이토카인과 염증인자의 mRNA 발현을 억제시켰다. 따라서, 본 연구는 Nrf2/HO-1 신호 전달 경로 활성을 통해 CSYJE의 항산화효과를 확인했으며 이는 CSYJE가 활성산소를 억제하여 항산화 화장품의 재료로서 사용될 수 있음을 시사한다.

Gene Expression Profiling of Genotoxicity Induced by MNNG in TK6 Cell

  • Suh, Soo-Kyung;Kim, Tae-Gyun;Kim, Hyun-Ju;Koo, Ye-Mo;Lee, Woo-Sun;Jung, Ki-Kyung;Jeong, Youn-Kyoung;Kang, Jin-Seok;Kim, Joo-Hwan;Lee, Eun-Mi;Park, Sue-Nie;Kim, Seung-Hee;Jung, Hai-Kwan
    • Molecular & Cellular Toxicology
    • /
    • 제3권2호
    • /
    • pp.98-106
    • /
    • 2007
  • Genotoxic stress triggers a variety of biological responses including the transcriptional activation of genes regulating DNA repair, cell survival and cell death. In this study, we investigated to examine gene expression profiles and genotoxic response in TK6 cells treated with DNA damaging agents MNNG (N-methyl-N'-nitrosoguanidine) and hydrogen peroxide $(H_2O_2)$. We extracted total RNA in three independent experiments and hybridized cRNA probes with oligo DNA chip (Applied Biosystems Human Genome Survey Microarray). We analyzed raw signal data with R program and AVADIS software and identified a number of deregulated genes with more than 1.5 log-scale fold change and statistical significancy. We indentified 14 genes including G protein alpha 12 showing deregulation by MNNG. The deregulated genes by MNNG represent the biological pathway regarding MAP kinase signaling pathway. Hydrogen peroxide altered 188 genes including sulfiredoxins. These results show that MNNG and $H_2O_2$ have both uniquely regulated genes that provide the potential to serve as biomarkers of exposure to DNA damaging agents.

단백질 인산화에 의해 매개되는 브라시노스테로이드 신호전달 연구의 최근 상황 (Update on Phosphorylation-Mediated Brassinosteroid Signaling Pathways)

  • 이유;김수환
    • 생명과학회지
    • /
    • 제22권3호
    • /
    • pp.428-436
    • /
    • 2012
  • 단백질 인산화는 세포의 활동을 조절하는 보편적인 과정이다. 브라시노스테로이드(brassinostreoid)에 의해 매개되는 신호전달은 브라시노스테로이드에 의해 활성화된 세포막상의 protein kinase 로부터 인산화되어 있는 전사인자들을 탈인산화하는 연속적인 인산화/탈인산화 과정이다. 브라시노스테로이드에 의해 매개되는 신호전달의 연구는 인산화에 관여하는 kinase 기질상의 아미노산을 밝히고, 그와 관련된 돌연변이체의 표현형을 알아봄으로써 급속하게 발전하였다. BRI1과 BAK1의 자기인산화(autophosphorylation), 상호인산화(transphosphorylation), 타이로신 인산화(tyrosine phosphorylation)를 밝힘으로써 그들의 조절작용을 식물의 생리학적, 발생학적 과정을 더 이해할 수 있는 장이 열렸다. 브라시노스테로이드에 의한 인산화는 수용체에 의해 매개되는 세포 내 함입(endocytosis)과 그에 뒤따르는 수용체의 파괴현상에서도 볼 수 있다. 인산화/탈인산화 과정에 관련하여 브라시노스테로이드에 의해 매개되는 신호전달은 더 연구할 여지가 많이 남아 있다. 이 총설은 단백질의 인산화/탈인산화 과정을 통한 브라시노스테로이드의 신호전달 연구의 최근 상황을 기술하였다.

Ventx1.1 as a Direct Repressor of Early Neural Gene zic3 in Xenopus laevis

  • Umair, Zobia;Kumar, Shiv;Kim, Daniel H.;Rafiq, Khezina;Kumar, Vijay;Kim, SungChan;Park, Jae-Bong;Lee, Jae-Yong;Lee, Unjoo;Kim, Jaebong
    • Molecules and Cells
    • /
    • 제41권12호
    • /
    • pp.1061-1071
    • /
    • 2018
  • From Xenopus embryo studies, the BMP4/Smad1-targeted gene circuit is a key signaling pathway for specifying the cell fate between the ectoderm and neuro-ectoderm as well as the ventral and dorsal mesoderm. In this context, several BMP4/Smad1 target transcriptional factors have been identified as repressors of the neuro-ectoderm. However, none of these direct target transcription factors in this pathway, including GATA1b, Msx1 and Ventx1.1 have yet been proven as direct repressors of early neuro-ectodermal gene expression. In order to demonstrate that Ventx1.1 is a direct repressor of neuro-ectoderm genes, a genome-wide Xenopus ChIP-Seq of Ventx1.1 was performed. In this study, we demonstrated that Ventx1.1 bound to the Ventx1.1 response cis-acting element 1 and 2 (VRE1 and VRE2) on the promoter for zic3, which is a key early neuro-ectoderm gene, and this Ventx1.1 binding led to repression of zic3 transcription. Site-directed mutagenesis of VRE1 and VRE2 within zic3 promoter completely abolished the repression caused by Ventx1.1. In addition, we found both the positive and negative regulation of zic3 promoter activity by FoxD5b and Xcad2, respectively, and that these occur through the VREs and via modulation of Ventx1.1 levels. Taken together, the results demonstrate that the BMP4/Smad1 target gene, Ventx1.1, is a direct repressor of neuro-ectodermal gene zic3 during early Xenopus embryogenesis.

Transforming Growth Factor β Inhibits MUC5AC Expression by Smad3/HDAC2 Complex Formation and NF-κB Deacetylation at K310 in NCI-H292 Cells

  • Lee, Su Ui;Kim, Mun-Ock;Kang, Myung-Ji;Oh, Eun Sol;Ro, Hyunju;Lee, Ro Woon;Song, Yu Na;Jung, Sunin;Lee, Jae-Won;Lee, Soo Yun;Bae, Taeyeol;Hong, Sung-Tae;Kim, Tae-Don
    • Molecules and Cells
    • /
    • 제44권1호
    • /
    • pp.38-49
    • /
    • 2021
  • Airway mucus secretion is an essential innate immune response for host protection. However, overproduction and hypersecretion of mucus, mainly composed of the gel-forming MUC5AC protein, are significant risk factors for patients with asthma and chronic obstructive pulmonary disease (COPD). The transforming growth factor β (TGFβ) signaling pathway negatively regulates MUC5AC expression; however, the underlying molecular mechanism is not fully understood. Here, we showed that TGFβ significantly reduces the expression of MUC5AC mRNA and its protein in NCI-H292 cells, a human mucoepidermoid carcinoma cell line. This reduced MUC5AC expression was restored by a TGFβ receptor inhibitor (SB431542), but not by the inhibition of NF-κB (BAY11-7082 or Triptolide) or PI3K (LY294002) activities. TGFβ-activated Smad3 dose-dependently bound to MUC5AC promoter. Notably, TGFβ-activated Smad3 recruited HDAC2 and facilitated nuclear translocation of HDAC2, thereby inducing the deacetylation of NF-κB at K310, which is essential for a reduction in NF-κB transcriptional activity. Both TGFβ-induced nuclear translocation of Smad3/HDAC2 and deacetylation of NF-κB at K310 were suppressed by a Smad3 inhibitor (SIS3). These results suggest that the TGFβ-activated Smad3/HDAC2 complex is an essential negative regulator for MUC5AC expression and an epigenetic regulator for NF-κB acetylation. Therefore, these results collectively suggest that modulation of the TGFβ1/Smad3/HDAC2/NF-κB pathway axis can be a promising way to improve lung function as a treatment strategy for asthma and COPD.

X선에 의한 연골세포 분화 억제 작용경로 (The mechanism of chondrogenesis inhibition by X-Irradiation)

  • 하종렬;임영빈;이윤애;손종경;이준일
    • 대한방사선기술학회지:방사선기술과학
    • /
    • 제26권1호
    • /
    • pp.91-97
    • /
    • 2003
  • 본 연구를 통해 X선에 의한 연골세포 분화 억제 작용경로를 조사하고자 하였다. 계배 limb bud 간충직세포를 배양하면서 여러 가지 선량(1-10Gy)의 X선을 조사하고 연골분화도를 조사한 결과, X선은 연골세포에 특이하게 발현되어 분화척도로 이용되는 typeII collagen의 발현과 proteoglycan의 축적을 저해하였다. 또한 세포내 신호전달 과정에서 중요한 매개자 역할을 하는 것으로 알려진 PKC 동위효소중 $PKC{\alpha}$의 발현을 저해하였다. 그러나 $PKC{\lambda}({\iota}),\;{\varepsilon}$ 등 다른 동위효소의 발현에는 별다른 영향을 미치지 못하였다. X선 조사에 의한 연골세포 분화 억제가 $PKC{\alpha}$의 downstream으로 알려져 있는 Erk-1을 통하여 이루어지는지 알아보기 위하여 Erk-1의 발현 및 인산화를 조사한 결과 X선은 그 발현에는 영향을 미치지 못했으나 인산화는 증가시켰다. 연골세포 분화 저해 효과가 Erk-1의 활성 변화에 의한 것인가를 확인하기 위하여 Erk-1을 인산화하는 MEK의 저해제인 PD98059를 처리하여 Erk-1의 인산화를 저해한 결과 X선 조사에 의한 연골분화 억제효과를 극복하는 것으로 나타났다. 또한 X선 조사가 분화 초기의 세포응집 과정에 어떤 영향을 미치는 지 알아보기 위하여 PNA 염색으로 조사한 결과 보선 조사는 세포응집을 저해하였다. 본 연구 결과를 종합하면 X선 조사는 분화 초기에 세포 응집을 억제하며 $PKC{\alpha}$의 발현을 저해하고 Erk-1의 인산화를 촉진하여 연골세포 분화를 억제하는 것으로 사료된다.

  • PDF