• Title/Summary/Keyword: MEK/ERK signaling

Search Result 72, Processing Time 0.021 seconds

Antitumor Activity of Combination Therapy with Metformin and Trametinib in Non-Small Cell Lung Cancer Cells

  • Ko, Eunjeong;Baek, Seungjae;Kim, Jiwon;Park, Deokbae;Lee, Youngki
    • Development and Reproduction
    • /
    • v.24 no.2
    • /
    • pp.113-123
    • /
    • 2020
  • Metformin has been widely used as an antidiabetic drug, and reported to inhibit cell proliferation in many cancers including non-small cell lung cancer (NSCLC). In NSCLC cells, metformin suppresses PI3K/AKT/mTOR signaling pathway, but effect of metformin on RAS/RAF/MEK/ERK signaling pathway is controversial; several studies showed the inhibition of ERK activity, while others demonstrated the activation of ERK in response to metformin exposure. Metformin-induced activation of ERK is therapeutically important, since metformin could enhance cell proliferation through RAS/RAF/MEK/ERK pathway and lead to impairment of its anticancer activity suppressing PI3K/AKT/mTOR pathway, requiring blockade of both signaling pathways for more efficient antitumor effect. The present study tested the combination therapy of metformin and trametinib by monitoring the alterations of regulatory effector proteins of cell signaling pathways and the effect of the combination on cell viability in NCI-H2087 NSCLC cells with NRAS and BRAF mutations. We show that metformin alone blocks PI3K/AKT/mTOR signaling pathway but induces the activation and phosphorylation of ERK. The combination therapy synergistically decreased cell viability in treatment with low doses of two drugs, while it gave antagonistic effect with high doses. These findings suggest that the efficacy of metformin and trametinib combination therapy may depend on the alteration of ERK activity induced by metformin and specific cellular context of cancer cells.

The Phospholipase-Protein Kinase C-MEK-ERK Pathway is Essential in Mycobacteria-induced CCL3 and CCL4 Expression in Human Monocytes (사람 단핵구에서 결핵균에 의해 유도되는 CCL3 및 CCL4 발현에 대한 Phospholipase-Protein Kinase C-MEK-ERK 경로의 역할 분석)

  • Yang, Chul-Su;Song, Chang-Hwa;Jung, Saet-Byel;Lee, Kil-Soo;Kim, Su-Young;Lee, Ji-Sook;Shin, A-Rum;Oh, Jae-Hee;Kwon, Yu-Mi;Kim, Hwa-Jung;Park, Jeong-Kyu;Paik, Tae-Hyun;Jo, Eun-Kyeong
    • IMMUNE NETWORK
    • /
    • v.5 no.4
    • /
    • pp.237-246
    • /
    • 2005
  • Background: Little information is available on the identification and characterization of the upstream regulators of the signal transduction cascades for Mycobacterium tuberculosis (M. tbc)-induced ERK 1/2 activation and chemokine expression. We investigated the signaling mechanisms involved in expression of CCL3 /MIP-1 and CCL4/MIP-1 in human primary monocytes infected with M. tbc. Methods: MAP kinase phosphorylation was determined using western blot analysis with specific primary antibodies (ERK 1/2, and phospho-ERK1/2), and the upstream signaling pathways were further investigated using specific inhibitors. Results: An avirulent strain, M. tbc H37Ra, induced greater and more sustained ERK 1/2 phosphorylation, and higher CCL3 and CCL4 production, than did M. tbc H37Rv. Specific inhibitors for mitogen-activated protein kinase (MAPK) kinase (MEK; U0126 and PD98059) significantly inhibited the expression of CCL3 and CCL4 in human monocytes. Mycobactetia-mediated expression of CCL3 and CCL4 was not inhibited by the Ras inhibitor manumycin A or the Raf-1 inhibitor GW 5074. On the other hand, phospholipase C (PLC) inhibitor (U73122) and protein kinase C (PKC)specific inhibitors ($G\ddot{o}6976$ and Ro31-8220) significantly reduced M. tbc-induced activation of ERK 1/2 and chemokine synthesis. Conclusion: These results are the first to demonstrate that the PLC-PKC-MEK-ERK, not the Ras-Raf-MEK-ERK, pathway is the major signaling pathway inducing M. tbc-mediated CCL3 and CCL4 expression in human primary monocytes.

Constitutively active Ras negatively regulates Erk MAP kinase through induction of MAP kinase phosphatase 3 (MKP3) in NIH3T3 cells

  • Park, Young Jae;Lee, Jong Min;Shin, Soon Young;Kim, Young Ho
    • BMB Reports
    • /
    • v.47 no.12
    • /
    • pp.685-690
    • /
    • 2014
  • The Ras/Raf/MEK/Erk signaling pathway is important for regulation of cell growth, proliferation, differentiation, survival, and apoptosis in response to a variety of extracellular stimuli. Lack of Erk MAPK activation is observed in several cancer cells despite active activation of Ras. However, little is known about the modulation of Erk1/2 activity by active Ras. Here, we show that overexpression of active H-Ras (H-RasG12R) in NIH3T3 fibroblasts impaired FGF2-induced Erk1/2 phosphorylation, as compared to wild-type cells. Northern blot analysis revealed that prolonged expression of active Ras increased MAP kinase phosphatase 3 (MKP3) mRNA expression, a negative regulator of Erk MAPK. Inhibition of the phosphatidylinositol 3-kinase (PI3K)/Akt pathway abrogated active Ras-induced up-regulation of MKP3 expression, leading to the rescue of Erk1/2 phosphorylation. Our results demonstrated that the Ras/Raf/MEK/Erk signaling cascade is negatively regulated by the PI3K/Aktdependent transcriptional activation of the MKP3 gene.

Induction of MAP kinase phosphatase 3 through Erk/MAP kinase activation in three oncogenic Ras (H-, K- and N-Ras)-expressing NIH/3T3 mouse embryonic fibroblast cell lines

  • Koo, JaeHyung;Wang, Sen;Kang, NaNa;Hur, Sun Jin;Bahk, Young Yil
    • BMB Reports
    • /
    • v.49 no.7
    • /
    • pp.370-375
    • /
    • 2016
  • Ras oncoproteins are small molecular weight GTPases known for their involvement in oncogenesis, which operate in a complex signaling network with multiple effectors. Approximately 25% of human tumors possess mutations in a member of this family. The Raf1/MEK/Erk1/2 pathway is one of the most intensively studied signaling mechanisms. Different levels of regulation account for the inactivation of MAP kinases by MAPK phosphatases in a cell type- and stimuli-dependent manner. In the present study, using three inducible Ras-expressing NIH/3T3 cell lines, we demonstrated that MKP3 upregulation requires the activation of the Erk1/2 pathway, which correlates with the shutdown of this pathway. We also demonstrated, by applying pharmacological inhibitors and effector mutants of Ras, that induction of MKP3 at the protein level is positively regulated by the oncogenic Ras/Raf/MEK/Erk1/2 signaling pathway.

Paraquat Induces Apoptosis through Cytochrome C Release and ERK Activation

  • Seo, Hong Joo;Choi, Sang Joon;Lee, Jung-Hee
    • Biomolecules & Therapeutics
    • /
    • v.22 no.6
    • /
    • pp.503-509
    • /
    • 2014
  • Paraquat has been suggested to induce apoptosis by generation of reactive oxygen species (ROS). However, little is known about the mechanism of paraquat-induced apoptosis. Here, we demonstrate that extracellular signal-regulated protein kinase (ERK) is required for paraquat-induced apoptosis in NIH3T3 cells. Paraquat treatment resulted in activation of ERK, and U0126, inhibitors of the MEK/ERK signaling pathway, prevented apoptosis. Moreover, paraquat-induced apoptosis was associated with cytochrome C release, which could be prevented by treatment with the MEK inhibitors. Taken together, our findings suggest that ERK activation plays an active role in mediating paraquat-induced apoptosis of NIH3T3 cells.

Isorhamnetin from Oenanthe javanica Attenuates Fibrosis in Rat Hepatic Stellate Cells via Inhibition of ERK Signaling Pathway

  • Lee, Mi-Kyeong;Yang, Hye-Kyung;Ha, Na-Ry;Sung, Sang-Hyun;Kim, Young-Choong
    • Natural Product Sciences
    • /
    • v.14 no.2
    • /
    • pp.81-85
    • /
    • 2008
  • Isorhamnetin isolated from Oenanthe javanica significantly inhibited proliferation and collagen production in HSC-T6 cells in concentration- and time-dependent manners. Pretreatment of HSC-T6 cells with isorhamnetin significantly inhibited serum-induced ERK phosphorylation, in a similar manner as PD98059, a known MEK inhibitor. These results suggested that isorhamnetin reduced collagen production in HSC-T6 cells, in part, via inhibition of ERK signaling pathway.

Rutin Suppresses Neoplastic Cell Transformation by Inhibiting ERK and JNK Signaling Pathways (Rutin의 ERK 및 JNK 신호전달체계 억제를 통한 암예방 효능)

  • Kang, Nam Joo
    • The Korean Journal of Food And Nutrition
    • /
    • v.28 no.4
    • /
    • pp.579-585
    • /
    • 2015
  • Rutin is a well-known flavonoid found in buckwheat. Recent studies have demonstrated that the biological actions of rutin include anti-inflammatory and anti-cancer effects, but the underlying molecular mechanisms of these actions are not yet fully understood. Neoplastic cell transformation is considered a major event that contributes to carcinogenesis, and the present study aimed to determine whether rutin would exert anti-tumor effects via the results suggest that rutin exerted a potent inhibitory influence on the molecular activity of the MEK/ERK and MKK4/JNK pathways and strongly attenuated EGF-induced neoplastic cell transformation. These findings provide insight into the biological actions of rutin and the molecular basis for the development of new chemoprotective agents.

Expression of Wee1 Gene in the Ascidian, Halocynthia roretzi Embryo

  • Lee, Won-Young;Ham, Hwa-Su;Kim, Gil-Jung
    • Development and Reproduction
    • /
    • v.15 no.1
    • /
    • pp.1-7
    • /
    • 2011
  • It is suggested that FGF/Ras/MEK/Erk signaling plays crucial roles in specification and cell division of the mesodermal precursor cells in ascidian embryos. To investigate how the number of cell division in tissue precursor cells is determined, we have characterized Wee1 homolog, Hr-Wee1 of the ascidian Halocynthia roretzi. We found that the Hr-Wee1 mRNA is expressed both maternally and zygotically. Maternal transcript is localized to the cytoplasm in the animal cells, while zygotic expression is seen in cells of the endoderm lineage from 32-cell to 110-cell stages. Zygotic in situ signal is detected in the A-line neural plate cells of neurulae, and in epidermal cells of the head region of tailbud embryos. Embryos treated with MEK signaling inhibitor showed a similar pattern to normal embryos in expression of Hr-Wee1. Therefore, it is likely that MEK signaling does not affect the maternal and zygotic expression of Hr-Wee1.

Mitogen-Activated Protein Kinase Signal Transduction in Solid Tumors

  • Lei, Yuan-Yuan;Wang, Wei-Jia;Mei, Jin-Hong;Wang, Chun-Liang
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.15 no.20
    • /
    • pp.8539-8548
    • /
    • 2014
  • Mitogen-activated protein kinase (MAPK) is an important signaling pathway in living beings in response to extracellular stimuli. There are 5 main subgroups manipulating by a set of sequential actions: ERK(ERK1/ERK2), c-Jun N(JNK/SAPK), p38 MAPK($p38{\alpha}$, $p38{\beta}$, $p38{\gamma}$ and $p38{\delta}$), and ERK3/ERK4/ERK5. When stimulated, factors of upstream or downstream change, and by interacting with each other, these groups have long been recognized to be related to multiple biologic processes such as cell proliferation, differentiation, death, migration, invasion and inflammation. However, once abnormally activated, cancer may occur. Several components of the MAPK network have already been proposed as targets in cancer therapy, such as p38, JNK, ERK, MEK, RAF, RAS, and DUSP1. Among them, alteration of the RAS-RAF-MEK-ERK-MAPK(RAS-MAPK) pathway has frequently been reported in human cancer as a result of abnormal activation of receptor tyrosine kinases or gain-of-function mutations in genes. The reported roles of MAPK signaling in apoptotic cell death are controversial, so that further in-depth investigations are needed to address these controversies. Based on an extensive analysis of published data, the goal of this review is to provide an overview on recent studies about the mechanism of MAP kinases, and how it generates certain tumors, as well as related treatments.

Expression of Hr-Erf Gene during Ascidian Embryogenesis

  • Kim, Jung Eun;Lee, Won Young;Kim, Gil Jung
    • Development and Reproduction
    • /
    • v.17 no.4
    • /
    • pp.389-397
    • /
    • 2013
  • FGF9/16/20 signaling pathway specify the developmental fates of notochord, mesenchyme, and neural cells in ascidian embryos. Although a conserved Ras/MEK/Erk/Ets pathway is known to be involved in this signaling, the detailed mechanisms of regulation of FGF signaling pathway have remained largely elusive. In this study, we have isolated Hr-Erf, an ascidian orthologue of vertebrate Erf, to elucidate interactions of transcription factors involved in FGF signaling of the ascidian embryo. The Hr-Erf cDNA encompassed 3110 nucleotides including sequence encoded a predicted polypeptide of 760 amino acids. The polypeptide had the Ets DNA-binding domain in its N-terminal region. In adult animals, Hr-Erf mRNA was predominantly detected in muscle, and at lower levels in ganglion, gills, gonad, hepatopancreas, and stomach by quantitative real-time PCR (QPCR) method. During embryogenesis, Hr-Erf mRNA was detected from eggs to early developmental stage embryos, whereas the transcript levels were decreased after neurula stage. Similar to the QPCR results, maternal transcripts of Hr-Erf was detected in the fertilized eggs by whole-mount in situ hybridization. Maternal mRNA of Hr-Erf was gradually lost from the neurula stage. Zygotic expression of Hr-Erf started in most blastomeres at the 8-cell stage. At gastrula stage, Hr-Erf was specifically expressed in the precursor cells of brain and mesenchyme. When MEK inhibitor was treated, embryos resulted in loss of Hr-Erf expression in mesenchyme cells, and in excess of Hr-Erf in a-line neural cells. These results suggest that zygotic Hr-Erf products are involved in specification of mesenchyme and neural cells.