• 제목/요약/키워드: ERK2 marker

검색결과 36건 처리시간 0.019초

Benzidine Induces Epithelial-Mesenchymal Transition of Human Bladder Cancer Cells through Activation of ERK5 Pathway

  • Sun, Xin;Zhang, Tao;Deng, Qifei;Zhou, Qirui;Sun, Xianchao;Li, Enlai;Yu, Dexin;Zhong, Caiyun
    • Molecules and Cells
    • /
    • 제41권3호
    • /
    • pp.188-197
    • /
    • 2018
  • Benzidine, a known carcinogen, is closely associated with the development of bladder cancer (BC). Epithelial-mesenchymal transition (EMT) is a critical pathophysiological process in BC progression. The underlying molecular mechanisms of mitogen-activated protein kinase (MAPK) pathway, especially extracellular regulated protein kinases 5 (ERK5), in regulating benzidine-induced EMT remains unclarified. Hence, two human bladder cell lines, T24 and EJ, were utilized in our study. Briefly, cell migration was assessed by wound healing assay, and cell invasion was determined by Transwell assay. Quantitative PCR and western blot were utilized to determine both gene expressions as well as protein levels of EMT and MAPK, respectively. Small interfering RNA (siRNA) was transfected to further determine ERK5 function. As a result, the migration and invasion abilities were enhanced, epithelial marker expression was decreased while mesenchymal marker expression was increased in human BC cell lines. Meanwhile, benzidine administration led to activation of ERK5 and activator protein 1 (AP-1) proteins, without effective stimulation of the Jun N-terminal kinase (JNK) or p38 pathways. Moreover, Benzidine-induced EMT and ERK5 activation were completely suppressed by XMD8-92 and siRNAs specific to ERK5. Of note, ERK1/2 was activated in benzidine-treated T24 cells, while benzidine-induced EMT could not be reversed by U0126, an ERK1/2 inhibitor, as indicated by further study. Collectively, our findings revealed that ERK5-mediated EMT was critically involved in benzidine-correlated BC progression, indicating the therapeutic significance of ERK5 in benzidine-related BC.

The Inactivation of ERK1/2, p38 and NF-kB Is Involved in the Down-Regulation of Osteoclastogenesis and Function by A2B Adenosine Receptor Stimulation

  • Kim, Bo Hyun;Oh, Ju Hee;Lee, Na Kyung
    • Molecules and Cells
    • /
    • 제40권10호
    • /
    • pp.752-760
    • /
    • 2017
  • A2B adenosine receptor (A2BAR) is known to be the regulator of bone homeostasis, but its regulatory mechanisms in osteoclast formation are less well-defined. Here, we demonstrate the effect of A2BAR stimulation on osteoclast differentiation and activity by RANKL. A2BAR was expressed in bone marrow-derived monocyte/macrophage (BMM) and RANKL increased A2BAR expression during osteoclastogenesis. A2BAR stimulation with its specific agonist BAY 60-6583 was sufficient to inhibit the activation of ERK1/2, p38 MAP kinases and $NF-{\kappa}B$ by RANKL as well as it abrogated cell-cell fusion in the late stage of osteoclast differentiation. Stimulation of A2BAR suppressed the expression of osteoclast marker genes, such as c-Fos, TRAP, Cathepsin-K and NFATc1, induced by RANKL, and transcriptional activity of NFATc1 was also inhibited by stimulation of A2BAR. A2BAR stimulation caused a notable reduction in the expression of Atp6v0d2 and DC-STAMP related to cell-cell fusion of osteoclasts. Especially, a decrease in bone resorption activity through suppression of actin ring formation by A2BAR stimulation was observed. Taken together, these results suggest that A2BAR stimulation inhibits the activation of ERK1/2, p38 and $NF-{\kappa}B$ by RANKL, which suppresses the induction of osteoclast marker genes, thus contributing to the decrease in osteoclast cell-cell fusion and bone resorption activity.

진귤 과피 추출물의 MMP-1 발현조절 효과 (Effects of Citrus sunki Peel Extract on Matrix Metalloproteinase-1 Expression)

  • 한구슬;이선령
    • 생명과학회지
    • /
    • 제23권12호
    • /
    • pp.1553-1556
    • /
    • 2013
  • 본 연구는 재래감귤종의 하나인 진귤 과피 추출물이 산화적 스트레스에 의한 MMP-1의 발현 조절에 미치는 효과를 확인하기 위해 수행되었다. $H_2O_2$를 피부세포에 처리하여 산화적 스트레스를 유도한 결과 노화 유발에 중요한 역할을 하는 것으로 알려진 MMP-1의 발현량이 증가하였고 진귤과피 추출물의 처리는 산화적 스트레스에 의해 증가된 MMP-1의 활성을 현저히 감소시켰다. 이러한 활성 조절이 ERK signaling을 통해 조절되는지 확인한 결과 산화적 스트레스에 의해 증가된 ERK의 인산화는 진귤과피 추출물의 처리로 억제되었고 MEK 억제재인 U0216을 처리하였을 경우 MMP-1의 활성도 또한 저해시키는 것을 확인하였다. 이상의 결과로 보아 $H_2O_2$에 의해 유도된 산화적 스트레스는 MMP-1의 발현을 촉진시켰고 진귤과피 추출물은 ERK 신호전달 경로를 통해 MMP-1의 발현을 조절하는 것으로 보여진다.

Obatoclax Regulates the Proliferation and Fusion of Osteoclast Precursors through the Inhibition of ERK Activation by RANKL

  • Oh, Ju Hee;Lee, Jae Yoon;Park, Jin Hyeong;No, Jeong Hyeon;Lee, Na Kyung
    • Molecules and Cells
    • /
    • 제38권3호
    • /
    • pp.279-284
    • /
    • 2015
  • Obatoclax, a pan-Bcl2 inhibitor, shows antitumor activities in various solid malignancies. Bcl2-deficient mice have shown the importance of Bcl2 in osteoclasts, as the bone mass of the mice was increased by the induced apoptosis of osteoclasts. Despite the importance of Bcl2, the effects of obatoclax on the proliferation and differentiation of osteoclast precursors have not been studied extensively. Here, we describe the anti-proliferative effects of obatoclax on osteoclast precursors and its negative role on fusion of the cells. Stimulation with low doses of obatoclax significantly suppressed the proliferation of osteoclast precursors in a dose-dependent manner while the apoptosis was markedly increased. Its stimulation was sufficient to block the activation of ERK MAP kinase by RANKL. The same was true when PD98059, an ERK inhibitor, was administered to osteoclast precursors. The activation of JNK1/2 and p38 MAP kinase, necessary for osteoclast differentiation, by RANKL was not affected by obatoclax. Interestingly, whereas the number of TRAP-positive mononuclear cells was increased by both obatoclax and PD98059, fused, multinucleated cells larger than $100{\pm}m$ in diameter containing more than 20 nuclei were completely reduced. Consistently, obatoclax failed to regulate the expression of osteoclast marker genes, including c-Fos, TRAP, RANK and CtsK. Instead, the expression of DC-STAMP and Atp6v0d2, genes that regulate osteoclast fusion, by RANKL was significantly abrogated by both obatoclax and PD98059. Taken together, these results suggest that obatoclax down-regulates the proliferation and fusion of osteoclast precursors through the inhibition of the ERK1/2 MAP kinase pathway.

High Dose of FGF-2 Induced Growth Retardation via ERK1/2 De-phosphorylation in Bone Marrow-derived Mesenchymal Stem Cells

  • Shim, Kwang Yong;Saima, Fatema Tuj;Eom, Young Woo
    • 대한의생명과학회지
    • /
    • 제23권2호
    • /
    • pp.49-56
    • /
    • 2017
  • Fibroblast growth factor (FGF)-2 is one of the most effective growth factors to increase the growth rate of mesenchymal stem cells (MSCs). Previously, we reported that low dose of FGF-2 (1 ng/ml) induced proliferation of bone marrow-derived mesenchymal stem cells (BMSCs) through AKT and ERK activation resulting in reduction of autophagy and senescence, but not at a high dose. In this study, we investigated the effects of high dose FGF-2 (10 ng/ml) on proliferation, autophagy and senescence of BMSCs for long term cultures (i.e., 2 months). FGF-2 increased the growth rate of BMSCs in a dose dependent manner for a short term (3 days), while during long term cultures (2 months), population doubling time was increased and accumulated cell number was lower than control in BMSCs when cultured with 10 ng/ml of FGF-2. 10 ng/ml of FGF-2 induced immediate de-phosphorylation of ERK1/2, expression of LC3-II, and increase of senescence associated ${\beta}$-galactosidase (SA-${\beta}$-Gal, senescence marker) expression. In conclusion, we showed that 10 ng/ml of FGF-2 was inadequate for ex vivo expansion of BMSCs because 10 ng/ml of FGF-2 induced growth retardation via ERK1/2 de-phosphorylation and induction of autophagy and senescence in BMSCs.

Modulation of Human Cardiac Progenitors via Hypoxia-ERK Circuit Improves their Functional Bioactivities

  • Jung, Seok Yun;Choi, Sung Hyun;Yoo, So Young;Baek, Sang Hong;Kwon, Sang Mo
    • Biomolecules & Therapeutics
    • /
    • 제21권3호
    • /
    • pp.196-203
    • /
    • 2013
  • Recent accumulating studies have reported that hypoxic preconditioning during ex vivo expansion enhanced the self-renewal or differentiation of various stem cells and provide an important strategy for the adequate modulation of oxygen in culture conditions, which might increase the functional bioactivity of these cells for cardiac regeneration. In this study, we proposed a novel priming protocol to increase the functional bioactivity of cardiac progenitor cells (CPCs) for the treatment of cardiac regeneration. Firstly, patient-derived c-$kit^+$ CPCs isolated from the atrium of human hearts by enzymatic digestion and secondly, pivotal target molecules identified their differentiation into specific cell lineages. We observed that hCPCs, in response to hypoxia, strongly activated ERK phosphorylation in ex vivo culture conditioning. Interestingly, pre-treatment with an ERK inhibitor, U0126, significantly enhanced cellular proliferation and tubular formation capacities of CPCs. Furthermore, we observed that hCPCs efficiently maintained the expression of the c-kit, a typical stem cell marker of CPCs, under both hypoxic conditioning and ERK inhibition. We also show that hCPCs, after preconditioning of both hypoxic and ERK inhibition, are capable of differentiating into smooth muscle cells (SMCs) and cardiomyocytes (CMs), but not endothelial cells (ECs), as demonstrated by the strong expression of ${\alpha}$-SMA, Nkx2.5, and cTnT, respectively. From our results, we conclude that the functional bioactivity of patient-derived hCPCs and their ability to differentiate into SMCs and CMs can be efficiently increased under specifically defined culture conditions such as short-term hypoxic preconditioning and ERK inhibition.

C-reactive protein accelerates DRP1-mediated mitochondrial fission by modulating ERK1/2-YAP signaling in cardiomyocytes

  • Suyeon Jin;Chan Joo Lee;Gibbeum Lim;Sungha Park;Sang-Hak Lee;Ji Hyung Chung;Jaewon Oh;Seok-Min Kang
    • BMB Reports
    • /
    • 제56권12호
    • /
    • pp.663-668
    • /
    • 2023
  • C-reactive protein (CRP) is an inflammatory marker and risk factor for atherosclerosis and cardiovascular diseases. However, the mechanism through which CRP induces myocardial damage remains unclear. This study aimed to determine how CRP damages cardiomyocytes via the change of mitochondrial dynamics and whether survivin, an anti-apoptotic protein, exerts a cardioprotective effect in this process. We treated H9c2 cardiomyocytes with CRP and found increased intracellular ROS production and shortened mitochondrial length. CRP treatment phosphorylated ERK1/2 and promoted increased expression, phosphorylation, and translocation of DRP1, a mitochondrial fission-related protein, from the cytoplasm to the mitochondria. The expression of mitophagy proteins PINK1 and PARK2 was also increased by CRP. YAP, a transcriptional regulator of PINK1 and PARK2, was also increased by CRP. Knockdown of YAP prevented CRP-induced increases in DRP1, PINK1, and PARK2. Furthermore, CRP-induced changes in the expression of DRP1 and increases in YAP, PINK1, and PARK2 were inhibited by ERK1/2 inhibition, suggesting that ERK1/2 signaling is involved in CRP-induced mitochondrial fission. We treated H9c2 cardiomyocytes with a recombinant TAT-survivin protein before CRP treatment, which reduced CRP-induced ROS accumulation and reduced mitochondrial fission. CRP-induced activation of ERK1/2 and increases in the expression and activity of YAP and its downstream mitochondrial proteins were inhibited by TAT-survivin. This study shows that mitochondrial fission occurs during CRP-induced cardiomyocyte damage and that the ERK1/2-YAP axis is involved in this process, and identifies that survivin alters these mechanisms to prevent CRP-induced mitochondrial damage.

Expression of Sodium-Iodide Symporter Depending on Mutational Status and Lymphocytic Thyroiditis in Papillary Thyroid Carcinoma

  • Song, Young Shin;Park, Young Joo
    • International journal of thyroidology
    • /
    • 제11권2호
    • /
    • pp.152-159
    • /
    • 2018
  • Background and Objectives: Sodium-iodine symporter (NIS) is a marker for the degree of differentiation in thyroid cancer. The genetic factors or microenvironment surrounding tumors can affect transcription of NIS. In this study, we investigated the NIS mRNA expression according to mutational status and coexistent lymphocytic thyroiditis in papillary thyroid cancer (PTC). Materials and Methods: The RNA expression levels of NIS in the samples from database of The Caner Genome Atlas (TCGA; n=494) and our institute (n=125) were analyzed. Results: The PTCs with the $BRAF^{V600E}$ mutation and the coexistence of $BRAF^{V600E}$ and TERT promoter mutations showed significantly lower expression of NIS (p<0.001, respectively), and those with BRAF-like molecular subtype also had reduced expression of NIS (p<0.001). NIS expression showed a positive correlation with thyroid differentiation score (r=0.593, p<0.001) and negative correlations with expressions of genes involved in ERK signaling (r=-0.164, p<0.001) and GLUT-1 gene (r=-0.204, p<0.001). The PTCs with lymphocytic thyroiditis showed significantly higher NIS expression (p=0.013), regardless of mutational status. Conclusion: The NIS expression was reduced by the $BRAF^{V600E}$ mutation and MAPK/ERK pathway activation, but restored by the presence of lymphocytic thyroiditis.

Involvement of ROS in Curcumin-induced Autophagic Cell Death

  • Lee, Youn-Ju;Kim, Nam-Yi;Suh, Young-Ah;Lee, Chu-Hee
    • The Korean Journal of Physiology and Pharmacology
    • /
    • 제15권1호
    • /
    • pp.1-7
    • /
    • 2011
  • Many anticancer agents as well as ionizing radiation have been shown to induce autophagy which is originally described as a protein recycling process and recently reported to play a crucial role in various disorders. In HCT116 human colon cancer cells, we found that curcumin, a polyphenolic phytochemical extracted from the plant Curcuma longa, markedly induced the conversion of microtubule-associated protein 1 light chain 3 (LC3)-I to LC3-II and degradation of sequestome-1 (SQSTM1) which is a marker of autophagosome degradation. Moreover, we found that curcumin caused GFP-LC3 formation puncta, a marker of autophagosome, and decrease of GFP-LC3 and SQSTM1 protein level in GFP-LC3 expressing HCT116 cells. It was further confirmed that treatment of cells with hydrogen peroxide induced increase of LC3 conversion and decrease of GFP-LC3 and SQSTM1 levels, but these changes by curcumin were almost completely blocked in the presence of antioxidant, N-acetylcystein (NAC), indicating that curcumin leads to reactive oxygen species (ROS) production, which results in autophagosome development and autolysosomal degradation. In parallel with NAC, SQSTM1 degradation was also diminished by bafilomycin A, a potent inhibitor of autophagosome-lysosome fusion, and cell viability assay was further confirmed that cucurmin-induced cell death was partially blocked by bafilomycin A as well as NAC. We also observed that NAC abolished curcumin-induced activation of extracelluar signal-regulated kinases (ERK) 112 and p38 mitogen-activated protein kinases (MAPK), but not Jun N-terminal kinase (JNK). However, the activation of ERK1/2 and p38 MAPK seemed to have no effect on the curcumin-induced autophagy, since both the conversion of LC3 protein and SQSTM1 degradation by curcumin was not changed in the presence of NAC. Taken together, our data suggest that curcumin induced ROS production, which resulted in autophagic activation and concomitant cell death in HCT116 human colon cancer cell. However, ROS-dependent activation of ERK1/2 and p38 MAPK, but not JNK, might not be involved in the curcumin-induced autophagy.