• 제목/요약/키워드: c-jun N-terminal Kinase

검색결과 280건 처리시간 0.025초

Therapeutic potential of targeting kinase inhibition in patients with idiopathic pulmonary fibrosis

  • Kim, Suji;Lim, Jae Hyang;Woo, Chang-Hoon
    • Journal of Yeungnam Medical Science
    • /
    • 제37권4호
    • /
    • pp.269-276
    • /
    • 2020
  • Fibrosis is characterized by excessive accumulation of extracellular matrix components. The fibrotic process ultimately leads to organ dysfunction and failure in chronic inflammatory and metabolic diseases such as pulmonary fibrosis, advanced kidney disease, and liver cirrhosis. Idiopathic pulmonary fibrosis (IPF) is a common form of progressive and chronic interstitial lung disease of unknown etiology. Pathophysiologically, the parenchyma of the lung alveoli, interstitium, and capillary endothelium becomes scarred and stiff, which makes breathing difficult because the lungs have to work harder to transfer oxygen and carbon dioxide between the alveolar space and bloodstream. The transforming growth factor beta (TGF-β) signaling pathway plays an important role in the pathogenesis of pulmonary fibrosis and scarring of the lung tissue. Recent clinical trials focused on the development of pharmacological agents that either directly or indirectly target kinases for the treatment of IPF. Therefore, to develop therapeutic targets for pulmonary fibrosis, it is essential to understand the key factors involved in the pathogenesis of pulmonary fibrosis and the underlying signaling pathway. The objective of this review is to discuss the role of kinase signaling cascades in the regulation of either TGF-β-dependent or other signaling pathways, including Rho-associated coiled-coil kinase, c-jun N-terminal kinase, extracellular signal-regulated kinase 5, and p90 ribosomal S6 kinase pathways, and potential therapeutic targets in IPF.

Valproic Acid Induces Transcriptional Activation of Human GD3 Synthase (hST8Sia I) in SK-N-BE(2)-C Human Neuroblastoma Cells

  • Kwon, Haw-Young;Dae, Hyun-Mi;Song, Na-Ri;Kim, Kyoung-Sook;Kim, Cheorl-Ho;Lee, Young-Choon
    • Molecules and Cells
    • /
    • 제27권1호
    • /
    • pp.113-118
    • /
    • 2009
  • In this study, we have shown the transcriptional regulation of the human GD3 synthase (hST8Sia I) induced by valproic acid (VPA) in human neuroblastoma SK-N-BE(2)-C cells. To elucidate the mechanism underlying the regulation of hST8Sia I gene expression in VPA-stimulated SK-N-BE(2)-C cells, we characterized the promoter region of the hST8Sia I gene. Functional analysis of the 5'-flanking region of the hST8Sia I gene by the transient expression method showed that the -1146 to -646 region, which contains putative binding sites for transcription factors c-Ets-1, CREB, AP-1 and NF-${\kappa}B$, functions as the VPA-inducible promoter of hST8Sia I in SK-N-BE(2)-C cells. Site-directed mutagenesis and electrophoretic mobility shift assay indicated that the NF-${\kappa}B$ binding site at -731 to -722 was crucial for the VPA-induced expression of hST8Sia I in SK-N-BE(2)-C cells. In addition, the transcriptional activity of hST8Sia I induced by VPA in SK-N-BE(2)-C cells was strongly inhibited by SP600125, which is a c-Jun N-terminal kinase (JNK) inhibitor, and $G{\ddot{O}}6976$, which is a protein kinase C (PKC) inhibitor, as determined by RT-PCR (reverse transcription-polymerase chain reaction) and luciferase assays. These results suggest that VPA markedly modulated transcriptional regulation of hST8Sia I gene expression through PKC/JNK signal pathways in SK-N-BE(2)-C cells.

혈관내피세포에서 c-Jun N-terminal kinase에 의해 조절되는 세포사멸에 고농도의 피노실빈이 미치는 효과 (Apoptotic Effect of Pinosylvin at a High Concentration Regulated by c-Jun N-Terminal Kinase in Bovine Aortic Endothelial Cells)

  • 송지나;박진선;정은실;소아영;피재호;박헌용
    • 생명과학회지
    • /
    • 제25권4호
    • /
    • pp.416-424
    • /
    • 2015
  • 피노실빈은 소나무 등에서 흔히 관찰되는 스틸벤노이드이다. 혈관내피세포에서 ~pM에서 ~nM 정도의 낮은 농도의 피노실빈은 세포성장, 세포이동, 항염증반응을 유도한다. 그러나 최근에 고농도의 피노실빈이 소 대동맥 내피세포의 세포사를 유발하고 있음이 보고되었으나, 그 자세한 기전이나 경로 연구가 미흡하여 이번 연구에서 고농도 피노실빈의 세포사 유발 경로를 규명하기 위한 실험을 수행하였다. 고농도의 피노실빈은 caspase-3 절단, 포스파티딜 세린의 플립플롭, 핵 분절 등을 유도하는 것으로 보아 세포사멸을 통한 세포사를 유발함을 알았다. 또한 혈청기아나 100 μM etoposide에 의하여 촉발하는 caspase-3 활성/핵분절이 추가적으로 증가하는 현상이 관찰되었다. 이는 피노실빈이 일으키는 세포사멸은 혈청기아나 etoposide에 의해 일어나는 세포사멸과는 다른 경로로 진행됨을 의미하므로 고농도의 피노실빈에 의해 촉발되는 세포신호전달을 탐색한 결과, JNK와 eNOS가 관여함을 알았다. 두 신호전달 물질 중에 어느 것이 피노실빈 유도 세포사에 중요한 지를 알기 위한 추가적인 실험을 진행하였다. 그 결과, JNK 억제자인 SP-600125는 피노실빈에 의한 세포사멸을 억제하였으나, eNOS 억제자인 L-NAME은 아무런 영향이 없는 것으로 보아 JNK가 피노실빈에 의해 유발되는 세포사멸에 관여하는 세포신호 전달물질임을 알았다.

Effects of Curcumin, the Active Ingredient of Turmeric(Curcuma longa), on Regulation of Glutamate-induced Toxicity and Activation of the Mitogen-activated Protein Kinase Phosphatase-1 (MKP-1) in HT22 Neuronal Cell

  • Lee, Sang-Hyun;Yun, Young-Gab
    • Natural Product Sciences
    • /
    • 제15권1호
    • /
    • pp.32-36
    • /
    • 2009
  • Glutamate causes neurotoxicity through formation of reactive oxygen species and activation of mitogen-activated protein kinase (MAPK) pathways. MAPK phosphatase-1 (MKP-1) is one of the phosphatases responsible for dephosphorylation/deactivation of three MAPK families: the extracellular signal-regulated kinase-1/2 (ERK-1/2), the c-Jun N-terminal kinase-1/2 (JNK-1/2), and the p38 MAPK. In this report, the potential involvement of MKP-1 in neuroprotective effects of curcumin, the active ingredient of turmeric (Curcuma longa), was examined using HT22 cells. Glutamate caused cell death and activation of ERK-1/2 but not p38 MAPK or JNK-1/2. Blockage of ERK-1/2 by its inhibitor protected HT22 cells against glutamate-induced toxicity. Curcumin attenuated glutamate-induced cell death and ERK-1/2 activation. Interestingly, curcumin induced MKP-1 activation. In HT22 cells transiently transfected with small interfering RNA against MKP-1, curcumin failed to inhibit glutamate-induced ERK-1/2 activation and to protect HT22 cells from glutamate-induced toxicity. These results suggest that curcumin can attenuate glutamate-induced neurotoxicity by activating MKP-1 which acts as the negative regulator of ERK-1/2. This novel pathway may contribute to and explain at least one of the neuroprotective actions of curcumin.

Shikonin Modulates Cell Proliferation by Inducing Apoptosis in LLC Cells via MAPK Regulation and Caspase Activation

  • 이수진;김성훈
    • 동의생리병리학회지
    • /
    • 제19권2호
    • /
    • pp.501-507
    • /
    • 2005
  • Shikonin is a chemically characterized component of traditional herbal medicine, the root of Lithospermum erythrorhizon and has been shown to possess antitumor activities. Here we investigated anticancer potential of shikonin and its possible mechanism of action in LLC cells. Shikonin inhibited the proliferation of LLC cells in a concentration-dependent manner. It was also demonstrated that shikonin induced apoptosis in LLC cells by Annexin V staining and TUNEL staining analysis. Shikonin treatment was caused that decrease of Bcl-2, activation of caspases and cleavage of PARP. And shikonin also induced that the activation of mitogen-activated protein kinases (MAPKs), such as extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK) and p38. Interestingly, the cell proliferation inhibition induced by shikonin was recovered by specific inhibitors of JNK and p38 but the inhibitor of MEK, the upstream kinase of ERK, did not recover. Additionally, shikonin administration at doses of 5 mg/kg in C57BL/6 mice strongly inhibited the primary tumor growth of LLC. Taken together, these results suggest that shikonin may suppress LLC cell proliferation by inducing an apoptotic process via activation of caspases and MAPKs

Protective effect of ginsenoside Rh3 against anticancer drug-induced apoptosis in LLC-PK1 kidney cells

  • Lee, Hye Lim;Kang, Ki Sung
    • Journal of Ginseng Research
    • /
    • 제41권2호
    • /
    • pp.227-231
    • /
    • 2017
  • Background: Ginsenosides are active components of Panax ginseng that exert various health benefits including kidney protection effect. The medicinal activity of ginsenosides can be enhanced by modulating their stereospecificity by heat processing. Ginsenosides Rk2 and Rh3 represent positional isomers of the double bond at C-20(21) or C-20(22). Methods: The present study investigated the kidney-protective effects of ginsenosides Rk2 and Rh3 against cisplatin, a platinum based anticancer drug, induced apoptotic damage in renal proximal LLC-PK1 cells. Results: As a result, ginsenoside Rh3 shows a stronger protective effect than that shown by Rk2. Cisplatin-induced elevated protein levels of phosphorylated c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinase (ERK), p38, and cleaved caspase-3 decreased after cotreatment with ginsenoside Rh3. The increase in the percentage of apoptotic LLC-PK1 cells induced by cisplatin treatment also significantly reduced after cotreatment with ginsenoside Rh3. Conclusion: These results demonstrate that inhibition of the JNK and ERK mitogen-activated protein kinase signaling cascade plays a critical role in mediating the renoprotective effect of ginsenoside Rh3.

Platycodon grandiflorum Extracts Exhibits Anti-inflammatory Properties by Down-regulating MAPK Signaling Pathways Lipopolysaccharide-treated RAW264.7 Cells

  • Kim, Hyeon Jin;Jeong, Seong-Yun;Kim, Jin-Kyung
    • 대한의생명과학회지
    • /
    • 제18권4호
    • /
    • pp.369-376
    • /
    • 2012
  • Platycodon grandiflorum is a medicinal herb that is used to treat pulmonary and respiratory allergic disorders. The objective of this study was to investigate the protective effects of ethyl acetate extract of Platycodon grandiflorum (PGEA) against inflammation and to discern the molecular mechanism of PGEA in lipopolysaccharide (LPS)-induced signal pathways in RAW264.7 macrophage cells. PGEA suppressed the generation of nitric oxide (NO) and the expression of inducible NO synthase induced by LPS in RAW264.7 cells, and inhibited the release of pro-inflammatory cytokines induced by LPS in RAW264.7 cells. Western blot analysis showed that PGEA suppressed LPS-induced phosphorylation of p38 and c-Jun N-terminal kinase (JNK) but not extracellular signal-regulated kinase and $I{\kappa}-B{\alpha}$ degradation. Inactivation of JNK and p38 was effectively alleviated by PGEA, which subsequently affected the activation of c-Jun and c-Fos, which are the essential components of the activator protein-1 (AP-1) transcription complex. Taken together, the results indicate PGEA suppress the activation of p38, JNK, and AP-1, thereby inhibiting the generation of NO and pro-inflammatory cytokines, which affect the regulation of inflammation. PGEA may be useful for the treatment of various inflammatory diseases.

Hologram Quantitative Structure Activity Relationship Analysis of JNK Antagonists

  • Kulkarni, Seema A.;Madhavan, Thirumurthy
    • 통합자연과학논문집
    • /
    • 제8권2호
    • /
    • pp.81-88
    • /
    • 2015
  • c-Jun N-terminal kinase-3 (JNK3) is a member of the mitogen-activated protein kinase family (MAPK), and plays an important role in neurological disorders. Therefore, identification of selective JNK3 inhibitor may contribute towards neuroprotection therapies. In this work, we performed hologram quantitative structure-activity relationship (HQSAR) on a series of thiophene trisubstituted derivatives. The best predictions were obtained for HQSAR model with $q^2=0.628$ and $r^2=0.986$. Statistical parameters from the generated QSAR models indicated the data is well fitted and have high predictive ability. HQSAR result showed that atom, bond and chirality descriptors play an important role in JNK3 activity and also shows that electronegative groups is highly favourble to enhance the biological activity. Our results could be useful to design novel and selective JNK3 inhibitors.

Shikonin Induced Apoptosis and Inhibited Angiogenesis on HSE Cells

  • Lee Soo-Jin;Kim Sung-Hoon
    • 동의생리병리학회지
    • /
    • 제19권5호
    • /
    • pp.1363-1369
    • /
    • 2005
  • Previously we have shown that shikonin has strong anti-tumor activities via inducing apoptosis and suppressing metastasis on LLC cells in vivo and in vitro. Here we have investigated anti-angiogenic potential of shikonin and its possible mechanism of action in HSE cells. Shikonin inhibited the proliferation of HSE cells in a concentration-dependent manner. It was shown that this proliferation inhibition was caused by apoptosis induced by shikonin via BrdU incorporation and Western blotting analysis. Shikonin treatment was caused that decrease of activation of caspases and cleavage of PARP. And shikonin induced that the activation of mitogen-activated protein kinases (MAPKs), such as extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK) and p38. Moreover, shikonin showed anti-angiogenic activities inhibiting tube-like formation of HSE cells in vitro and vascular formation of LLC cells in vivo. These findings suggest that shikonin may a possible candidate not only anti-metastatic agent but also anti-angiogenic agent.