Curcumin inhibits renal cyst formation and enlargement in vitro by regulating intracellular signaling pathways

2011 ◽  
Vol 654 (1) ◽  
pp. 92-99 ◽  
Author(s):  
Jinsheng Gao ◽  
Hong Zhou ◽  
Tianluo Lei ◽  
Li Zhou ◽  
Weidong Li ◽  
...  
Endocrinology ◽  
2012 ◽  
Vol 153 (9) ◽  
pp. 4502-4510 ◽  
Author(s):  
Isobelle Grant ◽  
Judith E. Cartwright ◽  
Brooke Lumicisi ◽  
Alison E. Wallace ◽  
Guy S. Whitley

Impaired trophoblast invasion is associated with pregnancy disorders such as early pregnancy loss and preeclampsia. There is evidence to suggest that the consumption of caffeine during pregnancy may increase the risk of pregnancy loss; however, little is known about the direct effect of caffeine on normal trophoblast biology. Our objectives were to examine the effect of caffeine on trophoblast migration and motility after stimulation with epidermal growth factor (EGF) and to investigate the intracellular signaling pathways involved in this process. Primary first-trimester extravillous trophoblasts (EVT) and the EVT-derived cell line SGHPL-4 were used to study the effect of caffeine on EGF-stimulated cellular motility using time-lapse microscopy. SGHPL-4 cells were further used to study the effect of caffeine and cAMP on EGF-stimulated invasion of fibrin gels. The influence of caffeine and cAMP on EGF-stimulated intracellular signaling pathways leading to the activation of Akt were investigated by Western blot analysis. Caffeine inhibits both EGF-stimulated primary EVT and SGHPL-4 cell motility. EGF stimulation activates phosphatidylinositol 3-kinase, and Akt and caffeine inhibit this activation. Although cAMP inhibits both motility and invasion, it does not inhibit the activation of Akt, indicating that the effects of caffeine seen in this study are independent of cAMP. Further investigation indicated a role for mammalian target of rapamycin complex 2 (mTORC2) as a target for the inhibitory effect of caffeine. In conclusion, we demonstrate that caffeine inhibits EGF-stimulated trophoblast invasion and motility in vitro and so could adversely influence trophoblast biology in vivo.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3723-3723
Author(s):  
Daniel B Lipka ◽  
Marie-Christine Blum ◽  
Florian H Heidel ◽  
Thomas Kindler ◽  
Thomas Fischer

Abstract In a multitude of cases, oncogenic mutations are gain of function mutations that confer a constitutively activated gene product. Currently, evidence from a large body of experimental studies suggests that oncogenic transformation induced by activating kinase mutations is not sufficiently explained by constitutive kinase activation alone but is a result of aberrantly activated signaling pathways in affected cells. The JAK2V617F-mutation is a highly prevalent molecular marker in Ph-negative myeloproliferative disease (MPD). In vitro, Ba/F3-cells expressing both erythropoietin receptor (EpoR) and the JAK2V617F-mutation show constitutive activation of the JAK-STAT pathway and cytokine independent growth. Multiple in-vitro and in-vivo studies demonstrate that the JAK2V617F-mutation mediates many of the phenotypic characteristics of the MPDs. Nevertheless, until now it is largely unclear, which signaling pathways in particular are involved in the process of malignant transformation in JAK2V617F-positive cells. Therefore, we applied a kinomics chip approach to screen for activated intracellular signaling pathways. We used a commercially available peptide chip containing 960 synthetic kinase substrate peptides spotted in triplicates and covering peptide substrates for approximately 50% of the human kinome. Peptides have been selected for their biologic relevance in physiologic processes such as stress response, growth and cell differentiation. With this approach, a broad spectrum of intracellular signaling pathways and kinases can be investigated simultaneously in a single experiment. As a proof of principle, we performed kinomics chip analysis of Ba/F3-cells stably transfected with EpoR and either the JAK2V617F-mutant (Ba/F3-EpoR-VF) or wildtype JAK2 (Ba/F3-EpoR-WT). In brief, cells were seeded and treated with erythropoietin. One chip per cell lysate was incubated with an activation buffer containing the cell lysate and radioactively labelled ATP for a defined time period and washed several times afterwards. The chip was then analyzed by means of autoradiography using a phospho-storage-screen and a phospho-imager. Chip analysis was performed using standard microarray software and Microsoft Excel software. Chip experiments were performed simultaneously for Ba/F3-EpoR-VF and Ba/F3-EpoR-WT and in duplicate. Analysis revealed differential activation of known pathways such as Ras/Raf/MEK/ERK, JAK/STAT, and PI3-Kinase with pronounced activation seen in Ba/F3-EpoR-VF cells as compared to Ba/F3-EpoR-WT cells. This was not a surprising result but strongly underlines the feasibility and validity of this approach and therefore served as an internal control. Differential regulation of a number of other signaling nodes that have not yet been described in the context of mutant JAK2 signaltransduction have been detected. To select for relevant hits among these potential targets, we first excluded all substrates from further analysis that are known to be involved in lymphocyte-specific pathways. For the remaining hits we performed a literature search to learn more about their known functions and their potential impact in JAK2V617F-positive MPD. Validation of selected signaling molecules by means of Western blotting analysis and functional investigations such as siRNA knock-down experiments are currently under way. In addition to the widely used lymphoid Ba/F3-model, we also established a novel cell culture model with simultaneous expression of EpoR and either mutant or wildtype JAK2 in a myeloid 32D-cell background. This model will be helpful to us to determine false-positive results due to cell-line specific changes. We conclude, that kinomic profiling using the above mentioned chip-technology is a valid method to comprehensively investigate differential activation of signaling pathways in cell lysates. In our cell line model, we were able to detect activation of well known signaling pathways in JAK2V617F-positive cells. Furthermore, we were able to identify candidate proteins that appear to be specifically involved in JAK2V617F-signaling.


2020 ◽  
Vol 19 ◽  
pp. 153473542097247
Author(s):  
Xue-Cong Zheng ◽  
Ze-Sheng Shi ◽  
Cheng-Zhi Qiu ◽  
Zhong-Shi Hong ◽  
Chun-Xiao Wang ◽  
...  

Protosappanin B (PSB) is a key active component of Lignum Sappan extract. Although the antiproliferative effects of Lignum Sappan extract have been demonstrated in various cancer cells, relatively little is known about the effects of PSB on tumor progression. The aim of this study was to explore the anti-tumor effects of PSB on human colon cancer cells by regulation of intracellular signaling pathways and Golgi phosphoprotein 3 (GOLPH3) expression in vitro and in vivo. Our results showed that PSB effectively inhibited the viability and migration of SW620 cells and induced apoptosis, but had poor effect on HCT116 cells. Furthermore, PSB significantly reduced the expression of p-AKT, p-p70S6K, β-catenin, and p-ERK1/2 proteins in SW620 cells, and this effect was reversed by the corresponding signaling pathway agonists. Interestingly, PSB could also suppress GOLPH3 expression of SW620 cells in a concentration-dependent manner, but SW620 cells transfected with lentiviral vectors overexpressing GOLPH3 can effectively resist the cytotoxic activity of PSB in vitro. The xenograft experiment of SW620 cells with LV-GOLPH3 confirmed that PSB distinctly inhibited the tumor growth via suppressing GOLPH3 expression. Collectively, these findings clarified a new anti-cancer mechanism of PSB through inhibition of GOLPH3 expression and intracellular signaling pathways in colon cancer cells. PSB may be a potential new drug for colon cancer.


2018 ◽  
Author(s):  
Serena Martinelli

A gain-of-function mutation in Janus kinase 2 (JAK2V617F) is at the basis of the majority of chronic myeloproliferative neoplasms (MPN). Enhanced activation of other downstream pathways including the PI3K/mTOR pathway has been documented as well. In this study we evaluated the effects of JAK1/2 inhibitors, alone and in combination with mTOR, with a dual mTOR/PI3K inhibitor and with a pan PI3K inhibitor in in-vitro and in-vivo MPN models. Our findings of strong synergy between the JAK2 inhibitors and mTOR/PI3K inhibitor suggested that we might be able to administer these drugs at lower concentrations than when the drugs are used individually. This provides a framework for combination trials using compounds in patients with myeloproliferative neoplasms


1994 ◽  
Vol 269 (18) ◽  
pp. 13162-13166
Author(s):  
Y. Konda ◽  
I. Gantz ◽  
J. DelValle ◽  
Y. Shimoto ◽  
H. Miwa ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document