Phenobarbital inhibits calpain activity and expression in mouse hepatoma cells

2016 ◽  
Vol 397 (1) ◽  
pp. 91-96 ◽  
Author(s):  
Nicola Groll ◽  
Ferdinand Kollotzek ◽  
Jens Goepfert ◽  
Thomas O. Joos ◽  
Michael Schwarz ◽  
...  

Abstract The antiepileptic drug phenobarbital (PB) exerts hepatic effects related to cell proliferation and tumorigenesis which are closely linked to the Wnt/β-catenin signaling pathway. This pathway is, amongst others, regulated by calpain proteases. We now identified PB as an inhibitor of Wnt/β-catenin signaling in mouse hepatoma cells. Further analyses revealed that PB inhibits calpain activity, an effect which is at least in parts mediated by a transcriptional regulation of calpain mRNA levels and which is furthermore independent of the constitutive androstane receptor, the known mediator of most effects of PB in liver cells.

2020 ◽  
Vol 48 (9) ◽  
pp. 030006052094379
Author(s):  
Yanshan Li ◽  
Yunxiuxiu Xu ◽  
Ruomei Wang ◽  
Wenxin Li ◽  
Wenguang He ◽  
...  

Objective To investigate whether the Notch–Hif-1α signaling pathway is involved in liver regeneration. Methods Rats were divided into two groups and treated with daily intraperitoneal injections of saline (control) or the gamma-secretase inhibitor, Fli-06, for 2 days. Two-thirds of the rat livers were resected and rats were later euthanized at specific time points post-resection to analyze the remnant livers. Each group's liver/body weight ratio was calculated, and immunostaining and western blotting were used to determine the cell proliferation marker, PCNA and Ki-67 expression. Real-time PCR and western blotting were used to compare the mRNA expression of Notch homolog-1 ( Notch1), hairy and enhancer of split-1 ( Hes1), and vascular endothelial growth factor ( Vegf), and the protein expression of NICD and HIF-1α, respectively. Results The liver/body weight ratios and number of Ki-67- and PCNA-positive cells were significantly lower in the experimental group than the control group, indicating lower levels of liver regeneration following the disruption of Notch signaling by Fli-06. The Hes1 and Vegf mRNA levels and NICD and HIF-1α protein expression levels were all down-regulated by Fli-06 treatment. Conclusion Notch–Hif-α signaling pathway activation plays an important role in liver regeneration, where it may contribute toward liver cell proliferation.


2010 ◽  
Vol 298 (3) ◽  
pp. F807-F817 ◽  
Author(s):  
Renfang Song ◽  
Melissa Spera ◽  
Colleen Garrett ◽  
Samir S. El-Dahr ◽  
Ihor V. Yosypiv

ANG II AT2 receptor (AT2R)-deficient mice exhibit abnormal ureteric bud (UB) budding, increased incidence of double ureters, and vesicoureteral reflux. However, the role of the AT2R during UB morphogenesis and the mechanisms by which aberrant AT2R signaling disrupts renal collecting system development have not been fully defined. In this study, we mapped the expression of the AT2R during mouse metanephric development, examined the impact of disrupted AT2R signaling on UB branching, cell proliferation, and survival, and investigated the cross talk of the AT2R with the glial-derived neurotrophic factor ( GDNF)/ c-Ret/Wnt11 signaling pathway. Embryonic mouse kidneys express AT2R in the branching UB and the mesenchyme. Treatment of embryonic day 12.5 ( E12.5) metanephroi with the AT2R antagonist PD123319 or genetic inactivation of the AT2R in mice inhibits UB branching, decreasing the number of UB tips compared with control (34 ± 1.0 vs. 43 ± 0.6, P < 0.01; 36 ± 1.8 vs. 48 ± 1.3, P < 0.01, respectively). In contrast, treatment of metanephroi with the AT2R agonist CGP42112 increases the number of UB tips compared with control (48 ± 1.8 vs. 39 ± 12.3, P < 0.05). Using real-time quantitative RT-PCR and whole mount in situ hybridization, we demonstrate that PD123319 downregulates the expression of GDNF, c-Ret, Wnt11, and Spry1 mRNA levels in E12.5 metanephroi grown ex vivo. AT2R blockade or genetic inactivation of AT2R stimulates apoptosis and inhibits proliferation of the UB cells in vivo. We conclude that AT2R performs essential functions during UB branching morphogenesis via control of the GDNF/c-Ret/Wnt11 signaling pathway, UB cell proliferation, and survival.


2020 ◽  
Vol 15 (1) ◽  
pp. 400-408
Author(s):  
Xin You ◽  
Hongyan Cui ◽  
Ning Yu ◽  
Qiuli Li

AbstractPreeclampsia (PE) is a serious disease during pregnancy associated with the dysfunction of trophoblast cell invasion. DDX46 is a kind of RNA helicase that has been found to regulate cancer cell metastasis. However, the role of DDX46 in PE remains unclear. Our results showed that the mRNA levels of DDX46 in placental tissues of pregnant women with PE were markedly lower than those in normal pregnancies. Loss-of-function assays showed that knockdown of DDX46 significantly suppressed cell proliferation of trophoblast cells. Besides, DDX46 knockdown decreased trophoblast cell migration and invasion capacity. In contrast, the overexpression of DDX46 promoted the migration and invasion of trophoblast cells. Furthermore, knockdown of DDX46 caused significant decrease in the levels of p-PI3K, p-Akt, and p-mTOR in HTR-8/SVneo cells. In addition, treatment with IGF-1 reversed the inhibitory effects of DDX46 knockdown on proliferation, migration, and invasion of HTR-8/SVneo cells. In conclusion, these data suggest that DDX46 might be involved in the progression of PE, which might be attributed to the regulation of PI3K/Akt/mTOR signaling pathway. Thus, DDX46 might serve as a therapeutic target for the treatment of PE.


Author(s):  
Mi Cheng ◽  
Zequn Liu ◽  
Wanqing Ji ◽  
Jie Zheng ◽  
Huiqian Zeng ◽  
...  

<b><i>Objectives:</i></b> The pathogenesis of preeclampsia (PE) is associated with impaired trophoblast invasion, which results in placental insufficiency. Our earlier studies demonstrated that tissue transglutaminase (tTG) is highly expressed in human PE serum. However, whether tTG participates in trophoblast invasion remains unclear. The aim of the present study was to determine the role and mechanism of tTG in regulating matrix metalloproteinase (MMP)-2/MMP-9 expression to reduce trophoblast invasiveness in PE. <b><i>Methods:</i></b> HTR-8/SVneo cells were transfected with a lentivirus vector and small interfering RNA targeting tTG. The protein level was detected by Western blotting. Cell proliferation and apoptosis were assessed by MTS and flow cytometry assays, respectively. Cell invasion was investigated by Transwell assay. In addition, the influence of tTG on PI3K and AKT mRNA levels in HTR-8/SVneo cells was evaluated using reverse transcription-quantitative PCR. <b><i>Results:</i></b> tTG-overexpression inhibited HTR-8/SVneo cell proliferation and invasion and promoted apoptosis. In addition, upregulation of tTG induced an increase of PI3K and phosphorylated AKT and a decrease of MMP-2 and MMP-9 expression. tTG-knockdown significantly promoted the proliferation and invasion of HTR-8/SVneo cells and inhibited the apoptosis. Furthermore, the PI3K expression level was reduced, and the MMP-2/MMP-9 protein levels were increased. <b><i>Conclusion:</i></b> Taken together, the present study demonstrated that tTG-overexpression inhibited HTR-8/SVneo cell invasion via reducing the expression of MMP-2 and MMP-9 by activating PI3K/AKT signaling pathway, which may lead to the occurrence or development of PE. The present data provide new insights into modulation of tTG expression as a potential therapeutic target for PE.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4809-4809
Author(s):  
Jin Yang ◽  
Dan Hong ◽  
Qi Zhou ◽  
Jun-jie Fan ◽  
Le Li ◽  
...  

Abstract Background and purpose: Chemokine (C-C motif) ligand 2(CCL2) is a member of the CC subfamily which displays chemotactic activity for monocytes and basophils. It has played a very important role in many solid tumors and changes in bone marrow microenvironment. However, its role in acute myeloid leukemia (AML) has not yet been clear. In this point, we established a cell line with CCL2 down-expression to explore the effect of CCL2 gene on leukemogenesis. Methods: Lentivirus with CCL2-knockdown was successfully constructed after screening effective CCL2 shRNA sequence and tranfected into HL-60 cells which was validated on the level of mRNA and protein by real-time PCR and Western blot. The cells coming from parental, sh-Vectors and shCCL2 were detected for cell growth viability by CCK-8 assay, cell cycles and apoptosis by Flow cytometry. We applied exon sequencing technology to identify the gene profiling between the CCL2 knockdown and the control, of which, Cyclin d1 was selected for further experiments as its expression level was significantly downregulated. Then we successfully down regulated cyclin d1 expression in HL-60 by means of RNA interference to detect the cell proliferation through CCK-8 assay, cell cycles and apoptosis through Flow cytometry. Results: HL-60 cell line expressed the highest level of CCL2 among acute leukemia cell lines (Figure 1). Among 4 pairs of CCL2 interference sequences, only pair 2 had the most efficient potential in knockdown CCL2 expression which was constructed into sh-Vector, GV248, and validated by real-time RT-PCR and Western blot(Figure 2). Low expression of CCL2 significantly decreased HL-60 cell growth. Meanwhile, the CCL2-shRNA-mediated HL-60 cells showed about 12% more cells arrested in G1 phase compared with controls (Table 1, Figure 3). The results of expression profiling showed that there were total 159 genes differentially expressed (Figure 4), of which, ten top pathways were illustrated in Table 2. Cyclin D1 was related to cell cycle, NOD-like receptor signaling pathway, TNF signaling pathway and NF-kappa B signaling pathway which was the lowest expression among cell cycle gene related in HL-60 cells transfect with shCCL2(Table 2, highlighted raw) and further validated by real-time RT-PCR and Western blot (Figure 5). After Cyclin D1 was decreased on the level of mRNA and protein of HL-60, the cell proliferation was evidently slow and cell cycle analysis also indicated a similar pattern of CCL2 (Figure 6). Conclusion: CCL2 involved in cell proliferation which was mediated by cyclin D1 via blocking more cells at G1 phase. Figure 3. Knockdown of CCL2 inhibits cell proliferation via G1 phase arrested. A: Down regulation of CCL2 influenced cell proliferation. From day 2 to 5, the proliferation rate of HL-60 cells transfected by shCCL2 grew significantly slower than controls. B: CCL2 played a role in cell cycle process. More cells transfected shCCL2 were arrested in G1 phase compared with controls. *Indicate significant differences with P-values <0.05 Figure 5. Cylin D1 was the most influenced gene among cell proliferation related genes profile. A: quantitative RT-PCR analysis showed that mRNA levels of preliferation related genes: PCNA, cyclin D1, c-jun, surviving, erk1, and erk2. Among them, Cyclin D1 was expressed lowest. B: Western blot analysis confirmed that the protein expression of total cyclin D1 was much lower compared with controls. Figure 6. The effect of Cyclin D1 on cell proliferation. A: Cyclin D1 was successfully knockdowned in HL-60. mRNA levels were determined by quantitative RT-PCR and revealed that Cyclin D1 expression was knockdowned by the vector of shccnd. B: Knockdown of cyclin D1 inhibits cell proliferation. Compared with control, HL-60 cells with low level of Cyclin D grew significant slowly. C: Down regulation of Cyclin D1 influences more cells arrested in G1 phase compared with control. *P-values <0.05 Figure 1. Figure 1. Figure 2. Figure 2. Figure 3. Figure 3. Disclosures No relevant conflicts of interest to declare.


Chemotherapy ◽  
2018 ◽  
Vol 63 (5) ◽  
pp. 293-300 ◽  
Author(s):  
Yong Cui ◽  
Yan Su ◽  
Liya Deng ◽  
Wenjing Wang

Background/Aims: Although the cure rate for retinoblastoma is high, surviving patients are at risk for developing secondary cancers and require life-long follow-up. It is imperative to discover and develop novel therapeutic agents with better efficiency and fewer adverse effects. Ginsenoside-Rg5 is an active derivate from ginseng and exerts anti-cancer activity in breast cancer cells. However, it is still unclear whether ginsenoside-Rg5 has similar anti-cancer functions in retinoblastoma. Methods: Retinoblastoma cells were treated with ginsenoside-Rg5, followed by MTT assay analysis of the cell viability, cell number assay and colony formation assay analyses of cell proliferation, and flow cytometric analysis of apoptosis. Gene mRNA levels and protein levels were determined by quantitative real-time PCR and Western blot, respectively. Results: Ginsenoside-Rg5 inhibited retinoblastoma cell viability in a dose-dependent and time-dependent manner via preventing cell proliferation and inducing cell apoptosis. BCL2 expression was downregulated by ginsenoside-Rg5 treatment via inactivating the AKT signaling pathway. BCL2 overexpression completely eliminated the inhibitory effect of ginsenoside-Rg5 on cancer cell viability. Conclusion: Ginsenoside-Rg5 inhibits cell proliferation and induces apoptosis in retinoblastoma cells by inactivating the AKT signaling pathway, thereby downregulating BCL2 expression.


2016 ◽  
Vol 39 (5) ◽  
pp. 2001-2013 ◽  
Author(s):  
Dongmei Yu ◽  
Yong Shang ◽  
Jian Yuan ◽  
Shuang Ding ◽  
Sai Luo ◽  
...  

Objectives: Our goal was to investigate the relationship between keloid and telomerase as well as clarifying the influence of Wnt/β-catenin signaling on keloid cell proliferation. Methods: Tissues from 18 keloid patients were collected for further study. Keloid progenitor cells (KPC) and skin progenitor cells (SKP) were both included in this study. Lenti-virus transfection was used to divide cells into different groups in which cells were treated with different substances: negative control (NC) group, wnt10a siRNA group, β-catenin siRNA group and TERT siRNA group. KPC cells were injected into 20 male BALB/c nude mice in order to build tumor models. Several experiments including immunohistochemistry, western blot and RT-PCR were conducted in order to detect the corresponding protein expressions and relative mRNA levels. MTT assay and flow cytometry were also conducted for assessing cell proliferation and apoptosis status. Results: β-catenin and telomerase expression levels in keloid tissues were elevated compared to normal tissues (all P < 0.05). KPC cells in keloid exhibited more dynamic telomerase activity than SKP cells (P < 0.05). Luciferase activity assay confirmed that β-catenin could directly interact with telomerase. After wnt10a/β-catenin signaling pathway was inhibited, the proliferation of KPC cells was significantly suppressed and the apoptosis rate was remarkably increased (all P < 0.05). Results from tumor models also validated that wnt10a/β-catenin signaling pathway influenced the activity and length of telomerase. Conclusions: Wnt/β-catenin signaling pathway is able to exacerbate keloid cell proliferation and inhibit the apoptosis of keloid cells through its interaction with telomerase.


Gene ◽  
2014 ◽  
Vol 546 (1) ◽  
pp. 50-55 ◽  
Author(s):  
Yohei Kanamori ◽  
Masaru Murakami ◽  
Tohru Matsui ◽  
Masayuki Funaba

2018 ◽  
Vol 50 (6) ◽  
pp. 2216-2228 ◽  
Author(s):  
Ting Zhang ◽  
Hongmei Wang ◽  
Qiang Li ◽  
Jianliang Fu ◽  
Jiankang Huang ◽  
...  

Background/Aims: This study focused on evaluating the effect of MALAT1 and MDM2 on ischemic stroke through regulation of the p53 signaling pathway. Materials: Bioinformatics analysis was performed to identify abnormally expressed lncRNAs, mRNAs and their associated pathways. Oxygen-glucose deprivation/reoxygenation (OGD/R) in cells and middle cerebral artery occlusion/reperfusion (MCAO/R) in mice were performed to simulate an ischemic stroke environment. Western blot and qRT-PCR were used to examine lncRNA expression and mRNA levels. Fluorescence in situ hybridization (FISH) LncRNA was used to locate mRNA. MTT and flow cytometry were performed to examine cell proliferation and apoptosis. Finally, immunohistochemistry was used to observe the expression of genes in vivo. Results: MALAT1 and MDM2, which exhibit strong expression in stroke tissues, were subjected to bioinformatics analysis, and the p53 pathway was chosen for further study. MALAT1, MDM2 and p53 signaling pathway-related proteins were all up regulated in OGD/R cells. Furthermore, Malat1, Mdm2 and p53 pathway related-proteins were also up regulated in MCAO/R mice. Both MALAT1 and MDM2 were localized in the nuclei. Down regulation of MALAT1 and MDM2 enhanced cell proliferation ability and reduced apoptosis, resulting in decreased infarct size in MCAO/R brains. Conclusion: These results indicate that MALAT1/MDM2/p53 signaling pathway axis may provide more effective clinical therapeutic strategy for patients with ischemic stroke.


Sign in / Sign up

Export Citation Format

Share Document