scholarly journals Capilliposide Isolated fromLysimachia capillipesHemsl. Induces ROS Generation, Cell Cycle Arrest, and Apoptosis in Human Nonsmall Cell Lung Cancer Cell Lines

2014 ◽  
Vol 2014 ◽  
pp. 1-11 ◽  
Author(s):  
Zheng-hua Fei ◽  
Kan Wu ◽  
Yun-liang Chen ◽  
Bing Wang ◽  
Shi-rong Zhang ◽  
...  

Several data has reported that capilliposide, extracted from a traditional Chinese medicine,Lysimachia capillipesHemsl. (LC) could exhibit inhibitory effect on cell proliferation in various cancers. The current study investigated the antitumor efficacy ofCapilliposideand elucidated its potential molecular mechanism involved in vivo and vitro. Our results indicated that LC capilliposide inhibited proliferation of lung cancer cells in a dose-dependent manner. LC capilliposide induced cell cycle arrest at the S stage and enhanced apoptosis in NSCLC cells. Treatment with LC capilliposide increased the intracellular level of ROS, which activated the mitochondrial apoptotic pathway. Blockage of ROS by NAC highly reversed the effect of LC capilliposide on apoptosis. Xenograft tumor growth was significantly lower in the LC-treated group compared with the untreated control group(P<0.05). The results also show that LC treatment does not produce any overt signs of acute toxicity in vivo. These findings demonstrate that LC capilliposide could exert an anti-tumor effect on NSCLC through mitochondrial-mediated apoptotic pathway and the activation of ROS is involved.

Marine Drugs ◽  
2020 ◽  
Vol 18 (10) ◽  
pp. 494
Author(s):  
Lan Wang ◽  
Yun Huang ◽  
Cui-hong Huang ◽  
Jian-chen Yu ◽  
Ying-chun Zheng ◽  
...  

Ascomylactam A was reported for the first time as a new 13-membered-ring macrocyclic alkaloid in 2019 from the mangrove endophytic fungus Didymella sp. CYSK-4 from the South China Sea. The aim of our study was to delineate the effects of ascomylactam A (AsA) on lung cancer cells and explore the antitumor molecular mechanisms underlying of AsA. In vitro, AsA markedly inhibited the cell proliferation with half-maximal inhibitory concentration (IC50) values from 4 to 8 μM on six lung cancer cell lines, respectively. In vivo, AsA suppressed the tumor growth of A549, NCI-H460 and NCI-H1975 xenografts significantly in mice. Furthermore, by analyses of the soft agar colony formation, 5-ethynyl-20-deoxyuridine (EdU) assay, reactive oxygen species (ROS) imaging, flow cytometry and Western blotting, AsA demonstrated the ability to induce cell cycle arrest in G1 and G1/S phases by increasing ROS generation and decreasing of Akt activity. Conversely, ROS inhibitors and overexpression of Akt could decrease cell growth inhibition and cell cycle arrest induced by AsA. Therefore, we believe that AsA blocks the cell cycle via an ROS-dependent Akt/Cyclin D1/Rb signaling pathway, which consequently leads to the observed antitumor effect both in vitro and in vivo. Our results suggest a novel leading compound for antitumor drug development.


2005 ◽  
Vol 25 (17) ◽  
pp. 7423-7431 ◽  
Author(s):  
Jenny S. L. Ho ◽  
Weili Ma ◽  
Daniel Y. L. Mao ◽  
Samuel Benchimol

ABSTRACT The ability of p53 to promote apoptosis and cell cycle arrest is believed to be important for its tumor suppression function. Besides activating the expression of cell cycle arrest and proapoptotic genes, p53 also represses a number of genes. Previous studies have shown an association between p53 activation and down-regulation of c-myc expression. However, the mechanism and physiological significance of p53-mediated c-myc repression remain unclear. Here, we show that c-myc is repressed in a p53-dependent manner in various mouse and human cell lines and mouse tissues. Furthermore, c-myc repression is not dependent on the expression of p21WAF1. Abrogating the repression of c-myc by ectopic c-myc expression interferes with the ability of p53 to induce G1 cell cycle arrest and differentiation but enhances the ability of p53 to promote apoptosis. We propose that p53-dependent cell cycle arrest is dependent not only on the transactivation of cell cycle arrest genes but also on the transrepression of c-myc. Chromatin immunoprecipitation assays indicate that p53 is bound to the c-myc promoter in vivo. We report that trichostatin A, an inhibitor of histone deacetylases, abrogates the ability of p53 to repress c-myc transcription. We also show that p53-mediated transcriptional repression of c-myc is accompanied by a decrease in the level of acetylated histone H4 at the c-myc promoter and by recruitment of the corepressor mSin3a. These data suggest that p53 represses c-myc transcription through a mechanism that involves histone deacetylation.


2017 ◽  
Vol 41 (1) ◽  
pp. 339-357 ◽  
Author(s):  
Qing Tang ◽  
JingJing Wu ◽  
Fang Zheng ◽  
Swei Sunny Hann ◽  
YuQing Chen

Background: Emodin has anti-neoplastic activities on multiple tumors. However, the molecular mechanisms underlying this effect still remain to be fully understood. Methods: Cell viability and cell cycle distribution were measured using 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assays and flow cytometry, respectively. Cell invasion and migration were examined by transwell invasion and wound healing assays. Western blot analysis was performed to examine the phosphorylation and protein expression of AMP-activated protein kinase alpha (AMPKα), extracellular signaling-regulated kinase 1/2 (ERK1/2), peroxisome proliferators-activated receptor gamma (PPARγ), insulin-like growth factor (IGF) binding protein 1 (IGFBP1) and the transcription factor Sp1. QRT-PCR was used to examine the mRNA levels of the IGFBP1 gene. Small interfering RNAs (siRNAs) were used to knockdown PPARγ and IGFBP1 genes. Exogenously expression of IGFBP1 and Sp1 was determined by transient transfection assays. IGFBP1 promoter activity was measured by Secrete-Pair Dual Luminescence Assay Kit. In vivo nude mice xenograft model and bioluminescent imaging system were used to confirm the findings. Results: We showed that emodin induced cell cycle arrest of NSCLC cells. Emodin increased PPARγ protein and luciferase reporter activity, which were abolished by inhibitors of MAPK extracellular signaling-regulated kinase (ERK) kinase (MEK)/ERK and AMPK. Silencing of PPARγ abrogated emodin-inhibited cell growth and cell cycle arrest. Furthermore, emodin elevated IGFBP1 mRNA, protein, and promoter activity through activation of PPARγ. Intriguingly, overexpressed Sp1 attenuated emodin-induced IGFBP1 expression, which was not observed in cells with silenced PPARγ gene. Moreover, silencing of IGFBP1 gene blunted emodin-induced inhibition of cell growth and cell cycle arrest. On the contrary, overexpressed IGFBP1 enhanced emodin-induced phosphorylation of AMPKα and ERK1/2, and restored emodin-inhibited growth in cells with silenced endogenous IGFBP1 gene. Emodin also inhibited growth of lung xenograft tumors and Sp1, and increased IGFBP1 and PPARγ protein expressions In vivo. Conclusion: Collectively, our results show that emodin inhibits growth of non-small-cell lung cancer (NSCLC) cells through ERK and AMPKα-mediated induction of PPARγ, followed by reduction of Sp1. This in turn induces IGFBP1 gene expression. Thus, the signaling cascades, positive feedback loop and cooperative interplay between transcription factors-induced the expression of IGFBP1 gene contribute to the overall responses of emodin. This study provides a novel mechanism by which emodin inhibits growth of human lung cancer cells.


2019 ◽  
Vol 18 ◽  
pp. 153473541983076 ◽  
Author(s):  
Hwa Jeong Kang ◽  
Jeehye Kim ◽  
Seong Hyeok Cho ◽  
So-Jung Park ◽  
Hwa-Seung Yoo ◽  
...  

Epidermal growth factor receptor mutation-positive non–small cell lung cancer is cared for mainly by target therapeutics in the clinical treatment at present. We investigated the antitumor effect of HangAmDan-B1 (HAD-B1) combined with afatinib on H1975 (L858R/T790M double mutation) lung cancer cells. The combined treatment of HAD-B1 with afatinib inhibited the proliferation of H1975 cells in a dose-dependent manner compared with the treatment of afatinib or HAD-B1 alone. The combined treatment group significantly induced early apoptosis and cell cycle arrest of the cells compared with afatinib- or HAD-B1-treated control group. Profile analysis of cell cycle proteins in H1975 cells treated with the combination of HAD-B1 and afatinib using InnoPharmaScreen antibody microarray showed downregulation of pERK1/2 and upregulation of p16 in the cells. In vivo tumor growth assay in xenograft animal model of human H1975 lung cancer cells revealed that the mean tumor volume in the group treated with the combination of HAD-B1 and afatinib showed a significant reduction compared with the control groups.


2013 ◽  
Vol 57 (12) ◽  
pp. 2103-2111 ◽  
Author(s):  
Noppawat Charoensinphon ◽  
Peiju Qiu ◽  
Ping Dong ◽  
Jinkai Zheng ◽  
Pearline Ngauv ◽  
...  

2021 ◽  
Vol 14 (11) ◽  
pp. 1108
Author(s):  
Hung-Tse Huang ◽  
Chia-Ching Liaw ◽  
Yu-Chi Lin ◽  
Geng-You Liao ◽  
Chih-Hua Chao ◽  
...  

Mesona procumbens is a popular material used in foods and herbal medicines in Asia for clearing heat and resolving toxins. However, phytochemical research on this plant is very rare. In this study, eleven new diterpenoids, mesonols A-K (1–11), comprising seven ent-kauranes, three ent-atisanes, and one sarcopetalane, were isolated from its methanolic extract. Structural elucidation of compounds 1–11 was performed by spectroscopic methods, especially 2D NMR, HRESIMS, and X-ray crystallographic analysis. All isolates were assessed for their antiproliferative activity, and compounds 1-4 showed potential antiproliferative activities against A549, Hep-3B, PC-3, HT29, and U937 cancer cells, with IC50 values ranging from 1.97 to 19.86 µM. The most active compounds, 1 and 2, were selected for further investigation of their effects on cell cycle progression, apoptosis, and ROS generation in U937 human leukemia cancer cells. Interestingly, it was found that compounds 1 and 2 induced antiproliferative effects in U937 cells through different mechanisms. Compound 1 caused cell cycle arrest at the G2/M phase and subsequent cell death in a dose- and time-dependent manner. However, 2-mediated antiproliferation of U937 cells triggered ROS-mediated mitochondrial-dependent apoptosis. These results provide insight into the molecular mechanism involved in the antiproliferative activities of compounds 1 and 2 in U937 cells. Altogether, the study showed that new diterpenoid compounds 1 and 2 from M. procumbens are potent and promising anticancer agents.


Gold Bulletin ◽  
2017 ◽  
Vol 50 (2) ◽  
pp. 177-189 ◽  
Author(s):  
V. Ramalingam ◽  
S. Revathidevi ◽  
T. S. Shanmuganayagam ◽  
L. Muthulakshmi ◽  
R. Rajaram

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 878-878
Author(s):  
Aristeidis Chaidos ◽  
Valentina Caputo ◽  
Ilaria Marigo ◽  
Binbin Liu ◽  
Suhail Chaudhry ◽  
...  

Abstract Despite the therapeutic advances that followed the introduction of new immunomodulatory drugs and proteasome inhibitors, multiple myeloma (MM) remains an incurable malignancy and eventually all patients develop and succumb to chemo-refractory disease. The recently developed bromodomain and extra terminal (BET) protein inhibitors are novel agents targeting the acetyl-binding pockets of the BET family proteins BRD2-4 and BRDT. BET proteins activate transcription through their ability to bind to acetyl-modified lysine residues of histone tails, thereby serving as chromatin scaffolds that recruit the P-TEFb and PAFc1 complexes to Polymerase II (RNA Pol II), ensuring transcriptional initiation and elongation. In preclinical models, two classes of BET inhibitors, benzodiazepines (e.g JQ1) and quinolones (e.g. I-BET151), have been shown to have significant anti-proliferative activity against a variety of hematologic tumours. However translation of these data to molecules suitable for clinical development has yet to be disclosed. Herein, we tested the anti-myeloma activity and extended the mechanistic insights on two BET inhibitors: the chemical probe molecule I-BET151 and I-BET762, an orally active benzodiazepine suitable for clinical development. I-BET151 was tested in vitro in 6 myeloma cell lines (MMCL) with cytogenetic profiles representative of the most common translocations found in MM. I-BET151 induces apoptosis and cell cycle arrest in all MMCL in a time-dependent manner, with IC50 ranging from 133nM to 411nM at 72hrs. With the exception of KMS11 cells, IC50 was similar in stroma-free conditions and in co-culture with MS5 stromal cells. Similarly, I-BET151 induces apoptosis and cell cycle arrest in primary MM cells (n=4) cultured in the presence of IL-6 and stroma. In a subcutaneous MM mouse model, as compared to placebo, treatment with I-BET151 30mg/Kg/day i.p for 21 days resulted in 4-5 fold reduction in tumour size (p<0.001). Gene expression profiling of H929 and KMS12BM MMCL following treatment with I-BET151 confirmed downregulation of oncogenic MYC and MYC-dependent transcriptional programmes, but also of MYC-independent molecular signatures. The latter include abrogation of a myeloma specific, IRF4-dependent oncogenic programme. To explore the transcriptional events implicated in MYC downregulation, we treated OPM-2 myeloma cells with I-BET151. We found a dose-dependent inhibition of cell proliferation with commensurate reduction in MYC mRNA levels. Using ChiP-RQ-PCR analysis we found that upon treatment with I-BET151, BRD2, 3 and 4 occupancy at the IgH1enhancer that drives overexpression of the juxtaposed MYC decreased in a time-dependent manner as early as 2hr post–treatment. Furthermore, recruitment of CDK9 and PAF, critical components of the P-TEFb and PAFc1 complexes respectively, and binding of RNA Pol II were almost abolished, suggesting that I-BET151-mediated transcriptional MYC silencing involves inhibition of BRD2-4 binding. Finally, we show that I-BET762, an orally active molecule suitable for clinical development, potently inhibited cell proliferation in vitro in 10 MMCL, with IC50 <1µM and kinetics profile similar to JQ1 and I-BET151. The anti-myeloma activity of I-BET762 was tested in vivo in a systemic xenograft model generated by injecting OPM-2 cells into NOD-SCID mice. Escalating I-BET762 doses from 10 mg/Kg od to 30 mg/Kg every other day, were well tolerated with no clear impact on body weight as compared to vehicle control. Plasma human light chain concentration was significantly reduced depending on dose (p<0.001). Human CD38+ bone marrow cells were <1% in mice treated with 10mg/Kg od or higher dose vs 10% in the vehicle-treated animals (p≤0.001). I-BET762 treatment resulted in a significant survival advantage observed in all I-BET762-treated groups of mice (p<0.002). In conclusion, I-BET151 and I-BET762 show potent anti-myeloma activity in vitro and in vivo. I-BET151 inhibition of BRD2-4 binding mediates MYC transcriptional silencing and cell cycle arrest, but MYC-independent mechanism are also likely to mediate the I-BET biological effects in MM. Our data is the first example of an orally active BET inhibitor significantly delaying MM progression in vivo and provides strong rationale for clinical testing in phase I/II trials. Disclosures: Tough: GlaxoSmithKline: Employment. Smithers:GlaxoSmithKline: Employment. Bassil:GlaxoSmithKline: Employment. Chapman:GlaxoSmithKline: Employment. Harker:GlaxoSmithKline: Employment. Barbash:GlaxoSmithKline: Employment. Tummino:GlaxoSmithKline: Employment. Al-Mahdi:GlaxoSmithKline: Employment. Haynes:GlaxoSmithKline: Employment. Cutler:GlaxoSmithKline: Employment. Le:GlaxoSmithKline: Employment. Witherington:GlaxoSmithKline: Employment. Parr:GlaxoSmithKline: Employment. Prinjha:GlaxoSmithKline: Employment.


Sign in / Sign up

Export Citation Format

Share Document