scholarly journals p53-Dependent Transcriptional Repression of c-myc Is Required for G1 Cell Cycle Arrest

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.

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.


2001 ◽  
Vol 21 (7) ◽  
pp. 2259-2268 ◽  
Author(s):  
Wen-Shu Wu ◽  
Sadeq Vallian ◽  
Edward Seto ◽  
Wen-Ming Yang ◽  
Diane Edmondson ◽  
...  

ABSTRACT The growth suppressor promyelocytic leukemia protein (PML) is disrupted by the chromosomal translocation t(15;17) in acute promyelocytic leukemia (APL). PML plays a key role in multiple pathways of apoptosis and regulates cell cycle progression. The present study demonstrates that PML represses transcription by functionally and physically interacting with histone deacetylase (HDAC). Transcriptional repression mediated by PML can be inhibited by trichostatin A, a specific inhibitor of HDAC. PML coimmunoprecipitates a significant level of HDAC activity in several cell lines. PML is associated with HDAC in vivo and directly interacts with HDAC in vitro. The fusion protein PML-RARα encoded by the t(15;17) breakpoint interacts with HDAC poorly. PML interacts with all three isoforms of HDAC through specific domains, and its expression deacetylates histone H3 in vivo. Together, the results of our study show that PML modulates histone deacetylation and that loss of this function in APL alters chromatin remodeling and gene expression. This event may contribute to the development of leukemia.


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.


2013 ◽  
Vol 2013 ◽  
pp. 1-7 ◽  
Author(s):  
Yan Sun ◽  
Hui-Juan Xu ◽  
Yan-Xia Zhao ◽  
Ling-Zhen Wang ◽  
Li-Rong Sun ◽  
...  

Crocin is a carotenoid of the saffron extract that exhibits antitumor activity against many human tumors. However, the effects of crocin on HL-60 cells in vivo have not been evaluated. This study aimed to examine the effects of crocin on HL-60 cells in vitro and in vivo and investigate the underlying mechanisms. HL-60 cells were treated by crocin, and cell proliferation, apoptosis, and cell cycle profiles were examined by MTT assay, AO/EB staining, and flow cytometry, respectively. Furthermore, HL-60 cells were xenografted into nude mice and treated by crocin, the tumor weight and size were calculated, and the expression of Bcl-2 and Bax in xenografts was detected by immunohistochemical staining. The results showed that crocin (0.625–5 mg/mL) inhibited HL-60 cell proliferation and induced apoptosis and cell cycle arrest at G0/G1 phase, in a concentration and time-dependent manner. In addition, crocin (6.25, 25 mg/kg) inhibited the tumor weight and size of HL-60 xenografts in nude mice, inhibited Bcl-2 expression, and increased Bax expression in xenografts. In summary, crocin inhibits the proliferation and tumorigenicity of HL-60 cells, which may be mediated by the induction of apoptosis and cell cycle arrest and the regulation of Bcl-2 and Bax expression.


2003 ◽  
Vol 23 (21) ◽  
pp. 7719-7731 ◽  
Author(s):  
Hasan Siddiqui ◽  
David A. Solomon ◽  
Ranjaka W. Gunawardena ◽  
Ying Wang ◽  
Erik S. Knudsen

ABSTRACT The retinoblastoma tumor suppressor protein (RB) is targeted for inactivation in the majority of human tumors, underscoring its critical role in attenuating cellular proliferation. RB inhibits proliferation by repressing the transcription of genes that are essential for cell cycle progression. To repress transcription, RB assembles multiprotein complexes containing chromatin-modifying enzymes, including histone deacetylases (HDACs). However, the extent to which HDACs participate in transcriptional repression and are required for RB-mediated repression has not been established. Here, we investigated the role of HDACs in RB-dependent cell cycle inhibition and transcriptional repression. We find that active RB mediates histone deacetylation on cyclin A, Cdc2, topoisomerase IIα, and thymidylate synthase promoters. We also demonstrate that this deacetylation is HDAC dependent, since the HDAC inhibitor trichostatin A (TSA) prevented histone deacetylation at each promoter. However, TSA treatment blocked RB repression of only a specific subset of genes, thereby demonstrating that the requirement of HDACs for RB-mediated transcriptional repression is promoter specific. The HDAC-independent repression was not associated with DNA methylation or gene silencing but was readily reversible. We show that this form of repression resulted in altered chromatin structure and was dependent on SWI/SNF chromatin remodeling activity. Importantly, we find that cell cycle inhibitory action of RB is not intrinsically dependent on the ability to recruit HDAC activity. Thus, while HDACs do play a major role in RB-mediated repression, they are dispensable for the repression of critical targets leading to cell cycle arrest.


2020 ◽  
Vol 20 (6) ◽  
pp. 734-750
Author(s):  
Wallax A.S. Ferreira ◽  
Rommel R. Burbano ◽  
Claudia do Ó. Pessoa ◽  
Maria L. Harada ◽  
Bárbara do Nascimento Borges ◽  
...  

Background: Pisosterol, a triterpene derived from Pisolithus tinctorius, exhibits potential antitumor activity in various malignancies. However, the molecular mechanisms that mediate the pisosterol-specific effects on glioma cells remain unknown. Objective: This study aimed to evaluate the antitumoral effects of pisosterol on glioma cell lines. Methods: The 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide (MTT) and trypan blue exclusion assays were used to evaluate the effect of pisosterol on cell proliferation and viability in glioma cells. The effect of pisosterol on the distribution of the cells in the cell cycle was performed by flow cytometry. The expression and methylation pattern of the promoter region of MYC, ATM, BCL2, BMI1, CASP3, CDK1, CDKN1A, CDKN2A, CDKN2B, CHEK1, MDM2, p14ARF and TP53 was analyzed by RT-qPCR, western blotting and bisulfite sequencing PCR (BSP-PCR). Results: Here, it has been reported that pisosterol markedly induced G2/M arrest and apoptosis and decreased the cell viability and proliferation potential of glioma cells in a dose-dependent manner by increasing the expression of ATM, CASP3, CDK1, CDKN1A, CDKN2A, CDKN2B, CHEK1, p14ARF and TP53 and decreasing the expression of MYC, BCL2, BMI1 and MDM2. Pisosterol also triggered both caspase-independent and caspase-dependent apoptotic pathways by regulating the expression of Bcl-2 and activating caspase-3 and p53. Conclusions: It has been, for the first time, confirmed that the ATM/ATR signaling pathway is a critical mechanism for G2/M arrest in pisosterol-induced glioma cell cycle arrest and suggests that this compound might be a promising anticancer candidate for further investigation.


Author(s):  
Shaikh Shohidul Islam ◽  
Md. Rezaul Karim ◽  
A. K. M. Asaduzzaman ◽  
A. H. M. Khurshid Alam ◽  
Zahid Hayat Mahmud ◽  
...  

Cancers ◽  
2021 ◽  
Vol 13 (15) ◽  
pp. 3708
Author(s):  
Bhaba K. Das ◽  
Aarthi Kannan ◽  
Quy Nguyen ◽  
Jyoti Gogoi ◽  
Haibo Zhao ◽  
...  

Merkel cell carcinoma (MCC) is an often-lethal skin cancer with increasing incidence and limited treatment options. Although immune checkpoint inhibitors (ICI) have become the standard of care in advanced MCC, 50% of all MCC patients are ineligible for ICIs, and amongst those treated, many patients develop resistance. There is no therapeutic alternative for these patients, highlighting the urgent clinical need for alternative therapeutic strategies. Using patient-derived genetic insights and data generated in our lab, we identified aurora kinase as a promising therapeutic target for MCC. In this study, we examined the efficacy of the recently developed and highly selective AURKA inhibitor, AK-01 (LY3295668), in six patient-derived MCC cell lines and two MCC cell-line-derived xenograft mouse models. We found that AK-01 potently suppresses MCC survival through apoptosis and cell cycle arrest, particularly in MCPyV-negative MCC cells without RB expression. Despite the challenge posed by its short in vivo durability upon discontinuation, the swift and substantial tumor suppression with low toxicity makes AK-01 a strong potential candidate for MCC management, particularly in combination with existing regimens.


Sign in / Sign up

Export Citation Format

Share Document