scholarly journals Wild-type p53 binds to MYC promoter G-quadruplex

2016 ◽  
Vol 36 (5) ◽  
Author(s):  
Marek Petr ◽  
Robert Helma ◽  
Alena Polášková ◽  
Aneta Krejčí ◽  
Zuzana Dvořáková ◽  
...  

We found that the p53 tumour suppressor protein binds specifically to the G-quadruplex DNA formed by MYC promoter sequence. We propose that p53 binding to G-quadruplexes can participate in regulation of p53 target genes.

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3825-3825
Author(s):  
Mariko Yoshimura ◽  
Jo Ishizawa ◽  
Archana Dilip ◽  
Sharon Shacham ◽  
Michael Kauffman ◽  
...  

Abstract We investigated the pathophysiological significance of MDM2 and the nuclear exporter Exportin 1 (XPO1/CRM1) on p53 transcription and their therapeutic targeting in mantle cell lymphoma (MCL). MCL is currently an incurable B-cell non-Hodgkin's lymphoma. Although TP53 mutations occur rarely (i.e., in 15 to 20% of the cases) in MCL, wild-type p53 is frequently inactivated by gene amplification of BMI1, homozygous deletion of CDKN2A (INK4a/ARF, over-expression of regulatory proteins, or gene deletion. For example, tumor cells may sequester p53 in the cytoplasm via the overexpression of proteins like MDM2 and XPO1 thereby abrogating its transcriptional anti-tumor activity. The selective MDM2 inhibitor Nutlin-3a (Vassilev, 2004) has been reported to induce p53-mediated apoptosis in MCL cells, and recently it was shown that the potent, small molecule Selective Inhibitor of Nuclear Export (SINE) induces apoptosis in MCL cells (Zhang, 2013). mRNA expression levels in MCL patient samples were determined using Oncomine (Compendia Bioscience, Ann Arbor, MI). MDM2 mRNA expression levels were not significantly higher in the MCL samples (n = 8) compared to normal B-cells ( n = 5) (P = 0.27). The MDM2 levels were also not associated with overall disease survival (P = 0.12) at 5 years (n = 63). In contrast, gene expression analysis on the same dataset demonstrated increased XPO1 mRNA expression in the MCL cells versus normal B-cells (P< 0.001). Furthermore, this higher XPO1 expression was associated with poorer prognosis in MCL patients (i.e., median overall survival: 3.2 years in low expression XPO1 cases and 1.9 years in high expression CRM1 cases, P = 0.033). mRNA levels of 11 representative p53 target genes were determined in 2 MCL (i.e., Z-138 and JVM-2) cells and were compared to those of four acute leukemia (i.e., OCI-AML3, MOLM-13, REH and NALM-6) cells. Cells from all six cell lines express wild-type p53. The MDM2 inhibitor Nutlin-3a (5 µM) poorly induced p53 target genes in the Z-138 (1.9 ± 0.12-fold) and JVM-2 cells (2.1 ± 0.23-fold) compared to acute leukemia cells (13.9 ± 5.2-fold) (P< 0.001 in both MCL cells), despite their increased p53 protein levels after Nutlin-3a treatment. The data suggest that p53-mediated transcription may be impaired in MCL via a Nutlin-induced increase in cytoplasmic p53 levels. Next, we treated Z-138 with 2 or 4 mM Nutlin-3a or with 80 or 160 nM KPT-185 for 18 hours and determined mRNA changes in p53 targets. KPT-185 induced p53 target genes more potently than Nutlin-3a (i.e., 2- to 4-times). To investigate the significance of p53-mediated apoptosis, Z-138 and JVM-2 were transduced with lentivirus encoding either negative control shRNA (ShC) or p53-specific shRNA (Shp53) and stable shRNA-expressing cells were generated. The p53-specific shRNA reduced p53 basal levels by 80 to 90%, and p53 knockdown cells were ∼80% less sensitive to Nutlin-3a and ∼60% to KPT-185, indicating that MDM2 or XPO1 inhibition rely on p53 pathway to induce apoptosis. Apoptotic induction by Nutlin-3a and KPT-185 in primary MCL cells were determined in 3 samples with wild-type p53. Treatment-specific Annexin V induction percentage in 2.5 µM Nutlin-3a-treated cells was 40.7 ± 8.0%, while it was 64.5 ± 15.0% in 100 nM KPT-185 treated cells. Treatment of 1 µM KPT-185 led to more prominent apoptosis, as evidenced by 81.8 ± 7.7% annexin V staining. Our data suggest that p53 is a component of MDM2 or XPO1 inhibition-induced apoptosis, and p53 transcription is more potently activated by XPO1 inhibition rather than MDM2 inhibition. Based on these data, we propose that active modulation of p53 by SINE XPO1 antagonism and/or MDM2 inhibition transcription would be beneficial in some cancer types to enhance p53-based cancer therapy. Disclosures: Shacham: Karyopharm Therapeutics Inc.: Employment. Kauffman:Karyopharm Therapeutics Inc.: Employment. Andreeff:Karyopharm Therapeutics Inc.: Research Funding.


Genes ◽  
2021 ◽  
Vol 12 (2) ◽  
pp. 277
Author(s):  
Paola Monti ◽  
Vaclav Brazda ◽  
Natália Bohálová ◽  
Otília Porubiaková ◽  
Paola Menichini ◽  
...  

P53, P63, and P73 proteins belong to the P53 family of transcription factors, sharing a common gene organization that, from the P1 and P2 promoters, produces two groups of mRNAs encoding proteins with different N-terminal regions; moreover, alternative splicing events at C-terminus further contribute to the generation of multiple isoforms. P53 family proteins can influence a plethora of cellular pathways mainly through the direct binding to specific DNA sequences known as response elements (REs), and the transactivation of the corresponding target genes. However, the transcriptional activation by P53 family members can be regulated at multiple levels, including the DNA topology at responsive promoters. Here, by using a yeast-based functional assay, we evaluated the influence that a G-quadruplex (G4) prone sequence adjacent to the p53 RE derived from the apoptotic PUMA target gene can exert on the transactivation potential of full-length and N-terminal truncated P53 family α isoforms (wild-type and mutant). Our results show that the presence of a G4 prone sequence upstream or downstream of the P53 RE leads to significant changes in the relative activity of P53 family proteins, emphasizing the potential role of structural DNA features as modifiers of P53 family functions at target promoter sites.


2012 ◽  
Vol 2012 ◽  
pp. 1-10 ◽  
Author(s):  
Rebecca Elston ◽  
Gareth J. Inman

Wild-type p53 and TGF-β are key tumour suppressors which regulate an array of cellular responses. TGF-β signals in part via the Smad signal transduction pathway. Wild-type p53 and Smads physically interact and coordinately induce transcription of a number of key tumour suppressive genes. Conversely mutant p53 generally subverts tumour suppressive TGF-β responses, diminishing transcriptional activation of key TGF-β target genes. Mutant p53 can also interact with Smads and this enables complex formation with the p53 family member p63 and blocks p63-mediated activation of metastasis suppressing genes to promote tumour progression. p53 and Smad function may also overlap during miRNA biogenesis as they can interact with the same components of the Drosha miRNA processing complex to promote maturation of specific subsets of miRNAs. This paper investigates the crosstalk between p53 and TGF-β signalling and the potential roles this plays in cancer biology.


2019 ◽  
Vol 21 (1) ◽  
pp. 127
Author(s):  
Otília Porubiaková ◽  
Natália Bohálová ◽  
Alberto Inga ◽  
Natália Vadovičová ◽  
Jan Coufal ◽  
...  

p53 is one of the most studied tumor suppressor proteins that plays an important role in basic biological processes including cell cycle, DNA damage response, apoptosis, and senescence. The human TP53 gene contains alternative promoters that produce N-terminally truncated proteins and can produce several isoforms due to alternative splicing. p53 function is realized by binding to a specific DNA response element (RE), resulting in the transactivation of target genes. Here, we evaluated the influence of quadruplex DNA structure on the transactivation potential of full-length and N-terminal truncated p53α isoforms in a panel of S. cerevisiae luciferase reporter strains. Our results show that a G-quadruplex prone sequence is not sufficient for transcription activation by p53α isoforms, but the presence of this feature in proximity to a p53 RE leads to a significant reduction of transcriptional activity and changes the dynamics between co-expressed p53α isoforms.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1484-1484 ◽  
Author(s):  
Leandro C.A. Cerchietti ◽  
Jose M. Polo ◽  
Gustavo F. Da Silva ◽  
Steve M. Dowdy ◽  
Catoretti M. Giorgio ◽  
...  

Abstract The BCL6 transcriptional repressor is an oncogene often constitutively expressed in diffuse large B-cell lymphomas (DLBCL). The oncogenic mechanism of action of BCL6 presumably involves repression of its direct target genes. We recently developed a targeted therapy agent (called BPI - BCL6 peptide inhibitor) that specifically blocks transcriptional repression by BCL6, and which causes apoptosis in lymphoma cells in vitro and in vivo. We present here potent and stable derivatives of BPI able to specifically eradicate lymphoma cells after a single dose in vitro. Expression array studies of BCL6 target genes reactivated by BPI revealed that one such gene is the p53 tumor suppressor. p53 was also recently shown to be BCL6 target gene by Phan et. al., Nature 2004. We find that BCL6 represses p53 in DLBCL cells through recruitment of the SMRT and N-CoR corepressors, which explains how BPI, which blocks recruitment of these corepressors, reactivates p53. We next wished to determine the contribution of BCL6-mediated repression of p53 to lymphomagenesis, and how p53 modulation might affect BCL6 targeted therapy strategies for DLBCL. We found that BPI could induce p53 target gene expression in DLBCL cells with wild-type p53 and that small molecules or peptides that block p53 rescue apoptosis induced by BPI. In contrast, although BPI also induces p53 in DLCBL cells with mutant p53, there was no activation of p53 target genes and no rescue by p53 blocking molecules. However BPI causes apoptosis of DLBCL cells regardless of p53 status indicating the BCL6 mediates its oncogenic actions through both p53 dependent and independent pathways. p53 is usually wild-type in DLBCL and our analysis of &gt;100 patients show that p53 protein is, surprisingly, still expressed in these tumors. These data suggest that p53 is not fully active in DLBCL cells, consistent with the fact that we found that BCL6 also directly represses upstream activators of p53 such as Chk1 and ATR. BCL6 blockade thus can fully restore activity of p53, both by increasing its expression levels and by enhancing its activation by upstream mediators. Accordingly, sequential administration of p53 activating molecules that enhance p53 activity, potently synergizes with BPI in killing lymphoma cells. BPI also synergizes with chemotherapy drugs that act in part through p53, such as doxorubicin. From these studies we conclude that i) BCL6 mediates lymphomagenesis by direct repression of p53 and upstream target gene pathways; ii) BCL6 positive lymphomas are dependent on BCL6 for their survival regardless of whether p53 is wild type or mutated; iii) Sequential targeting of BCL6 and p53 with BPI and a p53 activating molecule or doxorubicin is likely to be a highly effective therapeutic regimen for patients with DLBCL, especially for the majority who have wild-type p53; iv) The new BPI derivatives are sufficiently potent and stable to be tested in the clinical setting.


2009 ◽  
Vol 83 (6) ◽  
pp. 2491-2499 ◽  
Author(s):  
Eleonora Forte ◽  
Micah A. Luftig

ABSTRACT Epstein-Barr virus (EBV) growth transformation of primary B lymphocytes into indefinitely proliferating lymphoblastoid cell lines (LCLs) depends on the concerted activities of a subset of viral proteins expressed during latency. EBV drives quiescent B cells into S phase, and consequently, a host response is activated that includes expression of p53 and its target genes. Since LCLs retain wild-type p53, it was of interest to determine what contribution the p53 pathway may have in controlling established LCL growth and EBV-mediated transformation of primary B cells. We found that liberation of p53 through chemical antagonism of one of its major ubiquitin ligases, MDM2, using the small-molecule Nutlin-3 led to apoptosis of established LCLs and suppressed EBV-mediated transformation of primary B cells. The activation of latent p53 induced target genes associated with apoptosis. Furthermore, MDM2 antagonism synergized with NF-κB inhibition in killing LCLs. NF-κB was important to increase steady-state MDM2 protein levels rather than in affecting p53-dependent transcription, suggesting a unique mechanism by which LCLs survive in the presence of a primed p53 pathway. Nutlin sensitivity of EBV-infected cells provides a novel system for studying the pathways that dictate LCL survival and regulate EBV transformation. Finally, MDM2 antagonists may be considered for therapeutic intervention in EBV-associated malignancies expressing wild-type p53.


2020 ◽  
Author(s):  
Anne-Louise Latif ◽  
Ashley Newcombe ◽  
Sha Li ◽  
Kathryn Gilroy ◽  
Neil Robertson ◽  
...  

SummaryAcute Myeloid Leukemia (AML) is a typically-lethal molecularly heterogeneous disease, with few broad-spectrum therapeutic targets. Unusually, most AML retain wild-type TP53, encoding the pro-apoptotic tumor suppressor p53. MDM2 inhibitors (MDM2i), which activate wild-type p53, and BET inhibitors (BETi), targeting the BET-family co-activator BRD4, both show encouraging pre-clinical activity, but limited clinical activity as single agents. Here, we report synergistic toxicity of combined MDM2i and BETi towards AML cell lines, primary human blasts and mouse models, resulting from BETi’s ability to evict an unexpected repressive form of BRD4 from p53 target genes, and hence potentiate MDM2i-induced p53 activation. These results indicate that wild-type TP53 and a transcriptional repressor function of BRD4 together represent a potential broad-spectrum synthetic therapeutic vulnerability for AML.


Sign in / Sign up

Export Citation Format

Share Document