Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Oncogenesis ; 7(3): 33, 2018 Mar 29.
Article in English | MEDLINE | ID: mdl-29593251

ABSTRACT

Uveal melanoma (UM) is the most frequent ocular cancer in adults, accounting for ~5% of the total melanoma incidence. Although the primary tumor is well treatable, patients frequently develop metastases for which no curative therapy exists. Highly activated protein kinase C (PKC) is a common feature of UM and has shown potential as therapeutic intervention for UM patients. Unfortunately, PKC inhibition as single treatment appears to have only limited clinical benefit. Combining PKC inhibition with activation of p53, which is rarely mutated in UM, by MDM2 inhibitors has shown promising results in vitro and in vivo. However, clinical studies have shown strong adverse effects of MDM2 inhibition. Therefore, we investigated alternative approaches to achieve similar anticancer effects, but with potentially less adverse effects. We studied the potential of targeting MDMX, an essential p53 inhibitor during embryonal development but less universally expressed in adult tissues compared with MDM2. Therefore, targeting MDMX is predicted to have less adverse effects in patients. Depletion of MDMX, like the pharmacological activation of p53, inhibits the survival of UM cells, which is enhanced in combination with PKC inhibition. Also pan-PKC inhibitors elicit adverse effects in patients. As the PKC family consists of 10 different isoforms, it could be hypothesized that targeting a single PKC isoform would have less adverse effects compared with a pan-PKC inhibitor. Here we show that specifically depleting PKCδ inhibits UM cell growth, which can be further enhanced by p53 reactivation. In conclusion, our data show that the synergistic effects of p53 activation by MDM2 inhibition and broad spectrum PKC inhibition on survival of UM cells can also largely be achieved by the presumably less toxic combination of depletion of MDMX and targeting a specific PKC isoform, PKCδ.

2.
Cell Death Dis ; 6: e1821, 2015 Jul 16.
Article in English | MEDLINE | ID: mdl-26181202

ABSTRACT

The function of the tumor suppressor p53 is universally compromised in cancers. It is the most frequently mutated gene in human cancers (reviewed). In cases where p53 is not mutated, alternative regulatory pathways inactivate its tumor suppressive functions. This is primarily achieved through elevation in the expression of the key inhibitors of p53: Mdm2 or Mdmx (also called Mdm4) (reviewed). In breast cancer (BrCa), the frequency of p53 mutations varies markedly between the different subtypes, with basal-like BrCas bearing a high frequency of p53 mutations, whereas luminal BrCas generally express wild-type (wt) p53. Here we show that Mdmx is unexpectedly highly expressed in normal breast epithelial cells and its expression is further elevated in most luminal BrCas, whereas p53 expression is generally low, consistent with wt p53 status. Inducible knockdown (KD) of Mdmx in luminal BrCa MCF-7 cells impedes the growth of these cells in culture, in a p53-dependent manner. Importantly, KD of Mdmx in orthotopic xenograft transplants resulted in growth inhibition associated with prolonged survival, both in a preventative model and also in a treatment model. Growth impediment in response to Mdmx KD was associated with cellular senescence. The growth inhibitory capacity of Mdmx KD was recapitulated in an additional luminal BrCa cell line MPE600, which expresses wt p53. Further, the growth inhibitory capacity of Mdmx KD was also demonstrated in the wt p53 basal-like cell line SKBR7 line. These results identify Mdmx growth dependency in wt p53 expressing BrCas, across a range of subtypes. Based on our findings, we propose that Mdmx targeting is an attractive strategy for treating BrCas harboring wt p53.


Subject(s)
Apoptosis/genetics , Breast Neoplasms/genetics , Nuclear Proteins/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Tumor Suppressor Protein p53/genetics , Animals , Breast Neoplasms/pathology , Cell Cycle Proteins , Cell Proliferation/genetics , Epithelial Cells/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Mice , Mutation , Nuclear Proteins/genetics , Proto-Oncogene Proteins/genetics , Xenograft Model Antitumor Assays
3.
Cell Death Differ ; 19(6): 980-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22158418

ABSTRACT

Reactivation of the p53 tumor-suppressor protein by small molecules like Nutlin-3 and RITA (reactivation of p53 and induction of tumor cell apoptosis) is a promising strategy for cancer therapy. The molecular mechanisms involved in the responses to RITA remain enigmatic. Several groups reported the induction of a p53-dependent DNA damage response. Furthermore, the existence of a p53-dependent S-phase checkpoint has been suggested, involving the checkpoint kinase Chk1. We have recently shown synergistic induction of apoptosis by RITA in combination with Nutlin-3, and we observed concomitant Chk2 phosphorylation. Therefore, we investigated whether Chk2 contributes to the cellular responses to RITA. Strikingly, the induction of apoptosis seemed entirely Chk2 dependent. Transcriptional activity of p53 in response to RITA required the presence of Chk2. A partial rescue of apoptosis observed in Noxa knockdown cells emphasized the relevance of p53 transcriptional activity for RITA-induced apoptosis. In addition, we observed an early p53- and Chk2-dependent block of DNA replication upon RITA treatment. Replicating cells seemed more prone to entering RITA-induced apoptosis. Furthermore, the RITA-induced DNA damage response, which was not a secondary effect of apoptosis induction, was strongly attenuated in cells lacking p53 or Chk2. In conclusion, we identified Chk2 as an essential mediator of the cellular responses to RITA.


Subject(s)
Apoptosis/drug effects , Furans/pharmacology , Protein Serine-Threonine Kinases/metabolism , Cell Line, Tumor , Checkpoint Kinase 2 , G2 Phase Cell Cycle Checkpoints , HCT116 Cells , Humans , Imidazoles/pharmacology , Phosphorylation , Piperazines/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
4.
Oncogene ; 31(9): 1105-16, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-21765463

ABSTRACT

The prognosis of patients with uveal melanoma is poor. Because of the limited efficacy of current treatments, new therapeutic strategies need to be developed. Because p53 mutations are uncommon in uveal melanoma, reactivation of p53 may be used to achieve tumor regression. We investigated the use of combination therapies for intraocular melanoma, based on the p53 activators Nutlin-3 and reactivation of p53 and induction of tumor cell apoptosis (RITA) and the topoisomerase I inhibitor Topotecan. Nutlin-3 treatment induced p53-dependent growth inhibition in human uveal melanoma cell lines. The sensitivity to Nutlin-3 of the investigated cell lines did not correlate with basal Hdm2 or Hdmx levels. Nutlin-3 synergized with RITA and Topotecan to induce apoptosis in uveal melanoma cell lines and short-term cultures. Drug synergy correlated with enhanced induction of p53-Ser46 phosphorylation, which was attenuated by ATM inhibition. Nutlin-3 and Topotecan also significantly delayed tumor growth in vivo in a murine B16F10 model for ocular melanoma. Combination treatment appeared to inhibit tumor growth slightly more efficient than either drug alone. Nutlin-3, RITA and Topotecan lead to comparable p53 activation and growth inhibition under normoxia and hypoxia. Treatment with Nutlin-3 or RITA had no effect on HIF-1α induction by hypoxia, whereas the combination of these two drugs did inhibit hypoxia-induced HIF-1α. Also Topotecan, alone or in combination with Nutlin-3, reduced HIF-1α protein levels, suggesting that a certain level of DNA damage response is required for p53-mediated downregulation of HIF-1α. In conclusion, combination treatments based on small-molecule-induced p53 activation may have clinical potential for uveal melanoma.


Subject(s)
Antineoplastic Agents/therapeutic use , Melanoma/drug therapy , Tumor Suppressor Protein p53/agonists , Uveal Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , DNA-Binding Proteins/antagonists & inhibitors , Drug Synergism , Furans/pharmacology , Furans/therapeutic use , Humans , Hypoxia , Imidazoles/pharmacology , Imidazoles/therapeutic use , Melanoma/genetics , Melanoma, Experimental/drug therapy , Melanoma, Experimental/genetics , Mice , Phosphorylation/drug effects , Piperazines/pharmacology , Piperazines/therapeutic use , Protein Serine-Threonine Kinases/antagonists & inhibitors , Topotecan/pharmacology , Topotecan/therapeutic use , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/antagonists & inhibitors , Uveal Neoplasms/genetics
5.
Cell Death Differ ; 18(11): 1736-45, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21546907

ABSTRACT

Inactivation of the p53 tumour suppressor, either by mutation or by overexpression of its inhibitors Hdm2 and HdmX is the most frequent event in cancer. Reactivation of p53 by targeting Hdm2 and HdmX is therefore a promising strategy for therapy. However, Hdm2 inhibitors do not prevent inhibition of p53 by HdmX, which impedes p53-mediated apoptosis. Here, we show that p53 reactivation by the small molecule RITA leads to efficient HdmX degradation in tumour cell lines of different origin and in xenograft tumours in vivo. Notably, HdmX degradation occurs selectively in cancer cells, but not in non-transformed cells. We identified the inhibition of the wild-type p53-induced phosphatase 1 (Wip1) as the major mechanism important for full engagement of p53 activity accomplished by restoration of the ataxia telangiectasia mutated (ATM) kinase-signalling cascade, which leads to HdmX degradation. In contrast to previously reported transactivation of Wip1 by p53, we observed p53-dependent repression of Wip1 expression, which disrupts the negative feedback loop conferred by Wip1. Our study reveals that the depletion of both HdmX and Wip1 potentiates cell death due to sustained activation of p53. Thus, RITA is an example of a p53-reactivating drug that not only blocks Hdm2, but also inhibits two important negative regulators of p53 - HdmX and Wip1, leading to efficient elimination of tumour cells.


Subject(s)
Apoptosis/drug effects , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Furans/pharmacology , Phosphoprotein Phosphatases/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins , Cell Line, Tumor , Down-Regulation , Gene Expression Regulation/drug effects , Humans , Nuclear Proteins/metabolism , Phosphoprotein Phosphatases/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Phosphatase 2C , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Signal Transduction , Tumor Suppressor Protein p53/antagonists & inhibitors
6.
Oncogene ; 29(16): 2415-26, 2010 Apr 22.
Article in English | MEDLINE | ID: mdl-20140020

ABSTRACT

The p53 tumor suppressor protein is frequently mutated in human tumors. It is thought that the p53 pathway is indirectly impaired in the remaining tumors, for example by overexpression of its important regulators Mdm2 and Mdm4, making them attractive targets for the development of anti-cancer agents. Recent studies have suggested that Mdm4 levels determine the sensitivity of tumor cells for anti-cancer therapy. To investigate this possibility, we studied the drug sensitivity of several breast cancer cell lines containing wild-type p53, but expressing different Mdm4 levels. We show that endogenous Mdm4 levels can affect the sensitivity of breast cancer cells to anti-cancer agents, but in a cell line-dependent manner and depending on an intact apoptotic response. Furthermore, treatment with the non-genotoxic agent Nutlin-3 sensitizes cells for doxorubicin, showing that activation of p53 by targeting its regulators is an efficient strategy to decrease cell viability of breast cancer cells. These results confirm a function of Mdm4 in determining the efficacy of chemotherapeutic agents to induce apoptosis of cancer cells in a p53-dependent manner, although additional undetermined factors also influence the drug response. Targeting Mdm4 to sensitize tumor cells for chemotherapeutic drugs might be a strategy to effectively treat tumors harboring wild-type p53.


Subject(s)
Breast Neoplasms/drug therapy , Nuclear Proteins/physiology , Proto-Oncogene Proteins/physiology , Apoptosis/drug effects , Breast Neoplasms/pathology , Cell Cycle Proteins , Cell Line, Tumor , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Female , Humans , Imidazoles/pharmacology , Nuclear Proteins/analysis , Piperazines/pharmacology , Proto-Oncogene Proteins/analysis , Tumor Suppressor Protein p53/physiology
7.
J Gen Virol ; 78 ( Pt 2): 423-6, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9018065

ABSTRACT

The cellular transcription co-activators p300 and the CREB-binding protein CBP are cellular targets for transformation by the E1A proteins of non-oncogenic adenovirus 5 (Ad5). In this study, we show that the E1A proteins of oncogenic Ad12, like those of Ad5, can also bind to CBP and that this interaction is direct. In addition, we show that the Ad12 E1A proteins can also bind directly to p300. These results suggest that E1A can modulate the function of proteins of the p300 family via direct protein-protein interactions.


Subject(s)
Adenoviridae/metabolism , Adenovirus E1A Proteins/metabolism , Nuclear Proteins/metabolism , Trans-Activators , Transcription Factors/metabolism , Adenovirus E1A Proteins/genetics , Blotting, Western , CREB-Binding Protein , Glutathione Transferase/genetics , HeLa Cells , Humans , Oncogenic Viruses/metabolism , Protein Binding , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
8.
J Gen Virol ; 80 ( Pt 2): 381-390, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10073698

ABSTRACT

The cdk-inhibitor p21(CIP1/WAF1) inhibits the activities of cyclin-dependent kinases and proliferating cell nuclear antigen, thereby repressing cell-cycle progression and DNA replication. Transforming oncogenes, such as E1A of human adenovirus 5 (Ad5), may interfere with such growth-inhibitory proteins. In this study, we show that in various Ad5E1-transformed cells, p21(CIP1/WAF1) is expressed and that, in general, expression is not downregulated. In addition, colony-formation assays show that in Ad5E1-transformed cells highly overexpressed p21(CIP1/WAF1) can still cause growth inhibition. FACS experiments indicate, however, that a G1 arrest induced by moderate overexpression of p21(CIP1/WAF1) can be overcome by E1A. The E1A proteins may interfere with the function of p21(CIP1/WAF1) by binding. Indeed, p21(CIP1/WAF1) binds with its cyclin/cdk-binding N terminus to the transforming N-terminal and CR1 region of the E1A proteins. Together, these results lend support to the model that E1A can interfere directly with p21(CIP1/WAF1) function and thereby stimulates cell growth.


Subject(s)
Adenovirus E1A Proteins/physiology , Adenoviruses, Human/physiology , Cyclins/physiology , Adenovirus E1A Proteins/genetics , Adenoviruses, Human/genetics , Adenoviruses, Human/pathogenicity , Animals , Cell Division , Cell Line , Cell Transformation, Viral , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/genetics , Enzyme Inhibitors/metabolism , G1 Phase , Gene Expression , Humans , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL