Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
NPJ Breast Cancer ; 10(1): 5, 2024 Jan 06.
Article in English | MEDLINE | ID: mdl-38184660

ABSTRACT

The extracellular matrix (ECM) is an important component of the tumor microenvironment and undergoes extensive remodeling during both initiation and progression of breast cancer (BC). EMILIN1 is an ECM glycoprotein, whose function has been linked to cancer and metastasis. However, EMILIN1 role during mammary gland and BC development has never been investigated. In silico and molecular analyses of human samples from normal mammary gland and BC showed that EMILIN1 expression was lower in tumors than in healthy mammary tissue and it predicted poor prognosis, particularly in HER2-positive BC. HER2+ BC accounts for 15-20% of all invasive BC and is characterized by high aggressiveness and poor prognosis. The Δ16HER2 isoform, a splice variant with very high oncogenic potential, is frequently expressed in HER2+ BC and correlates with metastatic disease. To elucidate the role of EMILIN1 in BC, we analyzed the phenotype of MMTV-Δ16HER2 transgenic mice, developing spontaneous multifocal mammary adenocarcinomas, crossed with EMILIN1 knock-out (KO) animals. We observed that Δ16HER2/EMILIN1 KO female mice exhibited an accelerated normal mammary gland development and a significantly anticipated appearance of palpable tumors (13.32 vs 15.28 weeks). This accelerated tumor initiation was corroborated by an increased number of tumor foci observed in mammary glands from Δ16HER2/EMILIN1 KO mice compared to the wild-type counterpart. Altogether our results underscore the centrality of ECM in the process of BC initiation and point to a role for EMILIN1 during normal mammary gland development and in protecting from HER2-driven breast tumorigenesis.

3.
Front Oncol ; 12: 891580, 2022.
Article in English | MEDLINE | ID: mdl-35712501

ABSTRACT

The cyclin D-CDK4/6 complexes play a pivotal role in controlling the cell cycle. Deregulation in cyclin D-CDK4/6 pathway has been described in many types of cancer and it invariably leads to uncontrolled cell proliferation. Many efforts have been made to develop a target therapy able to inhibit CDK4/6 activity. To date, three selective CDK4/6 small inhibitors have been introduced in the clinic for the treatment of hormone positive advanced breast cancer patients, following the impressive results obtained in phase III clinical trials. However, since their approval, clinical evidences have demonstrated that about 30% of breast cancer is intrinsically resistant to CDK4/6 inhibitors and that prolonged treatment eventually leads to acquired resistance in many patients. So, on one hand, clinical and preclinical studies fully support to go beyond breast cancer and expand the use of CDK4/6 inhibitors in other tumor types; on the other hand, the question of primary and secondary resistance has to be taken into account, since it is now very clear that neoplastic cells rapidly develop adaptive strategies under treatment, eventually resulting in disease progression. Resistance mechanisms so far discovered involve both cell-cycle and non-cell-cycle related escape strategies. Full understanding is yet to be achieved but many different pathways that, if targeted, may lead to reversion of the resistant phenotype, have been already elucidated. Here, we aim to summarize the knowledge in this field, focusing on predictive biomarkers, to recognize intrinsically resistant tumors, and therapeutic strategies, to overcome acquired resistance.

4.
Cell Death Dis ; 12(10): 951, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34654798

ABSTRACT

In colorectal cancer, mutation of KRAS (RASMUT) reduces therapeutic options, negatively affecting prognosis of the patients. In this setting, administration of CDK4/6-inhibitors, alone or in combination with other drugs, is being tested as promising therapeutic strategy. Identifying sensitive patients and overcoming intrinsic and acquired resistance to CDK4/6 inhibition represent still open challenges, to obtain better clinical responses. Here, we investigated the role of the CDK inhibitor p27kip1 in the response to the selective CDK4/6-inhibitor Palbociclib, in colorectal cancer. Our results show that p27kip1 expression inversely correlated with Palbociclib response, both in vitro and in vivo. Generating a model of Palbociclib-resistant RASMUT colorectal cancer cells, we observed an increased expression of p27kip1, cyclin D, CDK4 and CDK6, coupled with an increased association between p27kip1 and CDK4. Furthermore, Palbociclib-resistant cells showed increased Src-mediated phosphorylation of p27kip1 on tyrosine residues and low doses of Src inhibitors re-sensitized resistant cells to Palbociclib. Since p27kip1 showed variable expression in RASMUT colorectal cancer samples, our study supports the possibility that p27kip1 could serve as biomarker to stratify patients who might benefit from CDK4/6 inhibition, alone or in combination with Src inhibitors.


Subject(s)
Colorectal Neoplasms/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Mutation/genetics , Piperazines/pharmacology , Proto-Oncogene Proteins p21(ras)/genetics , Pyridines/pharmacology , Adult , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Colorectal Neoplasms/pathology , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclin-Dependent Kinase 6/metabolism , Female , Humans , Male , Mice , Middle Aged , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Young Adult , src-Family Kinases/metabolism
5.
Cancers (Basel) ; 13(12)2021 Jun 17.
Article in English | MEDLINE | ID: mdl-34204543

ABSTRACT

Alterations in components of the cell-cycle machinery are present in essentially all tumor types. In particular, molecular alterations resulting in dysregulation of the G1 to S phase transition have been observed in almost all human tumors, including ovarian cancer. These alterations have been identified as potential therapeutic targets in several cancer types, thereby stimulating the development of small molecule inhibitors of the cyclin dependent kinases. Among these, CDK4 and CDK6 inhibitors confirmed in clinical trials that CDKs might indeed represent valid therapeutic targets in, at least some, types of cancer. CDK4 and CDK6 inhibitors are now used in clinic for the treatment of patients with estrogen receptor positive metastatic breast cancer and their clinical use is being tested in many other cancer types, alone or in combination with other agents. Here, we review the role of CDK4 and CDK6 complexes in ovarian cancer and propose the possible use of their inhibitors in the treatment of ovarian cancer patients with different types and stages of disease.

6.
J Pathol ; 253(2): 234-245, 2021 02.
Article in English | MEDLINE | ID: mdl-33140857

ABSTRACT

The CDKN1B gene, encoding for the CDK inhibitor p27kip1 , is mutated in defined human cancer subtypes, including breast, prostate carcinomas and small intestine neuroendocrine tumors. Lessons learned from small intestine neuroendocrine tumors suggest that CDKN1B mutations could be subclonal, raising the question of whether a deeper sequencing approach could lead to the identification of higher numbers of patients with mutations. Here, we addressed this question and analyzed human cancer biopsies from breast (n = 396), ovarian (n = 110) and head and neck squamous carcinoma (n = 202) patients, using an ultra-deep sequencing approach. Notwithstanding this effort, the mutation rate of CDKN1B remained substantially aligned with values from the literature, showing that essentially only hormone receptor-positive breast cancer displayed CDKN1B mutations in a relevant number of cases (3%). However, the analysis of copy number variation showed that another fraction of luminal breast cancer displayed loss (8%) or gain (6%) of the CDKN1B gene, further reinforcing the idea that the function of p27kip1 is important in this type of tumor. Intriguingly, an enrichment for CDKN1B alterations was found in samples from premenopausal luminal breast cancer patients (n = 227, 4%) and in circulating cell-free DNA from metastatic luminal breast cancer patients (n = 59, 8.5%), suggesting that CDKN1B alterations could correlate with tumor aggressiveness and/or occur later during disease progression. Notably, many of the identified somatic mutations resulted in p27kip1 protein truncation, leading to loss of most of the protein or of its C-terminal domain. Using a gene-editing approach in a luminal breast cancer cell line, MCF-7, we observed that the expression of p27kip1 truncating mutants that lose the C-terminal domains failed to rescue most of the phenotypes induced by CDKN1B gene knockout, indicating that the functions retained by the C-terminal portion are critical for its role as an oncosuppressor, at least in luminal breast cancer. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Breast Neoplasms/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , DNA Copy Number Variations , Intestinal Neoplasms/genetics , Neuroendocrine Tumors/genetics , Prostatic Neoplasms/genetics , Breast Neoplasms/pathology , Cyclin-Dependent Kinase Inhibitor p27/genetics , Female , Humans , Intestinal Neoplasms/pathology , MCF-7 Cells , Male , Mutation , Neuroendocrine Tumors/pathology , Prostatic Neoplasms/pathology
7.
Oncogene ; 39(40): 6370-6386, 2020 10.
Article in English | MEDLINE | ID: mdl-32848212

ABSTRACT

For many tumor types chemotherapy still represents the therapy of choice and many standard treatments are based on the use of platinum (PT) drugs. However, de novo or acquired resistance to platinum is frequent and leads to disease progression. In Epithelial Ovarian Cancer (EOC) patients, PT-resistant recurrences are very common and improving the response to treatment still represents an unmet clinical need. To identify new modulators of PT-sensitivity, we performed a loss-of-function screening targeting 680 genes potentially involved in the response of EOC cells to platinum. We found that SGK2 (Serum-and Glucocorticoid-inducible kinase 2) plays a key role in PT-response. We show here that EOC cells relay on the induction of autophagy to escape PT-induced death and that SGK2 inhibition increases PT sensitivity inducing a block in the autophagy cascade due to the impairment of lysosomal acidification. Mechanistically we demonstrate that SGK2 controls autophagy in a kinase-dependent manner by binding and inhibiting the V-ATPase proton pump. Accordingly, SGK2 phosphorylates the subunit V1H (ATP6V1H) of V-ATPase and silencing or chemical inhibition of SGK2, affects the normal autophagic flux and sensitizes EOC cells to platinum. Hence, we identified a new pathway that links autophagy to the survival of cancer cells under platinum treatment in which the druggable kinase SGK2 plays a central role. Our data suggest that blocking autophagy via SGK2 inhibition could represent a novel therapeutic strategy to improve patients' response to platinum.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Autophagy/drug effects , Carcinoma, Ovarian Epithelial/drug therapy , Immediate-Early Proteins/metabolism , Ovarian Neoplasms/drug therapy , Protein Serine-Threonine Kinases/metabolism , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Benzoates/pharmacology , Benzoates/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Carboplatin/pharmacology , Carboplatin/therapeutic use , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/pharmacology , Cisplatin/therapeutic use , Drug Resistance, Neoplasm/drug effects , Female , Humans , Immediate-Early Proteins/antagonists & inhibitors , Immediate-Early Proteins/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Phosphorylation/drug effects , Phosphorylation/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/metabolism , Vacuolar Proton-Translocating ATPases/metabolism
8.
Oncogene ; 39(22): 4390-4403, 2020 05.
Article in English | MEDLINE | ID: mdl-32332923

ABSTRACT

In epithelial ovarian cancer (EOC), response to platinum (PT)-based chemotherapy dictates subsequent treatments and predicts patients' prognosis. Alternative splicing is often deregulated in human cancers and can be altered by chemotherapy. Whether and how changes in alternative splicing regulation could impact on the response of EOC to PT-based chemotherapy is still not clarified. We identified the splicing factor proline and glutamine rich (SFPQ) as a critical mediator of response to PT in an unbiased functional genomic screening in EOC cells and, using a large cohort of primary and recurrent EOC samples, we observed that it is frequently overexpressed in recurrent PT-treated samples and that its overexpression correlates with PT resistance. At mechanistic level, we show that, under PT treatment, SFPQ, in complex with p54nrb, binds and regulates the activity of the splicing factor SRSF2. SFPQ/p54nrb complex decreases SRSF2 binding to caspase-9 RNA, favoring the expression of its alternative spliced antiapoptotic form. As a consequence, SFPQ/p54nrb protects cells from PT-induced death, eventually contributing to chemoresistance. Overall, our work unveils a previously unreported SFPQ/p54nrb/SRSF2 pathway that in EOC cells plays a central role in regulating alternative splicing and PT-induced apoptosis and that could result in the design of new possible ways of intervention to overcome PT resistance.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Cisplatin/pharmacology , DNA-Binding Proteins/physiology , Neoplasm Proteins/physiology , Ovarian Neoplasms/drug therapy , PTB-Associated Splicing Factor/physiology , RNA-Binding Proteins/physiology , Serine-Arginine Splicing Factors/physiology , Animals , Antineoplastic Agents, Alkylating/therapeutic use , Apoptosis , Caspase 8/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Caspase Inhibitors/pharmacology , Cell Line, Tumor , Cisplatin/therapeutic use , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Drug Resistance, Neoplasm/drug effects , Female , Gene Knockdown Techniques , Humans , Mice , Ovarian Neoplasms/metabolism , RNA Splicing , RNA, Messenger/metabolism , RNA, Neoplasm/metabolism , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , Recurrence , Spliceosomes/metabolism
9.
Sci Adv ; 5(5): eaav3235, 2019 05.
Article in English | MEDLINE | ID: mdl-31086816

ABSTRACT

Resistance to platinum-based chemotherapy is a common event in patients with cancer, generally associated with tumor dissemination and metastasis. Whether platinum treatment per se activates molecular pathways linked to tumor spreading is not known. Here, we report that the ubiquitin-specific protease 1 (USP1) mediates ovarian cancer cell resistance to platinum, by regulating the stability of Snail, which, in turn, promotes tumor dissemination. At the molecular level, we observed that upon platinum treatment, USP1 is phosphorylated by ATM and ATR and binds to Snail. Then, USP1 de-ubiquitinates and stabilizes Snail expression, conferring resistance to platinum, increased stem cell-like features, and metastatic ability. Consistently, knockout or pharmacological inhibition of USP1 increased platinum sensitivity and decreased metastatic dissemination in a Snail-dependent manner. Our findings identify Snail as a USP1 target and open the way to a novel strategy to overcome platinum resistance and more successfully treat patients with ovarian cancer.


Subject(s)
Apoptosis/drug effects , Coordination Complexes/pharmacology , Platinum/chemistry , Snail Family Transcription Factors/metabolism , Ubiquitin-Specific Proteases/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line, Tumor , Coordination Complexes/therapeutic use , Drug Resistance, Neoplasm , Female , Gene Editing , Humans , Mice , Mice, Nude , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Phosphorylation , RNA Interference , RNA, Small Interfering/metabolism , Snail Family Transcription Factors/antagonists & inhibitors , Snail Family Transcription Factors/genetics , Ubiquitin-Specific Proteases/antagonists & inhibitors , Ubiquitin-Specific Proteases/genetics , Ubiquitination , Xenograft Model Antitumor Assays
10.
EMBO Mol Med ; 9(10): 1415-1433, 2017 10.
Article in English | MEDLINE | ID: mdl-28778953

ABSTRACT

Epithelial ovarian cancer (EOC) is an infrequent but highly lethal disease, almost invariably treated with platinum-based therapies. Improving the response to platinum represents a great challenge, since it could significantly impact on patient survival. Here, we report that silencing or pharmacological inhibition of CDK6 increases EOC cell sensitivity to platinum. We observed that, upon platinum treatment, CDK6 phosphorylated and stabilized the transcription factor FOXO3, eventually inducing ATR transcription. Blockage of this pathway resulted in EOC cell death, due to altered DNA damage response accompanied by increased apoptosis. These observations were recapitulated in EOC cell lines in vitro, in xenografts in vivo, and in primary tumor cells derived from platinum-treated patients. Consistently, high CDK6 and FOXO3 expression levels in primary EOC predict poor patient survival. Our data suggest that CDK6 represents an actionable target that can be exploited to improve platinum efficacy in EOC patients. As CDK4/6 inhibitors are successfully used in cancer patients, our findings can be immediately transferred to the clinic to improve the outcome of EOC patients.


Subject(s)
Cyclin-Dependent Kinase 6/metabolism , Forkhead Box Protein O3/metabolism , Neoplasms, Glandular and Epithelial/drug therapy , Ovarian Neoplasms/drug therapy , Platinum/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Carcinoma, Ovarian Epithelial , Cell Death , Cell Line, Tumor , Cyclin-Dependent Kinase 6/genetics , DNA Damage , Female , Forkhead Box Protein O3/genetics , Humans , Mice , Mice, Nude , Neoplasms, Glandular and Epithelial/enzymology , Ovarian Neoplasms/enzymology , Piperazines/pharmacology , Piperazines/therapeutic use , Platinum/therapeutic use , Primary Cell Culture , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , Survival Analysis , Xenograft Model Antitumor Assays
11.
Sci Rep ; 7(1): 7104, 2017 08 02.
Article in English | MEDLINE | ID: mdl-28769043

ABSTRACT

Standard of care for Epithelial Ovarian Cancer (EOC) patients relies on platinum-based therapy. However, acquired resistance to platinum occurs frequently and predicts poor prognosis. To understand the mechanisms underlying acquired platinum-resistance, we have generated and characterized three platinum-resistant isogenic EOC cell lines. Resistant cells showed 3-to 5- folds increase in platinum IC50. Cross-resistance to other chemotherapeutic agents commonly used in the treatment of EOC patients was variable and dependent on the cell line utilized. Gene expression profiling (GEP) of coding and non-coding RNAs failed to identify a common signature that could collectively explain the mechanism of resistance. However, we observed that all resistant cell lines displayed a decreased level of DNA platination and a faster repair of damaged DNA. Furthermore, all platinum resistant cell lines displayed a change in their morphology and a higher ability to grown on mesothelium. Overall, we have established and characterized three new models of platinum-resistant EOC cell lines that could be exploited to further dissect the molecular mechanisms underlying acquired resistance to platinum. Our work also suggests that GEP studies alone, at least when performed under basal culture condition, do not represent the optimal way to identify molecular alterations linked to DNA repair pathway defects.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/metabolism , Drug Resistance, Neoplasm/genetics , Phenotype , Platinum/pharmacology , Cell Adhesion/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Female , Gene Expression Profiling , Humans , Signal Transduction , Tumor Cells, Cultured
12.
Sci Rep ; 7(1): 595, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28377607

ABSTRACT

Genomic instability represents a typical feature of aggressive cancers. Normal cells have evolved intricate responses to preserve genomic integrity in response to stress, such as DNA damage induced by γ-irradiation. Cyclin-dependent kinases (CDKs) take crucial part to these safeguard mechanisms, but involvement of CDK-inhibitors, such as p27Kip1, is less clear. We generated immortalized fibroblasts from p27kip1 knock-out (KO) mouse embryos and re-expressed p27kip1 WT, or its mutant forms, to identify the function of different domains. We γ-irradiated fibroblasts and observed that loss of p27Kip1 was associated to accumulation of residual DNA damage, increased number of mitotic aberration and, eventually, to survival advantage. Nuclear localization and cyclin/CDK-binding of p27Kip1 were critical to mediate proper response to DNA damage. In human luminal breast cancer (LBC) p27kip1 is frequently down-modulated and CDKN1B, p27Kip1 gene, sporadically mutated. We recapitulated results obtained in mouse fibroblasts in a LBC cell line genetically manipulated to be KO for CDKN1B gene. Following γ-irradiation, we confirmed that p27kip1 expression was necessary to preserve genomic integrity and to recognize and clear-out aberrant cells. Our study provides important insights into mechanisms underlying radio-resistance and unveils the possibility for novel treatment options exploiting DNA repair defects in LBC.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/deficiency , Genomic Instability , Radiation Tolerance/genetics , Animals , Breast Neoplasms , Cell Line, Tumor , Cell Survival/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , DNA Damage/radiation effects , Female , Gene Expression , Gene Knockout Techniques , Humans , MCF-7 Cells , Mice , Micronuclei, Chromosome-Defective , Mitosis/genetics , Mitosis/radiation effects , Mutation , NIH 3T3 Cells
13.
J Mol Cell Biol ; 8(1): 17-30, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26450989

ABSTRACT

Exposure of normal and tumor-derived cells to TGFß results in different outcomes, depending on the regulation of key targets. The CDK inhibitor p27(Kip1) is one of these TGFß targets and is essential for the TGFß-induced cell cycle arrest. TGFß treatment inhibits p27(Kip1) degradation and induces its nuclear translocation, through mechanisms that are still unknown. Recent evidences suggest that SUMOylation, a post-translational modification able to modulate the stability and subcellular localization of target proteins, critically modifies members of the TGFß signaling pathway. Here, we demonstrate that p27(Kip1) is SUMOylated in response to TGFß treatment. Using different p27(Kip1) point mutants, we identified lysine 134 (K134) as the residue modified by small ubiquitin-like modifier 1 (SUMO1) in response to TGFß treatment. TGFß-induced K134 SUMOylation increased protein stability and nuclear localization of both endogenous and exogenously expressed p27(Kip1). We observed that SUMOylation regulated p27(Kip1) binding to CDK2, thereby governing its nuclear proteasomal degradation through the phosphorylation of threonine 187. Importantly, p27(Kip1) SUMOylation was necessary for proper cell cycle exit following TGFß treatment. These data indicate that SUMOylation is a novel regulatory mechanism that modulates p27(Kip1) function in response to TGFß stimulation. Given the involvement of TGFß signaling in cancer cell proliferation and invasion, our data may shed light on an important aspect of this pathway during tumor progression.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/metabolism , Sumoylation/physiology , Cell Division/genetics , Cell Division/physiology , Cell Line , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/physiology , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , HeLa Cells , Humans , Immunoblotting , Immunoprecipitation , Mass Spectrometry , Phosphorylation/genetics , Phosphorylation/physiology , Protein Processing, Post-Translational , Protein Stability , Sumoylation/genetics , Transforming Growth Factor beta
14.
EMBO Mol Med ; 5(5): 707-22, 2013 May.
Article in English | MEDLINE | ID: mdl-23610071

ABSTRACT

Stathmin is a p53-target gene, frequently overexpressed in late stages of human cancer progression. Type II High Grade Epithelial Ovarian Carcinomas (HG-EOC) represents the only clear exception to this observation. Here, we show that stathmin expression is necessary for the survival of HG-EOC cells carrying a p53 mutant (p53(MUT) ) gene. At molecular level, stathmin favours the binding and the phosphorylation of p53(MUT) by DNA-PKCS , eventually modulating p53(MUT) stability and transcriptional activity. Inhibition of stathmin or DNA-PKCS impaired p53(MUT) -dependent transcription of several M phase regulators, resulting in M phase failure and EOC cell death, both in vitro and in vivo. In primary human EOC a strong correlation exists between stathmin, DNA-PKCS , p53(MUT) overexpression and its transcriptional targets, further strengthening the relevance of the new pathway here described. Overall our data support the hypothesis that the expression of stathmin and p53 could be useful for the identification of high risk patients that will benefit from a therapy specifically acting on mitotic cancer cells.


Subject(s)
Stathmin/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Cycle Checkpoints/drug effects , Cell Survival , Female , Humans , Mice , Mutation , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Phosphorylation , Protein Binding , Protein Stability , RNA Interference , RNA, Small Interfering/metabolism , Stathmin/antagonists & inhibitors , Stathmin/genetics , Transplantation, Heterologous , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...