Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
EMBO Mol Med ; 16(5): 1162-1192, 2024 May.
Article in English | MEDLINE | ID: mdl-38658801

ABSTRACT

Platinum (PT)-resistant Epithelial Ovarian Cancer (EOC) grows as a metastatic disease, disseminating in the abdomen and pelvis. Very few options are available for PT-resistant EOC patients, and little is known about how the acquisition of PT-resistance mediates the increased spreading capabilities of EOC. Here, using isogenic PT-resistant cells, genetic and pharmacological approaches, and patient-derived models, we report that Integrin α6 (ITGA6) is overexpressed by PT-resistant cells and is necessary to sustain EOC metastatic ability and adhesion-dependent PT-resistance. Using in vitro approaches, we showed that PT induces a positive loop that, by stimulating ITGA6 transcription and secretion, contributes to the formation of a pre-metastatic niche enabling EOC cells to disseminate. At molecular level, ITGA6 engagement regulates the production and availability of insulin-like growth factors (IGFs), over-stimulating the IGF1R pathway and upregulating Snail expression. In vitro data were recapitulated using in vivo models in which the targeting of ITGA6 prevents PT-resistant EOC dissemination and improves PT-activity, supporting ITGA6 as a promising druggable target for EOC patients.


Subject(s)
Drug Resistance, Neoplasm , Integrin alpha6 , Ovarian Neoplasms , Up-Regulation , Humans , Integrin alpha6/metabolism , Integrin alpha6/genetics , Female , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/drug effects , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Up-Regulation/drug effects , Animals , Cell Line, Tumor , Platinum/pharmacology , Platinum/therapeutic use , Carcinoma, Ovarian Epithelial/drug therapy , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/metabolism , Carcinoma, Ovarian Epithelial/pathology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects
2.
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.

4.
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
5.
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
6.
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
7.
Cancer Res ; 80(5): 1064-1077, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31862778

ABSTRACT

miR-223 is an anti-inflammatory miRNA that in cancer acts either as an oncosuppressor or oncopromoter, in a context-dependent manner. In breast cancer, we demonstrated that it dampens the activation of the EGF pathway. However, little is known on the role of miR-223 during breast cancer onset and progression. miR-223 expression was decreased in breast cancer of luminal and HER2 subtypes and inversely correlated with patients' prognosis. In normal luminal mammary epithelial cells, miR-223 acted cell autonomously in the control of their growth and morphology in three-dimensional context. In the MMTV-Δ16HER2 transgenic mouse model, oncogene transformation resulted in a timely abrogation of miR-223 expression, likely due to activation of E2F1, a known repressor of miR-223 transcription. Accordingly, treatment with CDK4/6 inhibitors, which eventually results in restraining E2F1 activity, restored miR-223 expression and miR-223 ablation induced luminal breast cancer resistance to CDK4/6 inhibition, both in vitro and in vivo. Notably, miR-223 expression was lost in microdissected ductal carcinoma in situ (DCIS) from patients with luminal and HER2-positive breast cancer. Altogether, these results identify downmodulation of miR-223 as an early step in luminal breast cancer onset and suggest that it could be used to identify aggressive DCIS and predict the response to targeted therapy. SIGNIFICANCE: miR-223 may represent a predictive biomarker of response to CDK4/6 inhibitors and its loss could identify DCIS lesions that are likely to progress into invasive breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/genetics , Carcinoma, Intraductal, Noninfiltrating/genetics , Cell Transformation, Neoplastic/genetics , MicroRNAs/metabolism , Adult , Aged , Aged, 80 and over , Animals , Antineoplastic Agents/therapeutic use , Breast/cytology , Breast/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Carcinoma, Intraductal, Noninfiltrating/drug therapy , Carcinoma, Intraductal, Noninfiltrating/mortality , Carcinoma, Intraductal, Noninfiltrating/pathology , Cell Culture Techniques , Cell Line, Tumor , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Disease Models, Animal , Disease Progression , Down-Regulation , Drug Resistance, Neoplasm/genetics , E2F1 Transcription Factor/metabolism , Epithelial Cells , Female , Gene Expression Regulation, Neoplastic , Humans , Mammary Glands, Animal/cytology , Mammary Glands, Animal/pathology , Mice, Knockout , MicroRNAs/genetics , Middle Aged , Neoplasm Invasiveness/genetics , Piperazines/pharmacology , Piperazines/therapeutic use , Prognosis , Pyridines/pharmacology , Pyridines/therapeutic use , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism
8.
Cells ; 9(1)2019 12 21.
Article in English | MEDLINE | ID: mdl-31877751

ABSTRACT

Platinum-based chemotherapy is the therapy of choice for epithelial ovarian cancer (EOC). Acquired resistance to platinum (PT) is a frequent event that leads to disease progression and predicts poor prognosis. To understand possible mechanisms underlying acquired PT-resistance, we have recently generated and characterized three PT-resistant isogenic EOC cell lines. Here, we more deeply characterize several PT-resistant clones derived from MDAH-2774 cells. We show that, in these cells, the increased PT resistance was accompanied by the presence of a subpopulation of multinucleated giant cells. This phenotype was likely due to an altered progression through the M phase of the cell cycle and accompanied by the deregulated expression of genes involved in M phase progression known to be target of mutant TP53. Interestingly, we found that PT-resistant MDAH cells acquired in the TP53 gene a novel secondary mutation (i.e., S185G) that accompanied the R273H typical of MDAH cells. The double p53S185G/R273H mutant increases the resistance to PT in a TP53 null EOC cellular model. Overall, we show how the selective pressure of PT is able to induce additional mutation in an already mutant TP53 gene in EOC and how this event could contribute to the acquisition of novel cellular phenotypes.


Subject(s)
Carcinoma, Ovarian Epithelial/genetics , Drug Resistance, Neoplasm/genetics , Tumor Suppressor Protein p53/genetics , Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Female , Humans , Mitosis/genetics , Mutation , Ovarian Neoplasms/genetics , Ovary/pathology , Platinum/pharmacology , Tumor Suppressor Protein p53/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.
Cancer Res ; 79(2): 397-409, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30478213

ABSTRACT

Postnatal development of the mammary gland relies on the maintenance of oriented cell division and apicobasal polarity, both of which are often deregulated in cancer. The microtubule (MT) network contributes to control these processes; however, very little is known about the impact of altered MT dynamics in the development of a complex organ and on the role played by MT-interacting proteins such as stathmin. In this study, we report that female stathmin knock-out (STM KO) mice are unable to nurse their litters due to frank impairment of mammary gland development. In mouse mammary epithelial cells, loss of stathmin compromised the trafficking of polarized proteins and the achievement of proper apicobasal polarity. In particular, prolactin receptor internalization and localization was altered in STM KO mammary epithelial cells, leading to decreased protein stability and downmodulation of the Prl/PrlR/STAT5 signaling pathway. Absence of stathmin induced alterations in mitotic spindle orientation, accumulation of mitotic defects, and apoptosis, overall contributing to tissue disorganization and further decreasing the expansion of the mammary epithelial compartment. Loss of stathmin in MMTV-Δ16HER2 transgenic mice decreased the incidence and increased the latency of these very aggressive mammary carcinomas. Collectively, these data identify the essential mammary protein stathmin as protumorigenic and suggest it may serve as a potential therapeutic target in breast cancer. SIGNIFICANCE: Stathmin expression is critical to maintain oriented cell division and apicobasal polarity in normal mammary glands and to establish a protumorigenic program that eventually sustains HER2-positive breast cancer formation in mice.


Subject(s)
Mammary Glands, Animal/growth & development , Mammary Neoplasms, Experimental/metabolism , Receptor, ErbB-2/metabolism , Stathmin/metabolism , Animals , Carcinogenesis , Female , HEK293 Cells , Humans , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Knockout , Mice, Transgenic , Prolactin/metabolism , Receptor, ErbB-2/genetics , Receptors, Prolactin/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , Stathmin/deficiency , Stathmin/genetics
11.
Mol Cancer Ther ; 18(2): 482-493, 2019 02.
Article in English | MEDLINE | ID: mdl-30425132

ABSTRACT

The CDK inhibitor, p27kip1, encoded by the Cdkn1b gene can negatively modulate cell proliferation. The control of p27 activity during the cell cycle is regulated at multiple levels, including transcription, translation, and protein stability. The last residue of p27 (threonine 198 in human, threonine 197 in mouse) is involved in the control of protein stability. We have generated a murine knock-in model (Cdkn1b T197A) in which threonine 197 is replaced by alanine, which renders p27 protein highly unstable due to a high rate of proteasomal degradation. Expectedly, Cdkn1b T197A/T197A mice present with increased body size and weight, organomegaly, and multiple organ hyperplasia, similar to what is observed in Cdkn1b KO/KO mice. We investigated the effects exerted by the restoration of normal levels of p27 protein in the tissue of Cdkn1b T197A/T197A mice. We found that proteasome inhibition with bortezomib rescues the hyperplasia induced by the lack of p27 expression in Cdkn1b T197A/T197A but not in Cdkn1b KO/KO mice. However, BAY 11-7082, a proteasome inhibitor that stabilizes IκB but not p27, fails to rescue hyperplasia in Cdkn1b T197A/T197A mice. Bortezomib increases p27 half-life and reduces the proliferation in MEFs derived from Cdkn1b T197A/T197A but not from Cdkn1b WT/WT mice, whereas BAY 11-7082 had no effect on the protein levels of p27 and on the proliferation rate of Cdkn1b T197A/T197A MEFs.The results presented here demonstrate that Cdkn1b T197A/T197A mice represent an attractive in vivo model to investigate whether the targeting of p27 degradation machinery might prove beneficial in the treatment of a variety of human proliferative disorders caused by increased turnover of p27 protein.


Subject(s)
Amino Acid Substitution , Bortezomib/pharmacology , Cyclin-Dependent Kinase Inhibitor p27/chemistry , Cyclin-Dependent Kinase Inhibitor p27/genetics , Models, Animal , Animals , Gene Knock-In Techniques , Hyperplasia , Mice , Nitriles/pharmacology , Proteasome Endopeptidase Complex/metabolism , Protein Stability , Proteolysis , Sulfones/pharmacology
12.
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
13.
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
14.
Clin Cancer Res ; 23(14): 3769-3780, 2017 Jul 15.
Article in English | MEDLINE | ID: mdl-28174235

ABSTRACT

Purpose: Head and neck squamous cell carcinomas (HNSCCs) cause more than 300,000 deaths worldwide each year. Locoregional and distant recurrences represent worse prognostic events and accepted surrogate markers of patients' overall survival. No valid biomarker and salvage therapy exist to identify and treat patients at high-risk of recurrence. We aimed to verify if selected miRNAs could be used as biomarkers of recurrence in HNSCC.Experimental Design: A NanoString array was used to identify miRNAs associated with locoregional recurrence in 44 patients with HNSCC. Bioinformatic approaches validated the signature and identified potential miRNA targets. Validation experiments were performed using an independent cohort of primary HNSCC samples and a panel of HNSCC cell lines. In vivo experiments validated the in vitro results.Results: Our data identified a four-miRNA signature that classified HNSCC patients at high- or low-risk of recurrence. These miRNAs collectively impinge on the epithelial-mesenchymal transition process. In silico and wet lab approaches showed that miR-9, expressed at high levels in recurrent HNSCC, targets SASH1 and KRT13, whereas miR-1, miR-133, and miR-150, expressed at low levels in recurrent HNSCC, collectively target SP1 and TGFß pathways. A six-gene signature comprising these targets identified patients at high risk of recurrences, as well. Combined pharmacological inhibition of SP1 and TGFß pathways induced HNSCC cell death and, when timely administered, prevented recurrence formation in a preclinical model of HNSCC recurrence.Conclusions: By integrating different experimental approaches and competences, we identified critical mediators of recurrence formation in HNSCC that may merit to be considered for future clinical development. Clin Cancer Res; 23(14); 3769-80. ©2017 AACR.


Subject(s)
Carcinoma, Squamous Cell/genetics , Head and Neck Neoplasms/genetics , MicroRNAs/genetics , Neoplasm Recurrence, Local/genetics , Adult , Aged , Animals , Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/pathology , Humans , Keratin-13/genetics , Male , Mice , Middle Aged , Neoplasm Recurrence, Local/pathology , Signal Transduction , Sp1 Transcription Factor/genetics , Squamous Cell Carcinoma of Head and Neck , Transforming Growth Factor beta/genetics , Tumor Suppressor Proteins/genetics , Xenograft Model Antitumor Assays
15.
Oncotarget ; 7(40): 64560-64574, 2016 Oct 04.
Article in English | MEDLINE | ID: mdl-27579539

ABSTRACT

The tumor suppressor protein p27Kip1 plays a pivotal role in the control of cell growth and metastasis formation.Several studies pointed to different roles for p27Kip1 in the control of Ras induced transformation, although no explanation has been provided to elucidate these differences. We recently demonstrated that p27kip1 regulates H-Ras activity via its interaction with stathmin.Here, using in vitro and in vivo models, we show that p27kip1 is an important regulator of Ras induced transformation. In H-RasV12 transformed cells, p27kip1 suppressed cell proliferation and tumor growth via two distinct mechanisms: 1) inhibition of CDK activity and 2) impairment of MT-destabilizing activity of stathmin. Conversely, in K-Ras4BV12 transformed cells, p27kip1 acted mainly in a CDK-dependent but stathmin-independent manner.Using human cancer-derived cell lines and primary breast and sarcoma samples, we confirmed in human models what we observed in mice.Overall, we highlight a pathway, conserved from mouse to human, important in the regulation of H-Ras oncogenic activity that could have therapeutic and diagnostic implication in patients that may benefit from anti-H-Ras therapies.


Subject(s)
Breast Neoplasms/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Sarcoma/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinogenesis , Cell Line, Tumor , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinases/metabolism , Female , Gene Expression Regulation, Neoplastic , Genes, ras/genetics , Humans , Mice , Mice, Nude , Phosphorylation , Sarcoma/genetics , Sarcoma/pathology , Stathmin/metabolism , Xenograft Model Antitumor Assays
16.
Proc Natl Acad Sci U S A ; 112(45): 13916-21, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26512117

ABSTRACT

The cyclin-dependent kinase (CDK) inhibitor p27(kip1) is a critical regulator of the G1/S-phase transition of the cell cycle and also regulates microtubule (MT) stability. This latter function is exerted by modulating the activity of stathmin, an MT-destabilizing protein, and by direct binding to MTs. We recently demonstrated that increased proliferation in p27(kip1)-null mice is reverted by concomitant deletion of stathmin in p27(kip1)/stathmin double-KO mice, suggesting that a CDK-independent function of p27(kip1) contributes to the control of cell proliferation. Whether the regulation of MT stability by p27(kip1) impinges on signaling pathway activation and contributes to the decision to enter the cell cycle is largely unknown. Here, we report that faster cell cycle entry of p27(kip1)-null cells was impaired by the concomitant deletion of stathmin. Using gene expression profiling coupled with bioinformatic analyses, we show that p27(kip1) and stathmin conjunctly control activation of the MAPK pathway. From a molecular point of view, we observed that p27(kip1), by controlling MT stability, impinges on H-Ras trafficking and ubiquitination levels, eventually restraining its full activation. Our study identifies a regulatory axis controlling the G1/S-phase transition, relying on the regulation of MT stability by p27(kip1) and finely controlling the spatiotemporal activation of the Ras-MAPK signaling pathway.


Subject(s)
Cell Cycle , Cyclin-Dependent Kinase Inhibitor p27/physiology , Microtubules/metabolism , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Enzyme Activation , Mice , Mice, Inbred C57BL , Protein Binding , Stathmin/metabolism
17.
Cell Cycle ; 13(19): 3100-11, 2014.
Article in English | MEDLINE | ID: mdl-25486569

ABSTRACT

The CDK inhibitor p27(kip1) is a critical regulator of cell cycle progression, but the mechanisms by which p27(kip1) controls cell proliferation in vivo are still not fully elucidated. We recently demonstrated that the microtubule destabilizing protein stathmin is a relevant p27(kip1) binding partner. To get more insights into the in vivo significance of this interaction, we generated p27(kip1) and stathmin double knock-out (DKO) mice. Interestingly, thorough characterization of DKO mice demonstrated that most of the phenotypes of p27(kip1) null mice linked to the hyper-proliferative behavior, such as the increased body and organ weight, the outgrowth of the retina basal layer and the development of pituitary adenomas, were reverted by co-ablation of stathmin. In vivo analyses showed a reduced proliferation rate in DKO compared to p27(kip1) null mice, linked, at molecular level, to decreased kinase activity of CDK4/6, rather than of CDK1 and CDK2. Gene expression profiling of mouse thymuses confirmed the phenotypes observed in vivo, showing that DKO clustered with WT more than with p27 knock-out tissue. Taken together, our results demonstrate that stathmin cooperates with p27(kip1) to control the early phase of G1 to S phase transition and that this function may be of particular relevance in the context of tumor progression.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/genetics , Stathmin/genetics , Animals , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cell Proliferation , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/genetics , Cyclin-Dependent Kinase 6/metabolism , Cyclin-Dependent Kinase Inhibitor p27/deficiency , Female , G1 Phase , Gene Expression Profiling , Gigantism/metabolism , Gigantism/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Pituitary Gland/metabolism , Pituitary Gland/pathology , S Phase , Stathmin/deficiency , Thymus Gland/metabolism , Thymus Gland/pathology
18.
Oncotarget ; 5(5): 1185-97, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24727437

ABSTRACT

MicroRNAs (miRs) are a large class of small regulatory RNAs that function as nodes of signaling networks. This implicates that miRs expression has to be finely tuned, as observed during cell cycle progression. Here, using an expression profiling approach, we provide evidence that the CDK inhibitor p27Kip1 regulates miRs expression following cell cycle exit. By using wild type and p27KO cells harvested in different phases of the cell cycle we identified several miRs regulated by p27Kip1 during the G1 to S phase transition. Among these miRs, we identified miR-223 as a miR specifically upregulated by p27Kip1 in G1 arrested cells. Our data demonstrate that p27Kip1 regulated the expression of miR-223, via two distinct mechanisms. p27Kip1 directly stabilized mature miR-223 expression, acting as a RNA binding protein and it controlled E2F1 expression that, in turn, regulated miR-223 promoter activity. The resulting elevated miR-223 levels ultimately participated to arresting cell cycle progression following contact inhibition. Importantly, this mechanism of growth control was conserved in human cells and deranged in breast cancers. Here, we identify a novel and conserved function of p27Kip1 that, by modulating miR-223 expression, contributes to proper regulation of cell cycle exit following contact inhibition. Thus we propose a new role for miR-223 in the regulation of breast cancer progression.


Subject(s)
Contact Inhibition/physiology , Cyclin-Dependent Kinase Inhibitor p27/genetics , Gene Expression Regulation , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Cell Line, Transformed , Cell Line, Tumor , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p27/metabolism , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Fibroblasts , G1 Phase Cell Cycle Checkpoints , Gene Expression Profiling , Gene Knockout Techniques , Half-Life , Humans , Mice , Promoter Regions, Genetic , Proteolysis , S Phase Cell Cycle Checkpoints , Transcription, Genetic , Up-Regulation
20.
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
...