Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
J Pathol ; 253(2): 234-245, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33140857

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Variaciones en el Número de Copia de ADN , Neoplasias Intestinales/genética , Tumores Neuroendocrinos/genética , Neoplasias de la Próstata/genética , Neoplasias de la Mama/patología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Femenino , Humanos , Neoplasias Intestinales/patología , Células MCF-7 , Masculino , Mutación , Tumores Neuroendocrinos/patología , Neoplasias de la Próstata/patología
2.
Proc Natl Acad Sci U S A ; 112(45): 13916-21, 2015 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-26512117

RESUMEN

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.


Asunto(s)
Ciclo Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/fisiología , Microtúbulos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Animales , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Activación Enzimática , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Estatmina/metabolismo
3.
NPJ Breast Cancer ; 10(1): 5, 2024 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-38184660

RESUMEN

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.
EMBO Mol Med ; 16(5): 1162-1192, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38658801

RESUMEN

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.


Asunto(s)
Resistencia a Antineoplásicos , Integrina alfa6 , Neoplasias Ováricas , Regulación hacia Arriba , Humanos , Integrina alfa6/metabolismo , Integrina alfa6/genética , Femenino , Resistencia a Antineoplásicos/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Neoplasias Ováricas/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Animales , Línea Celular Tumoral , Platino (Metal)/farmacología , Platino (Metal)/uso terapéutico , Carcinoma Epitelial de Ovario/tratamiento farmacológico , Carcinoma Epitelial de Ovario/genética , Carcinoma Epitelial de Ovario/metabolismo , Carcinoma Epitelial de Ovario/patología , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos
5.
Oncogene ; 39(22): 4390-4403, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32332923

RESUMEN

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.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Cisplatino/farmacología , Proteínas de Unión al ADN/fisiología , Proteínas de Neoplasias/fisiología , Neoplasias Ováricas/tratamiento farmacológico , Factor de Empalme Asociado a PTB/fisiología , Proteínas de Unión al ARN/fisiología , Factores de Empalme Serina-Arginina/fisiología , Animales , Antineoplásicos Alquilantes/uso terapéutico , Apoptosis , Caspasa 8/metabolismo , Caspasa 9/genética , Caspasa 9/metabolismo , Inhibidores de Caspasas/farmacología , Línea Celular Tumoral , Cisplatino/uso terapéutico , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Neoplasias Ováricas/metabolismo , Empalme del ARN , ARN Mensajero/metabolismo , ARN Neoplásico/metabolismo , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/genética , Recurrencia , Empalmosomas/metabolismo
6.
Oncogene ; 39(40): 6370-6386, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32848212

RESUMEN

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.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Autofagia/efectos de los fármacos , Carcinoma Epitelial de Ovario/tratamiento farmacológico , Proteínas Inmediatas-Precoces/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Proteínas Serina-Treonina Quinasas/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis/efectos de los fármacos , Benzoatos/farmacología , Benzoatos/uso terapéutico , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/uso terapéutico , Carboplatino/farmacología , Carboplatino/uso terapéutico , Carcinoma Epitelial de Ovario/genética , Carcinoma Epitelial de Ovario/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cisplatino/farmacología , Cisplatino/uso terapéutico , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Humanos , Proteínas Inmediatas-Precoces/antagonistas & inhibidores , Proteínas Inmediatas-Precoces/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Fosforilación/efectos de los fármacos , Fosforilación/genética , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , ARN Interferente Pequeño/metabolismo , ATPasas de Translocación de Protón Vacuolares/metabolismo
7.
Cancer Res ; 80(5): 1064-1077, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31862778

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/genética , Carcinoma Intraductal no Infiltrante/genética , Transformación Celular Neoplásica/genética , MicroARNs/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Antineoplásicos/uso terapéutico , Mama/citología , Mama/patología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Carcinoma Intraductal no Infiltrante/tratamiento farmacológico , Carcinoma Intraductal no Infiltrante/mortalidad , Carcinoma Intraductal no Infiltrante/patología , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Regulación hacia Abajo , Resistencia a Antineoplásicos/genética , Factor de Transcripción E2F1/metabolismo , Células Epiteliales , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/patología , Ratones Noqueados , MicroARNs/genética , Persona de Mediana Edad , Invasividad Neoplásica/genética , Piperazinas/farmacología , Piperazinas/uso terapéutico , Pronóstico , Piridinas/farmacología , Piridinas/uso terapéutico , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo
8.
Clin Cancer Res ; 14(5): 1325-32, 2008 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-18316551

RESUMEN

PURPOSE: After apparently successful excision of breast cancer, risk of local recurrence remains high mainly in the area surrounding the original tumor, indicating that wound healing processes may be implicated. The proportional reduction of this risk by radiotherapy does not depend on the extent of surgery, suggesting that radiotherapy, in addition to killing tumor cells, may influence the tumor microenvironment. EXPERIMENTAL DESIGN: We studied how normal and mammary carcinoma cell growth and motility are affected by surgical wound fluids (WF), collected over 24 h following breast-conserving surgery in 45 patients, 20 of whom had received additional TARGeted Intraoperative radioTherapy (TARGIT), immediately after the surgical excision. The proteomic profile of the WF and their effects on the activation of intracellular signal transduction pathways of breast cancer cells were also analyzed. RESULTS: WF stimulated proliferation, migration, and invasion of breast cancer cell lines. The stimulatory effect was almost completely abrogated when fluids from TARGIT-treated patients were used. These fluids displayed altered expression of several cytokines and failed to properly stimulate the activation of some intracellular signal transduction pathways, when compared with fluids harvested from untreated patients. CONCLUSIONS: Delivery of TARGIT to the tumor bed alters the molecular composition and biological activity of surgical WF. This novel antitumoral effect could, at least partially, explain the very low recurrence rates found in a large pilot study using TARGIT. It also opens a novel avenue for identifying new molecular targets and testing novel therapeutic agents.


Asunto(s)
Neoplasias de la Mama/patología , Neoplasias de la Mama/radioterapia , Proliferación Celular/efectos de la radiación , Animales , Mama/citología , Mama/efectos de la radiación , Neoplasias de la Mama/cirugía , Movimiento Celular , Células Cultivadas , Progresión de la Enfermedad , Endotelio Vascular/citología , Endotelio Vascular/efectos de la radiación , Femenino , Estudios de Seguimiento , Humanos , Cuidados Intraoperatorios , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/efectos de la radiación , Mastectomía Segmentaria , Ratones , Persona de Mediana Edad , Células 3T3 NIH , Invasividad Neoplásica , Recurrencia Local de Neoplasia/prevención & control , Proyectos Piloto , Proteómica , Dosificación Radioterapéutica , Venas Umbilicales/citología , Venas Umbilicales/efectos de la radiación
9.
Cells ; 9(1)2019 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-31877751

RESUMEN

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.


Asunto(s)
Carcinoma Epitelial de Ovario/genética , Resistencia a Antineoplásicos/genética , Proteína p53 Supresora de Tumor/genética , Antineoplásicos/farmacología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Humanos , Mitosis/genética , Mutación , Neoplasias Ováricas/genética , Ovario/patología , Platino (Metal)/farmacología , Proteína p53 Supresora de Tumor/metabolismo
10.
Mol Cancer Ther ; 18(2): 482-493, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30425132

RESUMEN

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.


Asunto(s)
Sustitución de Aminoácidos , Bortezomib/farmacología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/química , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Modelos Animales , Animales , Técnicas de Sustitución del Gen , Hiperplasia , Ratones , Nitrilos/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Estabilidad Proteica , Proteolisis , Sulfonas/farmacología
11.
Sci Adv ; 5(5): eaav3235, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31086816

RESUMEN

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.


Asunto(s)
Apoptosis/efectos de los fármacos , Complejos de Coordinación/farmacología , Platino (Metal)/química , Factores de Transcripción de la Familia Snail/metabolismo , Proteasas Ubiquitina-Específicas/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Línea Celular Tumoral , Complejos de Coordinación/uso terapéutico , Resistencia a Antineoplásicos , Femenino , Edición Génica , Humanos , Ratones , Ratones Desnudos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Fosforilación , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Factores de Transcripción de la Familia Snail/antagonistas & inhibidores , Factores de Transcripción de la Familia Snail/genética , Proteasas Ubiquitina-Específicas/antagonistas & inhibidores , Proteasas Ubiquitina-Específicas/genética , Ubiquitinación , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Cancer Res ; 79(2): 397-409, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30478213

RESUMEN

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.


Asunto(s)
Glándulas Mamarias Animales/crecimiento & desarrollo , Neoplasias Mamarias Experimentales/metabolismo , Receptor ErbB-2/metabolismo , Estatmina/metabolismo , Animales , Carcinogénesis , Femenino , Células HEK293 , Humanos , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Noqueados , Ratones Transgénicos , Prolactina/metabolismo , Receptor ErbB-2/genética , Receptores de Prolactina/metabolismo , Factor de Transcripción STAT5/metabolismo , Transducción de Señal , Estatmina/deficiencia , Estatmina/genética
14.
Sci Rep ; 7(1): 595, 2017 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-28377607

RESUMEN

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.


Asunto(s)
Inhibidor p27 de las Quinasas Dependientes de la Ciclina/deficiencia , Inestabilidad Genómica , Tolerancia a Radiación/genética , Animales , Neoplasias de la Mama , Línea Celular Tumoral , Supervivencia Celular/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Daño del ADN/efectos de la radiación , Femenino , Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Células MCF-7 , Ratones , Micronúcleos con Defecto Cromosómico , Mitosis/genética , Mitosis/efectos de la radiación , Mutación , Células 3T3 NIH
15.
EMBO Mol Med ; 9(10): 1415-1433, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28778953

RESUMEN

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.


Asunto(s)
Quinasa 6 Dependiente de la Ciclina/metabolismo , Proteína Forkhead Box O3/metabolismo , Neoplasias Glandulares y Epiteliales/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , Platino (Metal)/farmacología , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Carcinoma Epitelial de Ovario , Muerte Celular , Línea Celular Tumoral , Quinasa 6 Dependiente de la Ciclina/genética , Daño del ADN , Femenino , Proteína Forkhead Box O3/genética , Humanos , Ratones , Ratones Desnudos , Neoplasias Glandulares y Epiteliales/enzimología , Neoplasias Ováricas/enzimología , Piperazinas/farmacología , Piperazinas/uso terapéutico , Platino (Metal)/uso terapéutico , Cultivo Primario de Células , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Piridinas/farmacología , Piridinas/uso terapéutico , Análisis de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Clin Cancer Res ; 23(14): 3769-3780, 2017 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-28174235

RESUMEN

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.


Asunto(s)
Carcinoma de Células Escamosas/genética , Neoplasias de Cabeza y Cuello/genética , MicroARNs/genética , Recurrencia Local de Neoplasia/genética , Adulto , Anciano , Animales , Biomarcadores de Tumor/genética , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/patología , Humanos , Queratina-13/genética , Masculino , Ratones , Persona de Mediana Edad , Recurrencia Local de Neoplasia/patología , Transducción de Señal , Factor de Transcripción Sp1/genética , Carcinoma de Células Escamosas de Cabeza y Cuello , Factor de Crecimiento Transformador beta/genética , Proteínas Supresoras de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Oncotarget ; 7(40): 64560-64574, 2016 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-27579539

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Sarcoma/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Carcinogénesis , Línea Celular Tumoral , Proliferación Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Quinasas Ciclina-Dependientes/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Genes ras/genética , Humanos , Ratones , Ratones Desnudos , Fosforilación , Sarcoma/genética , Sarcoma/patología , Estatmina/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Cell Cycle ; 13(19): 3100-11, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25486569

RESUMEN

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.


Asunto(s)
Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Estatmina/genética , Animales , Proteína Quinasa CDC2/genética , Proteína Quinasa CDC2/metabolismo , Proliferación Celular , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/genética , Quinasa 6 Dependiente de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/deficiencia , Femenino , Fase G1 , Perfilación de la Expresión Génica , Gigantismo/metabolismo , Gigantismo/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Hipófisis/metabolismo , Hipófisis/patología , Fase S , Estatmina/deficiencia , Timo/metabolismo , Timo/patología
19.
Oncotarget ; 5(5): 1185-97, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24727437

RESUMEN

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.


Asunto(s)
Inhibición de Contacto/fisiología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Regulación de la Expresión Génica , MicroARNs/genética , MicroARNs/metabolismo , Animales , Línea Celular Transformada , Línea Celular Tumoral , Proliferación Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Factor de Transcripción E2F1/genética , Factor de Transcripción E2F1/metabolismo , Fibroblastos , Puntos de Control de la Fase G1 del Ciclo Celular , Perfilación de la Expresión Génica , Técnicas de Inactivación de Genes , Semivida , Humanos , Ratones , Regiones Promotoras Genéticas , Proteolisis , Puntos de Control de la Fase S del Ciclo Celular , Transcripción Genética , Regulación hacia Arriba
20.
EMBO Mol Med ; 5(5): 707-22, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23610071

RESUMEN

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.


Asunto(s)
Estatmina/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Supervivencia Celular , Femenino , Humanos , Ratones , Mutación , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Fosforilación , Unión Proteica , Estabilidad Proteica , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Estatmina/antagonistas & inhibidores , Estatmina/genética , Trasplante Heterólogo , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA