Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Adv Exp Med Biol ; 1227: 145-164, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32072504

RESUMEN

Notch is a ligand-receptor interaction-triggered signaling cascade highly conserved, that influences multiple lineage decisions within the hematopoietic and the immune system. It is a recognized model of intercellular communication that plays an essential role in embryonic as well as in adult immune cell development and homeostasis. Four members belong to the family of Notch receptors (Notch1-4), and each of them plays nonredundant functions at several developmental stages. Canonical and noncanonical pathways of Notch signaling are multifaceted drivers of immune cells biology. In fact, increasing evidence highlighted Notch as an important modulator of immune responses, also in cancer microenvironment. In these contexts, multiple transduction signals, including canonical and alternative NF-κB pathways, play a relevant role. In this chapter, we will first describe the critical role of Notch and NF-κB signals in lymphoid lineages developing in thymus: natural killer T cells, thymocytes, and thymic T regulatory cells. We will address also the role played by ligand expressing cells. Given the importance of Notch/NF-κB cross talk, its role in T-cell leukemia development and progression will be discussed.


Asunto(s)
Linaje de la Célula , Linfocitos/citología , Linfocitos/metabolismo , FN-kappa B/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Animales , Humanos
2.
J Oncol ; 2019: 8707053, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31379945

RESUMEN

Triple-negative breast cancer (TNBC) is a subgroup of 15%-20% of diagnosed breast cancer patients. It is generally considered to be the most difficult breast cancer subtype to deal with, due to the lack of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2), which usually direct targeted therapies. In this scenario, the current treatments of TNBC-affected patients rely on tumor excision and conventional chemotherapy. As a result, the prognosis is overall poor. Thus, the identification and characterization of targets for novel therapies are urgently required. The Notch signaling pathway has emerged to act in the pathogenesis and tumor progression of TNBCs. Firstly, Notch receptors are associated with the regulation of tumor-initiating cells (TICs) behavior, as well as with the aetiology of TNBCs. Secondly, there is a strong evidence that Notch pathway is a relevant player in mammary cancer stem cells maintenance and expansion. Finally, Notch receptors expression and activation strongly correlate with the aggressive clinicopathological and biological phenotypes of breast cancer (e.g., invasiveness and chemoresistance), which are relevant characteristics of TNBC subtype. The purpose of this up-to-date review is to provide a detailed overview of the specific role of all four Notch receptors (Notch1, Notch2, Notch3, and Notch4) in TNBCs, thus identifying the Notch signaling pathway deregulation/activation as a pathognomonic feature of this breast cancer subtype. Furthermore, this review will also discuss recent information associated with different therapeutic options related to the four Notch receptors, which may be useful to evaluate prognostic or predictive indicators as well as to develop new therapies aimed at improving the clinical outcome of TNBC patients.

3.
Oncogene ; 36(32): 4641-4652, 2017 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-28368412

RESUMEN

Aberrant Hedgehog/GLI signaling has been implicated in a diverse spectrum of human cancers, but its role in lung adenocarcinoma (LAC) is still under debate. We show that the downstream effector of the Hedgehog pathway, GLI1, is expressed in 76% of LACs, but in roughly half of these tumors, the canonical pathway activator, Smoothened, is expressed at low levels, possibly owing to epigenetic silencing. In LAC cells including the cancer stem cell compartment, we show that GLI1 is activated noncanonically by MAPK/ERK signaling. Different mechanisms can trigger the MAPK/ERK/GLI1 cascade including KRAS mutation and stimulation of NRP2 by VEGF produced by the cancer cells themselves in an autocrine loop or by stromal cells as paracrine cross talk. Suppression of GLI1, by silencing or drug-mediated, inhibits LAC cells proliferation, attenuates their stemness and increases their susceptibility to apoptosis in vitro and in vivo. These findings provide insight into the growth of LACs and point to GLI1 as a downstream effector for oncogenic pathways. Thus, strategies involving direct inhibition of GLI1 may be useful in the treatment of LACs.


Asunto(s)
Adenocarcinoma/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Neoplasias Pulmonares/metabolismo , Células Madre Neoplásicas/metabolismo , Proteína con Dedos de Zinc GLI1/metabolismo , Adenocarcinoma/patología , Animales , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Femenino , Humanos , Neoplasias Pulmonares/patología , Ratones , Ratones SCID , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Células Madre Neoplásicas/patología , Neuropilina-2/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Piridinas/farmacología , Pirimidinas/farmacología , Interferencia de ARN/fisiología , ARN Interferente Pequeño/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína con Dedos de Zinc GLI1/antagonistas & inhibidores , Proteína con Dedos de Zinc GLI1/genética
4.
Oncogene ; 36(33): 4682-4691, 2017 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-28394338

RESUMEN

High-risk and MYCN-amplified neuroblastomas are among the most aggressive pediatric tumors. Despite intense multimodality therapies, about 50% of these patients succumb to their disease, making the search for effective therapies an absolute priority. Due to the important functions of poly (ADP-ribose) polymerases, PARP inhibitors have entered the clinical settings for cancer treatment and are being exploited in a variety of preclinical studies and clinical trials. PARP inhibitors based combination schemes have also been tested in neuroblastoma preclinical models with encouraging results. However, the expression of PARP enzymes in human neuroblastoma and the biological consequences of their inhibition remained largely unexplored. Here, we show that high PARP1 and PARP2 expression is significantly associated with high-risk neuroblastoma cases and poor survival, highlighting its previously unrecognized prognostic value for human neuroblastoma. In vitro, PARP1 and 2 are abundant in MYCN amplified and MYCN-overexpressing cells. In this context, PARP inhibitors with high 'PARP trapping' potency, such as olaparib or talazoparib, yield DNA damage and cell death preceded by intense signs of replication stress. Notwithstanding the activation of a CHK1-CDC25A replication stress response, PARP-inhibited MYCN amplified and overexpressing cells fail to sustain a prolonged checkpoint and progress through mitosis in the presence of damaged DNA, eventually undergoing mitotic catastrophe. CHK1-targeted inhibition of the replication stress checkpoint exacerbated this phenotype. These data highlight a novel route for cell death induction by PARP inhibitors and support their introduction, together with CHK1 inhibitors, in therapeutic approaches for neuroblastomas with high MYC(N) activity.


Asunto(s)
Replicación del ADN/efectos de los fármacos , Mitosis/efectos de los fármacos , Proteína Proto-Oncogénica N-Myc/metabolismo , Neuroblastoma/tratamiento farmacológico , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Poli(ADP-Ribosa) Polimerasas/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Niño , Humanos , Estimación de Kaplan-Meier , Proteína Proto-Oncogénica N-Myc/genética , Poli(ADP-Ribosa) Polimerasa-1/genética , Inhibidores de Poli(ADP-Ribosa) Polimerasas/administración & dosificación , Poli(ADP-Ribosa) Polimerasas/genética
5.
Oncogene ; 35(36): 4741-51, 2016 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-26876201

RESUMEN

Deregulated Notch signaling is associated with T-cell Acute Lymphoblastic Leukemia (T-ALL) development and progression. Increasing evidence reveals that Notch pathway has an important role in the invasion ability of tumor cells, including leukemia, although the underlying molecular mechanisms remain mostly unclear. Here, we show that Notch3 is a novel target protein of the prolyl-isomerase Pin1, which is able to regulate Notch3 protein processing and to stabilize the cleaved product, leading to the increased expression of the intracellular domain (N3IC), finally enhancing Notch3-dependent invasiveness properties. We demonstrate that the combined inhibition of Notch3 and Pin1 in the Notch3-overexpressing human leukemic TALL-1 cells reduces their high invasive potential, by decreasing the expression of the matrix metalloprotease MMP9. Consistently, Pin1 depletion in a mouse model of Notch3-induced T-ALL, by reducing N3IC expression and signaling, impairs the expansion/invasiveness of CD4(+)CD8(+) DP cells in peripheral lymphoid and non-lymphoid organs. Notably, in in silico gene expression analysis of human T-ALL samples we observed a significant correlation between Pin1 and Notch3 expression levels, which may further suggest a key role of the newly identified Notch3-Pin1 axis in T-ALL aggressiveness and progression. Thus, combined suppression of Pin1 and Notch3 proteins may be exploited as an additional target therapy for T-ALL.


Asunto(s)
Progresión de la Enfermedad , Peptidilprolil Isomerasa de Interacción con NIMA/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Receptor Notch3/biosíntesis , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Leucémica de la Expresión Génica , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Invasividad Neoplásica/genética , Estadificación de Neoplasias , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Receptor Notch3/genética , Transducción de Señal/genética
6.
Cell Death Differ ; 23(2): 197-206, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26068589

RESUMEN

The MRE11/RAD50/NBS1 (MRN) complex is a major sensor of DNA double strand breaks, whose role in controlling faithful DNA replication and preventing replication stress is also emerging. Inactivation of the MRN complex invariably leads to developmental and/or degenerative neuronal defects, the pathogenesis of which still remains poorly understood. In particular, NBS1 gene mutations are associated with microcephaly and strongly impaired cerebellar development, both in humans and in the mouse model. These phenotypes strikingly overlap those induced by inactivation of MYCN, an essential promoter of the expansion of neuronal stem and progenitor cells, suggesting that MYCN and the MRN complex might be connected on a unique pathway essential for the safe expansion of neuronal cells. Here, we show that MYCN transcriptionally controls the expression of each component of the MRN complex. By genetic and pharmacological inhibition of the MRN complex in a MYCN overexpression model and in the more physiological context of the Hedgehog-dependent expansion of primary cerebellar granule progenitor cells, we also show that the MRN complex is required for MYCN-dependent proliferation. Indeed, its inhibition resulted in DNA damage, activation of a DNA damage response, and cell death in a MYCN- and replication-dependent manner. Our data indicate the MRN complex is essential to restrain MYCN-induced replication stress during neural cell proliferation and support the hypothesis that replication-born DNA damage is responsible for the neuronal defects associated with MRN dysfunctions.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proliferación Celular , Enzimas Reparadoras del ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Neuronas/fisiología , Proteínas Nucleares/metabolismo , Proteínas Nucleares/fisiología , Proteínas Oncogénicas/fisiología , Ácido Anhídrido Hidrolasas , Proteínas de Ciclo Celular/genética , Células Cultivadas , Enzimas Reparadoras del ADN/genética , Replicación del ADN , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Humanos , Proteína Homóloga de MRE11 , Proteína Proto-Oncogénica N-Myc , Proteínas Nucleares/genética , Transcripción Genética
9.
Cell Death Dis ; 5: e1472, 2014 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-25321479

RESUMEN

Numb asymmetrically segregates at mitosis to control cell fate choices during development. Numb inheritance specifies progenitor over differentiated cell fates, and, paradoxically, also promotes neuronal differentiation, thus indicating that the role of Numb may change during development. Here we report that Numb nuclear localization is restricted to early thymocyte precursors, whereas timed appearance of pre-T-cell receptor (pre-TCR) and activation of protein kinase Cθ promote phosphorylation-dependent Numb nuclear exclusion. Notably, nuclear localization of Numb in early thymocyte precursors favors p53 nuclear stabilization, whereas pre-TCR-dependent Numb nuclear exclusion promotes the p53 downmodulation essential for further differentiation. Accordingly, the persistence of Numb in the nucleus impairs the differentiation and promotes precursor cell death. This study reveals a novel regulatory mechanism for Numb function based on its nucleus-cytosol shuttling, coupling the different roles of Numb with different stages of T-cell development.


Asunto(s)
Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Células Precursoras de Linfocitos T/citología , Células Precursoras de Linfocitos T/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Transporte Activo de Núcleo Celular , Animales , Muerte Celular , Diferenciación Celular , Núcleo Celular/metabolismo , Células HEK293 , Humanos , Isoenzimas/metabolismo , Ratones , Modelos Biológicos , Fosforilación , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteína Quinasa C/metabolismo , Proteína Quinasa C-theta , Estabilidad Proteica , Proteolisis , Transducción de Señal , Fracciones Subcelulares/metabolismo
10.
Leukemia ; 28(12): 2324-35, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24727676

RESUMEN

Notch signaling deregulation is linked to the onset of several tumors including T-cell acute lymphoblastic leukemia (T-ALL). Deregulated microRNA (miRNA) expression is also associated with several cancers, including leukemias. However, the transcriptional regulators of miRNAs, as well as the relationships between Notch signaling and miRNA deregulation, are poorly understood. To identify miRNAs regulated by Notch pathway, we performed microarray-based miRNA profiling of several Notch-expressing T-ALL models. Among seven miRNAs, consistently regulated by overexpressing or silencing Notch3, we focused our attention on miR-223, whose putative promoter analysis revealed a conserved RBPjk binding site, which was nested to an NF-kB consensus. Luciferase and chromatin immunoprecipitation assays on the promoter region of miR-223 show that both Notch and NF-kB are novel coregulatory signals of miR-223 expression, being able to activate cooperatively the transcriptional activity of miR-223 promoter. Notably, the Notch-mediated activation of miR-223 represses the tumor suppressor FBXW7 in T-ALL cell lines. Moreover, we observed the inverse correlation of miR-223 and FBXW7 expression in a panel of T-ALL patient-derived xenografts. Finally, we show that miR-223 inhibition prevents T-ALL resistance to γ-secretase inhibitor (GSI) treatment, suggesting that miR-223 could be involved in GSI sensitivity and its inhibition may be exploited in target therapy protocols.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteínas F-Box/genética , Regulación Leucémica de la Expresión Génica , MicroARNs/genética , FN-kappa B/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Ubiquitina-Proteína Ligasas/genética , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Proliferación Celular , Análisis por Conglomerados , Dipéptidos/farmacología , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos/genética , Proteína 7 que Contiene Repeticiones F-Box-WD , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Silenciador del Gen , Humanos , Ratones Transgénicos , Interferencia de ARN , Transducción de Señal/efectos de los fármacos
11.
Cell Death Dis ; 5: e1100, 2014 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-24603328

RESUMEN

Childhood neuroblastic tumors are characterized by heterogeneous clinical courses, ranging from benign ganglioneuroma (GN) to highly lethal neuroblastoma (NB). Although a refined prognostic evaluation and risk stratification of each tumor patient is becoming increasingly essential to personalize treatment options, currently only few biomolecular markers (essentially MYCN amplification, chromosome 11q status and DNA ploidy) are validated for this purpose in neuroblastic tumors. Here we report that Galectin-3 (Gal-3), a ß-galactoside-binding lectin involved in multiple biological functions that has already acquired diagnostic relevance in specific clinical settings, is variably expressed in most differentiated and less aggressive neuroblastic tumors, such as GN and ganglioneuroblastoma, as well as in a subset of NB cases. Gal-3 expression is associated with the INPC histopathological categorization (P<0.001) and Shimada favorable phenotype (P=0.001), but not with other prognostically relevant features. Importantly, Gal-3 expression was associated with a better 5-year overall survival (P=0.003), and with improved cumulative survival in patient subsets at worse prognosis, such as older age at diagnosis, advanced stages or NB histopathological classification. In vitro, Gal-3 expression and nuclear accumulation accompanied retinoic acid-induced cell differentiation in NB cell lines. Forced Gal-3 overexpression increased phenotypic differentiation and substrate adherence, while inhibiting proliferation. Altogether, these findings suggest that Gal-3 is a biologically relevant player for neuroblastic tumors, whose determination by conventional immunohistochemistry might be used for outcome assessment and patient's risk stratification in the clinical setting.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Galectina 3/metabolismo , Ganglioneuroma/metabolismo , Neuroblastoma/metabolismo , Adolescente , Apoptosis , Biomarcadores de Tumor/genética , Proteínas Sanguíneas , Adhesión Celular , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Niño , Preescolar , Femenino , Galectina 3/genética , Galectinas , Ganglioneuroblastoma/metabolismo , Ganglioneuroblastoma/patología , Ganglioneuroma/genética , Ganglioneuroma/mortalidad , Ganglioneuroma/patología , Humanos , Inmunohistoquímica , Lactante , Recién Nacido , Estimación de Kaplan-Meier , Masculino , Estadificación de Neoplasias , Neuroblastoma/genética , Neuroblastoma/mortalidad , Neuroblastoma/patología , Valor Predictivo de las Pruebas , Factores de Riesgo , Factores de Tiempo , Transfección
12.
Oncogene ; 33(32): 4173-84, 2014 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-24213577

RESUMEN

The Polycomb group (PcG) proteins regulate stem cell differentiation via the repression of gene transcription, and their deregulation has been widely implicated in cancer development. The PcG protein Enhancer of Zeste Homolog 2 (EZH2) works as a catalytic subunit of the Polycomb Repressive Complex 2 (PRC2) by methylating lysine 27 on histone H3 (H3K27me3), a hallmark of PRC2-mediated gene repression. In skeletal muscle progenitors, EZH2 prevents an unscheduled differentiation by repressing muscle-specific gene expression and is downregulated during the course of differentiation. In rhabdomyosarcoma (RMS), a pediatric soft-tissue sarcoma thought to arise from myogenic precursors, EZH2 is abnormally expressed and its downregulation in vitro leads to muscle-like differentiation of RMS cells of the embryonal variant. However, the role of EZH2 in the clinically aggressive subgroup of alveolar RMS, characterized by the expression of PAX3-FOXO1 oncoprotein, remains unknown. We show here that EZH2 depletion in these cells leads to programmed cell death. Transcriptional derepression of F-box protein 32 (FBXO32) (Atrogin1/MAFbx), a gene associated with muscle homeostasis, was evidenced in PAX3-FOXO1 RMS cells silenced for EZH2. This phenomenon was associated with reduced EZH2 occupancy and H3K27me3 levels at the FBXO32 promoter. Simultaneous knockdown of FBXO32 and EZH2 in PAX3-FOXO1 RMS cells impaired the pro-apoptotic response, whereas the overexpression of FBXO32 facilitated programmed cell death in EZH2-depleted cells. Pharmacological inhibition of EZH2 by either 3-Deazaneplanocin A or a catalytic EZH2 inhibitor mirrored the phenotypic and molecular effects of EZH2 knockdown in vitro and prevented tumor growth in vivo. Collectively, these results indicate that EZH2 is a key factor in the proliferation and survival of PAX3-FOXO1 alveolar RMS cells working, at least in part, by repressing FBXO32. They also suggest that the reducing activity of EZH2 could represent a novel adjuvant strategy to eradicate high-risk PAX3-FOXO1 alveolar RMS.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Proteínas Musculares/antagonistas & inhibidores , Factores de Transcripción Paired Box/metabolismo , Complejo Represivo Polycomb 2/fisiología , Rabdomiosarcoma Alveolar/metabolismo , Proteínas Ligasas SKP Cullina F-box/antagonistas & inhibidores , Adolescente , Apoptosis , Diferenciación Celular , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proliferación Celular , Supervivencia Celular , Niño , Proteína Potenciadora del Homólogo Zeste 2 , Femenino , Proteína Forkhead Box O1 , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/metabolismo , Homeostasis , Humanos , Masculino , Proteínas Musculares/fisiología , Factor de Transcripción PAX3 , Proteínas Ligasas SKP Cullina F-box/fisiología
13.
Cell Death Differ ; 20(12): 1688-97, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24013724

RESUMEN

The Hedgehog (Hh) signaling regulates tissue development, and its aberrant activation is a leading cause of malignancies, including medulloblastoma (Mb). Hh-dependent tumorigenesis often occurs in synergy with other mechanisms, such as loss of p53, the master regulator of the DNA damage response. To date, little is known about mechanisms connecting DNA-damaging events to morphogen-dependent processes. Here, we show that genotoxic stress triggers a cascade of signals, culminating with inhibition of the activity of Gli1, the final transcriptional effector of Hh signaling. This inhibition is dependent on the p53-mediated elevation of the acetyltransferase p300/CBP-associated factor (PCAF). Notably, we identify PCAF as a novel E3 ubiquitin ligase of Gli1. Indeed PCAF, but not a mutant with a deletion of its ubiquitination domain, represses Hh signaling in response to DNA damage by promoting Gli1 ubiquitination and its proteasome-dependent degradation. Restoring Gli1 levels rescues the growth arrest and apoptosis effect triggered by genotoxic drugs. Consistently, DNA-damaging agents fail to inhibit Gli1 activity in the absence of either p53 or PCAF. Finally, Mb samples from p53-null mice display low levels of PCAF and upregulation of Gli1 in vivo, suggesting PCAF as potential therapeutic target in Hh-dependent tumors. Together, our data define a mechanism of inactivation of a morphogenic signaling in response to genotoxic stress and unveil a p53/PCAF/Gli1 circuitry centered on PCAF that limits Gli1-enhanced mitogenic and prosurvival response.


Asunto(s)
Daño del ADN , Factores de Transcripción de Tipo Kruppel/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Factores de Transcripción p300-CBP/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células HEK293 , Proteínas Hedgehog/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel/química , Ratones , Mitógenos/farmacología , Modelos Biológicos , Proteolisis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/química , Ubiquitinación/efectos de los fármacos , Proteína con Dedos de Zinc GLI1
14.
Leukemia ; 27(2): 485-8, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22846929
15.
Oncogene ; 32(41): 4892-902, 2013 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-23178494

RESUMEN

Notch signaling is frequently hyperactivated in breast cancer, but how the enhanced signaling contributes to the tumor process is less well understood. In this report, we identify the proinflammatory cytokine interleukin-6 (IL-6) as a novel Notch target in breast tumor cells. Enhanced Notch signaling upregulated IL-6 expression, leading to activation of autocrine and paracrine Janus kinase/signal transducers and activators of transcription signaling. IL-6 upregulation was mediated by non-canonical Notch signaling, as it could be effectuated by a cytoplasmically localized Notch intracellular domain and was independent of the DNA-binding protein CSL. Instead, Notch-mediated IL-6 upregulation was controlled by two proteins in the nuclear factor (NF)-κB signaling cascade, IKKα and IKKß (inhibitor of nuclear factor kappa-B kinase subunit alpha and beta, respectively), as well as by p53. Activation of IL-6 by Notch required IKKα/IKKß function, but interestingly, did not engage canonical NF-κB signaling, in contrast to IL-6 activation by inflammatory agents such as lipopolysaccharide. With regard to p53 status, IL-6 expression was upregulated by Notch when p53 was mutated or lost, and restoring wild-type p53 into p53-mutated or -deficient cells abrogated the IL-6 upregulation. Furthermore, Notch-induced transcriptomes from p53 wild-type and -mutated breast tumor cell lines differed extensively, and for a subset of genes upregulated by Notch in a p53-mutant cell line, this upregulation was reduced by wild-type p53. In conclusion, we identify IL-6 as a novel non-canonical Notch target gene, and reveal roles for p53 and IKKα/IKKß in non-canonical Notch signaling in breast cancer and in the generation of cell context-dependent diversity in the Notch signaling output.


Asunto(s)
Neoplasias de la Mama/patología , Quinasa I-kappa B/metabolismo , Interleucina-6/metabolismo , Quinasas Janus/metabolismo , Receptores Notch/metabolismo , Factores de Transcripción STAT/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Comunicación Autocrina , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Interleucina-6/genética , Macrófagos/patología , Comunicación Paracrina , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Transcriptoma , Regulación hacia Arriba
16.
Oncogene ; 31(33): 3807-17, 2012 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-22120716

RESUMEN

Post-translational modifications of Notch3 and their functional role with respect to Notch3 overexpression in T-cell leukemia are still poorly understood. We identify here a specific novel property of Notch3 that is acetylated and deacetylated at lysines 1692 and 1731 by p300 and HDAC1, respectively, a balance impaired by HDAC inhibitors (HDACi) that favor hyperacetylation. By using HDACi and a non-acetylatable Notch3 mutant carrying K/R(1692-1731) mutations in the intracellular domain, we show that Notch3 acetylation primes ubiquitination and proteasomal-mediated degradation of the protein. As a consequence, Notch3 protein expression and its transcriptional activity are decreased both in vitro and in vivo in Notch3 transgenic (tg) mice, thus impairing downstream signaling upon target genes. Consistently, Notch3-induced T-cell proliferation is inhibited by HDACi, whereas it is enhanced by the non-acetylatable Notch3-K/R(1692-1731) mutant. Finally, HDACi-induced Notch3 hyperacetylation prevents in vivo growth of T-cell leukemia/lymphoma in Notch3 tg mice. Together, our findings suggest a novel level of Notch signaling control in which Notch3 acetylation/deacetylation process represents a key regulatory switch, thus representing a suitable druggable target for Notch3-sustained T-cell acute lymphoblastic leukemia therapy.


Asunto(s)
Leucemia de Células T/etiología , Receptores Notch/fisiología , Acetilación , Animales , Células HEK293 , Inhibidores de Histona Desacetilasas/uso terapéutico , Humanos , Leucemia de Células T/tratamiento farmacológico , Activación de Linfocitos , Ratones , Complejo de la Endopetidasa Proteasomal/fisiología , Receptor Notch3 , Linfocitos T/inmunología , Ubiquitinación
17.
Oncogene ; 30(1): 65-76, 2011 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-20818436

RESUMEN

Hedgehog pathway regulates tissue patterning and cell proliferation. Gli1 transcription factor is the major effector of Hedgehog signaling and its deregulation is often associated to medulloblastoma formation. Proteolytic processes represent a critical mechanism by which this pathway is turned off. Here, we characterize the regulation of an ubiquitin-mediated mechanism of Gli1 degradation, promoted by the coordinated action of the E3 ligase Itch and the adaptor protein Numb. We show that Numb activates the catalytic activity of Itch, releasing it from an inhibitory intramolecular interaction between its homologous to E6-AP C-terminus and WW domains. The consequent activation of Itch, together with the recruitment of Gli1 through direct binding with Numb, allows Gli1 to enter into the complex, resulting in Gli1 ubiquitination and degradation. This process is mediated by a novel Itch-dependent degron, composed of a combination of two PPXYs and a phospho-serine/proline motifs, localized in Gli1 C-terminal region, indicating the role of two different WW docking sites in Gli1 ubiquitination. Remarkably, Gli1 protein mutated in these modules is no longer regulated by Itch and Numb, and determines enhanced Gli1-dependent medulloblastoma growth, migration and invasion abilities, as well as in vitro transforming activity. Our data reveal a novel mechanism of regulation of Gli1 stability and function, which influences Hedgehog/Gli1 oncogenic potential.


Asunto(s)
Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Proteínas Hedgehog/metabolismo , Humanos , Ratones , Células 3T3 NIH , Proteínas Represoras/metabolismo , Transducción de Señal , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/genética , Proteína con Dedos de Zinc GLI1
18.
Oncogene ; 29(10): 1463-74, 2010 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-19966856

RESUMEN

Notch3 and pTalpha signaling events are essential for T-cell leukemogenesis and characterize murine and human T-cell acute lymphoblastic leukemia. Genetic ablation of pTalpha expression in Notch3 transgenic mice abrogates tumor development, indicating that pTalpha signaling is crucial to the Notch3-mediated leukemogenesis. Here we report a novel direct interaction between Notch3 and pTalpha. This interaction leads to the recruitment and persistence of the E3 ligase protein c-Cbl to the lipid rafts in Notch3-IC transgenic thymocytes. Conversely, deletion of pTalpha in Notch3 transgenic mice leads to cytoplasmic retention of c-Cbl that targets Notch3 protein to the proteasomal-degradative pathway. It appears that protein kinase C theta (PKCtheta), by regulating tyrosine and serine phosphorylation of Cbl, is able to control its function. We report here that the increased Notch3-IC degradation correlates with higher levels of c-Cbl tyrosine phosphorylation in Notch3-IC/pTalpha(-/-) double-mutant thymocytes, which also display a decreased PKCtheta activity. Our data indicate that pTalpha/pre-T-cell receptor is able to regulate the different subcellular localization of c-Cbl and, by regulating PKCtheta activity, is also able to influence its ubiquitin ligase activity upon Notch3 protein.


Asunto(s)
Leucemia de Células T/metabolismo , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Animales , Western Blotting , Línea Celular , Espacio Intracelular/metabolismo , Isoenzimas/metabolismo , Leucemia de Células T/genética , Leucemia de Células T/patología , Microdominios de Membrana/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Modelos Biológicos , Fosforilación , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Proteína Quinasa C/metabolismo , Proteína Quinasa C-theta , Proteínas Proto-Oncogénicas c-cbl/genética , Interferencia de ARN , Receptor Notch3 , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Receptores Notch/genética , Timo/metabolismo , Timo/patología , Transfección , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
19.
Br J Dermatol ; 162(3): 518-26, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19903178

RESUMEN

BACKGROUND: Hailey-Hailey disease (HHD) is an autosomal dominant disorder characterized by suprabasal cutaneous cell separation (acantholysis) leading to the development of erosive and oozing skin lesions. While a strong relationship exists between mutations in the gene that encodes the Ca(2+)/Mn(2+)-adenosine triphosphatase ATP2C1 and HHD, we still have little understanding of how these mutations affect manifestations of the disease. OBJECTIVES: This study was designed to determine early signalling events that affect epithelial cell growth and differentiation during HHD development. METHODS: Expression of key regulatory signals important for maintaining skin homeostasis were evaluated by Western blot analysis and by reverse transcriptase-polymerase chain reaction in primary keratinocytes obtained from skin biopsies of patients with HHD. Reactive oxygen species accumulation in primary keratinocytes derived from lesional skin of patients with HHD was assessed by dihydrorhodamine 123 (DHR) assay. RESULTS: HHD-derived keratinocytes showed downregulation of both Notch1 and differential regulation of different p63 isoforms. Itch and p63 are co-expressed in the epidermis and in primary keratinocytes where Itch controls the p63 protein steady-state level. We found that the Itch protein was significantly decreased in HHD-derived keratinocytes whereas the expression of its target, c-Jun, remained unaffected. We also found that HHD-derived keratinocytes undergo oxidative stress, which may explain both Notch1 and Itch downregulation. CONCLUSIONS: Our attempt to explore the molecular mechanism underlying HHD indicates a complex puzzle in which multi-hit combinations of altered signal pathways may explain the wide spectrum of defects in HHD.


Asunto(s)
ATPasas Transportadoras de Calcio/genética , Estrés Oxidativo/genética , Pénfigo Familiar Benigno/genética , Calcio , ATPasas Transportadoras de Calcio/metabolismo , Análisis Mutacional de ADN , Homeostasis/genética , Humanos , Linaje , Pénfigo Familiar Benigno/metabolismo , Fenotipo , Receptores Notch/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos
20.
Oncogene ; 27(38): 5092-8, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18758477

RESUMEN

The Notch3 gene was identified, at the beginning of 90s, as the third mammalian Notch and was initially reported as being expressed in proliferating neuroepithelium. Since then, increasing evidence has demonstrated a number of structural and functional differences between Notch3 and both Notch1 and Notch2, which exhibit the highest structural similarity among the four mammalian Notch receptors. Possibly due to its more restricted tissue distribution, targeted deletion of murine Notch3 does not lead to embryonic lethality as is observed with targeted deletion of Notch1 and Notch2. However, genetic mutation, amplification and deregulated expression of Notch3 have been correlated with the disruption of cell differentiation in transgenic mice and to development of diseases in mice and humans. This review discusses the possible relationships between the structural differences and the nonredundant roles that Notch3 plays in the pathogenesis of the human disease cerebral autosomal-dominant arteriopathy with subcortical infarcts and leukoencephalopathy and in the regulation of murine T-cell differentiation and leukemogenesis.


Asunto(s)
CADASIL/genética , Transformación Celular Neoplásica/genética , Leucemia Experimental/genética , Receptores Notch/fisiología , Animales , CADASIL/fisiopatología , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Leucemia Experimental/fisiopatología , Ratones , Ratones Transgénicos , Mutación , Especificidad de Órganos , Unión Proteica , Dominios y Motivos de Interacción de Proteínas/genética , Dominios y Motivos de Interacción de Proteínas/fisiología , Receptor Notch3 , Receptores Notch/química , Receptores Notch/deficiencia , Receptores Notch/genética , Relación Estructura-Actividad , Especificidad por Sustrato , Transactivadores/metabolismo , Factores de Transcripción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA