Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(6)2023 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-36982482

RESUMEN

Neuroblastoma (NB) is one of the primary causes of death for pediatric malignancies. Given the high heterogeneity in NB's mutation landscape, optimizing individualized therapies is still challenging. In the context of genomic alterations, MYCN amplification is the most correlated event with poor outcomes. MYCN is involved in the regulation of several cellular mechanisms, including cell cycle. Thus, studying the influence of MYCN overexpression in the G1/S transition checkpoint of the cell cycle may unveil novel druggable targets for the development of personalized therapeutical approaches. Here, we show that high expression of E2F3 and MYCN correlate with poor prognosis in NB despite the RB1 mRNA levels. Moreover, we demonstrate through luciferase reporter assays that MYCN bypasses RB function by incrementing E2F3-responsive promoter activity. We showed that MYCN overexpression leads to RB inactivation by inducing RB hyperphosphorylation during the G1 phase through cell cycle synchronization experiments. Moreover, we generated two MYCN-amplified NB cell lines conditionally knockdown (cKD) for the RB1 gene through a CRISPRi approach. Indeed, RB KD did not affect cell proliferation, whereas cell proliferation was strongly influenced when a non-phosphorylatable RB mutant was expressed. This finding revealed the dispensable role of RB in regulating MYCN-amplified NB's cell cycle. The described genetic interaction between MYCN and RB1 provides the rationale for using cyclin/CDK complexes inhibitors in NBs carrying MYCN amplification and relatively high levels of RB1 expression.


Asunto(s)
Neuroblastoma , Niño , Humanos , Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 4 Dependiente de la Ciclina/metabolismo , Amplificación de Genes , Regulación Neoplásica de la Expresión Génica , Proteína Proto-Oncogénica N-Myc/genética , Proteína Proto-Oncogénica N-Myc/metabolismo , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/genética , Neuroblastoma/metabolismo , Proteínas de Unión a Retinoblastoma/genética , Proteínas de Unión a Retinoblastoma/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
2.
Int J Mol Sci ; 22(21)2021 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-34769459

RESUMEN

PRDM12 is a member of the PRDI-BF1 (positive regulatory domain I-binding factor 1) homologous domain (PRDM)-containing protein family, a subfamily of Kruppel-like zinc finger proteins, controlling key processes in the development of cancer. PRDM12 is expressed in a spatio-temporal manner in neuronal systems where it exerts multiple functions. PRDM12 is essential for the neurogenesis initiation and activation of a cascade of downstream pro-neuronal transcription factors in the nociceptive lineage. PRDM12 inactivation, indeed, results in a complete absence of the nociceptive lineage, which is essential for pain perception. Additionally, PRDM12 contributes to the early establishment of anorexigenic neuron identity and the maintenance of high expression levels of pro-opiomelanocortin, which impacts on the program bodyweight homeostasis. PRDMs are commonly involved in cancer, where they act as oncogenes/tumor suppressors in a "Yin and Yang" manner. PRDM12 is not usually expressed in adult normal tissues but its expression is re-activated in several cancer types. However, little information is currently available on PRDM12 expression in cancers and its mechanism of action has not been thoroughly described. In this review, we summarize the recent findings regarding PRDM12 by focusing on four main biological processes: neurogenesis, pain perception, oncogenesis and cell metabolism. Moreover, we wish to highlight the importance of future studies focusing on the PRDM12 signaling pathway(s) and its role in cancer onset and progression.


Asunto(s)
Proteínas Portadoras/metabolismo , Neoplasias/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Dolor/metabolismo , Animales , Humanos , Neoplasias/patología , Neurogénesis/fisiología , Dolor/patología
3.
Int J Mol Sci ; 22(23)2021 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-34884690

RESUMEN

Neuroblastoma (NB) is one of the most frequently occurring neurogenic extracranial solid cancers in childhood and infancy. Over the years, many pieces of evidence suggested that NB development is controlled by gene expression dysregulation. These unleashed programs that outline NB cancer cells make them highly dependent on specific tuning of gene expression, which can act co-operatively to define the differentiation state, cell identity, and specialized functions. The peculiar regulation is mainly caused by genetic and epigenetic alterations, resulting in the dependency on a small set of key master transcriptional regulators as the convergence point of multiple signalling pathways. In this review, we provide a comprehensive blueprint of transcriptional regulation bearing NB initiation and progression, unveiling the complexity of novel oncogenic and tumour suppressive regulatory networks of this pathology. Furthermore, we underline the significance of multi-target therapies against these hallmarks, showing how novel approaches, together with chemotherapy, surgery, or radiotherapy, can have substantial antineoplastic effects, disrupting a wide variety of tumorigenic pathways through combinations of different treatments.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Genes myc , Terapia Molecular Dirigida , Neuroblastoma/genética , Animales , Inestabilidad Cromosómica , Epigénesis Genética , Redes Reguladoras de Genes , Predisposición Genética a la Enfermedad , Humanos , Neuroblastoma/tratamiento farmacológico
5.
Haematologica ; 104(4): 717-728, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30409797

RESUMEN

The oncoprotein BCR-ABL1 triggers chronic myeloid leukemia. It is clear that the disease relies on constitutive BCR-ABL1 kinase activity, but not all the interactors and regulators of the oncoprotein are known. We describe and validate a Drosophila leukemia model based on inducible human BCR-ABL1 expression controlled by tissue-specific promoters. The model was conceived to be a versatile tool for performing genetic screens. BCR-ABL1 expression in the developing eye interferes with ommatidia differentiation and expression in the hematopoietic precursors increases the number of circulating blood cells. We show that BCR-ABL1 interferes with the pathway of endogenous dAbl with which it shares the target protein Ena. Loss of function of ena or Dab, an upstream regulator of dAbl, respectively suppresses or enhances both the BCR-ABL1-dependent phenotypes. Importantly, in patients with leukemia decreased human Dab1 and Dab2 expression correlates with more severe disease and Dab1 expression reduces the proliferation of leukemia cells. Globally, these observations validate our Drosophila model, which promises to be an excellent system for performing unbiased genetic screens aimed at identifying new BCR-ABL1 interactors and regulators in order to better elucidate the mechanism of leukemia onset and progression.


Asunto(s)
Animales Modificados Genéticamente , Proteínas de Fusión bcr-abl , Regulación Leucémica de la Expresión Génica , Leucemia Mielógena Crónica BCR-ABL Positiva , Animales , Animales Modificados Genéticamente/genética , Animales Modificados Genéticamente/metabolismo , Modelos Animales de Enfermedad , Drosophila melanogaster , Proteínas de Fusión bcr-abl/biosíntesis , Proteínas de Fusión bcr-abl/genética , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología
6.
Hum Mol Genet ; 25(18): 3887-3907, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27466189

RESUMEN

Cyclin-dependent kinase-like 5 (CDKL5) is a Ser/Thr protein kinase predominantly expressed in the brain. Mutations of the CDKL5 gene lead to CDKL5 disorder, a neurodevelopmental pathology that shares several features with Rett Syndrome and is characterized by severe intellectual disability. The phosphorylation targets of CDKL5 are largely unknown, which hampers the discovery of therapeutic strategies for improving the neurological phenotype due to CDKL5 mutations. Here, we show that the histone deacetylase 4 (HDAC4) is a direct phosphorylation target of CDKL5 and that CDKL5-dependent phosphorylation promotes HDAC4 cytoplasmic retention. Nuclear HDAC4 binds to chromatin as well as to MEF2A transcription factor, leading to histone deacetylation and altered neuronal gene expression. By using a Cdkl5 knockout (Cdkl5 -/Y) mouse model, we found that hypophosphorylated HDAC4 translocates to the nucleus of neural precursor cells, thereby reducing histone 3 acetylation. This effect was reverted by re-expression of CDKL5 or by inhibition of HDAC4 activity through the HDAC4 inhibitor LMK235. In Cdkl5 -/Y mice treated with LMK235, defective survival and maturation of neuronal precursor cells and hippocampus-dependent memory were fully normalized. These results demonstrate a critical role of HDAC4 in the neurodevelopmental alterations due to CDKL5 mutations and suggest the possibility of HDAC4-targeted pharmacological interventions.


Asunto(s)
Histona Desacetilasas/biosíntesis , Discapacidad Intelectual/genética , Proteínas Serina-Treonina Quinasas/genética , Síndrome de Rett/genética , Espasmos Infantiles/genética , Animales , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/administración & dosificación , Síndromes Epilépticos , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/crecimiento & desarrollo , Hipocampo/patología , Histona Desacetilasas/efectos de los fármacos , Histona Desacetilasas/genética , Humanos , Discapacidad Intelectual/tratamiento farmacológico , Discapacidad Intelectual/fisiopatología , Factores de Transcripción MEF2/genética , Ratones , Ratones Noqueados , Mutación , Células-Madre Neurales/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/patología , Fosforilación , Síndrome de Rett/tratamiento farmacológico , Síndrome de Rett/patología , Espasmos Infantiles/tratamiento farmacológico , Espasmos Infantiles/patología
9.
Chromosoma ; 124(2): 277-87, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25413176

RESUMEN

The centromere directs the segregation of chromosomes during mitosis and meiosis. It is a distinct genetic locus whose identity is established through epigenetic mechanisms that depend on the deposition of centromere-specific centromere protein A (CENP-A) nucleosomes. This important chromatin domain has so far escaped comprehensive molecular analysis due to its typical association with highly repetitive satellite DNA. In previous work, we discovered that the centromere of horse chromosome 11 is completely devoid of satellite DNA; this peculiar feature makes it a unique model to dissect the molecular architecture of mammalian centromeres. Here, we exploited this native satellite-free centromere to determine the precise localization of its functional domains in five individuals: We hybridized DNA purified from chromatin immunoprecipitated with an anti CENP-A antibody to a high resolution array (ChIP-on-chip) of the region containing the primary constriction of horse chromosome 11. Strikingly, each individual exhibited a different arrangement of CENP-A binding domains. We then analysed the organization of each domain using a single nucleotide polymorphism (SNP)-based approach and single molecule analysis on chromatin fibres. Examination of the ten instances of chromosome 11 in the five individuals revealed seven distinct 'positional alleles', each one extending for about 80-160 kb, were found across a region of about 500 kb. Our results demonstrate that CENP-A binding domains are autonomous relative to the underlying DNA sequence and are characterized by positional instability causing the sliding of centromere position. We propose that this dynamic behaviour may be common in mammalian centromeres and may determine the establishment of epigenetic alleles.


Asunto(s)
Centrómero/genética , Cromosomas de los Mamíferos/genética , Caballos/genética , Alelos , Animales , Autoantígenos/genética , Línea Celular , Proteína A Centromérica , Cromatina/genética , Proteínas Cromosómicas no Histona/genética , Clonación Molecular , ADN Satélite , Epigénesis Genética , Femenino , Masculino , Meiosis , Procedimientos Analíticos en Microchip , Mitosis , Nucleosomas/genética , Polimorfismo de Nucleótido Simple
10.
Genes Chromosomes Cancer ; 54(3): 156-67, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25421174

RESUMEN

Gene amplification is relatively common in tumors. In certain subtypes of sarcoma, it often occurs in the form of ring and/or giant rod-shaped marker (RGM) chromosomes whose mitotic stability is frequently rescued by ectopic novel centromeres (neocentromeres). Little is known about the origin and structure of these RGM chromosomes, including how they arise, their internal organization, and which sequences underlie the neocentromeres. To address these questions, 42 sarcomas with RGM chromosomes were investigated to detect regions prone to double strand breaks and possible functional or structural constraints driving the amplification process. We found nine breakpoint cluster regions potentially involved in the genesis of RGM chromosomes, which turned out to be significantly enriched in poly-pyrimidine traits. Some of the clusters were located close to genes already known to be relevant for sarcomas, thus indicating a potential functional constraint, while others mapped to transcriptionally inactive chromatin domains enriched in heterochromatic sites. Of note, five neocentromeres were identified after analyzing 13 of the cases by fluorescent in situ hybridization. ChIP-on-chip analysis with antibodies against the centromeric protein CENP-A showed that they were a patchwork of small genomic segments derived from different chromosomes, likely joint to form a contiguous sequence during the amplification process.


Asunto(s)
Puntos de Rotura del Cromosoma , Cromosomas en Anillo , Sarcoma/genética , Centrómero/genética , Epigénesis Genética , Amplificación de Genes/genética , Humanos , Hibridación Fluorescente in Situ , Sarcoma/ultraestructura
11.
J Biol Chem ; 288(29): 20817-20829, 2013 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-23740250

RESUMEN

Intellectual disability in Down syndrome (DS) appears to be related to severe proliferation impairment during brain development. Recent evidence shows that it is not only cellular proliferation that is heavily compromised in DS, but also cell fate specification and dendritic maturation. The amyloid precursor protein (APP), a gene that is triplicated in DS, plays a key role in normal brain development by influencing neural precursor cell proliferation, cell fate specification, and neuronal maturation. APP influences these processes via two separate domains, the APP intracellular domain (AICD) and the soluble secreted APP. We recently found that the proliferation impairment of neuronal precursors (NPCs) from the Ts65Dn mouse model for DS was caused by derangement of the Shh pathway due to overexpression of patched1(Ptch1), its inhibitory regulator. Ptch1 overexpression was related to increased levels within the APP/AICD system. The overall goal of this study was to determine whether APP contributes to neurogenesis impairment in DS by influencing in addition to proliferation, cell fate specification, and neurite development. We found that normalization of APP expression restored the reduced neuronogenesis, the increased astrogliogenesis, and the reduced neurite length of trisomic NPCs, indicating that APP overexpression underpins all aspects of neurogenesis impairment. Moreover, we found that two different domains of APP impair neuronal differentiation and maturation in trisomic NPCs. The APP/AICD system regulates neuronogenesis and neurite length through the Shh pathway, whereas the APP/secreted AP system promotes astrogliogenesis through an IL-6-associated signaling cascade. These results provide novel insight into the mechanisms underlying brain development alterations in DS.


Asunto(s)
Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/metabolismo , Diferenciación Celular , Síndrome de Down/genética , Células-Madre Neurales/patología , Neuritas/metabolismo , Trisomía/genética , Animales , Astrocitos/metabolismo , Linaje de la Célula , Forma de la Célula , Modelos Animales de Enfermedad , Femenino , Silenciador del Gen , Proteínas Hedgehog/metabolismo , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Interleucina-6/metabolismo , Masculino , Ratones , Ratones Transgénicos , Modelos Biológicos , Células-Madre Neurales/metabolismo , Neurogénesis , Receptores Patched , Receptor Patched-1 , Estructura Terciaria de Proteína , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Relación Estructura-Actividad
12.
J Biol Chem ; 288(12): 8332-8341, 2013 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-23362253

RESUMEN

CLU (clusterin) is a tumor suppressor gene that we have previously shown to be negatively modulated by the MYCN proto-oncogene, but the mechanism of repression was unclear. Here, we show that MYCN inhibits the expression of CLU by direct interaction with the non-canonical E box sequence CACGCG in the 5'-flanking region. Binding of MYCN to the CLU gene induces bivalent epigenetic marks and recruitment of repressive proteins such as histone deacetylases and Polycomb members. MYCN physically binds in vitro and in vivo to EZH2, a component of the Polycomb repressive complex 2, required to repress CLU. Notably, EZH2 interacts with the Myc box domain 3, a segment of MYC known to be essential for its transforming effects. The expression of CLU can be restored in MYCN-amplified cells by epigenetic drugs with therapeutic results. Importantly, the anticancer effects of the drugs are ablated if CLU expression is blunted by RNA interference. Our study implies that MYC tumorigenesis can be effectively antagonized by epigenetic drugs that interfere with the recruitment of chromatin modifiers at repressive E boxes of tumor suppressor genes such as CLU.


Asunto(s)
Neuroblastoma/tratamiento farmacológico , Proteínas Nucleares/metabolismo , Proteínas Oncogénicas/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Región de Flanqueo 5' , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Secuencia de Bases , Línea Celular Tumoral/efectos de los fármacos , Movimiento Celular , Proliferación Celular/efectos de los fármacos , Cromatina/metabolismo , Clusterina/genética , Clusterina/metabolismo , Elementos E-Box , Proteína Potenciadora del Homólogo Zeste 2 , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ácidos Hidroxámicos/farmacología , Datos de Secuencia Molecular , Proteína Proto-Oncogénica N-Myc , Proteínas Nucleares/fisiología , Proteínas Oncogénicas/fisiología , Regiones Promotoras Genéticas , Unión Proteica , Proto-Oncogenes Mas
13.
Am J Hum Genet ; 88(1): 115-20, 2011 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-21211618

RESUMEN

THC2, an autosomal-dominant thrombocytopenia described so far in only two families, has been ascribed to mutations in MASTL or ACBD5. Here, we show that ANKRD26, another gene within the THC2 locus, and neither MASTL nor ACBD5, is mutated in eight unrelated families. ANKRD26 was also found to be mutated in the family previously reported to have an ACBD5 mutation. We identified six different ANKRD26 mutations, which were clustered in a highly conserved 19 bp sequence located in the 5' untranslated region. Mutations were not detected in 500 controls and are absent from the 1000 Genomes database. Available data from an animal model and Dr. Watson's genome give evidence against haploinsufficiency as the pathogenetic mechanism for ANKRD26-mediated thrombocytopenia. The luciferase reporter assay suggests that these 5' UTR mutations might enhance ANKRD26 expression. ANKRD26 is the ancestor of a family of primate-specific genes termed POTE, which have been recently identified as a family of proapoptotic proteins. Dysregulation of apoptosis might therefore be the pathogenetic mechanism, as demonstrated for another thrombocytopenia, THC4. Further investigation is needed to provide evidence supporting this hypothesis.


Asunto(s)
Repetición de Anquirina/genética , Genes Dominantes , Mutación , Secuencia de Bases , Rotura Cromosómica , Trastornos de los Cromosomas/genética , Secuencia Conservada/genética , Femenino , Sitios Genéticos , Haploinsuficiencia , Humanos , Masculino , Datos de Secuencia Molecular , Linaje , Trombocitopenia/congénito , Trombocitopenia/genética
14.
Soc Psychiatry Psychiatr Epidemiol ; 49(5): 693-701, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24092521

RESUMEN

PURPOSE: Cancer mortality data allow assessing, at the same time, the risk of developing the disease and the quality of care provided to patients after the oncologic diagnosis. This study explores the risk of death caused by a single tumor site in a psychiatric population treated in a community-based psychiatric service. METHODS: All patients with an ICD-10 psychiatric diagnosis, seeking care in 1982-2006 (25 years), were included. Data were drawn from the South Verona Psychiatric Case Register (PCR). Mortality and cause of death were ascertained using different procedures and sources. Standardized mortality ratios (SMRs) were used to compare the observed number of deaths with the expected number using as reference a population in the Veneto region. RESULTS: Having been admitted to the hospital (SMR = 1.32), having a short interval from registration (1.52), having a diagnosis of alcoholism (2.03), and being a middle-aged male (1.83) were factors showing an increased risk of death from cancer. Increased SMRs were found for cancer of the oral cavity (22.93), lymphoma, leukemias, Hodgkin's lymphoma (8.01), and central nervous system (CNS) and cranial nerve tumors (4.75). The SMR decreased for stomach tumors (0.49). Patients with alcoholism (5.90 for larynx), affective disorders (20.00 for lymphomas), and personality disorders (28.00 for SNC) were found to be exposed to a high risk of cancer death in specific sites. CONCLUSIONS: Psychiatric patients showed different patterns of site-specific cancer mortality when compared with the general population. The 20-fold higher risk of dying from hematological neoplasms needs further investigation. Chronic use of phenothiazines could be involved in the relative protection from stomach and prostate cancer found in psychiatric patients.


Asunto(s)
Causas de Muerte , Servicios Comunitarios de Salud Mental/estadística & datos numéricos , Trastornos Mentales/mortalidad , Neoplasias/mortalidad , Neoplasias/psicología , Adulto , Distribución por Edad , Anciano , Alcoholismo/mortalidad , Comorbilidad , Femenino , Hospitalización , Humanos , Clasificación Internacional de Enfermedades , Italia/epidemiología , Masculino , Trastornos Mentales/diagnóstico , Trastornos Mentales/psicología , Persona de Mediana Edad , Trastornos de la Personalidad , Sistema de Registros , Distribución por Sexo , Factores Socioeconómicos , Factores de Tiempo
15.
PLoS Genet ; 7(6): e1002135, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21698133

RESUMEN

The N-Myc oncoprotein is a critical factor in neuroblastoma tumorigenesis which requires additional mechanisms converting a low-level to a high-level N-Myc expression. N-Myc protein is stabilized when phosphorylated at Serine 62 by phosphorylated ERK protein. Here we describe a novel positive feedback loop whereby N-Myc directly induced the transcription of the class III histone deacetylase SIRT1, which in turn increased N-Myc protein stability. SIRT1 binds to Myc Box I domain of N-Myc protein to form a novel transcriptional repressor complex at gene promoter of mitogen-activated protein kinase phosphatase 3 (MKP3), leading to transcriptional repression of MKP3, ERK protein phosphorylation, N-Myc protein phosphorylation at Serine 62, and N-Myc protein stabilization. Importantly, SIRT1 was up-regulated, MKP3 down-regulated, in pre-cancerous cells, and preventative treatment with the SIRT1 inhibitor Cambinol reduced tumorigenesis in TH-MYCN transgenic mice. Our data demonstrate the important roles of SIRT1 in N-Myc oncogenesis and SIRT1 inhibitors in the prevention and therapy of N-Myc-induced neuroblastoma.


Asunto(s)
Fosfatasa 6 de Especificidad Dual/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Sirtuina 1/metabolismo , Animales , Sitios de Unión/genética , Línea Celular Tumoral , Proliferación Celular , Fosfatasa 6 de Especificidad Dual/genética , Inhibidores Enzimáticos/farmacología , Retroalimentación Fisiológica , Regulación Neoplásica de la Expresión Génica , Ratones , Ratones Transgénicos , Naftalenos/farmacología , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroblastoma/patología , Fosforilación , Regiones Promotoras Genéticas , Estabilidad Proteica , Proteínas Proto-Oncogénicas c-myc/genética , Pirimidinonas/farmacología , Distribución Aleatoria , Sirtuina 1/genética , Factor de Transcripción Sp1/metabolismo , Carga Tumoral/efectos de los fármacos , Carga Tumoral/genética
16.
Nat Commun ; 15(1): 5585, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38992040

RESUMEN

MYCN oncogene amplification is frequently observed in aggressive childhood neuroblastoma. Using an unbiased large-scale mutagenesis screen in neuroblastoma-prone transgenic mice, we identify a single germline point mutation in the transcriptional corepressor Runx1t1, which abolishes MYCN-driven tumorigenesis. This loss-of-function mutation disrupts a highly conserved zinc finger domain within Runx1t1. Deletion of one Runx1t1 allele in an independent Runx1t1 knockout mouse model is also sufficient to prevent MYCN-driven neuroblastoma development, and reverse ganglia hyperplasia, a known pre-requisite for tumorigenesis. Silencing RUNX1T1 in human neuroblastoma cells decreases colony formation in vitro, and inhibits tumor growth in vivo. Moreover, RUNX1T1 knockdown inhibits the viability of PAX3-FOXO1 fusion-driven rhabdomyosarcoma and MYC-driven small cell lung cancer cells. Despite the role of Runx1t1 in MYCN-driven tumorigenesis neither gene directly regulates the other. We show RUNX1T1 forms part of a transcriptional LSD1-CoREST3-HDAC repressive complex recruited by HAND2 to enhancer regions to regulate chromatin accessibility and cell-fate pathway genes.


Asunto(s)
Carcinogénesis , Proteína Proto-Oncogénica N-Myc , Neuroblastoma , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroblastoma/patología , Animales , Proteína Proto-Oncogénica N-Myc/genética , Proteína Proto-Oncogénica N-Myc/metabolismo , Humanos , Ratones , Carcinogénesis/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Ratones Transgénicos , Ratones Noqueados , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Histona Demetilasas/metabolismo , Histona Demetilasas/genética , Proteínas Co-Represoras/metabolismo , Proteínas Co-Represoras/genética
17.
Biochim Biophys Acta ; 1819(11-12): 1173-85, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22921766

RESUMEN

Mutations in the CDKL5 (cyclin-dependent kinase-like 5) gene are associated with a severe epileptic encephalopathy (early infantile epileptic encephalopathy type 2, EIEE2) characterized by early-onset intractable seizures, infantile spasms, severe developmental delay, intellectual disability, and Rett syndrome (RTT)-like features. Despite the clear involvement of CDKL5 mutations in intellectual disability, the function of this protein during brain development and the molecular mechanisms involved in its regulation are still unknown. Using human neuroblastoma cells as a model system we found that an increase in CDKL5 expression caused an arrest of the cell cycle in the G(0)/G(1) phases and induced cellular differentiation. Interestingly, CDKL5 expression was inhibited by MYCN, a transcription factor that promotes cell proliferation during brain development and plays a relevant role in neuroblastoma biology. Through a combination of different and complementary molecular and cellular approaches we could show that MYCN acts as a direct repressor of the CDKL5 promoter. Overall our findings unveil a functional axis between MYCN and CDKL5 governing both neuron proliferation rate and differentiation. The fact that CDKL5 is involved in the control of both neuron proliferation and differentiation may help understand the early appearance of neurological symptoms in patients with mutations in CDKL5.


Asunto(s)
Puntos de Control del Ciclo Celular , Diferenciación Celular , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Oncogénicas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Animales , Encéfalo , Línea Celular Tumoral , Síndromes Epilépticos , Humanos , Ratones , Mutación , Proteína Proto-Oncogénica N-Myc , Proteínas del Tejido Nervioso/genética , Neuronas/patología , Proteínas Nucleares/genética , Proteínas Oncogénicas/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Represoras/genética , Síndrome de Rett/genética , Síndrome de Rett/metabolismo , Síndrome de Rett/patología , Espasmos Infantiles/genética , Espasmos Infantiles/metabolismo , Espasmos Infantiles/patología
18.
Hum Mol Genet ; 20(8): 1560-73, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21266456

RESUMEN

Mental retardation in Down syndrome (DS) appears to be related to severe neurogenesis impairment during critical phases of brain development. Recent lines of evidence in the cerebellum of a mouse model for DS (the Ts65Dn mouse) have shown a defective responsiveness to Sonic Hedgehog (Shh), a potent mitogen that controls cell division during brain development, suggesting involvement of the Shh pathway in the neurogenesis defects of DS. Based on these premises, we sought to identify the molecular mechanisms underlying derangement of the Shh pathway in neural precursor cells (NPCs) from Ts65Dn mice. By using an in vitro model of NPCs obtained from the subventricular zone and hippocampus, we found that trisomic NPCs had an increased expression of the Shh receptor Patched1 (Ptch1), a membrane protein that suppresses the action of a second receptor, Smoothened (Smo), thereby maintaining the pathway in a repressed state. Partial silencing of Ptch1 expression in trisomic NPCs restored cell proliferation, indicating that proliferation impairment was due to Ptch1 overexpression. The overexpression of Ptch1 in trisomic NPCs resulted from increased levels of AICD [a transcription-promoting fragment of amyloid precursor protein (APP)] and increased AICD binding to the Ptch1 promoter. Our data provide novel evidence that Ptch1 overexpression underlies derangement of the Shh pathway in trisomic NPCs with consequent proliferation impairment. The demonstration that Ptch1 overexpression in trisomic NPCs is due to an APP fragment provides a link between this trisomic gene and the defective neuronal production that characterizes the DS brain.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Síndrome de Down/genética , Células-Madre Neurales/fisiología , Neuronas/fisiología , Receptores de Superficie Celular/biosíntesis , Acetilación , Animales , Ciclo Celular/genética , Proliferación Celular , Ciclohexilaminas/farmacología , Metilación de ADN , Síndrome de Down/embriología , Síndrome de Down/metabolismo , Femenino , Proteína Forkhead Box M1 , Factores de Transcripción Forkhead/genética , Proteínas Hedgehog/genética , Proteínas Hedgehog/farmacología , Hipocampo/embriología , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Ventrículos Laterales/embriología , Ventrículos Laterales/metabolismo , Ventrículos Laterales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Receptores Patched , Receptor Patched-1 , Complejo Represivo Polycomb 1 , Regiones Promotoras Genéticas , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas/genética , Interferencia de ARN , Receptores de Superficie Celular/genética , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/genética , Proteínas Represoras/genética , Receptor Smoothened , Tiofenos/farmacología , Regulación hacia Arriba , Alcaloides de Veratrum/farmacología , Proteína con Dedos de Zinc GLI1 , Proteína Gli2 con Dedos de Zinc , Proteína Gli3 con Dedos de Zinc
19.
Haematologica ; 98(2): 193-200, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22875622

RESUMEN

Imatinib has so far been the first-choice treatment in chronic myeloid leukemia with excellent results. However, only a proportion of patients achieve major molecular response - hence the need to find biological predictors of outcome to select the optimal therapeutic strategy now that more potent inhibitors are available. We investigated a panel of 20 polymorphisms in seven genes, potentially associated with the pharmacogenetics of imatinib, in a subset of 189 patients with newly diagnosed chronic myeloid leukemia enrolled in the TOPS trial. The analysis included polymorphisms in the transporters hOCT1, MDR1, ABCG2, OCTN1, and OATP1A2, and in the metabolizing genes CYP3A4 and CYP3A5. In the overall population, the OCTN1 C allele (rs1050152), a simple combination of polymorphisms in the hOCT1 gene and another combination in the genes involved in imatinib uptake were significantly associated with major molecular response. The combination of polymorphisms in imatinib uptake was also significantly associated with complete molecular response. Analyses restricted to Caucasians highlighted the significant association of MDR1 CC (rs60023214) genotype with complete molecular response. We demonstrate the usefulness of a pharmacogenetic approach for stratifying patients with chronic myeloid leukemia according to their likelihood of achieving a major or complete molecular response to imatinib. This represents an attractive opportunity for therapy optimization, worth testing in clinical trials.


Asunto(s)
Antineoplásicos/uso terapéutico , Benzamidas/uso terapéutico , Proteínas de Transporte de Catión/genética , Sistema Enzimático del Citocromo P-450/genética , Genotipo , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Piperazinas/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinas/uso terapéutico , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Adolescente , Adulto , Anciano , Alelos , Antineoplásicos/metabolismo , Benzamidas/metabolismo , Proteínas de Transporte de Catión/metabolismo , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Femenino , Humanos , Mesilato de Imatinib , Masculino , Persona de Mediana Edad , Proteínas de Transporte de Catión Orgánico/genética , Proteínas de Transporte de Catión Orgánico/metabolismo , Piperazinas/metabolismo , Polimorfismo de Nucleótido Simple , Inhibidores de Proteínas Quinasas/metabolismo , Pirimidinas/metabolismo , Simportadores , Resultado del Tratamiento , Adulto Joven
20.
Int J Mol Sci ; 14(10): 19681-704, 2013 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-24084719

RESUMEN

ncRNAs are the most recently identified class of regulatory RNAs with vital functions in gene expression regulation and cell development. Among the variety of roles they play, their involvement in human diseases has opened new avenues of research towards the discovery and development of novel therapeutic approaches. Important data come from the field of hereditary muscle dystrophies, like Duchenne muscle dystrophy and Myotonic dystrophies, rare diseases affecting 1 in 7000-15,000 newborns and is characterized by severe to mild muscle weakness associated with cardiac involvement. Novel therapeutic approaches are now ongoing for these diseases, also based on splicing modulation. In this review we provide an overview about ncRNAs and their behavior in muscular dystrophy and explore their links with diagnosis, prognosis and treatments, highlighting the role of regulatory RNAs in these pathologies.


Asunto(s)
Distrofias Musculares/genética , ARN no Traducido/genética , Animales , Humanos , Distrofias Musculares/diagnóstico , Distrofias Musculares/patología , Distrofias Musculares/terapia , Pronóstico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA