Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
2.
Dev Biol ; 426(1): 43-55, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28434803

RESUMEN

Cytoplasmic polyadenylation is a mechanism of mRNA regulation prevalent in metazoan germ cells; it is largely dependent on Cytoplasmic Polyadenylation Element Binding proteins (CPEBs). Two CPEB homologs were identified in the planarian Schmidtea mediterranea. Smed-CPEB1 is expressed in ovaries and yolk glands of sexually mature planarians, and required for oocyte and yolk gland development. In contrast, Smed-CPEB2 is expressed in the testes and the central nervous system; its function is required for spermatogenesis as well as non-autonomously for development of ovaries and accessory reproductive organs. Transcriptome analysis of CPEB knockdown animals uncovered a comprehensive collection of molecular markers for reproductive structures in S. mediterranea, including ovaries, testes, yolk glands, and the copulatory apparatus. Analysis by RNA interference revealed contributions for a dozen of these genes during oogenesis, spermatogenesis, or capsule formation. We also present evidence suggesting that Smed-CPEB2 promotes translation of Neuropeptide Y-8, a prohormone required for planarian sexual maturation. These findings provide mechanistic insight into potentially conserved processes of germ cell development, as well as events involved in capsule deposition by flatworms.


Asunto(s)
Células Germinativas/citología , Oogénesis/fisiología , Ovario/crecimiento & desarrollo , Planarias/anatomía & histología , Planarias/crecimiento & desarrollo , Espermatogénesis/fisiología , Factores de Escisión y Poliadenilación de ARNm/genética , Animales , Diferenciación Celular/genética , Femenino , Perfilación de la Expresión Génica , Ovario/metabolismo , Poliadenilación , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptores de Neuropéptido Y/biosíntesis , Receptores de Neuropéptido Y/genética , Maduración Sexual/genética , Maduración Sexual/fisiología , Factores de Escisión y Poliadenilación de ARNm/biosíntesis
3.
Nucleic Acids Res ; 45(7): 4142-4157, 2017 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-27998933

RESUMEN

miRNAs play important roles during mammalian spermatogenesis. However, the function of most miRNAs in spermatogenesis and the underlying mechanisms remain unknown. Here, we report that miR-202 is highly expressed in mouse spermatogonial stem cells (SSCs), and is oppositely regulated by Glial cell-Derived Neurotrophic Factor (GDNF) and retinoic acid (RA), two key factors for SSC self-renewal and differentiation. We used inducible CRISPR-Cas9 to knockout miR-202 in cultured SSCs, and found that the knockout SSCs initiated premature differentiation accompanied by reduced stem cell activity and increased mitosis and apoptosis. Target genes were identified with iTRAQ-based proteomic analysis and RNA sequencing, and are enriched with cell cycle regulators and RNA-binding proteins. Rbfox2 and Cpeb1 were found to be direct targets of miR-202 and Rbfox2 but not Cpeb1, is essential for the differentiation of SSCs into meiotic cells. Accordingly, an SSC fate-regulatory network composed of signaling molecules of GDNF and RA, miR-202 and diverse downstream effectors has been identified.


Asunto(s)
Células Madre Germinales Adultas/metabolismo , Ciclo Celular/genética , MicroARNs/metabolismo , Factores de Empalme de ARN/biosíntesis , Células Madre Germinales Adultas/citología , Animales , Técnicas de Inactivación de Genes , Masculino , Meiosis/genética , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Proteómica , Análisis de Secuencia de ARN , Espermatogénesis/genética , Factores de Transcripción/biosíntesis , Factores de Escisión y Poliadenilación de ARNm/biosíntesis
4.
Mol Med Rep ; 14(5): 4271-4278, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27633853

RESUMEN

MicroRNA-181a (miR-181a) is upregulated in osteosarcoma, and its overexpression promotes the proliferation and inhibits the apoptosis of osteosarcoma cells. However, the mechanism of miR­181a as an oncogene remains to be fully elucidated in osteosarcoma. Cleavage factor (CF) Im25 links alternative polyadenylation to glioblastoma tumor suppression, however, its role in osteosarcoma has not been reported. In the present study, it was confirmed that the expression of miR­181a was upregulated in osteosarcoma, and that silencing miR­181a inhibited the proliferation and promoted the apoptosis of osteosarcoma cells. miRNAs are short non­coding RNAs, which regulate target mRNAs by binding predominantly to the 3'untranslated region (3'UTR), inducing either translational repression or degradation of the target. In the present study, target genes of miR­181a were screened using miRanda, which is a commonly used prediction algorithm. It was found that miR­181a targeted the 3'UTR of CFIm25 mRNA. Subsequent experiments confirmed that miR­181a downregulated the expression of CFIm25 in osteosarcoma cells. Finally, it was demonstrated that the CFIm25 protein was also downregulated in osteosarcoma tissues, and inhibited the proliferation and promoted the apoptosis of the cells. Elucidating the roles of miR­181a and CFIm25 in osteosarcoma not only assists in further understanding the pathogenesis and progression of this disease, but also offers novel targets for effective therapies.


Asunto(s)
Proliferación Celular/genética , MicroARNs/genética , Osteosarcoma/genética , Factores de Escisión y Poliadenilación de ARNm/biosíntesis , Apoptosis/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Osteosarcoma/patología , ARN Mensajero/genética , Factores de Escisión y Poliadenilación de ARNm/genética
5.
Methods Mol Biol ; 1255: 91-115, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25487207

RESUMEN

In the post-genomic era where gene sequences are available for many organisms, attention has shifted from DNA to the workhorses of the cell-RNA and protein. A number of proteins, as recent studies indicate, seem to possess RNA-binding and RNA cleavage activities. In order to understand the events that comprise RNA processing such as splicing, 3' end processing, and even RNA turnover, well established methods are necessary. Bacterial recombinant proteins afford an invaluable opportunity to produce proteins in an economical and reproducible fashion in order to study these activities. This chapter describes various experimental protocols to begin the elucidation of the many events that surround RNA processing at the 3' end of a transcript.


Asunto(s)
Proteínas de Plantas/aislamiento & purificación , Plantas/metabolismo , Poliadenilación , ARN Mensajero/metabolismo , Proteínas Recombinantes/aislamiento & purificación , Factores de Escisión y Poliadenilación de ARNm/aislamiento & purificación , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Plantas/biosíntesis , Proteínas de Plantas/genética , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Ribonucleasas/metabolismo , Factores de Escisión y Poliadenilación de ARNm/biosíntesis , Factores de Escisión y Poliadenilación de ARNm/genética
6.
Nucleus ; 5(6): 613-25, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25493544

RESUMEN

The scaffolding protein Symplekin is part of multiple complexes involved in generating and modifying the 3' end of mRNAs, including cleavage-polyadenylation, histone pre-mRNA processing and cytoplasmic polyadenylation. To study these functions in vivo, we examined the localization of Symplekin during development and generated mutations of the Drosophila Symplekin gene. Mutations in Symplekin that reduce Symplekin protein levels alter the efficiency of both poly A(+) and histone mRNA 3' end formation resulting in lethality or sterility. Histone mRNA synthesis takes place at the histone locus body (HLB) and requires a complex composed of Symplekin and several polyadenylation factors that associates with the U7 snRNP. Symplekin is present in the HLB in the early embryo when Cyclin E/Cdk2 is active and histone genes are expressed and is absent from the HLB in cells that have exited the cell cycle. During oogenesis, Symplekin is preferentially localized to HLBs during S-phase in endoreduplicating follicle cells when histone mRNA is synthesized. After the completion of endoreplication, Symplekin accumulates in the cytoplasm, in addition to the nucleoplasm, and localizes to tricellular junctions of the follicle cell epithelium. This localization depends on the RNA binding protein ypsilon schachtel. CPSF-73 and a number of mRNAs are localized at this same site, suggesting that Symplekin participates in cytoplasmic polyadenylation at tricellular junctions.


Asunto(s)
Proteínas de Drosophila/biosíntesis , Desarrollo Embrionario , Histonas/genética , Poliadenilación/genética , Factores de Escisión y Poliadenilación de ARNm/biosíntesis , Animales , Citoplasma/genética , Drosophila , Proteínas de Drosophila/genética , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica , Histonas/metabolismo , Cuerpos de Inclusión Intranucleares/genética , Cuerpos de Inclusión Intranucleares/metabolismo , Mutación , Nucleoplasminas/genética , Ribonucleoproteína Nuclear Pequeña U7/genética , Fase S/genética , Factores de Escisión y Poliadenilación de ARNm/genética
7.
Oncotarget ; 5(16): 6756-69, 2014 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-25216517

RESUMEN

Glioma stemness has been recognized as the most important reason for glioma relapse and drug resistance. Differentiation of glioma stem cells (GSCs) has been implicated as a novel approach to target recurrent glioma. However, the detailed molecular mechanism involved in the differentiation of GSCs has not yet been elucidated. This study identified CPEB1 as the key modulator that induces the differentiation of GSCs at the post-transcriptional level. Gain and loss of function experiments showed that CPEB1 expression reduced sphere formation ability and the expression of stemness markers such as Nestin and Notch. To elucidate the detailed molecular mechanism underlying the action of CPEB1, we investigated the interacting ribonome of the CPEB1 complex using a Ribonomics approach. CPEB1 specifically suppressed the translation of HES1 and SIRT1 by interacting with a cytoplasmic polyadenylation element. The expression profile of CPEB1 negatively correlated with overall survival in glioma patients. Overexpression of CPEB1 decreased the number of GSCs in an orthotopically implanted glioma animal model. These results suggest that CPEB1-mediated translational control is essential for the differentiation of GSCs and provides novel therapeutic concepts for differentiation therapy.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Glioma/patología , Proteínas de Homeodominio/biosíntesis , Células Madre Neoplásicas/patología , Sirtuina 1/biosíntesis , Factores de Transcripción/biosíntesis , Factores de Escisión y Poliadenilación de ARNm/biosíntesis , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Diferenciación Celular/fisiología , Regulación hacia Abajo , Femenino , Glioma/genética , Glioma/metabolismo , Células HEK293 , Xenoinjertos , Proteínas de Homeodominio/genética , Humanos , Ratones , Ratones Endogámicos BALB C , Sirtuina 1/genética , Factor de Transcripción HES-1 , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Transfección , Factores de Escisión y Poliadenilación de ARNm/genética , Factores de Escisión y Poliadenilación de ARNm/metabolismo
8.
Mol Reprod Dev ; 81(4): 376-87, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24474627

RESUMEN

In many species, there is little transcription in the mature oocyte, and zygotic transcription does not begin immediately after fertilization. In zebrafish, zygotic transcription is not initiated until the mid-blastula transition, thus the production of new proteins during oogenesis and early embryogenesis is dependent on the translation of maternal mRNAs. In a growing number of species, the translation of key maternal transcripts is coupled to their cytoplasmic polyadenylation. One family of RNA-binding proteins implicated in this process is the cytoplasmic polyadenylation element (CPE)-binding proteins (CPEBs), which bind to a sequence in the 3'-untranslated regions of regulated transcripts and mediate their storage/repression or translation. In several species, there is evidence for two classes of CPEBs, a larger oocyte-type and a smaller CPEB that functions during embryogenesis. This appears to be the case in zebrafish as well, and we now provide evidence suggesting that the oocyte-type CPEB (zorba) regulates the translation of the embryonic-type (ElrA) by keeping the ElrA transcript in storage until fertilization. When zorba levels fall, ElrA protein is then produced and available to regulate the translation of additional mRNAs during embryogenesis. We have also identified a potential target of ElrA, the maternal mRNA for hnRNPab, which is a potential homolog of the Drosophila gene squid, whose product plays a role in patterning the Drosophila oocyte and embryo. These data suggest that during zebrafish embryogenesis, cytoplasmic polyadenylation mediates a cascade of translational control whose final targets play central patterning roles during embryogenesis.


Asunto(s)
Proteínas ELAV/biosíntesis , Regulación del Desarrollo de la Expresión Génica , Oogénesis/genética , Procesamiento Postranscripcional del ARN , ARN Mensajero/biosíntesis , Proteínas de Unión al ARN/fisiología , Proteínas de Pez Cebra/biosíntesis , Proteínas de Pez Cebra/fisiología , Pez Cebra/genética , Factores de Escisión y Poliadenilación de ARNm/biosíntesis , Regiones no Traducidas 3' , Animales , Blástula/metabolismo , Tipificación del Cuerpo/genética , Citoplasma/metabolismo , Proteínas ELAV/genética , Embrión no Mamífero/metabolismo , Femenino , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , Oocitos/metabolismo , Poliadenilación , Biosíntesis de Proteínas , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Pez Cebra/embriología , Pez Cebra/fisiología , Proteínas de Pez Cebra/genética , Factores de Escisión y Poliadenilación de ARNm/genética , Factores de Escisión y Poliadenilación de ARNm/fisiología
9.
Mol Cell Biol ; 32(21): 4306-22, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22907757

RESUMEN

Histone mRNAs are rapidly degraded at the end of S phase, and a 26-nucleotide stem-loop in the 3' untranslated region is a key determinant of histone mRNA stability. This sequence is the binding site for stem-loop binding protein (SLBP), which helps to recruit components of the RNA degradation machinery to the histone mRNA 3' end. SLBP is the only protein whose expression is cell cycle regulated during S phase and whose degradation is temporally correlated with histone mRNA degradation. Here we report that chemical inhibition of the prolyl isomerase Pin1 or downregulation of Pin1 by small interfering RNA (siRNA) increases the mRNA stability of all five core histone mRNAs and the stability of SLBP. Pin1 regulates SLBP polyubiquitination via the Ser20/Ser23 phosphodegron in the N terminus. siRNA knockdown of Pin1 results in accumulation of SLBP in the nucleus. We show that Pin1 can act along with protein phosphatase 2A (PP2A) in vitro to dephosphorylate a phosphothreonine in a conserved TPNK sequence in the SLBP RNA binding domain, thereby dissociating SLBP from the histone mRNA hairpin. Our data suggest that Pin1 and PP2A act to coordinate the degradation of SLBP by the ubiquitin proteasome system and the exosome-mediated degradation of the histone mRNA by regulating complex dissociation.


Asunto(s)
Proteínas Nucleares/metabolismo , Isomerasa de Peptidilprolil/metabolismo , Proteína Fosfatasa 2/metabolismo , Estabilidad del ARN , ARN Mensajero/metabolismo , Factores de Escisión y Poliadenilación de ARNm/metabolismo , Ciclo Celular/genética , Línea Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Regulación hacia Abajo , Células HEK293 , Células HeLa , Histonas , Humanos , Peptidilprolil Isomerasa de Interacción con NIMA , Proteínas Nucleares/biosíntesis , Isomerasa de Peptidilprolil/genética , Interferencia de ARN , ARN Interferente Pequeño , Proteínas de Unión al ARN/metabolismo , Ubiquitinación , Factores de Escisión y Poliadenilación de ARNm/biosíntesis , Factores de Escisión y Poliadenilación de ARNm/genética
10.
Mol Reprod Dev ; 79(6): 380-91, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22467188

RESUMEN

During early embryo development, chromatin packaging is sustained by histones of maternal origin. Most histone messenger RNAs are not polyadenylated, but rather end in an evolutionarily conserved stem-loop that controls RNA processing, nucleocytoplasmic transport, stability, and translation via interactions with a specific protein named stem-loop-binding protein (SLBP). In mouse oocytes, mSLBP is synthesized abundantly during maturation and activates histone translation. In Xenopus, xSLBP is present in stage-VI oocytes, but histone mRNA is protected from premature translation by the oocyte-specific Xenopus SLBP2 (xSLBP2) protein; during maturation xSLBP2 replacement by xSLBP results in histone synthesis. Here, we report the first experimental evidence and characterization of a mammalian SLBP2 ortholog. Bovine bSLBP and bSLBP2 display distinct expression patterns throughout oocyte maturation and pre-implantation embryo development. From the immature oocyte to the morula, bSLBP2 is concentrated in the nucleus, while it is homogeneously distributed throughout the cytoplasm in mature oocytes. A putative SLBP2 gene is conserved in the genome of several mammalian species, and the corresponding transcripts were detected in rat, dog, horse, and pig oocytes. By contrast, a pseudogene is found in mouse, human, and rabbit. Altogether, our data suggest that the availability of histones in oocytes is regulated by an alternative mechanism in bovine and other species as compared to mouse and frog.


Asunto(s)
Proteínas Nucleares/biosíntesis , Oogénesis , ARN Mensajero/genética , Factores de Escisión y Poliadenilación de ARNm/biosíntesis , Animales , Secuencia de Bases , Sitios de Unión/genética , Bovinos , Perros , Desarrollo Embrionario , Histonas/genética , Caballos , Humanos , Ratones , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oocitos/citología , Proteínas de Unión al ARN , Conejos , Ratas , Alineación de Secuencia , Porcinos , Xenopus laevis , Factores de Escisión y Poliadenilación de ARNm/genética , Factores de Escisión y Poliadenilación de ARNm/metabolismo
11.
Gut ; 61(8): 1115-23, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22052064

RESUMEN

BACKGROUND: Gastric cancer (GC) is a highly prevalent disease, being the fourth most common cancer and the second leading cause of cancer-associated deaths worldwide. Although many genes have been implicated in its development, many cases remain genetically unexplained. Hence, there is an urgent need to find new disease-related genes. METHODS: A transgenic Drosophila model was used to screen for novel genes putatively involved in GC. The authors evaluated the expression of the most interesting candidates in GC cell lines and primary tumours by semi-quantitative reverse transcription PCR, dissected the molecular mechanisms responsible for the deregulation of the most relevant one, and analysed its functional role in vitro and in a chicken embryo model. RESULTS: Six candidate genes were identified, of which cytoplasmic polyadenylation element binding protein 1 (CPEB1) was downregulated in all GC cell lines and in 11 of 12 primary GC tumours. The pivotal CPEB1 promoter CpG site was determined, and it was found that methylation at this 79th CpG site was associated with CPEB1 silencing in GC cell lines and primary tumours. It was also discovered that methylation of this site was significantly more prevalent in diffuse type GC (p=0.007) and in cases with lymph node metastases (p=0.042). In vitro, CPEB1 impaired invasion. Its antiangiogenic role was also discovered, which was associated with downregulation of MMP14 and VEGFA. CONCLUSIONS: The first evidence of CPEB1 involvement in GC is presented, along with the molecular mechanism underlying the regulation of its expression and its potential role in invasion and angiogenesis.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila/genética , Regulación Neoplásica de la Expresión Génica , Mutación , Neoplasias Experimentales/genética , ARN Neoplásico/genética , Neoplasias Gástricas/genética , Factores de Transcripción/genética , Factores de Escisión y Poliadenilación de ARNm/genética , Animales , Western Blotting , Línea Celular Tumoral , Metilación de ADN , Proteínas de Drosophila/biosíntesis , Silenciador del Gen , Humanos , Inmunohistoquímica , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Regiones Promotoras Genéticas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Factores de Transcripción/biosíntesis , Factores de Escisión y Poliadenilación de ARNm/biosíntesis
12.
J Biol Chem ; 286(4): 2853-63, 2011 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-21098481

RESUMEN

Precise control of the timing of translational activation of dormant mRNAs stored in oocytes is required for normal progression of oocyte maturation. We previously showed that Pumilio1 (Pum1) is specifically involved in the translational control of cyclin B1 mRNA during Xenopus oocyte maturation, in cooperation with cytoplasmic polyadenylation element-binding protein (CPEB). It was reported that another Pumilio, Pumilio2 (Pum2), exists in Xenopus oocytes and that this protein regulates the translation of RINGO mRNA, together with Deleted in Azoospermia-like protein (DAZL). In this study, we characterized Pum1 and Pum2 biochemically by using newly produced antibodies that discriminate between them. Pum1 and Pum2 are bound to several key proteins involved in translational control of dormant mRNAs, including CPEB and DAZL, in immature oocytes. However, Pum1 and Pum2 themselves have no physical interaction. Injection of anti-Pum1 or anti-Pum2 antibody accelerated CPEB phosphorylation, cyclin B1 translation, and oocyte maturation. Pum1 phosphorylation coincides with the dissociation of CPEB from Pum1 and the translational activation of cyclin B1 mRNA, a target of Pum1, whereas Pum2 phosphorylation occurred at timing earlier than that for Pum1. Some, but not all, of cyclin B1 mRNAs release the deadenylase PARN during oocyte maturation, whereas Pum1 remains associated with the mRNA. On the basis of these findings, we discuss the functions of Pum1 and Pum2 in translational control of mRNAs during oocyte maturation.


Asunto(s)
Oocitos/metabolismo , Biosíntesis de Proteínas/fisiología , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas de Xenopus/metabolismo , Animales , Anticuerpos/química , Secuencia de Bases , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/genética , Proteínas de Ciclo Celular/biosíntesis , Proteínas de Ciclo Celular/genética , Ciclina B1/biosíntesis , Ciclina B1/genética , Datos de Secuencia Molecular , Fosforilación/fisiología , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Factores de Tiempo , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Proteínas de Xenopus/biosíntesis , Proteínas de Xenopus/genética , Xenopus laevis , Factores de Escisión y Poliadenilación de ARNm/biosíntesis , Factores de Escisión y Poliadenilación de ARNm/genética
14.
Nat Struct Mol Biol ; 17(10): 1218-25, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20890289

RESUMEN

During replicative aging of primary cells morphological transformations occur, the expression pattern is altered and chromatin changes globally. Here we show that chronic damage signals, probably caused by telomere processing, affect expression of histones and lead to their depletion. We investigated the abundance and cell cycle expression of histones and histone chaperones and found defects in histone biosynthesis during replicative aging. Simultaneously, epigenetic marks were redistributed across the phases of the cell cycle and the DNA damage response (DDR) machinery was activated. The age-dependent reprogramming affected telomeric chromatin itself, which was progressively destabilized, leading to a boost of the telomere-associated DDR with each successive cell cycle. We propose a mechanism in which changes in the structural and epigenetic integrity of telomeres affect core histones and their chaperones, enforcing a self-perpetuating pathway of global epigenetic changes that ultimately leads to senescence.


Asunto(s)
Senescencia Celular/fisiología , Cromatina/metabolismo , Daño del ADN , Fibroblastos/metabolismo , Histonas/biosíntesis , Telómero/fisiología , Bleomicina/toxicidad , Línea Celular , Inmunoprecipitación de Cromatina , Replicación del ADN , Epigénesis Genética , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Humanos , Metilación , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Proteínas Oncogénicas Virales/fisiología , Proteínas E7 de Papillomavirus/fisiología , Procesamiento Proteico-Postraduccional , Proteínas Represoras/fisiología , Proteína de Retinoblastoma/fisiología , Proteína p53 Supresora de Tumor/fisiología , Factores de Escisión y Poliadenilación de ARNm/biosíntesis , Factores de Escisión y Poliadenilación de ARNm/genética
15.
Intern Med ; 49(12): 1195-200, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20558942

RESUMEN

A 28-year-old man with marked eosinophilia is described. FIP1L1/PDGFRA mRNA showed multiple alternatively-spliced fusion transcripts. Sequencing analysis showed that the deduced DNA breakpoints were intron 10 in the FIP1L1 gene and exon 12 in the PDGFRA gene. Then, a diagnosis of chronic eosinophilic leukemia (CEL) was made. Whereas the response to the treatments with prednisolone and hydroxyurea were unsatisfactory, treatment with imatinib showed a rapid decrease of eosinophils. The hemoglobin level also dropped and bone marrow examination showed pure red cell aplasia. Continued administration of very low dose imatinib (100 mg every 5 days) led to and maintained complete molecular remission, with good tolerability.


Asunto(s)
Síndrome Hipereosinofílico/genética , Proteínas de Fusión Oncogénica/biosíntesis , Piperazinas/uso terapéutico , Pirimidinas/uso terapéutico , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Aplasia Pura de Células Rojas/genética , Factores de Escisión y Poliadenilación de ARNm/biosíntesis , Adulto , Benzamidas , Enfermedad Crónica , Humanos , Síndrome Hipereosinofílico/complicaciones , Síndrome Hipereosinofílico/tratamiento farmacológico , Mesilato de Imatinib , Masculino , Proteínas de Fusión Oncogénica/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Aplasia Pura de Células Rojas/tratamiento farmacológico , Aplasia Pura de Células Rojas/etiología , Factores de Escisión y Poliadenilación de ARNm/genética
16.
Haematologica ; 94(9): 1236-41, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19734416

RESUMEN

BACKGROUND: A T-cell clone, thought to be the source of eosinophilopoietic cytokines, identified by clonal rearrangement of the T-cell receptor and by the presence of aberrant T-cell immunophenotype in peripheral blood defines lymphocytic variant of hypereosinophilic syndrome (L-HES). DESIGN AND METHODS: Peripheral blood samples from 42 patients who satisfied the diagnostic criteria for HES were studied for T-cell receptor clonal rearrangement by polymerase chain reaction according to BIOMED-2. The T-cell immunophenotype population was assessed in peripheral blood by flow cytometry. The FIP1L1-PDGFRA fusion gene was detected by nested polymerase chain reaction. RESULTS: Forty-two HES patients (18 males and 24 females) with a median age at diagnosis of 56 years (range 17-84) were examined in this study. Their median white blood cell count was 12.9 x 10(9)/L (range 5.3-121), with an absolute eosinophil count of 4.5 x 10(9)/L (range 1.5-99) and a median eosinophilic bone marrow infiltration of 30% (range 11-64). Among the 42 patients, clonal T-cell receptor rearrangements were detected in 18 patients (42.8%). Patients with T-cell receptor clonality included: T-cell receptor beta in 15 patients (35%), T-cell receptor gamma in 9 (21%) and T-cell receptor delta in 9 (21%) patients, respectively. Clonality was detected in all three T-cell receptor loci in 4 cases, in two loci in 7 patients and in one T-cell receptor locus in the remaining 7 patients. The FIP1L1-PDGFRA fusion transcript was absent in all but 2 patients with T-cell receptor clonality. Three patients out of 42 revealed an aberrant T-cell immunophenotype. In some patients, an abnormal CD4:CD8 ratio was demonstrated. CONCLUSIONS: T-cell abnormalities are present at high frequencies in patients with HES.


Asunto(s)
Reordenamiento Génico de la Cadena beta de los Receptores de Antígenos de los Linfocitos T/inmunología , Reordenamiento Génico de la Cadena gamma de los Receptores de Antígenos de los Linfocitos T/inmunología , Síndrome Hipereosinofílico/inmunología , Proteínas de Fusión Oncogénica/inmunología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/inmunología , Linfocitos T/inmunología , Factores de Escisión y Poliadenilación de ARNm/inmunología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Relación CD4-CD8 , Femenino , Reordenamiento Génico de la Cadena beta de los Receptores de Antígenos de los Linfocitos T/genética , Reordenamiento Génico de la Cadena gamma de los Receptores de Antígenos de los Linfocitos T/genética , Humanos , Síndrome Hipereosinofílico/sangre , Síndrome Hipereosinofílico/genética , Masculino , Persona de Mediana Edad , Proteínas de Fusión Oncogénica/biosíntesis , Proteínas de Fusión Oncogénica/genética , Reacción en Cadena de la Polimerasa , Estudios Prospectivos , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Linfocitos T/metabolismo , Linfocitos T/patología , Factores de Escisión y Poliadenilación de ARNm/biosíntesis , Factores de Escisión y Poliadenilación de ARNm/genética
18.
Ann Hematol ; 86(12): 855-63, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17701174

RESUMEN

To study the clinical characteristics of hypereosionophilic syndrome and chronic eosinophilic leukemia (HES/CEL) in Japan, the clinical data of 29 HES/CEL patients throughout the country were surveyed. Moreover, the involvement of the FIP1L1-alphaPDGFR fusion gene resulting from a cryptic del (4)(q12q12) was examined in 24 cases. The FIP1L1-alphaPDGFR messenger RNA (mRNA) was detected in three patients (13% of patients fulfilled WHO criteria and 17% of Chusid criteria). One had a novel fusion transcript, which skipped the exon 12 of alphaPDGFR. The transcript appears to be generated by a splicing mechanism that is different from the previously reported splicing patterns. In silico analysis, the exon skipping was not related to a disruption of the exonic splicing enhancers within the exon but strongly associated with the loss of the vast majority of the FIP1L intron 8a where intronic splicing enhancers were accumulated. Unexpectedly, pseudo-chimera DNA fragments with some shared characteristic features were occasionally generated from healthy control samples by reverse transcriptase polymerase chain reaction (RT-PCR). Considering the relatively low incidence of the FIP1L1-alphaPDGFR transcript positive case, extreme care must therefore be taken when making a diagnosis using RT-PCR before imatinib therapy.


Asunto(s)
Síndrome Hipereosinofílico/genética , Proteínas de Fusión Oncogénica/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Factores de Escisión y Poliadenilación de ARNm/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Exones/genética , Femenino , Humanos , Síndrome Hipereosinofílico/epidemiología , Intrones/genética , Japón/epidemiología , Masculino , Persona de Mediana Edad , Proteínas de Fusión Oncogénica/análisis , Proteínas de Fusión Oncogénica/biosíntesis , Empalme del ARN , ARN Mensajero/análisis , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/análisis , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Eliminación de Secuencia , Factores de Escisión y Poliadenilación de ARNm/análisis , Factores de Escisión y Poliadenilación de ARNm/biosíntesis
19.
Int Arch Allergy Immunol ; 143 Suppl 1: 28-32, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17541273

RESUMEN

BACKGROUND: EoL-1 cells have a FIP1L1-PDGFRA fusion gene which causes the transformation of eosinophilic precursor cells into leukemia cells. Recently, we suggested that the induction of differentiation of EoL-1 cells into eosinophils by the HDAC inhibitors apicidin and n-butyrate is due to the continuous inhibition of HDACs. However, neither apicidin nor n-butyrate inhibited the expression of FIP1L1-PDGFRA mRNA, although both these inhibitors suppressed cell proliferation. Therefore, in this study, we analyzed whether the levels of FIP1L1-PDGFRalpha protein and phosphorylated-Stat5 involved in the signaling for the proliferation of EoL-1 cells are attenuated by HDAC inhibitors. METHODS: EoL-1 cells were incubated in the presence of apicidin, TSA or n-butyrate. FIP1L1-PDGFRalpha and phosphorylated-Stat5 were detected by Western blotting. RESULTS: Treatment of EoL-1 cells with apicidin at 100 nM or n-butyrate at 500 microM decreased the levels of FIP1L1-PDGFRalpha protein and phosphorylated-Stat5, while that with trichostatin A at 30 nM did not. CONCLUSIONS: The decrease in the level of FIP1L1-PDGFRalpha protein caused by apicidin and n-butyrate might be one of the mechanisms by which EoL-1 cells are induced to differentiate into eosinophils by these HDAC inhibitors.


Asunto(s)
Butiratos/farmacología , Eosinófilos/citología , Inhibidores de Histona Desacetilasas , Síndrome Hipereosinofílico/patología , Proteínas de Neoplasias/fisiología , Proteínas de Fusión Oncogénica/fisiología , Péptidos Cíclicos/farmacología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/fisiología , Factores de Escisión y Poliadenilación de ARNm/fisiología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Línea Celular Tumoral/citología , Línea Celular Tumoral/efectos de los fármacos , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Humanos , Ácidos Hidroxámicos/farmacología , Síndrome Hipereosinofílico/genética , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Proteínas de Fusión Oncogénica/biosíntesis , Proteínas de Fusión Oncogénica/genética , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Factor de Transcripción STAT5/metabolismo , Factores de Escisión y Poliadenilación de ARNm/biosíntesis , Factores de Escisión y Poliadenilación de ARNm/genética
20.
Cancer Res ; 67(8): 3759-66, 2007 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-17440089

RESUMEN

An interstitial deletion on chromosome 4q12 resulting in the formation of the FIP1L1-PDGFRA fusion protein is involved in the pathogenesis of imatinib-sensitive chronic eosinophilic leukemia. The molecular mechanisms underlying the development of disease are largely undefined. Human CD34(+) hematopoietic progenitor cells were used to investigate the role of FIP1L1-PDGFRA in modulating lineage development. FIP1L1-PDGFRA induced both proliferation and differentiation of eosinophils, neutrophils, and erythrocytes in the absence of cytokines, which could be inhibited by imatinib. Whereas expression of FIP1L1-PDGFRA in hematopoietic stem cells and common myeloid progenitors induced the formation of multiple myeloid lineages, expression in granulocyte-macrophage progenitors induced only the development of eosinophils, neutrophils, and myeloblasts. Deletion of amino acids 30 to 233 in the FIP1L1 gene [FIP1L1(1-29)-PDGFRA] gave rise to an intermediate phenotype, exhibiting a dramatic reduction in the number of erythrocytes. FIP1L1-PDGFRA and FIP1L1(1-29)-PDGFRA both induced the activation of p38 and extracellular signal-regulated kinase 1/2 (ERK1/2) in myeloid progenitors, whereas signal transducers and activators of transcription 5 (STAT5) and protein kinase B/c-akt were only activated by FIP1L1-PDGFRA. Dominant-negative STAT5 partially inhibited FIP1L1-PDGFRA-induced colony formation, whereas combined inhibition of phosphatidylinositol-3-kinase and ERK1/2 significantly reversed FIP1L1-PDGFRA-induced colony formation. Taken together, these results suggest that expression of FIP1L1-PDFGRA in human hematopoietic progenitors induce a myeloproliferative phenotype via activation of multiple signaling molecules including phosphatidylinositol-3-kinase, ERK1/2, and STAT5.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Síndrome Hipereosinofílico/metabolismo , Proteínas de Fusión Oncogénica/biosíntesis , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Factores de Escisión y Poliadenilación de ARNm/biosíntesis , Benzamidas , Diferenciación Celular/fisiología , Procesos de Crecimiento Celular/fisiología , Enfermedad Crónica , Activación Enzimática , Células Madre Hematopoyéticas/patología , Humanos , Síndrome Hipereosinofílico/enzimología , Síndrome Hipereosinofílico/patología , Mesilato de Imatinib , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Células Mieloides/metabolismo , Células Mieloides/patología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Proteínas de Fusión Oncogénica/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Piperazinas/farmacología , Pirimidinas/farmacología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factor de Transcripción STAT5/metabolismo , Transducción de Señal , Factores de Escisión y Poliadenilación de ARNm/antagonistas & inhibidores , Factores de Escisión y Poliadenilación de ARNm/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...