Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 150(5): 934-47, 2012 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-22939621

RESUMEN

The factors that sequester transcriptionally repressed heterochromatin at the nuclear periphery are currently unknown. In a genome-wide RNAi screen, we found that depletion of S-adenosylmethionine (SAM) synthetase reduces histone methylation globally and causes derepression and release of heterochromatin from the nuclear periphery in Caenorhabditis elegans embryos. Analysis of histone methyltransferases (HMTs) showed that elimination of two HMTs, MET-2 and SET-25, mimics the loss of SAM synthetase, abrogating the perinuclear attachment of heterochromatic transgenes and of native chromosomal arms rich in histone H3 lysine 9 methylation. The two HMTs target H3K9 in a consecutive fashion: MET-2, a SETDB1 homolog, mediates mono- and dimethylation, and SET-25, a previously uncharacterized HMT, deposits H3K9me3. SET-25 colocalizes with its own product in perinuclear foci, in a manner dependent on H3K9me3, but not on its catalytic domain. This colocalization suggests an autonomous, self-reinforcing mechanism for the establishment and propagation of repeat-rich heterochromatin.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/citología , Núcleo Celular/química , Heterocromatina/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Animales , Caenorhabditis elegans/embriología , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/análisis , Proteínas de Caenorhabditis elegans/genética , Cromosomas/metabolismo , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Genoma de los Helmintos , N-Metiltransferasa de Histona-Lisina/análisis , N-Metiltransferasa de Histona-Lisina/genética , Laminas/metabolismo , Metionina Adenosiltransferasa/metabolismo , Metilación , Mutación
2.
Br J Haematol ; 194(1): 174-178, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33843056

RESUMEN

Blinatumomab with subsequent haematopoietic stem cell transplantation was applied in 13 infants with acute lymphoblastic leukaemia (ALL). Eight patients were treated in first remission due to slow clearance of minimal residual disease (MRD); one for MRD-reappearance after long MRD negativity, one for primary refractory disease and three during relapse treatment. In slow MRD responders, complete MRD response was achieved prior to transplantation, with an 18-month event-free survival of 75%. In contrast, only one of five patients with relapsed/refractory ALL is still in complete remission. These data provide a basis for future studies of immunotherapy in very high-risk infant ALL.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Trasplante de Células Madre Hematopoyéticas , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamiento farmacológico , Terapia Recuperativa , Preescolar , Supervivencia sin Enfermedad , Femenino , N-Metiltransferasa de Histona-Lisina/análisis , Humanos , Lactante , Estimación de Kaplan-Meier , Masculino , Proteína de la Leucemia Mieloide-Linfoide/análisis , Neoplasia Residual , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Recurrencia
3.
Chembiochem ; 21(1-2): 256-264, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31612581

RESUMEN

The SMYD2 protein lysine methyltransferase methylates various histone and non-histone proteins and is overexpressed in several cancers. Using peptide arrays, we investigated the substrate specificity of the enzyme, revealing a recognition of leucine (or weaker phenylalanine) at the -1 peptide site and disfavor of acidic residues at the +1 to +3 sites. Using this motif, novel SMYD2 peptide substrates were identified, leading to the discovery of 32 novel peptide substrates with a validated target site. Among them, 19 were previously reported to be methylated at the target lysine in human cells, strongly suggesting that SMYD2 is the protein lysine methyltransferase responsible for this activity. Methylation of some of the novel peptide substrates was tested at the protein level, leading to the identification of 14 novel protein substrates of SMYD2, six of which were more strongly methylated than p53, the best SMYD2 substrate described so far. The novel SMYD2 substrate proteins are involved in diverse biological processes such as chromatin regulation, transcription, and intracellular signaling. The results of our study provide a fundament for future investigations into the role of this important enzyme in normal development and cancer.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/análisis , Dicroismo Circular , Células HEK293 , N-Metiltransferasa de Histona-Lisina/aislamiento & purificación , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Metilación , Especificidad por Sustrato
4.
Biochem Biophys Res Commun ; 513(2): 340-346, 2019 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-30955858

RESUMEN

Dysfunction of histone methylation is known to be related to cancer progression. The histone methyltransferase SMYD2 methylates histone protein H3 and non-histone proteins, including poly ADP ribose polymerase 1 (PARP1). There have been reports of SMYD2 overexpression in several types of cancers. However, there are no reports regarding its role in high-grade serous ovarian carcinomas (HGSOCs). Therefore, we investigated the expression profile and conducted functional analysis on SMYD2 in HGSOC cells. In addition, we verified whether SMYD2 inhibition increases the susceptibility of HGSOC cells to PARP inhibitors. We analyzed the expression of histone methyltransferase SMYD2 by quantitative real-time polymerase chain reaction and immunohistochemistry using HGSOC clinical tissues (n = 35). We performed functional analyses, including cell proliferation assay, cell cycle analysis, and immunoblotting, after treatment with SMYD2 siRNAs and SMYD2 selective inhibitor LLY-507 in HGSOC cells. We also performed colony-formation assay after combination treatment with LLY-507 and PARP inhibitor olaparib in HGSOC cells. The expression profiles of SMYD2 showed significant overexpression of SMYD2 in HGSOC clinical tissues. The knockdown or inhibition of SMYD2 by siRNAs or LLY-507, respectively, suppressed cell growth by increasing the proportion of apoptotic cells. LLY-507 showed additive effect with olaparib in the colony-formation assay. These findings suggest that LLY-507 can be used alone or in combination with a PARP inhibitor for the treatment of patients with HGSOC.


Asunto(s)
Antineoplásicos/farmacología , Benzamidas/farmacología , Cistadenocarcinoma Seroso/tratamiento farmacológico , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , Neoplasias Ováricas/tratamiento farmacológico , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Pirrolidinas/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cistadenocarcinoma Seroso/patología , Femenino , N-Metiltransferasa de Histona-Lisina/análisis , Humanos , Neoplasias Ováricas/patología
5.
Biochem Biophys Res Commun ; 498(3): 579-585, 2018 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-29522714

RESUMEN

SET domain containing protein 2 (SETD2, also known as HYPB) is a 230-kD protein which is located at cytogenetic band p21.31 of chromosome 3. SETD2 is usually transformed or eradicated in multiple forms of tumours in humans. However, its primary function in gastric cancer (GC) remains unclear. In the current study, we investigated the mRNA and protein expression levels of SETD2 using immunohistochemistry, qPCR, RT-PCR, and immunoblotting. The function of SETD2 in GC cells was investigated by MTT and transwell assays. Our results revealed remarkably lower levels of SETD2 mRNA and protein in the tumour samples than in tumour-adjacent tissues. Decreased expression of SETD2 mRNA was observed in 122 (79.7%) of 153 primary tumour tissue samples. On the basis of the overall survival analysis, we could interpret that a low expression of SETD2 was correlated with a poor prognosis (P < 0.001, log-rank test). Multivariate survival analysis indicated that SETD2 was an obvious prognostic factor in patients with GC. SETD2 overexpression in GC cell lines significantly inhibited cell proliferation, migration, and invasion. Altogether, the investigation demonstrated the clinical significance of SETD2 expression, supporting the fundamental principle that a decrease in its level is an unfavourable event in the progression and prognosis of GC. Therefore, down-regulated SETD2 can presumably be a potential negative prognostic and progression marker for GC.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/genética , Invasividad Neoplásica/genética , Neoplasias Gástricas/genética , Anciano , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Regulación hacia Abajo , Femenino , Mucosa Gástrica/metabolismo , N-Metiltransferasa de Histona-Lisina/análisis , Humanos , Masculino , Persona de Mediana Edad , Invasividad Neoplásica/diagnóstico , Invasividad Neoplásica/patología , Pronóstico , Estómago/patología , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/patología
6.
Reproduction ; 154(1): 23-34, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28420800

RESUMEN

Histone methyltransferase SETDB1 suppresses gene expression and modulates heterochromatin formation through H3K9me2/3. Previous studies have revealed that SETDB1 catalyzes lysine 9 of histone H3 tri-methylation and plays essential roles in maintaining the survival of embryonic stem cells and spermatogonial stem cells in mice. However, the function of SETDB1 in porcine male germ cells remains unclear. The aim of the present study was to reveal the expression profile and function of SETDB1 in porcine germ cells. SETDB1 expression gradually increased during testis development. SETDB1 was strongly localized in gonocytes. Knockdown of SETDB1 gene expression led to gonocyte apoptosis and a decrease in H3K27me3, but no significant change in H3K9me3. These observations suggested that SETDB1 is a novel epigenetic regulator of porcine male germ cells, and contributes to the maintenance of gonocyte survival in pigs, probably due to the regulation of H3K27me3 rather than H3K9me3. These findings will provide a theoretical basis for the future study of epigenetic regulation of spermatogenesis.


Asunto(s)
Células Madre Germinales Adultas/fisiología , Supervivencia Celular/fisiología , N-Metiltransferasa de Histona-Lisina/fisiología , Sus scrofa , Animales , Animales Recién Nacidos , Apoptosis , Epigénesis Genética , Expresión Génica , Técnicas de Silenciamiento del Gen , Histona Demetilasas/análisis , Histona Demetilasas/fisiología , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/análisis , N-Metiltransferasa de Histona-Lisina/genética , Masculino , Espermatogénesis/fisiología , Testículo/crecimiento & desarrollo
7.
Biochem Biophys Res Commun ; 458(3): 525-530, 2015 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-25677622

RESUMEN

Cleft lip with or without palate (CL/P) is a common congenital anomaly in humans and is thought to be caused by genetic and environmental factors. However, the epigenetic mechanisms underlying orofacial clefts are not fully understood. Here, we investigate how the overdose of retinoic acid (RA), which can induce cleft palate in mice and humans, regulates histone methyltransferase, Wolf-Hirschhorn syndrome candidate 1 (WHSC1) during palatal development in mice. We treated mouse embryonic fibroblasts (MEFs) with 1 µM all-trans RA and discovered that the global level of H3K36me3 was downregulated and that expression of the H3K36 methyltransferase gene, Whsc1, was reduced. The expression level of WHSC1 in embryonic palatal shelves was reduced during palatogenesis, following maternal administration of 100 mg/kg body weight of RA by gastric intubation. Furthermore, the expression of WHSC1 in palatal shelves was observed in epithelial and mesenchymal cells at all stages, suggesting an important role for palatal development. Our results suggest that the pathogenesis of cleft palate observed after excessive RA exposure is likely to be associated with a reduction in the histone methyltransferase, WHSC1.


Asunto(s)
Fisura del Paladar/inducido químicamente , Sobredosis de Droga/complicaciones , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , N-Metiltransferasa de Histona-Lisina/genética , Hueso Paladar/embriología , Tretinoina/efectos adversos , Animales , Línea Celular , Fisura del Paladar/genética , Fisura del Paladar/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Femenino , N-Metiltransferasa de Histona-Lisina/análisis , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Metilación/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Hueso Paladar/anomalías , ARN Mensajero/genética
8.
Histochem Cell Biol ; 143(3): 259-66, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25294562

RESUMEN

Tissue-specific gene expression is subjected to epigenetic and genetic regulation. Posttranslational modifications of histone tails alter the accessibility of nuclear proteins to DNA, thus affecting the activity of the regulatory complex of nuclear proteins. Methylation at histone 3 lysine 9 (H3K9) is a crucial modification that affects gene expression and cell differentiation. H3K9 is known to have 0-3 methylation states, and these four methylated states are determined by the expression of sets of histone methyltransferases. During development, teeth are formed through mutual interactions between the mesenchyme and epithelium via a process that is subjected to the epigenetic regulation. In this study, we examined the expression of all H3K9 methyltransferases (H3K9MTases) during mouse tooth development. We found that four H3K9MTases-G9a, Glp, Prdm2, and Suv39h1-were highly expressed in the tooth germ, with expression peaks at around embryonic days 16.5 and 17.5 in mice. Immunohistochemical and in situ hybridization analyses revealed that all four H3K9MTases were enriched in the mesenchyme more than in the epithelium. Substrates of H3K9MTases, H3K9me1, H3K9me2, and H3K9me3 were also enriched in the mesenchyme. Taken together, these data suggested that coordinated expression of four H3K9MTases in the dental mesenchyme might play important roles in tooth development.


Asunto(s)
Perfilación de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/biosíntesis , N-Metiltransferasa de Histona-Lisina/genética , Germen Dentario/enzimología , Germen Dentario/crecimiento & desarrollo , Animales , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/análisis , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Endogámicos C57BL
9.
Biochem Biophys Res Commun ; 444(4): 634-7, 2014 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-24486544

RESUMEN

Pituitary homeobox 2 (PITX2), a Paired-like homeodomain transcription factor and a downstream effector of Wnt/ß-catenin signaling, plays substantial roles in embryonic development and human disorders. The mechanism of its functions, however, is not fully understood. In this study, we demonstrated that PITX2 associated with histone H3 lysine 4 (H3K4) methyltransferase (HKMT) mixed-lineage leukemia 4 (MLL4/KMT2D), Pax transactivation domain-interacting protein (PTIP), and other H3K4·HKMT core subunits. This association of PITX2 with H3K4·HKMT complex was dependent on PITX2's homeodomain. Consistently, the PITX2 protein complex was shown to possess H3K4·HKMT activity. Furthermore, the chromatin immunoprecipitation result revealed co-occupancy of PITX2 and PTIP on the promoter of the PITX2's transcriptional target. Taken together, our data provide new mechanistic perspectives on PITX2's functions and its related biological processes.


Asunto(s)
Proteínas Portadoras/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Proteínas de Homeodominio/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Proteínas Portadoras/análisis , Proteínas de Unión al ADN/metabolismo , Células HEK293 , N-Metiltransferasa de Histona-Lisina/análisis , Histonas/metabolismo , Proteínas de Homeodominio/análisis , Humanos , Proteínas Nucleares/análisis , Regiones Promotoras Genéticas , Mapas de Interacción de Proteínas , Factores de Transcripción/análisis , Proteína del Homeodomínio PITX2
10.
Blood ; 120(8): e17-27, 2012 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-22802335

RESUMEN

We recently generated 2 phenotypically similar Hoxa9+Meis1 overexpressing acute myeloid leukemias that differ by their in vivo biologic behavior. The first leukemia, named FLA2, shows a high frequency of leukemia stem cells (LSCs; 1 in 1.4 cells), whereas the second, FLB1, is more typical with a frequency of LSCs in the range of 1 per several hundred cells. To gain insights into possible mechanisms that determine LSC self-renewal, we profiled and compared the abundance of nuclear and cytoplasmic proteins and phosphoproteins from these leukemias using quantitative proteomics. These analyses revealed differences in proteins associated with stem cell fate, including a hyperactive p38 MAP kinase in FLB1 and a differentially localized Polycomb group protein Ezh2, which is mostly nuclear in FLA2 and predominantly cytoplasmic in FLB1. Together, these newly documented proteomes and phosphoproteomes represent a unique resource with more than 440 differentially expressed proteins and 11 543 unique phosphopeptides, of which 80% are novel and 7% preferentially phosphorylated in the stem cell-enriched leukemia.


Asunto(s)
Leucemia Mieloide Aguda/metabolismo , Células Madre Neoplásicas/metabolismo , Proteoma/análisis , Proteoma/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas de Unión al ADN/análisis , Proteínas de Unión al ADN/metabolismo , Proteína Potenciadora del Homólogo Zeste 2 , Activación Enzimática , N-Metiltransferasa de Histona-Lisina/análisis , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Ratones , Datos de Secuencia Molecular , Fosforilación , Complejo Represivo Polycomb 2 , Proteínas del Grupo Polycomb , Mapas de Interacción de Proteínas , Procesamiento Proteico-Postraduccional , Proteínas Represoras/análisis , Proteínas Represoras/metabolismo , Factores de Transcripción/análisis , Factores de Transcripción/metabolismo , Células Tumorales Cultivadas , Proteínas Quinasas p38 Activadas por Mitógenos/análisis , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
Anal Biochem ; 467: 14-21, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25172130

RESUMEN

S-Adenosyl-l-methionine (SAM) is recognized as an important cofactor in a variety of biochemical reactions. As more proteins and pathways that require SAM are discovered, it is important to establish a method to quickly identify and characterize SAM binding proteins. The affinity of S-adenosyl-l-homocysteine (SAH) for SAM binding proteins was used to design two SAH-derived capture compounds (CCs). We demonstrate interactions of the proteins COMT and SAHH with SAH-CC with biotin used in conjunction with streptavidin-horseradish peroxidase. After demonstrating SAH-dependent photo-crosslinking of the CC to these proteins, we used a CC labeled with a fluorescein tag to measure binding affinity via fluorescence anisotropy. We then used this approach to show and characterize binding of SAM to the PR domain of PRDM2, a lysine methyltransferase with putative tumor suppressor activity. We calculated the Kd values for COMT, SAHH, and PRDM2 (24.1 ± 2.2 µM, 6.0 ± 2.9 µM, and 10.06 ± 2.87 µM, respectively) and found them to be close to previously established Kd values of other SAM binding proteins. Here, we present new methods to discover and characterize SAM and SAH binding proteins using fluorescent CCs.


Asunto(s)
Catecol O-Metiltransferasa/análisis , Proteínas de Unión al ADN/análisis , Polarización de Fluorescencia/métodos , N-Metiltransferasa de Histona-Lisina/análisis , Hidrolasas/análisis , Proteínas Nucleares/análisis , S-Adenosilhomocisteína/metabolismo , S-Adenosilmetionina/metabolismo , Factores de Transcripción/análisis , Catecol O-Metiltransferasa/metabolismo , Proteínas de Unión al ADN/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Hidrolasas/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo
12.
Anal Biochem ; 443(2): 214-21, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24018340

RESUMEN

Epigenetic modifications of the genome, such as DNA methylation and posttranslational modifications of histone proteins, contribute to gene regulation. Growing evidence suggests that histone methyltransferases are associated with the development of various human diseases, including cancer, and are promising drug targets. High-quality generic assays will facilitate drug discovery efforts in this area. In this article, we present a liquid chromatography/mass spectrometry (LC/MS)-based S-adenosyl homocysteine (SAH) detection assay for histone methyltransferases (HMTs) and its applications in HMT drug discovery, including analyzing the activity of newly produced enzymes, developing and optimizing assays, performing focused compound library screens and orthogonal assays for hit confirmations, selectivity profiling against a panel of HMTs, and studying mode of action of select hits. This LC/MS-based generic assay has become a critical platform for our methyltransferase drug discovery efforts.


Asunto(s)
Cromatografía Liquida/métodos , Descubrimiento de Drogas/métodos , N-Metiltransferasa de Histona-Lisina/análisis , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Pruebas de Enzimas/métodos , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , S-Adenosilhomocisteína/análisis , S-Adenosilhomocisteína/metabolismo
13.
J Surg Oncol ; 107(4): 428-32, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22886632

RESUMEN

BACKGROUND AND OBJECTIVES: Endometrial cancer is a common gynecologic malignancy. It has been reported that overexpression of multiple myeloma SET (MMSET) promoted cellular adhesion, clonogenic growth, and tumorigenicity in other carcinomas. Therefore, the authors expected to investigate whether MMSET overexpression is an independent prognostic marker in endometrial cancer. METHODS: Immunohistochemistry was performed to examine the expression of MMSET in 161 endometrial cancer specimens and 62 normal endometrium specimens. The correlation of MMSET expression with clinicopathological parameters was assessed using χ(2) analysis. Patients' survival was analyzed using the Kaplan-Meier and log-rank tests. Cox regression was used for the multivariate analysis of prognostic factors. RESULTS: MMSET immunoreactivity was overexpressed in endometrial cancer compared with normal endometrium (P < 0.001). Moreover, MMSET expression was correlated with FIGO stage, histological grade, depth of myometrial invasion, lymph node metastasis, and vascular/lymphatic invasion. Furthermore, Patients with positive MMSET expression had significantly poorer overall survival and disease-free survival compared with patients with negative expression of MMSET (both P < 0.001, log-rank test). Multivariate Cox regression analysis revealed that positive MMSET expression was an independent prognostic factor for both OS and DFS of patients with endometrial cancer (P = 0.008 and P = 0.048, respectively). CONCLUSIONS: Overexpression of MMSET may contribute to the progression of endometrial cancer and also may serve as a new biomarker to predict the prognosis of endometrial cancer.


Asunto(s)
Biomarcadores de Tumor/análisis , Neoplasias Endometriales/química , Neoplasias Endometriales/patología , N-Metiltransferasa de Histona-Lisina/análisis , Proteínas Represoras/análisis , Adulto , Anciano , Supervivencia sin Enfermedad , Neoplasias Endometriales/mortalidad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Metástasis Linfática , Persona de Mediana Edad , Análisis Multivariante , Clasificación del Tumor , Invasividad Neoplásica , Estadificación de Neoplasias , Pronóstico , Modelos de Riesgos Proporcionales , Factores de Riesgo , Regulación hacia Arriba
14.
Nucleic Acids Res ; 38(21): 7500-12, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20671031

RESUMEN

Polycomb group (PcG) proteins are key regulators of stem-cell and cancer biology. They mainly act as repressors of differentiation and tumor-suppressor genes. One key silencing step involves the trimethylation of histone H3 on Lys27 (H3K27) by EZH2, a core component of the Polycomb Repressive Complex 2 (PRC2). The mechanism underlying the initial recruitment of mammalian PRC2 complexes is not well understood. Here, we show that NIPP1, a regulator of protein Ser/Thr phosphatase-1 (PP1), forms a complex with PP1 and PRC2 components on chromatin. The knockdown of NIPP1 or PP1 reduced the association of EZH2 with a subset of its target genes, whereas the overexpression of NIPP1 resulted in a retargeting of EZH2 from fully repressed to partially active PcG targets. However, the expression of a PP1-binding mutant of NIPP1 (NIPP1m) did not cause a redistribution of EZH2. Moreover, mapping of the chromatin binding sites with the DamID technique revealed that NIPP1 was associated with multiple PcG target genes, including the Homeobox A cluster, whereas NIPP1m showed a deficient binding at these loci. We propose that NIPP1 associates with a subset of PcG targets in a PP1-dependent manner and thereby contributes to the recruitment of the PRC2 complex.


Asunto(s)
Cromatina/metabolismo , Proteínas de Unión al ADN/análisis , Endorribonucleasas/metabolismo , N-Metiltransferasa de Histona-Lisina/análisis , Fosfoproteínas Fosfatasas/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/análisis , Sitios de Unión , Línea Celular , Cromatina/química , Cromatina/enzimología , Endorribonucleasas/análisis , Endorribonucleasas/antagonistas & inhibidores , Proteína Potenciadora del Homólogo Zeste 2 , Histona Metiltransferasas , Humanos , Fosfoproteínas Fosfatasas/análisis , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Complejo Represivo Polycomb 2 , Proteínas del Grupo Polycomb , Regiones Promotoras Genéticas , Proteína Fosfatasa 1/antagonistas & inhibidores , Proteína Fosfatasa 1/metabolismo , Proteína Fosfatasa 1/fisiología , Interferencia de ARN , Proteínas de Unión al ARN/análisis , Proteínas de Unión al ARN/antagonistas & inhibidores
15.
Cancer Sci ; 102(1): 260-6, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21054678

RESUMEN

HTLV-1 Tax deregulates signal transduction pathways, transcription of genes, and cell cycle regulation of host cells, which is mainly mediated by its protein-protein interactions with host cellular factors. We previously reported an interaction of Tax with a histone methyltransferase (HMTase), SUV39H1. As the interaction was mediated by the SUV39H1 SET domain that is shared among HMTases, we examined the possibility of Tax interaction with another HMTase, SMYD3, which methylates histone H3 lysine 4 and activates transcription of genes, and studied the functional effects. Expression of endogenous SMYD3 in T cell lines and primary T cells was confirmed by immunoblotting analysis. Co-immuno-precipitaion assays and in vitro pull-down assay indicated interaction between Tax and SMYD3. The interaction was largely dependent on the C-terminal 180 amino acids of SMYD3, whereas the interacting domain of Tax was not clearly defined, although the N-terminal 108 amino acids were dispensable for the interaction. In the cotransfected cells, colocalization of Tax and SMYD3 was indicated in the cytoplasm or nuclei. Studies using mutants of Tax and SMYD3 suggested that SMYD3 dominates the subcellular localization of Tax. Reporter gene assays showed that nuclear factor-κB activation promoted by cytoplasmic Tax was enhanced by the presence of SMYD3, and attenuated by shRNA-mediated knockdown of SMYD3, suggesting an increased level of Tax localization in the cytoplasm by SMYD3. Our study revealed for the first time Tax-SMYD3 direct interaction, as well as apparent tethering of Tax by SMYD3, influencing the subcellular localization of Tax. Results suggested that SMYD3-mediated nucleocytoplasmic shuttling of Tax provides one base for the pleiotropic effects of Tax, which are mediated by the interaction of cellular proteins localized in the cytoplasm or nucleus.


Asunto(s)
Productos del Gen tax/fisiología , N-Metiltransferasa de Histona-Lisina/fisiología , Transporte Activo de Núcleo Celular , Células Cultivadas , Productos del Gen tax/análisis , N-Metiltransferasa de Histona-Lisina/análisis , N-Metiltransferasa de Histona-Lisina/química , Humanos , FN-kappa B/metabolismo , Estructura Terciaria de Proteína
16.
Nucleic Acids Res ; 37(21): 7047-58, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19778927

RESUMEN

Dot1 is a conserved histone methyltransferase that methylates histone H3 on lysine 79. We previously observed that in Saccharomyces cerevisiae, a single DOT1 gene encodes two Dot1 protein species. Here, we show that the relative abundance of the two isoforms changed under nutrient-limiting conditions. A mutagenesis approach showed that the two Dot1 isoforms are produced from two alternative translation start sites as a result of leaky scanning by the ribosome. The leaky scanning was not affected by the 5'- or 3'-untranslated regions of DOT1, indicating that translation initiation is determined by the DOT1 coding sequence. Construction of yeast strains expressing either one of the isoforms showed that both were sufficient for Dot1's role in global H3K79 methylation and telomeric gene silencing. However, the absence of the long isoform of Dot1 altered the resistance of yeast cells to the chitin-binding drug Calcofluor White, suggesting that the two Dot1 isoforms have a differential function in cell wall biogenesis.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/genética , Proteínas Nucleares/genética , Iniciación de la Cadena Peptídica Traduccional , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Pared Celular/enzimología , N-Metiltransferasa de Histona-Lisina/análisis , N-Metiltransferasa de Histona-Lisina/metabolismo , Proteínas Nucleares/análisis , Proteínas Nucleares/metabolismo , Isoformas de Proteínas/análisis , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ribosomas/metabolismo , Saccharomyces cerevisiae/enzimología , Proteínas de Saccharomyces cerevisiae/análisis , Proteínas de Saccharomyces cerevisiae/metabolismo
17.
Hum Pathol ; 108: 1-11, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33121982

RESUMEN

Mesonephric carcinomas (MEs) and female adnexal tumors of probable Wolffian origin (FATWO) are derived from embryologic remnants of Wolffian/mesonephric ducts. Mesonephric-like carcinomas (MLCs) show identical morphology to ME of the cervix but occur in the uterus and ovary without convincing mesonephric remnants. ME, MLC, and FATWO are challenging to diagnose due to their morphologic similarities to Müllerian/paramesonephric tumors, contributing to a lack of evidence-based and tumor-specific treatments. We performed whole-proteomic analysis on 9 ME/MLC and 56 endometrial carcinomas (ECs) to identify potential diagnostic biomarkers. Although there were no convincing differences between ME and MLC, 543 proteins showed increased expression in ME/MLC relative to EC. From these proteins, euchromatic histone lysine methyltransferase 2 (EHMT2), glutathione S-transferase Mu 3 (GSTM3), eukaryotic translation elongation factor 1 alpha 2 (EEF1A2), and glycogen synthase kinase 3 beta were identified as putative biomarkers. Immunohistochemistry was performed on these candidates and GATA3 in 14 ME/MLC, 8 FATWO, 155 EC, and normal tissues. Of the candidates, only GATA3 and EHMT2 were highly expressed in mesonephric remnants and mesonephric-derived male tissues. GATA3 had the highest sensitivity and specificity for ME/MLC versus EC (93% and 99%) but was absent in FATWO. EHMT2 was 100% sensitive for ME/MLC & FATWO but was not specific (65%). Similarly, EEF1A2 was reasonably sensitive to ME/MLC (92%) and FATWO (88%) but was the least specific (38%). GSTM3 performed intermediately (sensitivity for ME/MLC and FATWO: 83% and 38%, respectively; specificity 67%). Although GATA3 remained the best diagnostic biomarker for ME/MLC, we have identified EHMT2, EEF1A2, and GSTM3 as proteins of interest in these cancers. FATWO's cell of origin is uncertain and remains an area for future research.


Asunto(s)
Biomarcadores de Tumor/análisis , Glutatión Transferasa/análisis , Antígenos de Histocompatibilidad/análisis , N-Metiltransferasa de Histona-Lisina/análisis , Mesonefroma/diagnóstico , Factor 1 de Elongación Peptídica/análisis , Femenino , Humanos , Proteómica/métodos
18.
Sci Rep ; 10(1): 20598, 2020 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-33244033

RESUMEN

Mechanical cues from the cellular microenvironment are converted into biochemical signals controlling diverse cell behaviours, including growth and differentiation. But it is still unclear how mechanotransduction ultimately affects nuclear readouts, genome function and transcriptional programs. Key signaling pathways and transcription factors can be activated, and can relocalize to the nucleus, upon mechanosensing. Here, we tested the hypothesis that epigenetic regulators, such as methyltransferase enzymes, might also contribute to mechanotransduction. We found that the SMYD3 lysine methyltransferase is spatially redistributed dependent on cell geometry (cell shape and aspect ratio) in murine myoblasts. Specifically, elongated rectangles were less permissive than square shapes to SMYD3 nuclear accumulation, via reduced nuclear import. Notably, SMYD3 has both nuclear and cytoplasmic substrates. The distribution of SMYD3 in response to cell geometry correlated with cytoplasmic and nuclear lysine tri-methylation (Kme3) levels, but not Kme2. Moreover, drugs targeting cytoskeletal acto-myosin induced nuclear accumulation of Smyd3. We also observed that square vs rectangular geometry impacted the nuclear-cytoplasmic relocalisation of several mechano-sensitive proteins, notably YAP/TAZ proteins and the SETDB1 methyltransferase. Thus, mechanical cues from cellular geometric shapes are transduced by a combination of transcription factors and epigenetic regulators shuttling between the cell nucleus and cytoplasm. A mechanosensitive epigenetic machinery could potentially affect differentiation programs and cellular memory.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/análisis , Mioblastos/citología , Animales , Línea Celular , Núcleo Celular/metabolismo , Forma de la Célula , Citoplasma/metabolismo , Citoesqueleto/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Ratones , Mioblastos/metabolismo , Transporte de Proteínas
19.
Int J Oncol ; 57(3): 756-766, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32705243

RESUMEN

SET and MYND domain­containing protein 3 (SMYD3) is a lysine methyltransferase, and its aberrant expression has been implicated in several malignancies. However, its clinical and biological roles in non­small cell lung cancer (NSCLC) remain unclear. In the present study, it was revealed that SMYD3 was significantly upregulated in NSCLC tissues, as compared with paired adjacent normal tissues. A high SMYD3 expression was associated with aggressive clinicopathological characteristics, as well as poor disease­free survival and overall survival (OS) in NSCLC patients. Multivariate analysis revealed that SMYD3 overexpression was an independent predictor of poor OS in NSCLC patients. In addition, SMYD3 knockdown inhibited cell proliferation, triggered apoptosis, and blocked migration and invasion in NSCLC cells in vitro, whereas stable SMYD3 overexpression promoted NSCLC cell proliferation. Furthermore, the SMYD3­silenced NSCLC cells became more sensitive, whereas the SMYD3­overexpressed NSCLC cells became more resistant to the apoptosis induced by cisplatin. Mechanistic analysis revealed that SMYD3 knockdown led to the upregulation of Bim, Bak and Bax, and the downregulation of Bcl­2, Bcl­xl, MMP­2 and MMP­9 in NSCLC cells. In combination, the present findings indicated that SMYD3 has oncogenic potential in the context of NSCLC, providing evidence that may be exploited for both prognostic and therapeutic purposes in the future.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Regulación Neoplásica de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/genética , Neoplasias Pulmonares/genética , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Apoptosis/genética , Carcinogénesis/genética , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Cisplatino/farmacología , Cisplatino/uso terapéutico , Resistencia a Antineoplásicos/genética , Femenino , Técnicas de Silenciamiento del Gen , N-Metiltransferasa de Histona-Lisina/análisis , Humanos , Estimación de Kaplan-Meier , Pulmón/patología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Invasividad Neoplásica/genética , Pronóstico , Activación Transcripcional , Regulación hacia Arriba
20.
Anal Chem ; 81(15): 6300-9, 2009 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20337398

RESUMEN

A novel method to improve the detection of protein ions using a linear ion trap mass spectrometer is presented. A scan function combining charge separation with segmented transmission of multiply charged ions was developed to enhance the sensitivity and resolution of the linear ion trap for the nanoLC-MS analysis of intact proteins. The analytical benefits of the present method are particularly apparent in protein analyses, where the increased proportion of multiply charged ions can exacerbate space-charge effects and compromise the dynamic range of the linear ion trap instrument. The enhanced ion storage and charge separation capabilities of our targeted and enhanced multiply charged scan mode provided a 4-fold increase in signal-to-noise and 5-fold increase in resolution, thus enabling the detection of closely related protein isoforms. The application of this method is demonstrated for low femtomole detection of protein standards and nuclear extracts enriched in histone proteins. The enhanced resolution of this scan mode also enabled us to monitor subtle changes in the methylation of a subpopulation of histone H3 that occurs in chicken DT40 cells lacking specific methyltransferase activity. The extent of the fold change and PTM site localization was performed using predictive software tools and targeted multiple reaction monitoring analysis of histone peptides. Monomethylation of Lys 79 in histone H3 (H3K79me1) was down regulated by 240-fold in methyltransferase deficient cells.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/análisis , Histonas/análisis , Lisina/análisis , Espectrometría de Masas , Animales , Células Cultivadas , Pollos , Cromatografía Líquida de Alta Presión , Histona Metiltransferasas , Metilación , Nanotecnología , Procesamiento Proteico-Postraduccional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA