Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Biol Reprod ; 106(1): 132-144, 2022 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-34849582

RESUMEN

The mechanisms by which sex is determined, and how a sexual phenotype is stably maintained during adulthood, have been the focus of vigorous scientific inquiry. Resources common to the biomedical field (automated staining and imaging platforms) were leveraged to provide the first immunofluorescent data for a reptile species with temperature induced sex reversal. Two four-plex immunofluorescent panels were explored across three sex classes (sex reversed ZZf females, normal ZWf females, and normal ZZm males). One panel was stained for chromatin remodeling genes JARID2 and KDM6B, and methylation marks H3K27me3, and H3K4me3 (Jumonji Panel). The other CaRe panel stained for environmental response genes CIRBP and RelA, and H3K27me3 and H3K4me3. Our study characterized tissue specific expression and cellular localization patterns of these proteins and histone marks, providing new insights to the molecular characteristics of adult gonads in a dragon lizard Pogona vitticeps. The confirmation that mammalian antibodies cross react in P. vitticeps paves the way for experiments that can take advantage of this new immunohistochemical resource to gain a new understanding of the role of these proteins during embryonic development, and most importantly for P. vitticeps, the molecular underpinnings of sex reversal.


Asunto(s)
Epigénesis Genética/fisiología , Lagartos/fisiología , Procesos de Determinación del Sexo/fisiología , Temperatura , Animales , Ensamble y Desensamble de Cromatina/genética , Femenino , Gónadas/química , Histonas/análisis , Inmunohistoquímica/métodos , Inmunohistoquímica/veterinaria , Histona Demetilasas con Dominio de Jumonji/análisis , Lagartos/genética , Masculino , Metilación , Proteínas de Unión al ARN/análisis , Procesos de Determinación del Sexo/genética
2.
J Neuropathol Exp Neurol ; 81(1): 54-60, 2022 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-34875075

RESUMEN

Patients with IDH-wildtype glioblastoma (GBM) generally have a poor prognosis. However, there is an increasing need of novel robust biomarkers in the daily clinico-pathological setting to identify and support treatment in patients who become long-time survivors. Jumonji domain-containing protein 6 (JMJD6) is involved in epigenetic regulation of demethylation of histones and has been associated with GBM aggressiveness. We investigated the expression and prognostic potential of JMJD6 tumor fraction score in 184 IDH-wildtype GBMs. Whole-slides were double-stained with an antibody against JMJD6 and an exclusion-cocktail consisting of 4 antibodies (CD31, SMA, CD45, and Iba-1), enabling evaluation of tumor cells only. Stainings were quantified with a combined software- and scoring-based approach. For comparison, IDH-mutated WHO grade II, III and IV astrocytic gliomas were also stained, and the JMJD6 tumor fraction score increased with increasing WHO grade, although not significantly. In multivariate analysis including age, gender, performance status and post-surgical treatment high JMJD6 tumor fraction score was associated with longer overall survival in IDH-wildtype GBMs (p = 0.03), but the effect disappeared when MGMT promoter status was included (p = 0.34). We conclude that JMJD6 is highly expressed in IDH-wildtype GBM but it has no independent prognostic value.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/patología , Glioblastoma/patología , Histona Demetilasas con Dominio de Jumonji/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/análisis , Neoplasias Encefálicas/genética , Femenino , Glioblastoma/genética , Humanos , Isocitrato Deshidrogenasa/genética , Histona Demetilasas con Dominio de Jumonji/análisis , Masculino , Persona de Mediana Edad , Pronóstico
3.
J Neuropathol Exp Neurol ; 79(7): 754-762, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32447376

RESUMEN

The diagnosis of anaplastic meningioma (AM) (WHO grade III) is based on the presence of a high mitotic index (MI) and/or overt anaplasia. Only few data exist about the reproducibility and prognostic value of overt anaplasia. Additionally, the prognostic value of H3K27me3 loss in AM has not yet been demonstrated. Our objectives were to evaluate the reproducibility and prognostic value of WHO criteria and H3K27me3 loss in a multicenter series of 66 AM. Interobserver reproducibility was good for the determination of WHO grade (Kappa = 0.671) and MI (intraclass correlation coefficient [ICC] = 0.649), and fair for assessment of overt anaplasia (Kappa = 0.366). Patients with meningiomas showing high MI had significantly shorter overall survival (OS) than patients with meningiomas showing overt anaplasia without high MI (p = 0.009). OS was significantly lower in case of overt anaplasia with low MI (<20/1.6 mm2) than in atypical meningiomas (p = 0.008). H3K27me3 loss was present in 10/47 (21%) of AM and independently associated with shorter OS (p = 0.036; Cox multivariate analysis), with a good reproducibility (Kappa = 0.643). In conclusion, the presence of overt anaplasia could give additional prognostic information in tumors lacking high MI. Finally, loss of H3K27me3 is an easy-to-use and reproducible marker of poorer prognosis.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Neoplasias Meníngeas/metabolismo , Neoplasias Meníngeas/patología , Meningioma/metabolismo , Meningioma/patología , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/análisis , Femenino , Estudios de Seguimiento , Humanos , Histona Demetilasas con Dominio de Jumonji/análisis , Masculino , Neoplasias Meníngeas/diagnóstico , Meningioma/diagnóstico , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos , Tasa de Supervivencia/tendencias , Adulto Joven
4.
Inflammation ; 42(4): 1389-1400, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31041569

RESUMEN

Chronic nasal sinusitis with nasal polyps (CRSwNP) is a reversible nasal mucosal remodeling disease caused by persistent inflammation and structural changes in chronic nasal mucosa. Although there have been many studies on the inflammation of the nasal mucosa epithelium, the mechanism remains unclear. Our study found that H3K4me3 histone demethylase KDM2B (also known as JHDM1B) and transcriptional regulator Brg1 (also called SNF2-ß or Smarca4) were significantly decreased in nasal mucosa of CRSwNP patients, and they were positively correlated. Brg1 and KDM2B co-localize in the epithelial cells of nasal mucosa. We used poly(I:C)-treated nasal mucosal epithelial cells (HNECs) to find that the expression of KDM2B and Brg1 was also decreased, and the main expression position transferred from the nucleus to the nuclear membrane. We used small interfering RNA to knock down the expression of KDM2B and Brg1 in nasal epithelial cells. It was interesting to find that the decreased expression of KDM2B and Brg1 produced similar effects to that of poly(I:C)-treated cells, which could promote inflammatory response of nasal mucosal epithelial cells. And Brg1 appears to play a role in KDM2B regulating gene promoters of IL-6 and TNF-α inflammatory. This study shows that KDM2B and Brg1 may have an inhibitory effect on the development of CRSwNP nasal mucosal epithelial inflammation. This study will provide a new perspective for gene targeting therapy of CRSwNPs.


Asunto(s)
ADN Helicasas/fisiología , Proteínas F-Box/fisiología , Inflamación/patología , Histona Demetilasas con Dominio de Jumonji/fisiología , Mucosa Nasal/patología , Pólipos Nasales/etiología , Proteínas Nucleares/fisiología , Sinusitis/complicaciones , Factores de Transcripción/fisiología , Enfermedad Crónica , ADN Helicasas/análisis , Células Epiteliales/química , Células Epiteliales/patología , Proteínas F-Box/análisis , Regulación de la Expresión Génica , Humanos , Histona Demetilasas con Dominio de Jumonji/análisis , Mucosa Nasal/química , Proteínas Nucleares/análisis , Factores de Transcripción/análisis
5.
Biochem Biophys Res Commun ; 508(2): 576-582, 2019 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-30514438

RESUMEN

Hepatocellular carcinoma (HCC) is the fifth most common cancer and the third most lethal cancer worldwide. Although gene mutations associated with HCC development have been intensively studied, how epigenetic factors specifically modulate the functional properties of HCC by regulating target gene expression is unclear. Here we demonstrated the overexpression of KDM3B in liver tissue of HCC patients using public RNA-seq data. Ablation of KDM3B by CRISPR/Cas9 retarded the cell cycle and proliferation of hepatocarcinoma HepG2 cells. Approximately 30% of KDM3B knockout cells exhibited mitotic spindle multipolarity as a chromosome instability (CIN) phenotype. RNA-seq analysis of KDM3B knockout revealed significantly down-regulated expression of cell cycle related genes, especially cell proliferation factor CDC123. Furthermore, the expression level of Cyclin D1 was reduced in KDM3B knockout by proteosomal degradation without any change in the expression of CCND1, which encodes Cyclin D1. The results implicate KDM3B as a crucial epigenetic factor in cell cycle regulation that manipulates chromatin dynamics and transcription in HCC, and identifies a potential gene therapy target for effective treatment of HCC.


Asunto(s)
Carcinoma Hepatocelular/genética , Redes Reguladoras de Genes , Genes cdc/genética , Histona Demetilasas con Dominio de Jumonji/fisiología , Neoplasias Hepáticas/genética , Carcinoma Hepatocelular/metabolismo , Proteínas de Ciclo Celular/metabolismo , Ciclina D1/metabolismo , Regulación Neoplásica de la Expresión Génica , Células Hep G2 , Humanos , Histona Demetilasas con Dominio de Jumonji/análisis , Histona Demetilasas con Dominio de Jumonji/genética , Hígado/metabolismo , Neoplasias Hepáticas/metabolismo , Transcripción Genética
6.
Biochem Biophys Res Commun ; 485(1): 62-68, 2017 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-28189690

RESUMEN

Inflammatory mediators and adhesion molecules have been implicated in a variety of diseases including atherosclerosis. As both the mediator-releasing and targeted cells, vascular endothelial cells play key role in pathological processes. NF-κB signaling regulates a cluster of inflammatory factors in LPS-activated vascular endothelial cells but the underlying mechanisms remain largely unknown. Here, we investigated the epigenetic regulation of LPS upon the expression of inflammatory mediators and adhesion molecules. We found that LPS treatment promoted jmjd3 expression, enhanced Jmjd3 nuclear accumulation in human vascular endothelial cells. In addition, LPS enhanced the demethylation of H3K27me3, a specific substrate of Jmjd3. LPS treatment recruited Jmjd3 and NF-κB to the promoter region of target genes, suggesting Jmjd3 synergizes with NF-κB to activate the expression of target genes. We further found that Jmjd3 attenuated the methylation status in promoter region of target genes, culminating in target gene expression. Our findings unveil epigenetic regulations of LPS upon NF-κB pathway and identify Jmjd3 as a critical modulator of NF-κB pathway and potential therapeutic target for NF-κB related diseases including atherosclerosis.


Asunto(s)
Células Endoteliales/inmunología , Epigénesis Genética , Histona Demetilasas con Dominio de Jumonji/genética , Lipopolisacáridos/inmunología , FN-kappa B/inmunología , Regulación hacia Arriba , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/inmunología , Citocinas/genética , Citocinas/inmunología , Células Endoteliales/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inflamación/genética , Inflamación/inmunología , Histona Demetilasas con Dominio de Jumonji/análisis , Histona Demetilasas con Dominio de Jumonji/inmunología , Regiones Promotoras Genéticas
7.
Tumour Biol ; 37(9): 12535-12546, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27350366

RESUMEN

The role and clinical implication of ZRF1 in breast cancer are poorly understood. So this study is aimed to explore the role of ZRF1 in breast cancer progression. With this context, we first assessed its expression pattern in FFPE primary and metastasis breast tissue samples as well as from publicly available databases. Moreover, we also explored the survival status of patients from the publicly available database and interestingly discover that high expression of ZRF1 decreases the survival of estrogen-positive breast cancer patients more than estrogen-negative status patients. In the perspective of this, we evaluated the role ZRF1 in MCF-7 breast cancer cells and found that it's silencing by knockdown results in decreased cell proliferation as well as cell viability. Results also show that expression of ZRF1 is down regulated in the presence of estrogen-depleted conditions but independent of RAS/MEK as well as AKT axes. Moreover, the decrease in viability of MCF-7 cells was accompanied by induction of apoptosis and DNA damage, well-marked with upregulation of cleaved PARP and downregulation of BCL2 and H2AUbK119 levels. Furthermore, we also explored that knockdown of ZRF1 sensitises the effect of curcumin, observed with decrease in cell viability and dropping of IC50 value from 25 to 15 µM. This investigation thus shed a new light on the role on ZRF1 in breast cancer cells and hence can be exploited to design better therapeutic intervention.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Curcumina/farmacología , Proteínas de Unión al ADN/fisiología , Proteínas Oncogénicas/fisiología , Receptores de Estrógenos/análisis , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/química , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Movimiento Celular , Proliferación Celular , Proteínas de Unión al ADN/genética , Femenino , Humanos , Histona Demetilasas con Dominio de Jumonji/análisis , Células MCF-7 , Chaperonas Moleculares , Proteínas Oncogénicas/genética , Proteínas Proto-Oncogénicas c-akt/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/análisis , Proteínas de Unión al ARN
8.
Am J Surg Pathol ; 40(4): 479-89, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26645727

RESUMEN

Most malignant peripheral nerve sheath tumors (MPNSTs) exhibit combined inactivation of NF1, CDKN2A, and polycomb repressive complex 2 component genes (Embryonic Ectoderm Development [EED] and Suppressor of Zeste 12 [SUZ12]). Mutations in EED and SUZ12 induce loss of trimethylation at lysine 27 of histone 3 (H3K27me3), with subsequent aberrant transcriptional activation of polycomb repressive complex 2-repressed homeobox master regulators. These findings prompted us to investigate the performance of an anti-H3K27me3 monoclonal antibody clone C36B11 as an immunohistochemical marker for MPNSTs. We assessed the C36B11 reactivity pattern in a pathologically and genetically well-characterized cohort of 68 MPNSTs, spanning various clinical presentations, such as type 1 neurofibromatosis (NF1), radiotherapy, and sporadic MPNSTs. We found that 69% (n=47) of all MPNSTs demonstrated loss of H3K27me3 expression, with 42 (61%) showing complete loss and 5 (7%) showing partial loss, whereas 31% (n=21) retained H3K27me3 expression. Among the NF1-related high-grade MPNSTs, 60% demonstrated loss of expression. In contrast, the majority of both sporadic (95%) and radiotherapy-related (91%) MPNSTs showed loss of H3K27me3 expression. Two of the 3 low-grade MPNSTs and all neurofibromas showed retained expression. Furthermore, all 5 epithelioid MPNSTs retained H3K27me3 labeling. The specificity of H3K27me3 loss as a marker for MPNSTs was studied by testing a large spectrum of lesions included in MPNST differential diagnosis, such as spindle/desmoplastic melanomas, synovial sarcomas, myoepithelial tumors, and other mesenchymal neoplasms, all of which retained expression of H3K27me3. We conclude that immunohistochemical analysis of H3K27me3 has good sensitivity and robust specificity for the diagnosis of MPNST, particularly outside of NF1 clinical history, which represents the most challenging diagnostic setting.


Asunto(s)
Biomarcadores de Tumor/análisis , Histona Demetilasas con Dominio de Jumonji/análisis , Neurilemoma/diagnóstico , Adolescente , Adulto , Anciano , Anticuerpos Monoclonales , Niño , Preescolar , Metilación de ADN , Femenino , Histonas , Humanos , Masculino , Persona de Mediana Edad , Sensibilidad y Especificidad , Análisis de Matrices Tisulares , Adulto Joven
9.
Oncotarget ; 6(14): 12723-39, 2015 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-25909289

RESUMEN

JARID1B is a member of the family of JmjC domain-containing proteins that removes methyl residues from methylated lysine 4 on histone H3 lysine 4 (H3K4). JARID1B has been proposed as an oncogene in many types of tumors; however, its role and underlying mechanisms in hepatocellular carcinoma (HCC) remain unknown. Here we show that JARID1B is elevated in HCC and its expression level is positively correlated with metastasis. In addition Kaplan-Meier survival analysis showed that high expression of JARID1B was associated with decreased overall survival of HCC patients. Overexpression of JARID1B in HCC cells increased proliferation, epithelial-mesenchymal transition, migration and invasion in vitro, and enhanced tumorigenic and metastatic capacities in vivo. In contrast, silencing JARID1B in aggressive and invasive HCC cells inhibited these processes. Mechanistically, we found JARID1B exerts its function through modulation of H3K4me3 at the PTEN gene promoter, which was associated with inactive PTEN transcription. PTEN overexpression blocked JARID1B-driven proliferation, EMT, and metastasis. Our results, for the first time, portray a pivotal role of JARID1B in stimulating metastatic behaviors of HCC cells. Targeting JARID1B may thus be a useful strategy to impede HCC cell invasion and metastasis.


Asunto(s)
Carcinoma Hepatocelular/patología , Transición Epitelial-Mesenquimal/genética , Histona Demetilasas con Dominio de Jumonji/biosíntesis , Neoplasias Hepáticas/patología , Invasividad Neoplásica/genética , Proteínas Nucleares/biosíntesis , Proteínas Represoras/biosíntesis , Adulto , Animales , Biomarcadores de Tumor/análisis , Western Blotting , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/mortalidad , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Femenino , Xenoinjertos , Humanos , Inmunohistoquímica , Histona Demetilasas con Dominio de Jumonji/análisis , Estimación de Kaplan-Meier , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/mortalidad , Masculino , Ratones , Microscopía Confocal , Persona de Mediana Edad , Invasividad Neoplásica/patología , Proteínas Nucleares/análisis , Fosfohidrolasa PTEN/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Represoras/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/fisiología , Transcriptoma
10.
Acta Med Okayama ; 69(1): 17-27, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25703167

RESUMEN

Sentinel lymph node (SLN) biopsies have widely been used for the detection of occult LN metastasis of malignant melanoma (MM). In addition to conventional biomarkers, we assessed the diagnostic and prognostic significance of melanoma-initiating cell (MIC) markers in SLNs of MM. We examined the expressions of gp100, MART-1 and tyrosinase mRNA for routine diagnosis and those of ABCB5, CD133, nestin, KDM5B, NGFR and RANK mRNA as MIC markers. The presence of micrometastasis was confirmed immunohistochemically using antibodies to S-100, HMB-45, MART-1, and tyrosinase. Discordance between immunohistochemical and molecular data was observed in 14 of 70 (20.0%) patients, among whom five (7.1%) were positive for only molecular markers;two of these five patients tested positive for micrometastasis by repeated immunohistochemical stainings. The quantitative expression levels of gp100, MART-1, and tyrosinase mRNA were significantly higher in the metastatic LNs;the cut-off values remain to be elucidated. ABCB5 mRNA expression was detected more frequently in the metastatic SLNs (p<0.05) and in the group of patients with recurrence. To make a definite diagnosis of metastasis, we still need a combination of immunohistochemical and molecular probes. ABCB5 might be a suitable molecular marker for the detection of melanoma-initiating cells in SLNs.


Asunto(s)
Melanoma/patología , Células Madre Neoplásicas/metabolismo , Biopsia del Ganglio Linfático Centinela , Subfamilia B de Transportador de Casetes de Unión a ATP , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/análisis , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Anciano , Anciano de 80 o más Años , Biomarcadores , Femenino , Humanos , Inmunohistoquímica , Histona Demetilasas con Dominio de Jumonji/análisis , Histona Demetilasas con Dominio de Jumonji/genética , Antígeno MART-1/genética , Masculino , Persona de Mediana Edad , Proteínas Nucleares/análisis , Proteínas Nucleares/genética , ARN Mensajero/análisis , Proteínas Represoras/análisis , Proteínas Represoras/genética
11.
Int J Clin Exp Pathol ; 8(10): 12922-8, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26722485

RESUMEN

KDM4A, KDM4B and KDM4D are lysine demethylases which demethylate H3 at lysine K9 and K36 sites, additionally KDM4D also the H1.4 linker histone at K26 lysine. Lysine methylation changes can repress or induce gene expression at specific sites thus influencing cellular functions. We analysed the immunohistochemical expression of KDM4A, KDM4B and KDM4D in a clinical material of 188 patients with lung carcinomas. There were 132 (70%) squamous cell carcinomas, 53 (28%) adenocarcinomas and 3 (2%) large cell carcinomas in the study. Additionally, the trimethylated state of chromatin was detected with an antibody to trimethylated H3K9 residue. Nuclear KDM4A and KDM4D were associated with the presence of lymph node metastases in tumors. Cytoplasmic KDM4A was associated with poor survival of the patients (P = 0.015) and with a shorter recurrence free interval (P = 0.028). KDM4A and KDM4D appear to have a significant role in the metastatic spread of lung carcinomas. The findings are also in line with their proposed involvement in mechanisms associated with cell proliferation, apoptosis and DNA repair.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , Histona Demetilasas con Dominio de Jumonji/biosíntesis , Neoplasias Pulmonares/patología , Anciano , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Metilación de ADN/fisiología , Femenino , Humanos , Inmunohistoquímica , Histona Demetilasas con Dominio de Jumonji/análisis , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/mortalidad , Masculino , Persona de Mediana Edad , Análisis de Supervivencia
12.
Int J Oral Sci ; 6(4): 205-11, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25394593

RESUMEN

Histone methylation is one of the most widely studied post-transcriptional modifications. It is thought to be an important epigenetic event that is closely associated with cell fate determination and differentiation. To explore the spatiotemporal expression of histone H3 lysine 4 trimethylation (H3K4me3) and histone H3 lysine 27 trimethylation (H3K27me3) epigenetic marks and methylation or demethylation transferases in tooth organ development, we measured the expression of SET7, EZH2, KDM5B and JMJD3 via immunohistochemistry and quantitative polymerase chain reaction (qPCR) analysis in the first molar of BALB/c mice embryos at E13.5, E15.5, E17.5, P0 and P3, respectively. We also measured the expression of H3K4me3 and H3K27me3 with immunofluorescence staining. During murine tooth germ development, methylation or demethylation transferases were expressed in a spatial-temporal manner. The bivalent modification characterized by H3K4me3 and H3K27me3 can be found during the tooth germ development, as shown by immunofluorescence. The expression of SET7, EZH2 as methylation transferases and KDM5B and JMJD3 as demethylation transferases indicated accordingly with the expression of H3K4me3 and H3K27me3 respectively to some extent. The bivalent histone may play a critical role in tooth organ development via the regulation of cell differentiation.


Asunto(s)
Histonas/metabolismo , Odontogénesis/fisiología , Procesamiento Proteico-Postraduccional/fisiología , Animales , Diferenciación Celular/fisiología , Proteínas de Unión al ADN/análisis , Papila Dental/embriología , Embrión de Mamíferos , Órgano del Esmalte/embriología , Proteína Potenciadora del Homólogo Zeste 2 , Epigénesis Genética/fisiología , Regulación del Desarrollo de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/análisis , Histona Demetilasas con Dominio de Jumonji/análisis , Lisina/metabolismo , Metilación , Ratones , Ratones Endogámicos BALB C , Complejo Represivo Polycomb 2/análisis , Germen Dentario/embriología
13.
J Endod ; 40(8): 1132-8, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25069920

RESUMEN

INTRODUCTION: Dental pulp has limited capability to regenerate, which happens in the early stage of pulpitis. An ambiguous relationship exists; inflammation may impair or support pulp regeneration. Epigenetics, which is involved in cell proliferation and inflammation, could regulate human dental pulp cell (HDPCs) regeneration. The aim of this study was to determine the role of the epigenetic mark, enhancer of zeste homolog 2 (EZH2), in the inflammation, proliferation, and regeneration of dental pulp. We used trimethylated histone H3 lysine 27(H3K27me3) and its lysine demethylase 6B (KDM6B) to monitor functional effects of altered EZH2 levels. METHODS: We detected epigenetic marks (EZH2, H3K27me3, and KDM6B) in pulp tissue by immunohistochemistry and immunofluorescence. EZH2 levels in HDPCs in inflammatory responses or differentiation were analyzed by quantitative polymerase chain reaction and Western blot. Quantitative polymerase chain reaction was used to assess the effects of EZH2 inhibition on interleukins in HDPCs upon tumor necrosis factor alpha stimulation. Cell proliferation was tested by cell counting kit-8, cell cycle, and apoptosis analysis. HDPC differentiation was investigated by quantitative polymerase chain reaction, alkaline phosphatase activity, and oil red O staining. RESULTS: EZH2 and H3K27me3 were decreased, whereas KDM6B was increased in infected pulp tissue and cells, which were similar to HDPC differentiation. EZH2 inhibition suppressed IL-1b, IL-6, and IL-8 messenger RNA (mRNA) in HDPCs upon inflammatory stimuli and impeded HDPC proliferation by decreasing cell number, arresting cell cycle, and increasing apoptosis. Suppressed EZH2 impaired adipogenesis, peroxisome proliferator-activated receptor r (PPAR-r), and CCAAT-enhancer binding protein a (CEBP/a) mRNA in adipogenic induction while enhancing alkaline phosphatase activity, Osx, and bone sialoprotein (BSP) mRNA in mineralization induction of HDPCs. CONCLUSIONS: EZH2 inhibited HDPC osteogenic differentiation while enhancing inflammatory response and proliferation, suggesting its role in pulp inflammation, proliferation, and regeneration.


Asunto(s)
Pulpa Dental/fisiología , Complejo Represivo Polycomb 2/fisiología , Pulpitis/fisiopatología , Regeneración/fisiología , Adipogénesis/fisiología , Fosfatasa Alcalina/análisis , Apoptosis/fisiología , Proteína alfa Potenciadora de Unión a CCAAT/análisis , Calcificación Fisiológica/fisiología , Diferenciación Celular/fisiología , Proliferación Celular , Células Cultivadas , Pulpa Dental/citología , Pulpa Dental/efectos de los fármacos , Proteína Potenciadora del Homólogo Zeste 2 , Epigénesis Genética/fisiología , Histonas/análisis , Humanos , Sialoproteína de Unión a Integrina/análisis , Interleucina-1beta/análisis , Interleucina-6/análisis , Interleucina-8/análisis , Histona Demetilasas con Dominio de Jumonji/análisis , Osteogénesis/fisiología , Receptores Activados del Proliferador del Peroxisoma/análisis , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Factor de Transcripción Sp7 , Factores de Transcripción/análisis , Factor de Necrosis Tumoral alfa/farmacología
14.
Biochem Biophys Res Commun ; 449(1): 1-7, 2014 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-24802408

RESUMEN

A number of JmjC domain-containing histone demethylases have been identified and biochemically characterized in mammalian. JMJD2A is a transcriptional cofactor and enzyme that catalyzes demethylation of histone H3 lysines 9 and 36. Here in this study, we aim to explore the role of JMJD2A in human gastric cancer. Quantitative real-time PCR, Western blot and immunohistochemistry analyses reveal higher expression of JMJD2A in clinical gastric cancer tissues than that in normal gastric mucosa. JMJD2A expression is associated with tumor stage and nodal status, and high level of JMJD2A predicts poor overall and disease-free survival. Univariate and multivariate survival analyses demonstrate that JMJD2A could serve as an independent prognostic factor. Furthermore, we show that inhibition the expression of JMJD2A attenuates the growth and transformation of three lines of gastric cancer cells. Mechanically, JMJD2A knockdown induces apoptosis of gastric cancer cells by up-regulating the expression of pro-apoptotic proteins and by down-regulating anti-apoptotic protein. Finally, we show that JMJD2A level is correlated with the level of the pro-apoptotic microRNA miR-34a in gastric cancer tissues and JMJD2A represses the expression of miR-34a by decreasing its promoter activity. Those findings demonstrate that JMJD2A regulates gastric cancer growth and serves as an independent prognostic factor, and implicate that JMJD2A may be a promising target for intervention.


Asunto(s)
Biomarcadores de Tumor/análisis , Histona Demetilasas con Dominio de Jumonji/análisis , Neoplasias Gástricas/química , Anciano , Anciano de 80 o más Años , Animales , Proliferación Celular , China/epidemiología , Supervivencia sin Enfermedad , Femenino , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Metástasis Linfática , Masculino , Ratones , Persona de Mediana Edad , Estadificación de Neoplasias , Prevalencia , Pronóstico , Medición de Riesgo , Neoplasias Gástricas/genética , Neoplasias Gástricas/mortalidad , Neoplasias Gástricas/patología , Tasa de Supervivencia , Carga Tumoral , Células Tumorales Cultivadas
15.
Nucleic Acids Res ; 42(10): 6168-82, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24728997

RESUMEN

Various types of human cancers exhibit amplification or deletion of KDM4A-D members, which selectively demethylate H3K9 and H3K36, thus implicating their activity in promoting carcinogenesis. On this basis, it was hypothesized that dysregulated expression of KDM4A-D family promotes chromosomal instabilities by largely unknown mechanisms. Here, we show that unlike KDM4A-B, KDM4C is associated with chromatin during mitosis. This association is accompanied by a decrease in the mitotic levels of H3K9me3. We also show that the C-terminal region, containing the Tudor domains of KDM4C, is essential for its association with mitotic chromatin. More specifically, we show that R919 residue on the proximal Tudor domain of KDM4C is critical for its association with chromatin during mitosis. Interestingly, we demonstrate that depletion or overexpression of KDM4C, but not KDM4B, leads to over 3-fold increase in the frequency of abnormal mitotic cells showing either misaligned chromosomes at metaphase, anaphase-telophase lagging chromosomes or anaphase-telophase bridges. Furthermore, overexpression of KDM4C demethylase-dead mutant has no detectable effect on mitotic chromosome segregation. Altogether, our findings implicate KDM4C demethylase activity in regulating the fidelity of mitotic chromosome segregation by a yet unknown mechanism.


Asunto(s)
Cromatina/enzimología , Segregación Cromosómica , Histona Demetilasas con Dominio de Jumonji/metabolismo , Mitosis/genética , Línea Celular , Histona Demetilasas con Dominio de Jumonji/análisis , Histona Demetilasas con Dominio de Jumonji/química , Estructura Terciaria de Proteína
16.
Int J Cancer ; 132(6): 1249-59, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23180591

RESUMEN

Heterogeneity of tumor tissue has been accounted for in recent years by a hierarchy-based model in which cancer stem cells (CSCs) have the ability both to self-renew and to give rise to differentiated tumor cells and are responsible for the overall organization of a tumor. Research into CSCs has progressed rapidly and concomitantly with recent advances in the biology of normal tissue stem cells, resulting in the identification of CSCs in a wide range of human tumors. Studies of mouse models of human cancer have provided further insight into the characteristics of CSCs as well as a basis for the development of novel therapies targeted to these cells. However, recent studies have revealed complexities, such as plasticity of stem cell properties and clonal diversity of CSCs, in certain tumor types that have led to revision of the original CSC model. In this review, we summarize the history of the discovery and characterization of CSCs, as well as address recent advances that have revealed the complexity of these cells and their therapeutic implications.


Asunto(s)
Células Madre Neoplásicas/fisiología , Animales , Separación Celular , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Histona Demetilasas con Dominio de Jumonji/análisis , Neoplasias/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Proteínas Nucleares/análisis , Proteínas Represoras/análisis
17.
BMC Cancer ; 12: 516, 2012 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-23148692

RESUMEN

BACKGROUND: The histone demethylase GASC1 (JMJD2C) is an epigenetic factor suspected of involvement in development of different cancers, including breast cancer. It is thought to be overexpressed in the more aggressive breast cancer types based on mRNA expression studies on cell lines and meta analysis of human breast cancer sets. This study aimed to evaluate the prognostic and predictive value of GASC1 for women with invasive breast cancer. METHODS: All the 355 cases were selected from a cohort enrolled in the Kuopio Breast Cancer Project between April 1990 and December 1995. The expression of GASC1 was studied by immunohistochemistry (IHC) on tissue microarrays. Additionally relative GASC1 mRNA expression was measured from available 57 cases. RESULTS: In our material, 56% of the cases were GASC1 negative and 44% positive in IHC staining. Women with GASC1 negative tumors had two years shorter breast cancer specific survival and time to relapse than the women with GASC1 positive tumors (p=0.017 and p=0.034 respectively). The majority of GASC1 negative tumors were ductal cases (72%) of higher histological grade (84% of grade II and III altogether). When we evaluated estrogen receptor negative and progesterone receptor negative cases separately, there was 2 times more GASC1 negative than GASC1 positive tumors in each group (chi2, p= 0.033 and 0.001 respectively). In the HER2 positive cases, there was 3 times more GASC1 negative cases than GASC1 positives (chi2, p= 0.029). Patients treated with radiotherapy (n=206) and hormonal treatment (n=62) had better breast cancer specific survival, when they were GASC1 positive (Cox regression: HR=0.49, p=0.007 and HR=0.33, p=0.015, respectively). The expression of GASC1 mRNA was in agreement with the protein analysis. CONCLUSIONS: This study indicates that the GASC1 is both a prognostic and a predictive factor for women with invasive breast cancer. GASC1 negativity is associated with tumors of more aggressive histopathological types (ductal type, grade II and III, ER negative, PR negative). Patients with GASC1 positive tumors have better breast cancer specific survival and respond better to radiotherapy and hormonal treatment.


Asunto(s)
Biomarcadores de Tumor/análisis , Neoplasias de la Mama/enzimología , Carcinoma Ductal de Mama/enzimología , Histona Demetilasas con Dominio de Jumonji/biosíntesis , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/mortalidad , Carcinoma Ductal de Mama/patología , Supervivencia sin Enfermedad , Moduladores de los Receptores de Estrógeno/uso terapéutico , Femenino , Humanos , Inmunohistoquímica , Histona Demetilasas con Dominio de Jumonji/análisis , Estimación de Kaplan-Meier , Persona de Mediana Edad , Pronóstico , Modelos de Riesgos Proporcionales , ARN Mensajero/análisis , Radioterapia , Análisis de Matrices Tisulares
18.
Neoplasma ; 58(2): 153-7, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21275466

RESUMEN

BACKGROUND: Hypoxia-inducible factor 1α has been shown to play a central role in RCC tumorigenesis by acting as a transcription factor. Histone demethylase JMJD1A is an iron- and 2-oxoglutarate-dependent dioxygenase which catalyze the demethylation of mono- and dimethylated H3K9. JMJD1A can be upregulated by hypoxia via HIF-1 and associated with cancer. The expression of JMJD1A was determined in 10 kidney cancer tissue and adjacent tissue by quantitative polymerase chain reaction, western blotting and immunohistochemistry. Furthermore, the expression of JMJD1A was investigated in cell line 786-0 through adding nickle or cobalt ion to mimic hypoxic environment. The expression of JMJD1A was higher in cancer tissue than adjacent tissue, and in hypoxic environment than normal environment. In cancer tissue, the JMJD1A mainly located around blood vessels which indicated that JMJD1A is involved tumor angiogenesis. CONCLUSION: the increased expression of JMJD1A might be associated with the progression of kidney cancer. KEYWORDS: renal cell carcinoma, histone demethylase, JMJD1A, hypoxia-inducible factor, iron.


Asunto(s)
Carcinoma de Células Renales/enzimología , Histona Demetilasas con Dominio de Jumonji/fisiología , Neoplasias Renales/enzimología , Carcinoma de Células Renales/etiología , Hipoxia de la Célula , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Histona Demetilasas con Dominio de Jumonji/análisis , Histona Demetilasas con Dominio de Jumonji/genética , Neoplasias Renales/etiología , ARN Mensajero/análisis
19.
EMBO J ; 28(21): 3341-52, 2009 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-19779457

RESUMEN

Jmjd3, a JmjC family histone demethylase, is induced by the transcription factor NF-kB in response to microbial stimuli. Jmjd3 erases H3K27me3, a histone mark associated with transcriptional repression and involved in lineage determination. However, the specific contribution of Jmjd3 induction and H3K27me3 demethylation to inflammatory gene expression remains unknown. Using chromatin immunoprecipitation-sequencing we found that Jmjd3 is preferentially recruited to transcription start sites characterized by high levels of H3K4me3, a marker of gene activity, and RNA polymerase II (Pol_II). Moreover, 70% of lipopolysaccharide (LPS)-inducible genes were found to be Jmjd3 targets. Although most Jmjd3 target genes were unaffected by its deletion, a few hundred genes, including inducible inflammatory genes, showed moderately impaired Pol_II recruitment and transcription. Importantly, most Jmjd3 target genes were not associated with detectable levels of H3K27me3, and transcriptional effects of Jmjd3 absence in the window of time analysed were uncoupled from measurable effects on this histone mark. These data show that Jmjd3 fine-tunes the transcriptional output of LPS-activated macrophages in an H3K27 demethylation-independent manner.


Asunto(s)
Regulación de la Expresión Génica , Histona Demetilasas con Dominio de Jumonji/metabolismo , Lipopolisacáridos/inmunología , Activación de Macrófagos , Macrófagos/inmunología , Animales , Células Cultivadas , Femenino , Técnicas de Inactivación de Genes , Histona Demetilasas con Dominio de Jumonji/análisis , Histona Demetilasas con Dominio de Jumonji/genética , Macrófagos/metabolismo , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA