Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
1.
Biol Reprod ; 103(3): 548-559, 2020 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-32507875

RESUMO

Numerous oocytes are retrieved during in vitro fertilization from patients with polycystic ovary syndrome (PCOS). The poor quality of these oocytes leads to lower fertilization and decreases in cleavage and implantation. MiR-155 is one of the microRNA (miRNA) that is increased in serum and granulosa cells of PCOS patients. In this study, we investigate the effects of miR-155 expression and its target genes on oocyte maturation and embryo development. We used the calcium phosphate protocol to transfect vectors that contained miR-155 or miR-off 155 and alone eGFP into cumulus oophorus complex (COCs) of B6D2F1 female mice for in vitro maturation. Cumulus expansion, nuclear, and cytoplasmic maturation, as well as cleavage rates were determined in groups transfected and compared with the control groups. Quantitative real-time polymerase chain reaction was performed to analyze expression levels of miR-155 and the target genes in the cumulus cells, oocytes, and blastocysts. MiR-155 overexpression in COCs suppressed cumulus expansion, oocyte maturation, and inhibition of endogenous miR-155 by miR-off 155 improved cumulus expansion and oocyte maturation by downregulation and expression increase of the Smad2 and Bcl2 genes. On the other hand, overexpression and downregulation of miR-155 in the COCs led to increase and decrease in cleavage rates by changes in expressions of the Mecp2, Jarid2, and Notch1 genes, respectively (P < 0.05). These results suggested that miR-155 overexpression in granulosa cells of PCOS patients can negatively affect nuclear and cytoplasmic maturation, but this miRNA expression has a positive impact on embryo development.


Assuntos
Blastocisto/fisiologia , Células do Cúmulo/fisiologia , MicroRNAs/genética , Oócitos/crescimento & desenvolvimento , Oócitos/fisiologia , Animais , Biologia Computacional , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Glutationa/metabolismo , Técnicas de Maturação in Vitro de Oócitos , Proteína 2 de Ligação a Metil-CpG/biossíntese , Proteína 2 de Ligação a Metil-CpG/genética , Camundongos , Oogênese , Complexo Repressor Polycomb 2/biossíntese , Complexo Repressor Polycomb 2/genética , Gravidez , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptor Notch1/biossíntese , Receptor Notch1/genética , Proteína Smad2/genética , Proteína Smad2/metabolismo
2.
Biosci Biotechnol Biochem ; 84(2): 314-320, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31589096

RESUMO

Glycyrrhiza glabra is considered as potential drug for nasopharyngeal carcinoma (NPC). However, whether the long noncoding RNAs' (lncRNAs) contributes to the anti-cancer function of this herb is unknown. In present study, we analyzed the differential expression of lncRNA between G. glabra-treated and untreated C666-1 cells. Out of those tumor-related lncRNAs, AK027294 had a strongest down-regulation upon G. glabra treatment. Knockdown of AK027294 suppresses the proliferation of C666-1 cells by inducing the apoptosis. Moreover, either G. glabra treatment or knockdown of AK027294 significantly increases the production of EZH1 (Enhancer of zeste 1 polycomb repressive complex 2 subunit). Collectively, we have identified a potential mechanism that the down-regulation of AK027294 contributes to the anti-cancer function of G. glabra and also provide the potential inter-relationship between AK027294 and EZH1.


Assuntos
Proliferação de Células/genética , Glycyrrhiza , Carcinoma Nasofaríngeo/patologia , Neoplasias Nasofaríngeas/patologia , RNA Longo não Codificante/genética , Apoptose/genética , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Humanos , Carcinoma Nasofaríngeo/genética , Carcinoma Nasofaríngeo/metabolismo , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/metabolismo , Complexo Repressor Polycomb 2/biossíntese
3.
J Exp Clin Cancer Res ; 38(1): 277, 2019 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-31234902

RESUMO

BACKGROUND: The deubiquitinating enzyme ubiquitin-specific protease 3 (USP3) plays a crucial role in numerous biological processes. The aberrant expression of USP3 may have an important role in tumor development. However, the mechanism by which USP3 promotes gastric cancer (GC) metastasis remains largely unknown. METHODS: Effects of USP3 on the progression of GC in vivo and in vitro and the potential underlying mechanisms have been investigated utilizing proteomics, RT-PCR, western blotting, immunohistochemistry, immunofluorescence, cell invasion and migration assays and xenograft tumor models. RESULTS: USP3 expression was upregulated in GC compared with matched normal tissues and was predictive of poor survival. USP3 also promoted migration and epithelial-to-mesenchymal transition (EMT) in GC cells. Moreover, TGF-ß1 induced USP3 expression, and USP3 knockdown inhibited TGF-ß1-induced EMT. Furthermore, we utilized Isobaric Tag for Relative and Absolute Quantitation (iTRAQ) to identify differentially expressed proteins in USP3-overexpressing cells compared with control cells. Importantly, we found that SUZ12 is indispensable for USP3-mediated oncogenic activity in GC. We observed that USP3 interacted with and stabilized SUZ12 via deubiquitination. SUZ12 knockdown inhibited USP3-induced migration and invasion, as well as EMT in GC cells. Examination of clinical samples confirmed that USP3 expression was positively correlated with SUZ12 protein expression and that the levels of USP3 or SUZ12 protein were negatively correlated with the levels of E-cadherin protein. CONCLUSIONS: These findings identify USP3 as a critical regulator. The USP3-SUZ12 axis might promote tumor progression and could be a potential therapeutic candidate for human GC.


Assuntos
Transição Epitelial-Mesenquimal , Complexo Repressor Polycomb 2/metabolismo , Neoplasias Gástricas/metabolismo , Proteases Específicas de Ubiquitina/metabolismo , Animais , Caderinas/metabolismo , Carcinoma/patologia , Carcinoma/secundário , Linhagem Celular Tumoral , Movimento Celular , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos , Invasividade Neoplásica , Metástase Neoplásica , Proteínas de Neoplasias , Complexo Repressor Polycomb 2/biossíntese , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Fatores de Transcrição , Fator de Crescimento Transformador beta1/metabolismo , Proteases Específicas de Ubiquitina/genética , Ubiquitinação , Regulação para Cima , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Dev Biol ; 450(1): 34-46, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30851270

RESUMO

Many organisms both undergo dramatic morphological changes during post-embryonic development and also regenerate lost structures, but the roles of epigenetic regulators in such processes are only beginning to be understood. In the present study, the functions of two histone modifiers were examined during metamorphosis and larval limb regeneration in the red flour beetle Tribolium castaneum. Polycomb (Pc), a member of Polycomb repressive complex 1 (PRC1), and Enhancer of zeste (E(z)), a member of Polycomb repressive complex 2 (PRC2), were silenced in larvae using RNA interference. In the absence of Pc, the head appendages of adults transformed into a leg-like morphology, and the legs and wings assumed a metathoracic identity, indicating that Pc acts to specify proper segmental identity. Similarly, silencing of E(z) led to homeotic transformation of legs and wings. Additional defects were also observed in limb patterning as well as eye morphogenesis, indicating that PcG proteins play critical roles in imaginal precursor cells. In addition, larval legs and antennae failed to re-differentiate when either Pc or E(z) was knocked down, indicating that histone modification is necessary for proper blastema growth and differentiation. These findings indicate that PcG proteins play extensive roles in postembryonic plasticity of imaginal precursor cells.


Assuntos
Membro Posterior/fisiologia , Proteínas de Insetos/biossíntese , Morfogênese , Complexo Repressor Polycomb 1/biossíntese , Complexo Repressor Polycomb 2/biossíntese , Regeneração , Tribolium/metabolismo , Animais , Proteínas de Insetos/genética , Larva/genética , Larva/metabolismo , Complexo Repressor Polycomb 1/genética , Complexo Repressor Polycomb 2/genética , Tribolium/genética
5.
Med Sci Monit ; 25: 801-810, 2019 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-30688289

RESUMO

BACKGROUND Acute kidney injury (AKI) involves the renal tubular epithelium. The enhancer of zeste homolog 1 (EZH1) gene has a role in cell development and differentiation. This study aimed to investigate the effect of overexpression of the EZH1 gene on aristolochic acid-induced injury in HK-2 human kidney proximal tubule epithelial cells in vitro. MATERIAL AND METHODS The HK-2 cells were cultured and treated with aristolochic acid and the effects of aristolochic acid-injury were evaluated using a cell counting kit-8 (CCK-8) assay. Overexpression of EZH1 used gene plasmid transfection into HK-2 cells. The cell apoptosis rate and levels of intracellular reactive oxygen species (ROS) were measured using flow cytometry. Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot were performed to determine the expressions of inflammatory cytokines including interleukin (IL)-1ß, IL-6, tumor necrosis factor-α (TNF-α), apoptosis-related genes, and the downstream target genes of NF-κB signaling pathway, including NFKBIA, CXCL8, and cyclin D1. RESULTS Aristolochic acid inhibited HK-2 cell viability, induced cell apoptosis, increased the levels of ROS and inflammatory cytokines, including IL-1ß, IL-6, TNF-α, and activated the NF-κB pathway. Overexpression the EZH1 gene inhibited HK-2 cell apoptosis, reduced ROS levels, and down-regulated the expressions of IL-1ß, IL-6, TNF-α, Bax and Cyt C mRNA and protein, and increased the expressions of Bcl-2 and NFKBIA, CXCL8 and cyclin D1, indicating that overexpression of EZH1 suppressed NF-κB signaling in aristolochic acid-injured HK-2 cells. CONCLUSIONS Overexpression of EZH1 reduced HK-2 cell injury induced by aristolochic acid in vitro by inhibition of NF-κB signaling.


Assuntos
Injúria Renal Aguda/metabolismo , Túbulos Renais Proximais/efeitos dos fármacos , Complexo Repressor Polycomb 2/metabolismo , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/patologia , Apoptose/efeitos dos fármacos , Ácidos Aristolóquicos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Citocinas/metabolismo , Células Epiteliais/metabolismo , Humanos , Inflamação/patologia , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Rim/efeitos dos fármacos , Rim/patologia , Túbulos Renais Proximais/patologia , NF-kappa B/metabolismo , Complexo Repressor Polycomb 2/biossíntese , Complexo Repressor Polycomb 2/genética , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo
6.
Neurobiol Dis ; 119: 149-158, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30099093

RESUMO

BACKGROUND: With the capacity to modulate gene networks in an environmentally-sensitive manner, the role of epigenetic systems in mental disorders has come under intense investigation. Dysregulation of epigenetic effectors, including microRNAs and histone-modifying enzymes, may better explain the role of environmental risk factors and the observed heritability rate that cannot be fully attributed to known genetic risk alleles. Here, we aimed to identify novel epigenetic targets of the schizophrenia-associated microRNA 132 (miR-132). METHODS: Histone modifications were quantified by immunodetection in response to viral-mediated overexpression of miR-132 while a luminescent reporter system was used to validate targets of miR-132 in vitro. Genome-wide profiling, quantitative PCR and NanoSting were used to quantify gene expression in post-mortem human brains, neuronal cultures and prefrontal cortex (PFC) of mice chronically exposed to antipsychotics. Following viral-mediated depletion of Enhancer of Zeste 1 (EZH1) in the murine PFC, behaviors including sociability and motivation were assessed using a 3-chambered apparatus and forced-swim test, respectively. RESULTS: Overexpression of miR-132 decreased global histone 3 lysine 27 tri-methylation (H3K27me3), a repressive epigenetic mark. Moreover, the polycomb-associated H3K27 methyltransferase, EZH1, is regulated by miR-132 and upregulated in the PFC of schizophrenics. Unlike its homolog EZH2, expression of EZH1 in the murine PFC decreased following chronic exposure to antipsychotics. Viral-mediated depletion of EZH1 in the mouse PFC attenuated sociability, enhanced motivational behaviors, and affected gene expression pathways related to neurotransmission and behavioral phenotypes. CONCLUSIONS: EZH1 is dysregulated in schizophrenia, sensitive to antipsychotic medications, and a brain-enriched miR-132 target that controls neurobehavioral phenotypes.


Assuntos
Antipsicóticos/uso terapêutico , Epigênese Genética/fisiologia , Motivação/fisiologia , Complexo Repressor Polycomb 2/biossíntese , Esquizofrenia/metabolismo , Comportamento Social , Adulto , Idoso , Animais , Antipsicóticos/farmacologia , Linhagem Celular Tumoral , Estudos de Coortes , Epigênese Genética/efeitos dos fármacos , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Motivação/efeitos dos fármacos , Complexo Repressor Polycomb 2/genética , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Esquizofrenia/tratamento farmacológico , Esquizofrenia/genética
7.
Development ; 145(14)2018 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-29945864

RESUMO

Epigenetic regulation of gene expression has a crucial role allowing for the self-renewal and differentiation of stem and progenitor populations during organogenesis. The mammalian kidney maintains a population of self-renewing stem cells that differentiate to give rise to thousands of nephrons, which are the functional units that carry out filtration to maintain physiological homeostasis. The polycomb repressive complex 2 (PRC2) epigenetically represses gene expression during development by placing the H3K27me3 mark on histone H3 at promoter and enhancer sites, resulting in gene silencing. To understand the role of PRC2 in nephron differentiation, we conditionally inactivated the Eed gene, which encodes a nonredundant component of the PRC2 complex, in nephron progenitor cells. Resultant kidneys were smaller and showed premature loss of progenitor cells. The progenitors in Eed mutant mice that were induced to differentiate did not develop into properly formed nephrons. Lhx1, normally expressed in the renal vesicle, was overexpressed in kidneys of Eed mutant mice. Thus, PRC2 has a crucial role in suppressing the expression of genes that maintain the progenitor state, allowing nephron differentiation to proceed.


Assuntos
Diferenciação Celular/fisiologia , Epigênese Genética/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Néfrons/embriologia , Complexo Repressor Polycomb 2/biossíntese , Células-Tronco/metabolismo , Animais , Proteínas com Homeodomínio LIM/biossíntese , Proteínas com Homeodomínio LIM/genética , Camundongos , Camundongos Transgênicos , Mutação , Néfrons/citologia , Complexo Repressor Polycomb 2/genética , Células-Tronco/citologia , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética
8.
Ann Hematol ; 97(7): 1193-1208, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29560522

RESUMO

Primary myelofibrosis (PMF) is one of the BCR/ABL-negative myeloproliferative neoplasms (MPNs), characterized by the diffuse fibrous hyperproliferation, bone marrow osteosclerosis, extramedullary hematopoiesis, and marked splenomegaly. The patients with PMF have an insidious onset, a long duration of clinical course, and the deteriorated quality of life. It has been reported that the CALR gene 9 exon mutations were detected in 25-30% PMF patients, particularly as high as 80% in the JAK2/MPL-negative ones. As the second most common mutation in BCR/ABL-negative MPNs, CALR mutation has been included in the latest World Health Organization (WHO) classification criteria as one of the main diagnostic criteria for both essential thrombocythemia (ET) and PMF. Moreover, the CALR mutations indicated a favorable prognosis, which the mechanism is still under investigation. It was demonstrated that a characterized high expression of EZH2 and SUZ12 in CALR-mutated patients. Taking EZH2 as the research entry point, we initially discussed the mechanism that the CALR-positive patients with PMF exhibited a better prognosis in the current study.


Assuntos
Calreticulina/genética , Proteína Potenciadora do Homólogo 2 de Zeste/biossíntese , Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/biossíntese , Mielofibrose Primária/genética , Adulto , Idoso , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Feminino , Células HEK293 , Células HL-60 , Humanos , Janus Quinase 2/genética , Masculino , MicroRNAs/biossíntese , MicroRNAs/sangue , MicroRNAs/genética , Pessoa de Meia-Idade , Mutação , Proteínas de Neoplasias/genética , Complexo Repressor Polycomb 2/biossíntese , Complexo Repressor Polycomb 2/genética , Mielofibrose Primária/metabolismo , Prognóstico , Interferência de RNA , RNA Neoplásico/biossíntese , RNA Neoplásico/sangue , RNA Neoplásico/genética , Proteínas Recombinantes/metabolismo , Fatores de Transcrição , Transdução Genética
9.
Gynecol Oncol ; 149(2): 388-393, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29544705

RESUMO

OBJECTIVE: Low-grade endometrial stromal sarcomas (LGESS) harbor chromosomal translocations that affect proteins associated with chromatin remodeling Polycomb Repressive Complex 2 (PRC2), including SUZ12, PHF1 and EPC1. Roughly half of LGESS also demonstrate nuclear accumulation of ß-catenin, which is a hallmark of Wnt signaling activation. However, the targets affected by the fusion proteins and the role of Wnt signaling in the pathogenesis of these tumors remain largely unknown. METHODS: Here we report the results of a meta-analysis of three independent gene expression profiling studies on LGESS and immunohistochemical evaluation of nuclear expression of ß-catenin and Lef1 in 112 uterine sarcoma specimens obtained from 20 LGESS and 89 LMS patients. RESULTS: Our results demonstrate that 143 out of 310 genes overexpressed in LGESS are known to be directly regulated by SUZ12. In addition, our gene expression meta-analysis shows activation of multiple genes implicated in Wnt signaling. We further emphasize the role of the Wnt signaling pathway by demonstrating concordant nuclear expression of ß-catenin and Lef1 in 7/16 LGESS. CONCLUSIONS: Based on our findings, we suggest that LGESS-specific fusion proteins disrupt the repressive function of the PRC2 complex similar to the mechanism seen in synovial sarcoma, where the SS18-SSX fusion proteins disrupt the mSWI/SNF (BAF) chromatin remodeling complex. We propose that these fusion proteins in LGESS contribute to overexpression of Wnt ligands with subsequent activation of Wnt signaling pathway and formation of an active ß-catenin/Lef1 transcriptional complex. These observations could lead to novel therapeutic approaches that focus on the Wnt pathway in LGESS.


Assuntos
Neoplasias do Endométrio/genética , Proteínas de Fusão Oncogênica/genética , Sarcoma do Estroma Endometrial/genética , Via de Sinalização Wnt/genética , Neoplasias do Endométrio/metabolismo , Neoplasias do Endométrio/patologia , Feminino , Perfilação da Expressão Gênica , Humanos , Imuno-Histoquímica , Fator 1 de Ligação ao Facilitador Linfoide/biossíntese , Fator 1 de Ligação ao Facilitador Linfoide/genética , Gradação de Tumores , Proteínas de Neoplasias , Proteínas de Fusão Oncogênica/metabolismo , Complexo Repressor Polycomb 2/biossíntese , Complexo Repressor Polycomb 2/genética , Sarcoma do Estroma Endometrial/metabolismo , Sarcoma do Estroma Endometrial/patologia , Análise Serial de Tecidos , Fatores de Transcrição , beta Catenina/biossíntese , beta Catenina/genética
10.
BMC Cancer ; 18(1): 158, 2018 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-29415665

RESUMO

BACKGROUND: Polycomb repressive complex 2 (PRC2; formed by EZH2, SUZ12, and EED protein subunits) and PRC1 (BMI1 protein) induce gene silencing through histone modification by H3K27me3. In the present study, we characterized the PRC expression pattern and its clinical implication in sarcoma. METHODS: Using immunohistochemistry, we analyzed PRC expression in 105 sarcoma patients with 5 subtypes: synovial sarcoma (n = 18), rhabdomyosarcoma (n = 28), Ewing sarcoma (n = 15), osteosarcoma (n = 30), and others (n = 14). RESULTS: The median age at diagnosis in the patient cohort was 26.8 years (range: 1-78 years) and the male-to-female ratio was 1:4. Initial disease presentation was locoregional disease in 83% of patients and initial metastatic disease in the remaining 17%. PRC expression was not significantly different according to histologic subtype (P = 0.400). Overall survival (OS) was significantly poor for SUZ12 high (P = 0.001), EED1 high (P = 0.279), and H3K27me3 high (P = 0.009). Ultimately, patients with PRC2high had significantly inferior OS than the no expression group (P = 0.009). In the Cox proportional hazard model adjusted for stage, histologic grade, surgery, margin and initial metastasis, SUZ12 expression (P = 0.020, HR 29.069, 95% CI 1.690-500.007), H3K27me3 (P = 0.010, HR 3.743, 95% CI 1.370-10.228) expression was significantly associated with shorter OS. CONCLUSION: We detected PRC expression in various sarcomas and demonstrated its independent negative prognostic role, suggesting the PRC axis as promising therapeutic target for treating sarcoma.


Assuntos
Proteína Potenciadora do Homólogo 2 de Zeste/biossíntese , Código das Histonas , Complexo Repressor Polycomb 2/biossíntese , Sarcoma/metabolismo , Adolescente , Adulto , Idoso , Criança , Pré-Escolar , Feminino , Humanos , Imuno-Histoquímica , Lactente , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Proteínas de Neoplasias , Prognóstico , Sarcoma/patologia , Fatores de Transcrição , Adulto Jovem
11.
Sci Rep ; 8(1): 1720, 2018 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-29379063

RESUMO

Human mesenchymal stem/stromal cells (hMSCs) provide support for cancer progression, partly through their secretome that includes extracellular vesicles (EVs). Based on deep-sequencing of small RNA from EVs of MSCs, we now report the characterization of novel small RNA, named n-miR-G665, which exhibits typical properties of miRNAs. n-miR-G665 sequence is conserved and expressed in most cell types. Knockdown studies using anti-agomirs and shRNA studies demonstrated that n-miR-G665 plays an important role in cell proliferation. Functional assays to reveal the targets of n-miR-G665 showed that polycomb protein Suz12 is regulated by n-miR-G665, which in turn regulates the expression of n-miR-G665 through feedback loop mechanism. These data shed light on a previously unknown novel feedback regulatory mechanism for controlling Suz12 expression regulated by previously not described miRNA, which may highlight a new therapeutic approach to control the polycomb repressor complex 2 activity in cancers.


Assuntos
Regulação da Expressão Gênica , Células-Tronco Mesenquimais/química , Células-Tronco Mesenquimais/fisiologia , MicroRNAs/metabolismo , Complexo Repressor Polycomb 2/biossíntese , Linhagem Celular , Proliferação de Células , Vesículas Extracelulares/química , Técnicas de Silenciamento de Genes , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , MicroRNAs/genética , MicroRNAs/isolamento & purificação , Proteínas de Neoplasias , Fatores de Transcrição
12.
Circ Res ; 121(2): 106-112, 2017 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-28512107

RESUMO

RATIONALE: Polycomb repressive complex 2 is a major epigenetic repressor that deposits methylation on histone H3 on lysine 27 (H3K27me) and controls differentiation and function of many cells, including cardiac myocytes. EZH1 and EZH2 are 2 alternative catalytic subunits with partial functional redundancy. The relative roles of EZH1 and EZH2 in heart development and regeneration are unknown. OBJECTIVE: We compared the roles of EZH1 versus EZH2 in heart development and neonatal heart regeneration. METHODS AND RESULTS: Heart development was normal in Ezh1-/- (Ezh1 knockout) and Ezh2f/f::cTNT-Cre (Ezh2 knockout) embryos. Ablation of both genes in Ezh1-/-::Ezh2f/f::cTNT-Cre embryos caused lethal heart malformations, including hypertrabeculation, compact myocardial hypoplasia, and ventricular septal defect. Epigenome and transcriptome profiling showed that derepressed genes were upregulated in a manner consistent with total EZH dose. In neonatal heart regeneration, Ezh1 was required, but Ezh2 was dispensable. This finding was further supported by rescue experiments: cardiac myocyte-restricted re-expression of EZH1 but not EZH2 restored neonatal heart regeneration in Ezh1 knockout. In myocardial infarction performed outside of the neonatal regenerative window, EZH1 but not EZH2 likewise improved heart function and stimulated cardiac myocyte proliferation. Mechanistically, EZH1 occupied and activated genes related to cardiac growth. CONCLUSIONS: Our work unravels divergent mechanisms of EZH1 in heart development and regeneration, which will empower efforts to overcome epigenetic barriers to heart regeneration.


Assuntos
Desenvolvimento Embrionário/fisiologia , Coração/embriologia , Coração/fisiologia , Complexo Repressor Polycomb 2/biossíntese , Regeneração/fisiologia , Animais , Animais Recém-Nascidos , Coração/crescimento & desenvolvimento , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos Cardíacos/metabolismo , Complexo Repressor Polycomb 2/deficiência
13.
Cancer Discov ; 6(11): 1237-1247, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27630126

RESUMO

Tyrosine kinase inhibitors (TKI) have revolutionized chronic myelogenous leukemia (CML) management. Disease eradication, however, is hampered by innate resistance of leukemia-initiating cells (LIC) to TKI-induced killing, which also provides the basis for subsequent emergence of TKI-resistant mutants. We report that EZH2, the catalytic subunit of Polycomb Repressive Complex 2 (PRC2), is overexpressed in CML LICs and required for colony formation and survival and cell-cycle progression of CML cell lines. A critical role for EZH2 is supported by genetic studies in a mouse CML model. Inactivation of Ezh2 in conventional conditional mice and through CRISPR/Cas9-mediated gene editing prevents initiation and maintenance of disease and survival of LICs, irrespective of BCR-ABL1 mutational status, and extends survival. Expression of the EZH2 homolog EZH1 is reduced in EZH2-deficient CML LICs, creating a scenario resembling complete loss of PRC2. EZH2 dependence of CML LICs raises prospects for improved therapy of TKI-resistant CML and/or eradication of disease by addition of EZH2 inhibitors. SIGNIFICANCE: This work defines EZH2 as a selective vulnerability for CML cells and their LICs, regardless of BCR-ABL1 mutational status. Our findings provide an experimental rationale for improving disease eradication through judicious use of EZH2 inhibitors within the context of standard-of-care TKI therapy. Cancer Discov; 6(11); 1237-47. ©2016 AACR.See related article by Scott et al., p. 1248This article is highlighted in the In This Issue feature, p. 1197.


Assuntos
Proteína Potenciadora do Homólogo 2 de Zeste/biossíntese , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Células-Tronco Neoplásicas/metabolismo , Complexo Repressor Polycomb 2/biossíntese , Animais , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteínas de Fusão bcr-abl/genética , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Humanos , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Camundongos , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia , Complexo Repressor Polycomb 2/genética , Inibidores de Proteínas Quinases/administração & dosagem , Transdução de Sinais
14.
Pathology ; 48(5): 467-82, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27311868

RESUMO

The Polycomb repressive complex-2 members (EZH2, EED, SUZ12 and EZH1) are important regulators of haematopoiesis, cell cycle and differentiation. Over-expression of EZH2 has been linked to cancer metastases and poor prognosis. Detailed information on the expression of other members in normal and neoplastic lymphoid tissue remains to be elucidated. Immunohistochemical and immunofluorescent analyses of 156 samples from haematopoietic neoplasms patients and 27 haematopoietic cell lines were used. B-cell neoplasms showed a significant over-expression of EZH2, EED and SUZ12 in the aggressive subtypes compared to the indolent subtypes and normal tissue (p = 0.000-0.046) while expression of EZH1 was decreased in mantle cell lymphoma compared to normal tissue (p = 0.011). T/NK-cell neoplasms also showed significant over-expression of EZH2, EED and SUZ12 (p = 0.000-0.002) and decreased expression of EZH1 (p = 0.001) compared to normal cells. EZH2 and EZH1 have opposite expression patterns both in normal and neoplastic lymphoid tissues as well as an opposite relation to Ki-67. These results were supported by western blotting analyses. Immunofluorescent staining revealed a difference in the intracellular localisation of EZH1 compared to other members. These evidences suggest that EZH2 and EZH1 are important in the counter-balancing mechanisms controlling proliferation/resting of lymphoid cells. The disruption of the balanced EZH2/EZH1 ratio may play important roles in the pathogenesis of lymphomas.


Assuntos
Biomarcadores Tumorais/análise , Proteína Potenciadora do Homólogo 2 de Zeste/biossíntese , Linfoma de Células B/patologia , Linfoma de Células T/patologia , Complexo Repressor Polycomb 2/biossíntese , Western Blotting , Proteína Potenciadora do Homólogo 2 de Zeste/análise , Humanos , Imuno-Histoquímica , Linfoma de Células B/metabolismo , Linfoma de Células T/metabolismo , Complexo Repressor Polycomb 2/análise
15.
BMC Genomics ; 17: 20, 2016 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-26728506

RESUMO

BACKGROUND: RNA interference (RNAi) is a powerful platform utilized to target transcription of specific genes and downregulate the protein product. To achieve effective silencing, RNAi is usually applied to cells or tissue with a transfection reagent to enhance entry into cells. A commonly used control is the same transfection reagent plus a "noncoding RNAi". However, this does not control for the genomic response to the transfection reagent alone or in combination with the noncoding RNAi. These control effects while not directly targeting the gene in question may influence expression of other genes that in turn alter expression of the target. The current study was prompted by our work focused on prevention of vascular bypass graft failure and our experience with gene silencing in human aortic smooth muscle cells (HAoSMCs) where we suspected that off target effects through this mechanism might be substantial. We have used Next Generation Sequencing (NGS) technology and bioinformatics analysis to examine the genomic response of HAoSMCs to the transfection reagent alone (polyethyleneimine (PEI)) or in combination with commercially obtained control small interfering RNA (siRNAs) (Dharmacon and Invitrogen). RESULTS: Compared to untreated cells, global gene expression of HAoSMcs after transfection either with PEI or in combination with control siRNAs displayed significant alterations in gene transcriptome after 24 h. HAoSMCs transfected by PEI alone revealed alterations of 213 genes mainly involved in inflammatory and immune responses. HAoSMCs transfected by PEI complexed with siRNA from either Dharmacon or Invitrogen showed substantial gene variation of 113 and 85 genes respectively. Transfection of cells with only PEI or with PEI and control siRNAs resulted in identification of 20 set of overlapping altered genes. Further, systems biology analysis revealed key master regulators in cells transfected with control siRNAs including the cytokine, Interleukin (IL)-1, transcription factor GATA Binding Protein (GATA)-4 and the methylation enzyme, Enhancer of zeste homolog 2 (EZH-2) a cytokine with an apical role in initiating the inflammatory response. CONCLUSIONS: Significant off-target effects in HAoSMCs transfected with PEI alone or in combination with control siRNAs may lead to misleading conclusions concerning the effectiveness of a targeted siRNA strategy. The lack of structural information about transfection reagents and "non coding" siRNA is a hindrance in the development of siRNA based therapeutics.


Assuntos
Aorta/efeitos dos fármacos , Biologia Computacional , Regulação da Expressão Gênica/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Aorta/metabolismo , Proteína Potenciadora do Homólogo 2 de Zeste , Fator de Transcrição GATA4/biossíntese , Regulação da Expressão Gênica/genética , Inativação Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Interleucina-1/biossíntese , Músculo Liso Vascular/metabolismo , Complexo Repressor Polycomb 2/biossíntese , Polietilenoimina/administração & dosagem , RNA Interferente Pequeno/genética , Transfecção/métodos
16.
Molecules ; 22(1)2016 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-28042859

RESUMO

Silibinin, extracted from milk thistle (Silybum marianum L.), has exhibited considerable preclinical activity against prostate carcinoma. Its antitumor and chemopreventive activities have been associated with diverse effects on cell cycle, apoptosis, and receptor-dependent mitogenic signaling pathways. Here we hypothesized that silibinin's pleiotropic effects may reflect its interference with epigenetic mechanisms in human prostate cancer cells. More specifically, we have demonstrated that silibinin reduces gene expression levels of the Polycomb Repressive Complex 2 (PRC2) members Enhancer of Zeste Homolog 2 (EZH2), Suppressor of Zeste Homolog 12 (SUZ12), and Embryonic Ectoderm Development (EED) in DU145 and PC3 human prostate cancer cells, as evidenced by Real Time Polymerase Chain Reaction (RT-PCR). Furthermore immunoblot and immunofluorescence analysis revealed that silibinin-mediated reduction of EZH2 levels was accompanied by an increase in trimethylation of histone H3 on lysine (Κ)-27 residue (H3K27me3) levels and that such response was, in part, dependent on decreased expression levels of phosphorylated Akt (ser473) (pAkt) and phosphorylated EZH2 (ser21) (pEZH2). Additionally silibinin exerted other epigenetic effects involving an increase in total DNA methyltransferase (DNMT) activity while it decreased histone deacetylases 1-2 (HDACs1-2) expression levels. We conclude that silibinin induces epigenetic alterations in human prostate cancer cells, suggesting that subsequent disruptions of central processes in chromatin conformation may account for some of its diverse anticancer effects.


Assuntos
Antineoplásicos/farmacologia , Antioxidantes/farmacologia , Proteína Potenciadora do Homólogo 2 de Zeste/biossíntese , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Complexo Repressor Polycomb 2/biossíntese , Neoplasias da Próstata/tratamento farmacológico , Silimarina/farmacologia , Linhagem Celular Tumoral , DNA (Citosina-5-)-Metiltransferases , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Epigênese Genética , Histona Desacetilase 1/biossíntese , Histona Desacetilase 2/biossíntese , Histonas/metabolismo , Humanos , Masculino , Metilação/efeitos dos fármacos , Proteínas de Neoplasias , Fosforilação/efeitos dos fármacos , Complexo Repressor Polycomb 2/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Silibina , Fatores de Transcrição
17.
Stem Cells ; 34(2): 322-33, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26523946

RESUMO

Identification of a gene set capable of driving rapid and proper reprogramming to induced pluripotent stem cells (iPSCs) is an important issue. Here we show that the efficiency and kinetics of iPSC reprogramming are dramatically improved by the combined expression of Jarid2 and genes encoding its associated proteins. We demonstrate that forced expression of JARID2 promotes iPSC reprogramming by suppressing the expression of Arf, a known reprogramming barrier, and that the N-terminal half of JARID2 is sufficient for such promotion. Moreover, JARID2 accelerated silencing of the retroviral Klf4 transgene and demethylation of the Nanog promoter, underpinning the potentiating activity of JARID2 in iPSC reprogramming. We further show that JARID2 physically interacts with ESRRB, SALL4A, and PRDM14, and that these JARID2-associated proteins synergistically and robustly facilitate iPSC reprogramming in a JARID2-dependent manner. Our findings provide an insight into the important roles of JARID2 during reprogramming and suggest that the JARID2-associated protein network contributes to overcoming reprogramming barriers.


Assuntos
Técnicas de Reprogramação Celular/métodos , Proteínas de Ligação a DNA , Expressão Gênica , Células-Tronco Pluripotentes Induzidas/metabolismo , Complexo Repressor Polycomb 2 , Receptores de Estrogênio , Fatores de Transcrição , Animais , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Fator 4 Semelhante a Kruppel , Camundongos , Complexo Repressor Polycomb 2/biossíntese , Complexo Repressor Polycomb 2/genética , Proteínas de Ligação a RNA , Receptores de Estrogênio/biossíntese , Receptores de Estrogênio/genética , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética
18.
Int J Oncol ; 48(1): 145-52, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26648239

RESUMO

Pancreatic cancer has a poor prognosis because of its high invasiveness and recurrence, and these properties closely link to the phenomenon of epithelial-mesenchymal transition (EMT). Recently, it has been reported that Sox4 is indispensable for EMT in vitro and in vivo and regulates various master regulators of EMT including Zeb, Twist and Snail. Moreover, Sox4 induces the transcription of Ezh2 which is the histone methyltransferase, and reprograms the cancer epigenome to promote EMT and metastasis. Therefore, the present study evaluated the importance of Sox4, Ezh2 and miR-335, which regulate Sox4 expression epigenetically, in clinical samples with pancreatic cancer. This retrospective analysis included data from 36 consecutive patients who underwent complete surgical resection for pancreatic cancer and did not undergo any preoperative therapies. We assessed the clinical significance of Sox4/Ezh2 axis and miR-335 expression, using immunohistochemistry and qRT-PCR with laser captured microdissection (LCM). The Sox4 positive patients had significantly worse prognosis as for disease-free survival (DFS) (P=0.0154) and the Ezh2-positive patients had significantly worse prognosis as for overall survival (OS) (P=0.0347). The miR-335 expression was inversely correlated with Sox4 expression in the identical clinical specimens, but it was not related to the prognosis. Sox4/Ezh2 axis was closely associated with the prognosis in pancreatic cancer patients.


Assuntos
MicroRNAs/biossíntese , Neoplasias Pancreáticas/genética , Complexo Repressor Polycomb 2/biossíntese , Fatores de Transcrição SOXC/biossíntese , Idoso , Intervalo Livre de Doença , Proteína Potenciadora do Homólogo 2 de Zeste , Transição Epitelial-Mesenquimal , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , MicroRNAs/genética , Pessoa de Meia-Idade , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/cirurgia , Complexo Repressor Polycomb 2/genética , Prognóstico , Fatores de Transcrição SOXC/genética , Resultado do Tratamento
19.
Cancer Res ; 76(3): 675-85, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26676756

RESUMO

EZH2 overexpression promotes cancer by increasing histone methylation to silence tumor suppressor genes, but how EZH2 levels become elevated in cancer is not understood. In this study, we investigated the mechanisms by which EZH2 expression is regulated in non-small cell lung carcinoma cells by oncogenic KRAS. In cells harboring KRAS(G12C) and KRAS(G12D) mutations, EZH2 expression was modulated by MEK-ERK and PI3K/AKT signaling, respectively. Accordingly, MEK-ERK depletion decreased EZH2 expression in cells harboring the KRAS(G12C) mutation, whereas PI3K/AKT depletion decreased EZH2 expression, EZH2 phosphorylation, and STAT3 activity in KRAS(G12D)-mutant cell lines. Combined inhibition of EZH2 and MEK-ERK or PI3K/AKT increased the sensitivity of cells with specific KRAS mutations to MEK-ERK and PI3K/AKT-targeted therapies. Our work defines EZH2 as a downstream effector of KRAS signaling and offers a rationale for combining EZH2 inhibitory strategies with MEK-ERK- or PI3K/AKT-targeted therapies to treat lung cancer patients, as stratified into distinct treatment groups based on specific KRAS mutations.


Assuntos
Adenocarcinoma/metabolismo , Genes ras , Neoplasias Pulmonares/metabolismo , Sistema de Sinalização das MAP Quinases , Mutação , Fosfatidilinositol 3-Quinases/metabolismo , Complexo Repressor Polycomb 2/biossíntese , Complexo Repressor Polycomb 2/genética , Adenocarcinoma/genética , Adenocarcinoma de Pulmão , Animais , Linhagem Celular Tumoral , Proteína Potenciadora do Homólogo 2 de Zeste , Feminino , Xenoenxertos , Humanos , Neoplasias Pulmonares/genética , Camundongos , Camundongos Nus , Fosforilação , Complexo Repressor Polycomb 2/metabolismo , Transfecção , Proteínas ras/genética , Proteínas ras/metabolismo
20.
J Biol Chem ; 291(8): 3785-95, 2016 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-26655220

RESUMO

Hereditary forms of Wilms arise from developmentally arrested clones of renal progenitor cells with biallelic mutations of WT1; recently, it has been found that Wilms tumors may also be associated with biallelic mutations in DICER1 or DROSHA, crucial for miRNA biogenesis. We have previously shown that a critical role for WT1 during normal nephrogenesis is to suppress transcription of the Polycomb group protein, EZH2, thereby de-repressing genes in the differentiation cascade. Here we show that WT1 also suppresses translation of EZH2. All major WT1 isoforms induce an array of miRNAs, which target the 3' UTR of EZH2 and other Polycomb-associated transcripts. We show that the WT1(+KTS) isoform binds to the 5' UTR of EZH2 and interacts directly with the miRNA-containing RISC to enhance post-transcriptional inhibition. These observations suggest a novel mechanism through which WT1 regulates the transition from resting stem cell to activated progenitor cell during nephrogenesis. Our findings also offer a plausible explanation for the fact that Wilms tumors can arise either from loss of WT1 or loss of miRNA processing enzymes.


Assuntos
Células-Tronco Mesenquimais/metabolismo , MicroRNAs/metabolismo , Complexo Repressor Polycomb 2/biossíntese , Biossíntese de Proteínas , RNA Neoplásico/metabolismo , Proteínas WT1/metabolismo , Regiões 3' não Traduzidas , Regiões 5' não Traduzidas , Proteína Potenciadora do Homólogo 2 de Zeste , Humanos , Células-Tronco Mesenquimais/patologia , MicroRNAs/genética , Complexo Repressor Polycomb 2/genética , RNA Neoplásico/genética , Proteínas WT1/genética , Tumor de Wilms/genética , Tumor de Wilms/metabolismo , Tumor de Wilms/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...