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










Base de dados
Intervalo de ano de publicação
1.
Br J Haematol ; 194(1): 174-178, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33843056

RESUMO

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.


Assuntos
Anticorpos Biespecíficos/uso terapêutico , Antineoplásicos Imunológicos/uso terapêutico , Transplante de Células-Tronco Hematopoéticas , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamento farmacológico , Terapia de Salvação , Pré-Escolar , Intervalo Livre de Doença , Feminino , Histona-Lisina N-Metiltransferase/análise , Humanos , Lactente , Estimativa de Kaplan-Meier , Masculino , Proteína de Leucina Linfoide-Mieloide/análise , Neoplasia Residual , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patologia , Recidiva
2.
Hum Pathol ; 108: 1-11, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33121982

RESUMO

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.


Assuntos
Biomarcadores Tumorais/análise , Glutationa Transferase/análise , Antígenos de Histocompatibilidade/análise , Histona-Lisina N-Metiltransferase/análise , Mesonefroma/diagnóstico , Fator 1 de Elongação de Peptídeos/análise , Feminino , Humanos , Proteômica/métodos
3.
Sci Rep ; 10(1): 20598, 2020 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-33244033

RESUMO

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.


Assuntos
Histona-Lisina N-Metiltransferase/análise , Mioblastos/citologia , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Forma Celular , Citoplasma/metabolismo , Citoesqueleto/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Camundongos , Mioblastos/metabolismo , Transporte Proteico
4.
Int J Oncol ; 57(3): 756-766, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32705243

RESUMO

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.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Regulação Neoplásica da Expressão Gênica , Histona-Lisina N-Metiltransferase/genética , Neoplasias Pulmonares/genética , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Apoptose/genética , Carcinogênese/genética , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Técnicas de Silenciamento de Genes , Histona-Lisina N-Metiltransferase/análise , Humanos , Estimativa de Kaplan-Meier , Pulmão/patologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica/genética , Prognóstico , Ativação Transcricional , Regulação para Cima
5.
Chembiochem ; 21(1-2): 256-264, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31612581

RESUMO

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.


Assuntos
Histona-Lisina N-Metiltransferase/análise , Dicroísmo Circular , Células HEK293 , Histona-Lisina N-Metiltransferase/isolamento & purificação , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Metilação , Especificidade por Substrato
6.
Biochem Biophys Res Commun ; 513(2): 340-346, 2019 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-30955858

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Benzamidas/farmacologia , Cistadenocarcinoma Seroso/tratamento farmacológico , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Neoplasias Ovarianas/tratamento farmacológico , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Pirrolidinas/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cistadenocarcinoma Seroso/patologia , Feminino , Histona-Lisina N-Metiltransferase/análise , Humanos , Neoplasias Ovarianas/patologia
7.
Biochem Biophys Res Commun ; 498(3): 579-585, 2018 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-29522714

RESUMO

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.


Assuntos
Regulação Neoplásica da Expressão Gênica , Histona-Lisina N-Metiltransferase/genética , Invasividade Neoplásica/genética , Neoplasias Gástricas/genética , Idoso , Biomarcadores Tumorais/análise , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Regulação para Baixo , Feminino , Mucosa Gástrica/metabolismo , Histona-Lisina N-Metiltransferase/análise , Humanos , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica/diagnóstico , Invasividade Neoplásica/patologia , Prognóstico , Estômago/patologia , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/patologia
8.
J Mol Cell Biol ; 10(2): 161-174, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28992061

RESUMO

Myogenic differentiation is accompanied by alterations in the chromatin states, which permit or restrict the transcriptional machinery and thus impact distinctive gene expression profiles. The mechanisms by which higher-order chromatin remodeling is associated with gene activation and silencing during differentiation is not fully understood. In this study, we provide evidence that the euchromatic lysine methyltransferase GLP regulates heterochromatin organization and myogenic differentiation. Interestingly, GLP represses expression of the methyl-binding protein MeCP2 that induces heterochromatin clustering during differentiation. Consequently, MeCP2 and HP1γ localization at major satellites are altered upon modulation of GLP expression. In GLP knockdown cells, depletion of MeCP2 restored both chromatin organization and myogenic differentiation. These results identify a novel regulatory axis between a histone methylation writer and DNA methylation reader, which is important for heterochromatin organization during differentiation.


Assuntos
Heterocromatina/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Proteína 2 de Ligação a Metil-CpG/metabolismo , Desenvolvimento Muscular , Animais , Repetição de Anquirina , Diferenciação Celular , Linhagem Celular , Heterocromatina/ultraestrutura , Histona-Lisina N-Metiltransferase/análise , Proteína 2 de Ligação a Metil-CpG/análise , Camundongos , Domínios Proteicos
9.
Reproduction ; 154(1): 23-34, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28420800

RESUMO

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.


Assuntos
Células-Tronco Germinativas Adultas/fisiologia , Sobrevivência Celular/fisiologia , Histona-Lisina N-Metiltransferase/fisiologia , Sus scrofa , Animais , Animais Recém-Nascidos , Apoptose , Epigênese Genética , Expressão Gênica , Técnicas de Silenciamento de Genes , Histona Desmetilases/análise , Histona Desmetilases/fisiologia , Histona Metiltransferases , Histona-Lisina N-Metiltransferase/análise , Histona-Lisina N-Metiltransferase/genética , Masculino , Espermatogênese/fisiologia , Testículo/crescimento & desenvolvimento
10.
J Natl Cancer Inst ; 109(6)2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28131992

RESUMO

BACKGROUND: Pancreatic cancer is one of the cancers where anti-PD-L1/PD-1 immunotherapy has been unsuccessful. What confers pancreatic cancer resistance to checkpoint immunotherapy is unknown. The aim of this study is to elucidate the underlying mechanism of PD-L1 expression regulation in the context of pancreatic cancer immune evasion. METHODS: Pancreatic cancer mouse models and human specimens were used to determine PD-L1 and PD-1 expression and cancer immune evasion. Histone methyltransferase inhibitors, RNAi, and overexpression were used to elucidate the underlying molecular mechanism of PD-L1 expression regulation. All statistical tests were two-sided. RESULTS: PD-L1 is expressed in 60% to 90% of tumor cells in human pancreatic carcinomas and in nine of 10 human pancreatic cancer cell lines. PD-1 is expressed in 51.2% to 52.1% of pancreatic tumor-infiltrating cytotoxic T lymphocytes (CTLs). Tumors grow statistically significantly faster in FasL-deficient mice than in wild-type mice (P = .03-.001) and when CTLs are neutralized (P = .03-<.001). H3K4 trimethylation (H3K4me3) is enriched in the cd274 promoter in pancreatic tumor cells. MLL1 directly binds to the cd274 promoter to catalyze H3K4me3 to activate PD-L1 transcription in tumor cells. Inhibition or silencing of MLL1 decreases the H3K4me3 level in the cd274 promoter and PD-L1 expression in tumor cells. Accordingly, inhibition of MLL1 in combination with anti-PD-L1 or anti-PD-1 antibody immunotherapy effectively suppresses pancreatic tumor growth in a FasL- and CTL-dependent manner. CONCLUSIONS: The Fas-FasL/CTLs and the MLL1-H3K4me3-PD-L1 axis play contrasting roles in pancreatic cancer immune surveillance and evasion. Targeting the MLL1-H3K4me3 axis is an effective approach to enhance the efficacy of checkpoint immunotherapy against pancreatic cancer.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Carcinoma/metabolismo , Carcinoma/terapia , Histona-Lisina N-Metiltransferase/metabolismo , Proteína de Leucina Linfoide-Mieloide/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/terapia , Receptor de Morte Celular Programada 1/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Antígeno B7-H1/análise , Antígeno B7-H1/imunologia , Carcinoma/genética , Carcinoma/imunologia , Linhagem Celular Tumoral , Metilação de DNA/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Epigênese Genética , Proteína Ligante Fas/genética , Feminino , Histona-Lisina N-Metiltransferase/análise , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Histona-Lisina N-Metiltransferase/genética , Humanos , Imunoterapia , Indóis/farmacologia , Indóis/uso terapêutico , Camundongos , Camundongos Endogâmicos C57BL , Proteína de Leucina Linfoide-Mieloide/análise , Proteína de Leucina Linfoide-Mieloide/antagonistas & inibidores , Proteína de Leucina Linfoide-Mieloide/genética , Transplante de Neoplasias , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/imunologia , Piperazinas/farmacologia , Receptor de Morte Celular Programada 1/análise , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Linfócitos T Citotóxicos/química , Evasão Tumoral , Microambiente Tumoral/imunologia
11.
Mol Oncol ; 10(10): 1497-1515, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27717710

RESUMO

Histone methyltransferases (HMTs) catalyze the methylation of lysine and arginine residues on histone tails and non-histone targets. These important post-translational modifications are exquisitely regulated and affect chromatin compaction and transcriptional programs leading to diverse biological outcomes. There is accumulating evidence that genetic alterations of several HMTs impinge on oncogenic or tumor-suppressor functions and influence both cancer initiation and progression. HMTs therefore represent an opportunity for therapeutic targeting in those patients with tumors in which HMTs are dysregulated, to reverse the histone marks and transcriptional programs associated with aggressive tumor behavior. In this review, we describe the known histone methyltransferases and their emerging roles in breast cancer tumorigenesis.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Mama/patologia , Regulação Neoplásica da Expressão Gênica , Histona-Lisina N-Metiltransferase/genética , Proteína-Arginina N-Metiltransferases/genética , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Mama/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Descoberta de Drogas , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Epigênese Genética/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Código das Histonas/efeitos dos fármacos , Histona-Lisina N-Metiltransferase/análise , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Terapia de Alvo Molecular , Proteína-Arginina N-Metiltransferases/análise , Proteína-Arginina N-Metiltransferases/antagonistas & inibidores , Proteína-Arginina N-Metiltransferases/metabolismo , Ativação Transcricional/efeitos dos fármacos
12.
Biochem Biophys Res Commun ; 458(3): 525-530, 2015 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-25677622

RESUMO

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.


Assuntos
Fissura Palatina/induzido quimicamente , Overdose de Drogas/complicações , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Histona-Lisina N-Metiltransferase/genética , Palato/embriologia , Tretinoína/efeitos adversos , Animais , Linhagem Celular , Fissura Palatina/genética , Fissura Palatina/metabolismo , Regulação para Baixo/efeitos dos fármacos , Feminino , Histona-Lisina N-Metiltransferase/análise , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Metilação/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Palato/anormalidades , RNA Mensageiro/genética
13.
Histochem Cell Biol ; 143(3): 259-66, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25294562

RESUMO

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.


Assuntos
Perfilação da Expressão Gênica , Regulação Enzimológica da Expressão Gênica , Histona-Lisina N-Metiltransferase/biossíntese , Histona-Lisina N-Metiltransferase/genética , Germe de Dente/enzimologia , Germe de Dente/crescimento & desenvolvimento , Animais , Histona Metiltransferases , Histona-Lisina N-Metiltransferase/análise , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Endogâmicos C57BL
14.
J Vis Exp ; (93): e52203, 2014 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-25489813

RESUMO

Lysine methylation is an emerging post-translation modification and it has been identified on several histone and non-histone proteins, where it plays crucial roles in cell development and many diseases. Approximately 5,000 lysine methylation sites were identified on different proteins, which are set by few dozens of protein lysine methyltransferases. This suggests that each PKMT methylates multiple proteins, however till now only one or two substrates have been identified for several of these enzymes. To approach this problem, we have introduced peptide array based substrate specificity analyses of PKMTs. Peptide arrays are powerful tools to characterize the specificity of PKMTs because methylation of several substrates with different sequences can be tested on one array. We synthesized peptide arrays on cellulose membrane using an Intavis SPOT synthesizer and analyzed the specificity of various PKMTs. Based on the results, for several of these enzymes, novel substrates could be identified. For example, for NSD1 by employing peptide arrays, we showed that it methylates K44 of H4 instead of the reported H4K20 and in addition H1.5K168 is the highly preferred substrate over the previously known H3K36. Hence, peptide arrays are powerful tools to biochemically characterize the PKMTs.


Assuntos
Histona-Lisina N-Metiltransferase/análise , Histona-Lisina N-Metiltransferase/metabolismo , Oligopeptídeos/química , Análise Serial de Proteínas/métodos , Sequência de Aminoácidos , Histonas/química , Lisina/química , Metilação , Oligopeptídeos/síntese química , Especificidade por Substrato
15.
Int J Oral Sci ; 6(4): 205-11, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25394593

RESUMO

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.


Assuntos
Histonas/metabolismo , Odontogênese/fisiologia , Processamento de Proteína Pós-Traducional/fisiologia , Animais , Diferenciação Celular/fisiologia , Proteínas de Ligação a DNA/análise , Papila Dentária/embriologia , Embrião de Mamíferos , Órgão do Esmalte/embriologia , Proteína Potenciadora do Homólogo 2 de Zeste , Epigênese Genética/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Histona-Lisina N-Metiltransferase/análise , Histona Desmetilases com o Domínio Jumonji/análise , Lisina/metabolismo , Metilação , Camundongos , Camundongos Endogâmicos BALB C , Complexo Repressor Polycomb 2/análise , Germe de Dente/embriologia
16.
Anal Biochem ; 467: 14-21, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25172130

RESUMO

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.


Assuntos
Catecol O-Metiltransferase/análise , Proteínas de Ligação a DNA/análise , Polarização de Fluorescência/métodos , Histona-Lisina N-Metiltransferase/análise , Hidrolases/análise , Proteínas Nucleares/análise , S-Adenosil-Homocisteína/metabolismo , S-Adenosilmetionina/metabolismo , Fatores de Transcrição/análise , Catecol O-Metiltransferase/metabolismo , Proteínas de Ligação a DNA/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Hidrolases/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo
17.
Int J Clin Exp Pathol ; 7(5): 2496-507, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24966962

RESUMO

Hepatocellular carcinoma (HCC) is the world's fifth most common cancer and second leading cause of cancer-related death in Taiwan. Over 600,000 HCC patients die each year worldwide despite recent advances in surgical techniques and medical treatments. Epigenetic regulations including DNA methylation and histone modification control gene expressions and play important roles during tumorigenesis. This study evaluates association between histone-modifying genes and prognosis of HCC to ferret out new diagnostic markers. We collected 50 paired HCC and adjacent non-cancerous tissues from Taiwanese patients for survey by RT-qPCR and tissue microarray-based immunohistochemistry (TMA-based IHC) staining. RT-qPCR data showed four of twenty-four genes over eightfold up-regulated in tumor tissues: e.g., histone phosphorylation gene-ARK2, methylation genes-G9a, SUV39H2, and EZH2 (n=50, all p<0.0001). Results of TMA-based IHC staining showed proteins of ARK2, EZH2, G9a, and SUV39H2 also overexpressed in tumor tissues. Staining intensity of SUV39H2 correlated with HCV infection (p=0.025). We further restricted the analysis only in tumor tissues, we found EZH2 staining intensity associated with tumor stage (p=0.016) and survival (p=0.007); SUV39H2 intensity associated with tumor stage (p=0.044). Our findings indicate overexpression of histone-modifying genes EZH2 and SUV39H2 associated with prognosis of HCC cases. EZH2 expression can serve as a novel prognostic biomarker during HCC progression among Taiwanese.


Assuntos
Biomarcadores Tumorais/análise , Carcinoma Hepatocelular/enzimologia , Histona-Lisina N-Metiltransferase/análise , Histonas/metabolismo , Neoplasias Hepáticas/enzimologia , Complexo Repressor Polycomb 2/análise , Biomarcadores Tumorais/genética , Biópsia , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/mortalidade , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/virologia , Proteína Potenciadora do Homólogo 2 de Zeste , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Hepacivirus/isolamento & purificação , Histona-Lisina N-Metiltransferase/genética , Humanos , Imuno-Histoquímica , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/mortalidade , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/virologia , Metilação , Gradação de Tumores , Estadiamento de Neoplasias , Fosforilação , Complexo Repressor Polycomb 2/genética , Valor Preditivo dos Testes , Processamento de Proteína Pós-Traducional , RNA Mensageiro/análise , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Risco , Taiwan , Fatores de Tempo , Análise Serial de Tecidos , Regulação para Cima
18.
Biochem Biophys Res Commun ; 444(4): 634-7, 2014 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-24486544

RESUMO

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.


Assuntos
Proteínas de Transporte/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Proteínas de Homeodomínio/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Proteínas de Transporte/análise , Proteínas de Ligação a DNA/metabolismo , Células HEK293 , Histona-Lisina N-Metiltransferase/análise , Histonas/metabolismo , Proteínas de Homeodomínio/análise , Humanos , Proteínas Nucleares/análise , Regiões Promotoras Genéticas , Mapas de Interação de Proteínas , Fatores de Transcrição/análise , Proteína Homeobox PITX2
19.
ChemMedChem ; 9(3): 554-9, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23671032

RESUMO

New protein lysine methyltransferase (PKMT) assays are needed to facilitate screening for improved PKMT inhibitors, because PKMTs are mutated or overexpressed in several cancers. In cells, methylated lysine residues are recognized by reading domains such as the chromodomain of HP1ß, which bind to target proteins in a lysine-methylation-specific manner. Herein we describe a sensitive, robust, and non-radioactive high-throughput PKMT assay that employs the HP1ß chromodomain to detect the methylation of peptide substrates by the human SUV39H1 and SUV39H2 PKMTs. The assay has a very good dynamic range and high signal-to-noise ratio. It can be used to screen for PKMT inhibitors, as illustrated by analyzing the inhibition of SUV39H1 by chaetocin. The IC50 value of this inhibition was found to be 480 nM, which is close to its published value. Our data indicate that natural reading domains can be used as alternates to methyl-specific antibodies in PKMT assays. Reading domains can be produced recombinantly in E. coli at low cost and consistent quality, and they are accessible to protein design.


Assuntos
Ensaios Enzimáticos/métodos , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Histona-Lisina N-Metiltransferase/metabolismo , Homólogo 5 da Proteína Cromobox , Histona-Lisina N-Metiltransferase/análise , Humanos , Concentração Inibidora 50 , Piperazinas/química , Piperazinas/farmacologia , Relação Estrutura-Atividade
20.
Anal Biochem ; 443(2): 214-21, 2013 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-24018340

RESUMO

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.


Assuntos
Cromatografia Líquida/métodos , Descoberta de Drogas/métodos , Histona-Lisina N-Metiltransferase/análise , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Ensaios Enzimáticos/métodos , Histona Metiltransferases , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , S-Adenosil-Homocisteína/análise , S-Adenosil-Homocisteína/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...