Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Cardiovasc Pharmacol ; 81(2): 150-164, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36607630

RESUMO

ABSTRACT: Uric acid (UA) accumulation triggers endothelial dysfunction, oxidative stress, and inflammation. Histone deacetylase (HDAC) plays a vital role in regulating the pathological processes of various diseases. However, the influence of HDAC inhibitor on UA-induced vascular endothelial cell injury (VECI) remains undefined. Hence, this study aimed to investigate the effect of HDACs inhibition on UA-induced vascular endothelial cell dysfunction and its detailed mechanism. UA was used to induce human umbilical vein endothelial cell (HUVEC) injury. Meanwhile, potassium oxonate-induced and hypoxanthine-induced hyperuricemia mouse models were also constructed. A broad-spectrum HDAC inhibitor trichostatin A (TSA) or selective HDAC6 inhibitor TubastatinA (TubA) was given to HUVECs or mice to determine whether HDACs can affect UA-induced VECI. The results showed pretreatment of HUVECs with TSA or HDAC6 knockdown-attenuated UA-induced VECI and increased FGF21 expression and phosphorylation of AKT, eNOS, and FoxO3a. These effects could be reversed by FGF21 knockdown. In vivo, both TSA and TubA reduced inflammation and tissue injury while increased FGF21 expression and phosphorylation of AKT, eNOS, and FoxO3a in the aortic and renal tissues of hyperuricemia mice. Therefore, HDACs, especially HDAC6 inhibitor, alleviated UA-induced VECI through upregulating FGF21 expression and then activating the PI3K/AKT pathway. This suggests that HDAC6 may serve as a novel therapeutic target for treating UA-induced endothelial dysfunction.


Assuntos
Inibidores de Histona Desacetilases , Hiperuricemia , Animais , Humanos , Camundongos , Desacetilase 6 de Histona/metabolismo , Desacetilase 6 de Histona/farmacologia , Inibidores de Histona Desacetilases/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Células Endoteliais da Veia Umbilical Humana , Inflamação/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Ácido Úrico
2.
Molecules ; 27(14)2022 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-35889201

RESUMO

In this study, we report on the synthesis of new organoselenium derivatives, including nonsteroidal anti-inflammatory drugs (NSAIDs) scaffolds and Se functionalities (isoselenocyanate and selenourea), which were evaluated against four types of cancer cell line: SW480 (human colon adenocarcinoma cells), HeLa (human cervical cancer cells), A549 (human lung carcinoma cells), MCF-7 (human breast adenocarcinoma cells). Among these compounds, most of the investigated compounds reduced the viability of different cancer cell lines. The most promising compound 6b showed IC50 values under 10 µM against the four cancer cell lines, particularly to HeLa and MCF-7, with IC50 values of 2.3 and 2.5 µM, respectively. Furthermore, two compounds, 6b and 6f, were selected to investigate their ability to induce apoptosis in MCF-7 cells via modulation of the expression of anti-apoptotic Bcl-2 protein, pro-inflammatory cytokines (IL-2) and proapoptotic caspase-3 protein. The redox properties of the NSAIDs-Se derivatives were conducted by 2, 2-didiphenyl-1-picrylhydrazyl (DPPH), bleomycin-dependent DNA damage and glutathione peroxidase (GPx)-like assays. Finally, a molecular docking study revealed that an interaction with the active site of thioredoxin reductase 1 (TrxR1) predicted the antiproliferative activity of the synthesized candidates. Overall, these results could serve as a promising launch point for further designs of NSAIDs-Se derivatives as potential antiproliferative agents.


Assuntos
Adenocarcinoma , Antineoplásicos , Neoplasias do Colo , Anti-Inflamatórios não Esteroides/farmacologia , Antineoplásicos/química , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Simulação de Acoplamento Molecular , Estrutura Molecular , Compostos Organosselênicos , Relação Estrutura-Atividade , Ureia/análogos & derivados
3.
Cytometry A ; 101(3): 254-263, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34448526

RESUMO

The potential of flow cytometry for the study of changes in prodigiosin on the cell surface of Serratia marcescens is of academic and practical interest. This is because S. marcescens can produce prodigiosin, a secondary metabolite, with potential use as a cancer-cell inhibitor. In this study, three groups of bacterial cultures with different carbon sources were compared, and the effect of the addition of cAMP to the sucrose-based culture was studied. Both cellular morphology and DNA content were detected by flow cytometry, rendering a broad description of the bacterial behavior. It is the first use of flow cytometry to investigate the dynamics of prodigiosin on the surface of S. marcescens during growth in different media. The fluorescence intensity is related to the DNA content, the forward-scattered light is related to cell volume, and the side-scattered light is related to the surface morphology, especially the surface prodigiosin. These may contribute to the potential development of a bacterial metabolic monitoring strategy using both DNA content analysis and bacterial morphology based on flow cytometry technique.


Assuntos
Prodigiosina , Serratia marcescens , Meios de Cultura/metabolismo , DNA/metabolismo , Citometria de Fluxo , Prodigiosina/metabolismo , Prodigiosina/farmacologia , Serratia marcescens/genética , Serratia marcescens/metabolismo
4.
Nat Commun ; 12(1): 2693, 2021 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-33976158

RESUMO

Notch signaling represents a key mechanism mediating cancer metastasis and stemness. To understand how Notch signaling is overactivated to couple tumor metastasis and self-renewal in NSCLC cells, we performed the current study and showed that RFC4, a DNA replication factor amplified in more than 40% of NSCLC tissues, directly binds to the Notch1 intracellular domain (NICD1) to competitively abrogate CDK8/FBXW7-mediated degradation of NICD1. Moreover, RFC4 is a functional transcriptional target gene of Notch1 signaling, forming a positive feedback loop between high RFC4 and NICD1 levels and sustained overactivation of Notch signaling, which not only leads to NSCLC tumorigenicity and metastasis but also confers NSCLC cell resistance to treatment with the clinically tested drug DAPT against NICD1 synthesis. Furthermore, together with our study, analysis of two public datasets involving more than 1500 NSCLC patients showed that RFC4 gene amplification, and high RFC4 and NICD1 levels were tightly correlated with NSCLC metastasis, progression and poor patient prognosis. Therefore, our study characterizes the pivotal roles of the positive feedback loop between RFC4 and NICD1 in coupling NSCLC metastasis and stemness properties and suggests its therapeutic and diagnostic/prognostic potential for NSCLC therapy.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/genética , Receptor Notch1/genética , Proteína de Replicação C/genética , Transdução de Sinais/genética , Células A549 , Animais , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/terapia , Linhagem Celular Tumoral , Retroalimentação Fisiológica , Feminino , Células HEK293 , Humanos , Estimativa de Kaplan-Meier , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/terapia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Metástase Neoplásica , Receptor Notch1/metabolismo , Proteína de Replicação C/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
5.
Oncogene ; 40(19): 3449-3459, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33859372

RESUMO

Long non-coding RNAs (lncRNAs) are emerging as a new class of regulators for a variety of biological processes and have been suggested to play pivotal roles in cancer development and progression. Our current study found that a lncRNA, designated enhancing IL-6/STAT3 signaling activation (LEISA, ENST00000603468), functioned as an oncogenic lncRNA in lung adenocarcinoma (LAD), a major form of non-small cell lung carcinoma, which is one of the most frequently diagnosed malignancies with high morbidity and mortality worldwide, and was involved in the regulation of STAT3 induced IL-6 transcription. Our data showed that LEISA was highly expressed in, and correlated with the clinical progression and prognosis of LAD. Ectopic expression of LEISA promoted the proliferation and suppressed apoptosis of LAD cells in vitro and in vivo. Mechanistically, we demonstrated that LEISA recruited STAT3 to bind the promoter of IL-6 and upregulated IL-6 expression. Taken together, our work identifies LEISA as a potential diagnostic biomarker and therapeutic target for LAD.


Assuntos
Adenocarcinoma de Pulmão/genética , Interleucina-6/genética , Neoplasias Pulmonares/genética , RNA Longo não Codificante/genética , Fator de Transcrição STAT3/genética , Adenocarcinoma de Pulmão/metabolismo , Adenocarcinoma de Pulmão/patologia , Animais , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Progressão da Doença , Xenoenxertos , Humanos , Interleucina-6/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Regiões Promotoras Genéticas , Fator de Transcrição STAT3/metabolismo , Taxa de Sobrevida
6.
Chem Biodivers ; 17(5): e1900603, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32198823

RESUMO

In the present study, twenty-four selenocyanate and diselenide compounds were synthesized and characterized, and their anticancer activities against the human cancer cell lines Caco2, BGC-823, MCF-7 and PC-3 were determined. Interestingly, most of the new compounds were active in reducing the viability of different cancer cell lines. Two compounds exhibited higher promising activities than other derivatives. The most active compound showed the least IC50 values against the four cancer cell lines, particularly to PC-3 with IC50 values below 5 µm. Two compounds were selected to monitor the expression levels of Bcl-2, IL-2 and caspase-3 molecular biomarkers. Interestingly, the two compounds downregulated the Bcl-2 expression levels and upregulated the expression of IL-2 and caspase-3 in PC-3 cells compared to untreated cells. Moreover, most of the synthesized organoselenides exhibited good Gpx-like activities comparable to ebselen. These results appear that introduction of selenocyanate (-SeCN) or diselenides (-Se-Se-) moiety to some carboxy derivatives could serve as a promising launch point for the further design of this type of organic selenium anticancer agent.


Assuntos
Antineoplásicos/farmacologia , Cianatos/farmacologia , Compostos Organosselênicos/farmacologia , Compostos de Selênio/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/química , Proliferação de Células/efeitos dos fármacos , Cianatos/síntese química , Cianatos/química , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Estrutura Molecular , Compostos Organosselênicos/síntese química , Compostos Organosselênicos/química , Compostos de Selênio/síntese química , Compostos de Selênio/química , Relação Estrutura-Atividade , Células Tumorais Cultivadas
7.
Bioresour Technol ; 147: 307-314, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24001560

RESUMO

Oil palm empty fruit bunch (OPEFB), contains abundant cellulose and hemicelluloses and can be used as a renewable resource for fuel and chemical production. This study, as the first attempt, aims to convert OPEFB derived sugars to polyhydroxybutyrate (PHB). OPEFB collected from a Malaysia palm oil refinery plant was chemically pretreated and enzymatically hydrolyzed by an in-house prepared cellulase cocktail. The PHB producer, Bacillus megaterium R11, was isolated in Singapore and could accumulate PHB up to 51.3% of its cell dry weight (CDW) from both glucose and xylose. Tryptone was identified as its best nitrogen source. PHB content and production reached 58.5% and 9.32 g/L, respectively, for an overall OPEFB sugar concentration of 45 g/L. These respectively reached 51.6% and 12.48 g/L for OPEFB hydrolysate containing 60 g/L sugar with a productivity of 0.260 g/L/h.


Assuntos
Arecaceae/metabolismo , Bacillus megaterium/metabolismo , Butiratos/metabolismo , Poli-Hidroxialcanoatos/metabolismo , Meios de Cultura
8.
Nat Commun ; 4: 1662, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23575666

RESUMO

Toll-like receptor 4 (Tlr4) has a pivotal role in innate immune responses, and the transcription factor CCAAT/enhancer binding protein delta (C/EBPδ, Cebpd) is a Tlr4-induced gene. Here we identify a positive feedback loop in which C/EBPδ activates Tlr4 gene expression in macrophages and tumour cells. In addition, we discovered a negative feedback loop whereby the tumour suppressor FBXW7α (FBW7, Cdc4), whose gene expression is inhibited by C/EBPδ, targets C/EBPδ for degradation when C/EBPδ is phosphorylated by GSK-3ß. Consequently, FBXW7α suppresses Tlr4 expression and responses to the ligand lipopolysaccharide. FBXW7α depletion alone is sufficient to augment pro-inflammatory signalling in vivo. Moreover, as inflammatory pathways are known to modulate tumour biology, Cebpd null mammary tumours, which have reduced metastatic potential, show altered expression of inflammation-associated genes. Together, these findings reveal a role for C/EBPδ upstream of Tlr4 signalling and uncover a function for FBXW7α as an attenuator of inflammatory signalling.


Assuntos
Proteína delta de Ligação ao Facilitador CCAAT/fisiologia , Regulação para Baixo , Proteínas F-Box/fisiologia , Inflamação/fisiopatologia , Transdução de Sinais/fisiologia , Receptor 4 Toll-Like/fisiologia , Ubiquitina-Proteína Ligases/fisiologia , Animais , Linhagem Celular Tumoral , Proteínas F-Box/genética , Proteína 7 com Repetições F-Box-WD , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Inflamação/genética , Camundongos , RNA Mensageiro/genética , Ubiquitina-Proteína Ligases/genética
9.
Proc Natl Acad Sci U S A ; 107(20): 9210-5, 2010 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-20439707

RESUMO

The transcription factor CCAAT/enhancer binding protein delta (C/EBPdelta, CEBPD, NFIL-6beta) has tumor suppressor function; however, the molecular mechanism(s) by which C/EBPdelta exerts its effect are largely unknown. Here, we report that C/EBPdelta induces expression of the Cdc27 (APC3) subunit of the anaphase promoting complex/cyclosome (APC/C), which results in the polyubiquitination and degradation of the prooncogenic cell cycle regulator cyclin D1, and also down-regulates cyclin B1, Skp2, and Plk-1. In C/EBPdelta knockout mouse embryo fibroblasts (MEF) Cdc27 levels were reduced, whereas cyclin D1 levels were increased even in the presence of activated GSK-3beta. Silencing of C/EBPdelta, Cdc27, or the APC/C coactivator Cdh1 (FZR1) in MCF-10A breast epithelial cells increased cyclin D1 protein expression. Like C/EBPdelta, and in contrast to cyclin D1, Cdc27 was down-regulated in several breast cancer cell lines, suggesting that Cdc27 itself may be a tumor suppressor. Cyclin D1 is a known substrate of polyubiquitination complex SKP1/CUL1/F-box (SCF), and our studies show that Cdc27 directs cyclin D1 to alternative degradation by APC/C. These findings shed light on the role and regulation of APC/C, which is critical for most cellular processes.


Assuntos
Neoplasias da Mama/metabolismo , Proteína delta de Ligação ao Facilitador CCAAT/metabolismo , Proteínas de Ciclo Celular/metabolismo , Ciclina D1/metabolismo , Regulação Neoplásica da Expressão Gênica/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Animais , Subunidade Apc3 do Ciclossomo-Complexo Promotor de Anáfase , Western Blotting , Proteína delta de Ligação ao Facilitador CCAAT/genética , Linhagem Celular Tumoral , Ciclina B1/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Imunoprecipitação , Camundongos , Camundongos Knockout , Microscopia de Fluorescência , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Quinases Associadas a Fase S/metabolismo , Quinase 1 Polo-Like
10.
Zhonghua Zhong Liu Za Zhi ; 31(2): 90-4, 2009 Feb.
Artigo em Chinês | MEDLINE | ID: mdl-19538881

RESUMO

OBJECTIVE: To investigate the inhibitory effects of an antisense PC cell derived growth factor (PCDGF) vector on proliferation and invasion of highly malignant ovarian cancer cell lines Sw626 and A2780 cells, and preliminarily explore the related mechanisms. METHODS: MTT assay and Boyden chamber in vitro invasion assay were employed to detect the changes of proliferation and invasion ability in the Sw626 and A2780 cells transfected with anti-sense PCDGF. The expression levels of cyclin D1 and CDK4 proteins before and after transfection were detected by Western blotting. The effects on the expression and activity of MMP-2 were evaluated by quantitative RT-PCR and zymography, respectively. RESULTS: Comparing with the blank group, the proliferation inhibition rate of the Sw626 and A2780 cells transfected with anti-sense PCDGF was 72.9% and 70.9%, respectively, and the invasion ability was inhibited by 62.9% and 59.0%, respectively. The levels of cyclin D1 and CDK4 protein expression in antisense PCDGF transfected cells were 0.38 +/- 0.08 and 0.37 +/- 0.13, respectively, all significantly lower than 0.84 +/- 0.11 and 0.64 +/- 0.11, respectively, in the blank group (P < 0.01). The MMP-2 mRNA expression level in antisense PCDGF transfected cell group was 0.66 +/- 0.11, not significantly decreased in comparison with 0.89 +/- 0.09 in the blank group (P > 0.05), but the activity of MMP-2 was inhibited significantly. CONCLUSION: The antisense PCDGF vector may inhibit markedly the proliferation and invasion of highly malignant ovarian cancer cells, and partially reverses their malignant phenotype. It seems to be related with down-regulating the expression of cyclin D1 and CDK4 and inhibiting the activity of MMP-2. Our findings indicate that PCDGF may become a new target for antisense gene therapy of ovarian cancer.


Assuntos
Adesão Celular , Proliferação de Células , DNA Antissenso , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neoplasias Ovarianas/patologia , Linhagem Celular Tumoral , Ciclina D1/metabolismo , Quinase 4 Dependente de Ciclina/metabolismo , Regulação para Baixo , Feminino , Vetores Genéticos , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Invasividade Neoplásica , Neoplasias Ovarianas/metabolismo , Progranulinas , RNA Mensageiro/metabolismo , Transfecção
11.
Leuk Res ; 31(6): 839-51, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17023045

RESUMO

The acute myeloid leukemia 1 (AML1, RUNX1) transcription factor is a key regulator of hematopoietic differentiation both in embryonic stem cells and mature hematopoietic progenitors. The RUNX1 protein is thought to play a role in the control of progression through the cell cycle. We have shown that post-transcriptional regulation of RUNX1 activity occurs, in part, through phosphorylation. To investigate whether transit through the cell cycle is associated with changes in the phosphorylation of RUNX1, we have derived phospho-specific antibodies against three of the five major phosphorylation sites in the transcriptional activation domain of RUNX1, S276, S303 and S462. Using these antibodies we demonstrate that treatment of Jurkat T-cells with nocodazole, a G2/M blocking compound, causes an increase in phosphorylation of these three amino acids. By elutriating the Jurkat cells, we are able to demonstrate that these amino acids are normally phosphorylated at the G2/M phase of the cell cycle. Using in vivo inhibitors and in vitro assays this phosphorylation appears to be dependent on Cdk1. We find that RUNX1 degradation occurs at the G2/M-G1 transition and is regulated by both Cdc20 and phosphoryation, suggesting that the anaphase promoting complex plays a role in modifying the level of this protein. Regulation of the extent of phosphorylation of RUNX1 may play a role in controlling the degradation of the protein, implying that additional E3 ligases may also be involved.


Assuntos
Divisão Celular/fisiologia , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Fase G2/fisiologia , Processamento de Proteína Pós-Traducional/fisiologia , Antineoplásicos/farmacologia , Proteínas Cdc20 , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Divisão Celular/efeitos dos fármacos , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Fase G2/efeitos dos fármacos , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Células Jurkat , Nocodazol/farmacologia , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Ubiquitina-Proteína Ligases/metabolismo
12.
Mol Cell Biol ; 26(20): 7420-9, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17015473

RESUMO

AML1 (RUNX1) regulates hematopoiesis, angiogenesis, muscle function, and neurogenesis. Previous studies have shown that phosphorylation of AML1, particularly at serines 276 and 303, affects its transcriptional activation. Here, we report that phosphorylation of AML1 serines 276 and 303 can be blocked in vivo by inhibitors of the cyclin-dependent kinases (CDKs) Cdk1 and Cdk2. Furthermore, these residues can be phosphorylated in vitro by purified Cdk1/cyclin B and Cdk2/cyclin A. Mutant AML1 protein which cannot be phosphorylated at these sites (AML1-4A) is more stable than wild-type AML1. AML-4A is resistant to degradation mediated by Cdc20, one of the substrate-targeting subunits of the anaphase-promoting complex (APC). However, Cdh1, another targeting subunit used by the APC, can mediate the degradation of AML1-4A. A phospho-mimic protein, AML1-4D, can be targeted by Cdc20 or Cdh1. These observations suggest that both Cdc20 and Cdh1 can target AML1 for degradation by the APC but that AML1 phosphorylation may affect degradation mediated by Cdc20-APC to a greater degree.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Quinases Ciclina-Dependentes/metabolismo , Complexos Ubiquitina-Proteína Ligase/metabolismo , Ciclossomo-Complexo Promotor de Anáfase , Animais , Caderinas/genética , Caderinas/metabolismo , Ciclo Celular , Linhagem Celular , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Quinases Ciclina-Dependentes/antagonistas & inibidores , Ciclinas/metabolismo , Humanos , Camundongos , Mutação/genética , Fosforilação/efeitos dos fármacos , Ligação Proteica , Inibidores de Proteínas Quinases/farmacologia , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Proteínas Quinases Associadas a Fase S/genética , Proteínas Quinases Associadas a Fase S/metabolismo
13.
Mol Cancer Res ; 3(7): 391-401, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16046550

RESUMO

The acute myeloid leukemia 1 (AML1) transcription factors are key regulators of hematopoietic differentiation. Cellular AML1c protein is found in the nucleus and can be separated into two fractions, one soluble in buffers containing salt and nonionic detergent and the other insoluble and tightly bound to the nuclear matrix. We find that the AML1c protein is modified by both phosphorylation and ubiquitination. Our studies show that the majority of the ubiquitinated AML1c is associated with the insoluble nuclear matrix. Treatment of cells with the proteasome inhibitor PS341 (Velcade, Bortezomib) increases the levels of ubiquitinated AML1c. Mutation of the four phosphorylation sites necessary for transcriptional regulation (serine 276, serine 293, serine 303, and threonine 300) mimics the effects of the proteasome inhibitor, increasing the levels of ubiquitinated, matrix-bound AML1c. We find that the soluble and insoluble forms of AML1c are degraded at a similar rate. However, mutation of these four serine/threonine residues statistically increases the half-life of the matrix-associated AML1c. Thus, phosphorylation of AML1c on specific serine/threonine residues controls both transcriptional activity and rate of degradation.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Matriz Nuclear/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Animais , Ácidos Borônicos/farmacologia , Bortezomib , Subunidade alfa 2 de Fator de Ligação ao Core , Proteínas de Ligação a DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/genética , Imunoprecipitação , Camundongos , Camundongos Knockout , Mutação , Fosforilação , Inibidores de Proteases/farmacologia , Proteínas Proto-Oncogênicas/efeitos dos fármacos , Proteínas Proto-Oncogênicas/genética , Pirazinas/farmacologia , Serina/genética , Treonina/genética , Fatores de Transcrição/efeitos dos fármacos , Fatores de Transcrição/genética , Ubiquitina/metabolismo
14.
J Biol Chem ; 279(51): 53116-25, 2004 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-15475366

RESUMO

We find that phorbol ester (PE) treatment of K562 cells greatly stimulates promoters (T cell receptor beta, myeloperoxidase, macrophage colony-stimulating factor receptor, and granulocyte macrophage colony-stimulating factor receptor) containing AML1 transcription factor binding sites. This stimulation of AML1c transcriptional activity is mediated by direct phosphorylation of the AML1c molecule on multiple phosphorylation sites. Eleven AML1c (S/T)P sites in the transcriptional activating domain are phosphorylated at a basal level in untreated K562 cells; treatment of the K562 cells with PE results in increased phosphorylation at five of these sites (serines 276, 293, 303, 462, and threonine 300). Mutation of these five sites to alanine inhibits PE-induced transcriptional activity; mutation of the sites to an acidic amino acid, aspartic acid, stimulates constitutive activity. Single mutations in four amino acids or double mutations (serines 276 and 293 or threonine 300 and serine 303) have little effect on AML1c transcriptional activity. Inhibitor assays suggest that the ERK family of protein kinases is activated by PEs to phosphorylate the (S/T)P sites within the AML1c molecule and markedly enhance the transcriptional activity of AML1c.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Ésteres de Forbol/farmacologia , Proteínas Proto-Oncogênicas/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Ácido Aspártico/química , Sítios de Ligação , Western Blotting , Diferenciação Celular , Subunidade alfa 2 de Fator de Ligação ao Core , Vetores Genéticos , Humanos , Imunoprecipitação , Células K562 , Luciferases/metabolismo , Mutação , Fosforilação , Regiões Promotoras Genéticas , Estrutura Terciária de Proteína , Serina/química , Treonina/química , Fatores de Tempo , Ativação Transcricional , Células U937
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA