Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Mol Biol ; 434(5): 167426, 2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-34973238

RESUMO

FOXM1 is an essential proliferation-associated transcription factor that controls the activation of a number of cell cycle regulatory genes. Unlike other forkhead box (FOX) transcription factors, FOXM1 has been shown to prefer binding tandem regulatory DNA sites. However, the underlying reason for such preference is not clear. Here, we showed that the tandem DNA motif, named DIV2, is widely distributed in the promoter region of FOXM1 target genes. The binding of FOXM1 on the DIV2 site differs dramatically from other sites, which is in a highly cooperative fashion, with a much enhanced thermal stability and can be clearly detected by EMSA. The crystal structure of FOXM1 in complex with the DIV2 DNA reveals that the cooperative binding is likely to be driven by intermolecular protein-protein interactions (PPIs). Further half-site spacer insertion assays showed that FOXM1 can bind another site, DIV0, in a similar manner to the DIV2 site. Given the high occurrence of the DIV2 and DIV0 sites in FOXM1 target genes, our results suggest that FOXM1 prefers tandem DNA sites to enable cooperative DNA recognition, and such binding characteristics may further confer its specificity during transcriptional regulation.


Assuntos
Proteína Forkhead Box M1 , Regiões Promotoras Genéticas , Sítios de Ligação , Cristalografia , DNA/química , Proteína Forkhead Box M1/química , Proteína Forkhead Box M1/genética , Regulação Neoplásica da Expressão Gênica , Regiões Promotoras Genéticas/genética
2.
J Exp Clin Cancer Res ; 38(1): 313, 2019 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-31311575

RESUMO

BACKGROUND: Breast cancer is the most common malignancy in women worldwide. Among the breast cancer subtypes, triple-negative breast cancer (TNBC) is the most aggressive and the most difficult to treat. One of the master regulators in TNBC progression is the architectural transcription factor HMGA1. This study aimed to further explore the HMGA1 molecular network to identify molecular mechanisms involved in TNBC progression. METHODS: RNA from the MDA-MB-231 cell line, silenced for HMGA1 expression, was sequenced and, with a bioinformatic analysis, molecular partners HMGA1 could cooperate with in regulating common downstream gene networks were identified. Among the putative partners, the FOXM1 transcription factor was selected. The relationship occurring between HMGA1 and FOXM1 was explored by qRT-PCR, co-immunoprecipitation and protein stability assays. Subsequently, the transcriptional activity of HMGA1 and FOXM1 was analysed by luciferase assay on the VEGFA promoter. The impact on angiogenesis was assessed in vitro, evaluating the tube formation ability of endothelial cells exposed to the conditioned medium of MDA-MB-231 cells silenced for HMGA1 and FOXM1 and in vivo injecting MDA-MB-231 cells, silenced for the two factors, in zebrafish larvae. RESULTS: Here, we discover FOXM1 as a novel molecular partner of HMGA1 in regulating a gene network implicated in several breast cancer hallmarks. HMGA1 forms a complex with FOXM1 and stabilizes it in the nucleus, increasing its transcriptional activity on common target genes, among them, VEGFA, the main inducer of angiogenesis. Furthermore, we demonstrate that HMGA1 and FOXM1 synergistically drive breast cancer cells to promote tumor angiogenesis both in vitro in endothelial cells and in vivo in a zebrafish xenograft model. Moreover, using a dataset of breast cancer patients we show that the co-expression of HMGA1, FOXM1 and VEGFA is a negative prognostic factor of distant metastasis-free survival and relapse-free survival. CONCLUSIONS: This study reveals FOXM1 as a crucial interactor of HMGA1 and proves that their cooperative action supports breast cancer aggressiveness, by promoting tumor angiogenesis. Therefore, the possibility to target HMGA1/FOXM1 in combination should represent an attractive therapeutic option to counteract breast cancer angiogenesis.


Assuntos
Núcleo Celular/metabolismo , Proteína Forkhead Box M1/metabolismo , Proteína HMGA1a/metabolismo , Neoplasias de Mama Triplo Negativas/genética , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Linhagem Celular Tumoral , Meios de Cultivo Condicionados/farmacologia , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Feminino , Proteína Forkhead Box M1/química , Proteína Forkhead Box M1/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Células HEK293 , Proteína HMGA1a/genética , Humanos , Prognóstico , Regiões Promotoras Genéticas , Estabilidade Proteica , Análise de Sequência de RNA , Análise de Sobrevida , Transcrição Gênica , Neoplasias de Mama Triplo Negativas/metabolismo , Peixe-Zebra
3.
Elife ; 82019 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-31134895

RESUMO

Intrinsically disordered transcription factor transactivation domains (TADs) function through structural plasticity, adopting ordered conformations when bound to transcriptional co-regulators. Many transcription factors contain a negative regulatory domain (NRD) that suppresses recruitment of transcriptional machinery through autoregulation of the TAD. We report the solution structure of an autoinhibited NRD-TAD complex within FoxM1, a critical activator of mitotic gene expression. We observe that while both the FoxM1 NRD and TAD are primarily intrinsically disordered domains, they associate and adopt a structured conformation. We identify how Plk1 and Cdk kinases cooperate to phosphorylate FoxM1, which releases the TAD into a disordered conformation that then associates with the TAZ2 or KIX domains of the transcriptional co-activator CBP. Our results support a mechanism of FoxM1 regulation in which the TAD undergoes switching between disordered and different ordered structures.


Assuntos
Ativação Enzimática , Proteína Forkhead Box M1/química , Proteína Forkhead Box M1/metabolismo , Proteínas de Ciclo Celular/metabolismo , Fragmentos de Peptídeos/metabolismo , Fosforilação , Ligação Proteica , Conformação Proteica , Domínios Proteicos , Processamento de Proteína Pós-Traducional , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Sialoglicoproteínas/metabolismo , Quinase 1 Polo-Like
4.
J Mol Graph Model ; 80: 197-210, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29414039

RESUMO

The FOXM1 protein is a relevant transcription factor involved in cancer cell proliferation. The direct or indirect inhibition of this protein's transcriptional activity by small molecule drugs correlates well with a potentially significant anti-cancer profile, making this macro molecule a promising drug target. There are a few drug molecules reported to interact with (and inhibit) the FOXM1 DNA binding domain (FOXM1-BD), causing downregulation of protein expression and cancer cell proliferation inhibition. Among these drug molecules are the proteasome inhibitor thiostrepton, the former antidiabetic drug troglitazone, and the new FDI-6 molecule. Despite their structural differences, these drugs exert a similar inhibitory profile, and this observation prompted us to study a possible similar mechanism of action. Using a series of molecular dynamics simulations and docking protocols, we identified essential binding interactions exerted by all three classes of drugs, among which, a π-sulfur interaction (between a His287 and a sulfur-containing heterocycle) was the most important. In this report, we describe the preliminary evidence suggesting the presence of a drug-binding pocket within FOXM1 DNA binding domain, in which inhibitors fit to dissociate the protein-DNA complex. This finding suggests a common mechanism of action and a basic framework to design new FOXM1 inhibitors.


Assuntos
Desenho de Fármacos , Proteína Forkhead Box M1/química , Modelos Moleculares , Relação Quantitativa Estrutura-Atividade , Sítios de Ligação , Proteína Forkhead Box M1/farmacologia , Humanos , Ligantes , Conformação Molecular , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Estrutura Molecular , Ligação Proteica , Tioestreptona/química , Tioestreptona/farmacologia
5.
Drug Deliv ; 25(1): 226-240, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29313393

RESUMO

The Forkhead box M1 (FoxM1) transcription factor is an important anti-tumor target. A novel targeted ultrasound (US)-sensitive nanobubble that is likely to make use of the physical energy of US exposure for the improvement of delivery efficacy to target tumors and specifically silence FoxM1 expression appears as among the most potential nanocarriers in respect of drug delivery. In this study, we synthesized a promising anti-tumor targeted FoxM1 siRNA-loaded cationic nanobubbles (CNBs) conjugated with an A10-3.2 aptamer (siFoxM1-Apt-CNBs), which demonstrate high specificity when binding to prostate-specific membrane antigen (PSMA) positive LNCaP cells. Uniform nanoscaled siFoxM1-Apt-CNBs were developed using a thin-film hydration sonication, carbodiimide chemistry approaches, and electrostatic adsorption methods. Fluorescence imaging as well as flow cytometry evidenced the fact that the siFoxM1-Apt-CNBs were productively developed and that they specifically bound to PSMA-positive LNCaP cells. siFoxM1-Apt-CNBs combined with ultrasound-mediated nanobubble destruction (UMND) significantly improved transfection efficiency, cell apoptosis, and cell cycle arrest in vitro while reducing FoxM1 expression. In vivo xenografts tumors in nude-mouse model results showed that siFoxM1-Apt-CNBs combined with UMND led to significant inhibition of tumor growth and prolonged the survival of the mice, with low toxicity, an obvious reduction in FoxM1 expression, and a higher apoptosis index. Our study suggests that siFoxM1-Apt-CNBs combined with UMND might be a promising targeted gene delivery strategy for therapy of prostate cancer.


Assuntos
Aptâmeros de Nucleotídeos/administração & dosagem , Cátions/administração & dosagem , Nanopartículas/administração & dosagem , Neoplasias da Próstata/tratamento farmacológico , RNA Interferente Pequeno/administração & dosagem , Animais , Antígenos de Superfície/química , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Aptâmeros de Nucleotídeos/química , Cátions/química , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos/métodos , Proteína Forkhead Box M1/administração & dosagem , Proteína Forkhead Box M1/química , Técnicas de Transferência de Genes , Terapia Genética/métodos , Glutamato Carboxipeptidase II/química , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanopartículas/química , RNA Interferente Pequeno/química , Transfecção/métodos , Ondas Ultrassônicas
6.
Biochem Biophys Res Commun ; 495(2): 1681-1687, 2018 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-29196265

RESUMO

O-GlcNAcylation is a dynamic post-translational modification of cytonuclear proteins for intracellular signaling. Elevated O-GlcNAcylation is a general feature of cancer and contributes to cancer progression, and recent studies indicate the contribution to increasing incidence of various types of cancer in diabetic patients. However, the role of O-GlcNAcylation in tumor progression is not fully elucidated. Forkhead box M1 (FOXM1), a master mitotic transcription factor, has been implicated in all major hallmarks of cancer, and is wildly expressed in solid tumors. Given that FOXM1 expression was reported to be elevated in gastric cancer, we examined the effect of high glucose or an inhibitor of O-GlcNAc hydrolase, Thiamet G (TMG), on FOXM1 protein expression in a human gastric cancer cell line, MKN45 cells, and confirmed that FOXM1 protein level and the cell proliferation were upregulated. To investigate the molecular mechanisms by which FOXM1 protein expression is regulated by O-GlcNAcylation, the effect of high glucose and TMG on FOXM1 ubiquitination was examined in MKN45 cells. As a result, the ubiquitination and degradation of FOXM1 protein were both suppressed by high glucose and TMG treatment. However, the O-GlcNAcylation was not detected on FOXM1 but not on GSK-3ß. High glucose and TMG treatment increased phospho-serine 9 GSK-3ß, an inactive form, and the degradation of FOXM1 protein was suppressed by treatment of GSK-3ß inhibitors in MKN45 cells. Taken together, we suggest that high glucose and elevated O-GlcNAcylation stabilize FOXM1 protein by its reduced degradation via GSK-3ß inactivation in MKN45 cells, suggesting that the higher risk of gastric cancer in diabetic patients could be partially due to O-GlcNAcylation-mediated FOXM1 stabilization.


Assuntos
Proteína Forkhead Box M1/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Neoplasias Gástricas/metabolismo , Acetilglucosamina/metabolismo , Acilação , Linhagem Celular Tumoral , Proliferação de Células , Complicações do Diabetes/etiologia , Complicações do Diabetes/metabolismo , Complicações do Diabetes/patologia , Inibidores Enzimáticos/farmacologia , Proteína Forkhead Box M1/química , Glucose/metabolismo , Glicogênio Sintase Quinase 3 beta/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta/química , Humanos , Processamento de Proteína Pós-Traducional , Estabilidade Proteica/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Piranos/farmacologia , Neoplasias Gástricas/etiologia , Neoplasias Gástricas/patologia , Tiazóis/farmacologia , Ubiquitinação/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , beta-N-Acetil-Hexosaminidases/antagonistas & inibidores , beta-N-Acetil-Hexosaminidases/metabolismo
7.
Clin Cancer Res ; 23(22): 6923-6933, 2017 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-28899970

RESUMO

Purpose: Prostate cancer was recently classified to three clinically relevant subtypes (PCS) demarcated by unique pathway activation and clinical aggressiveness. In this preclinical study, we investigated molecular targets and therapeutics for PCS1, the most aggressive and lethal subtype, with no treatment options available in the clinic.Experimental Design: We utilized the PCS1 gene set and our model of enzalutamide (ENZR) castration-resistant prostate cancer (CRPC) to identify targetable pathways and inhibitors for PCS1. The findings were evaluated in vitro and in the ENZR CRPC xenograft model in vivoResults: The results revealed that ENZR CRPC cells are enriched with PCS1 signature and that Forkhead box M1 (FOXM1) pathway is the central driver of this subtype. Notably, we identified Monensin as a novel FOXM1-binding agent that selectively targets FOXM1 to reverse the PCS1 signature and its associated stem-like features and reduces the growth of ENZR CRPC cells and xenograft tumors.Conclusions: Our preclinical data indicate FOXM1 pathway as a master regulator of PCS1 tumors, namely in ENZR CRPC, and targeting FOXM1 reduces cell growth and stemness in ENZR CRPC in vitro and in vivo These preclinical results may guide clinical evaluation of targeting FOXM1 to eradicate highly aggressive and lethal PCS1 prostate cancer tumors. Clin Cancer Res; 23(22); 6923-33. ©2017 AACR.


Assuntos
Biomarcadores Tumorais , Proteína Forkhead Box M1/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Benzamidas , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Proteína Forkhead Box M1/química , Proteína Forkhead Box M1/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos , Modelos Moleculares , Terapia de Alvo Molecular , Células-Tronco Neoplásicas , Nitrilas , Feniltioidantoína/análogos & derivados , Feniltioidantoína/farmacologia , Prognóstico , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/patologia , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Neoplasias de Próstata Resistentes à Castração/genética , Neoplasias de Próstata Resistentes à Castração/metabolismo , Neoplasias de Próstata Resistentes à Castração/patologia , Ligação Proteica , Relação Estrutura-Atividade , Transcriptoma , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Methods Mol Biol ; 1647: 129-138, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28808999

RESUMO

The study of protein-protein interactions represents a key aspect of biological research. Identifying unknown protein binding partners using mass spectrometry (MS)-based proteomics has evolved into an indispensable strategy in drug discovery. The classic approach of immunoprecipitation with specific antibodies against the proteins of interest has limitations, such as the need for immunoprecipitation-qualified antibody. The biotin AP-tag pull-down system has the advantage of high specificity, ease of use, and no requirement for antibody. It is based on the high specificity, high affinity interaction between biotin and streptavidin. After pulldown, in-gel tryptic digestion and tandem mass spectrometry (MS/MS) analysis of sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) protein bands can be performed. In this work, we provide protocols that can be used for the identification of proteins that interact with FOXM1, a protein that has recently emerged as a potential biomarker and drug target in oncotherapy, as an example. We focus on the pull-down procedure and assess the efficacy of the pulldown with known FOXM1 interactors such as ß-catenin. We use a high performance LTQ Orbitrap MSn system that combines rapid LTQ ion trap data acquisition with high mass accuracy Orbitrap analysis to identify the interacting proteins.


Assuntos
Biotina/química , Descoberta de Drogas/métodos , Mapeamento de Interação de Proteínas/métodos , Proteômica/métodos , Espectrometria de Massas em Tandem/métodos , Biomarcadores Tumorais/análise , Biomarcadores Tumorais/química , Biotina/metabolismo , Proteína Forkhead Box M1/química , Proteína Forkhead Box M1/metabolismo , Células HEK293 , Humanos , Ligação Proteica , Estreptavidina/química , Estreptavidina/metabolismo , beta Catenina/química , beta Catenina/metabolismo
9.
Sci Rep ; 7: 46017, 2017 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-28387346

RESUMO

FoxM1b is a cell cycle-regulated transcription factor, whose over-expression is a marker for poor outcome in cancers. Its transcriptional activation function requires phosphorylation by Cdk1 or Cdk2 that primes FoxM1b for phosphorylation by Plk1, which triggers association with the co-activator CBP. FoxM1b also possesses transcriptional repression function. It represses the mammary differentiation gene GATA3 involving DNMT3b and Rb. We investigated what determines the two distinct functions of FoxM1b: activation and repression. We show that Rb binds to the C-terminal activation domain of FoxM1b. Analyses with phospho-defective and phospho-mimetic mutants of FoxM1b identified a critical role of the Plk1 phosphorylation sites in regulating the binding of FoxM1b to Rb and DNMT3b. That is opposite of what was seen for the interaction of FoxM1b with CBP. We show that, in addition to GATA3, FoxM1b also represses the mammary luminal differentiation marker FoxA1 by promoter-methylation, and that is regulated by the Plk1 phosphorylation sites in FoxM1b. Our results show that the Plk1 phosphorylation sites in FoxM1b serve as a regulator for its repressor function, and they provide insights into how FoxM1b inhibits differentiation genes and activates proliferation genes during cancer progression.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteína Forkhead Box M1/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Repressoras/metabolismo , Proteína do Retinoblastoma/metabolismo , Sítios de Ligação , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA/genética , Proteína Forkhead Box M1/química , Fator de Transcrição GATA3/genética , Humanos , Células MCF-7 , Mutação/genética , Fragmentos de Peptídeos/metabolismo , Fosforilação , Regiões Promotoras Genéticas , Ligação Proteica , Domínios Proteicos , Sialoglicoproteínas/metabolismo , DNA Metiltransferase 3B , Quinase 1 Polo-Like
10.
Neuro Oncol ; 19(3): 352-363, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-27923917

RESUMO

Background: Forkhead box M1 (FOXM1) is overexpressed and activates numerous oncoproteins in tumors. However, the mechanism by which the FOXM1 protein aberrantly accumulates in human cancer remains uncertain. This study was designed to clarify the upstream signaling pathway(s) that regulate FOXM1 protein stability and transcriptional activity. Methods: Mass spectrometry and immunoprecipitation were performed to identify the FOXM-metadherin (MTDH) interaction. In vivo and in vitro ubiquitination assays were conducted to test the effect of MTDH on FOXM1 stability. Chromatin immunoprecipitation assays were used to determine the involvement of MTDH in FOXM1 transcriptional activity. Cell invasion assays, tube formation assays, and in vivo tumor formation assays were performed to evaluate the cooperative activities of FOXM1 and MTDH during tumorigenesis. Results: MTDH directly interacts with FOXM1 via the N-terminal inhibitory domain of MTDH, and this interaction disrupted the binding of cadherin-1 to FOXM1, thus protecting FOXM1 from subsequent proteasomal degradation. Deleting the MTDH-binding sites of FOXM1 abolished the MTDH overexpression-mediated stabilization of FOXM1. MTDH also bound to FOXM1 target gene promoters and enhanced FOXM1 transcriptional activity. MTDH knockdown destabilized FOXM1 and attenuated its transcriptional activity, consequently inhibiting cell cycle progression, angiogenesis, and cancer cell invasion in vitro and in vivo; these effects were abolished via forced overexpression of a stabilized mutant form of FOXM1. Thus, MTDH stabilized FOXM1 and supported the sustained activation of FOXM1 target genes. Conclusion: These findings highlight a novel MTDH-regulated mechanism of FOXM1 stabilization and provide profound insight into the tumorigenic events simultaneously mediated by FOXM1 and MTDH.


Assuntos
Astrócitos/patologia , Moléculas de Adesão Celular/metabolismo , Transformação Celular Neoplásica/patologia , Proteína Forkhead Box M1/genética , Regulação Neoplásica da Expressão Gênica , Glioblastoma/patologia , Animais , Apoptose , Astrócitos/metabolismo , Caderinas/genética , Caderinas/metabolismo , Moléculas de Adesão Celular/genética , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Proteína Forkhead Box M1/química , Proteína Forkhead Box M1/metabolismo , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Proteínas de Membrana , Camundongos , Estabilidade Proteica , Proteínas de Ligação a RNA , Transdução de Sinais , Células Tumorais Cultivadas , Ubiquitinação , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Bioorg Chem ; 70: 12-16, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27881238

RESUMO

The oncogenic transcription factor forkhead box M (Foxm) is overexpressed in human colorectal cancer (CRC). Targeting the protein interaction with its cognate DNA has been established as an attractive approach to anti-CRC chemotherapy. State-of-the-art molecular dynamics (MD) simulations revealed that the Foxm adopts considerably different conformations to interact with and without its DNA partner; the holo conformation is tightly packed as a typical globulin configuration, whereas the apo form is locally unstructured that exhibits intrinsic disorder in DNA recognition helix, indicating that DNA binding can help the Foxm refolding. With this finding, the MD equilibrium structure of DNA-free Foxm was utilized to perform molecular docking virtual screening against a natural organic compound library. Consequently, six hit compounds were identified as potential small-molecule mediators of Foxm-DNA interaction; their binding affinities (KD) to Foxm DNA-binding domain were then determined to range between 3.8 and 230µM by using isothermal titration calorimetry. These compounds were suggested to recognize and stabilize the apo conformation of Foxm, thus shifting the binding reaction equilibrium of Foxm from DNA-bound to DNA-free states to disrupt the formation of Foxm-DNA adduct.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Neoplasias Colorretais/tratamento farmacológico , DNA/metabolismo , Descoberta de Drogas , Proteína Forkhead Box M1/metabolismo , Sítios de Ligação/efeitos dos fármacos , Neoplasias Colorretais/metabolismo , DNA/química , Proteína Forkhead Box M1/química , Humanos , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Conformação de Ácido Nucleico/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos
12.
Oncotarget ; 7(37): 60366-60382, 2016 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-27542221

RESUMO

Forkhead box transcription factor M1 (FOXM1) plays crucial roles in a wide array of biological processes, including cell proliferation and differentiation, the cell cycle, and tumorigenesis by regulating the expression of its target genes. Elevated expression of FOXM1 is frequently observed in a multitude of malignancies. Here we show that FOXM1 can be acetylated by p300/CBP at lysines K63, K422, K440, K603 and K614 in vivo. This modification is essential for its transactivation on the target genes. Acetylation of FOXM1 increases during the S phase and remains high throughout the G2 and M phases, when FOXM1 transcriptional activity is required. We find that the acetylation-deficient FOXM1 mutant is less active and exhibits significantly weaker tumorigenic activities compared to wild-type FOXM1. Mechanistically, the acetylation of FOXM1 enhances its transcriptional activity by increasing its DNA binding affinity, protein stability, and phosphorylation sensitivity. In addition, we demonstrate that NAD-dependent histone deacetylase SIRT1 physically binds to and deacetylates FOXM1 in vivo. The deacetylation of FOXM1 by SIRT1 attenuates its transcriptional activity and decreases its protein stability. Together, our findings demonstrate that the reversible acetylation of FOXM1 by p300/CBP and SIRT1 modulates its transactivation function.


Assuntos
Acetilação , Proteína Forkhead Box M1/metabolismo , Neoplasias/metabolismo , Sirtuína 1/metabolismo , Fatores de Transcrição de p300-CBP/metabolismo , Animais , Ciclo Celular , Feminino , Proteína Forkhead Box M1/química , Proteína Forkhead Box M1/genética , Regulação da Expressão Gênica , Células HEK293 , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/patologia , Estabilidade Proteica , Ativação Transcricional , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...