Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
J Phys Chem B ; 126(45): 9176-9186, 2022 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-36331868

RESUMO

Structural characterization of intrinsically disordered proteins (IDPs) requires a concerted effort between experiments and computations by accounting for their conformational heterogeneity. Given the diversity of experimental tools providing local and global structural information, constructing an experimental restraint-satisfying structural ensemble remains challenging. Here, we use the disordered N-terminal domain (NTD) of the estrogen receptor alpha (ERalpha) as a model system to combine existing small-angle X-ray scattering (SAXS) and hydroxyl radical protein footprinting (HRPF) data and newly acquired solvent accessibility data via D2O-induced fluorine chemical shifting (DFCS) measurements. A new set of DFCS data for the solvent exposure of a set of 12 amino acid positions were added to complement previously acquired HRPF measurements for the solvent exposure of the other 16 nonoverlapping amino acids, thereby improving the NTD ensemble characterization considerably. We also found that while choosing an initial ensemble of structures generated from a different atomic-level force field or sampling/modeling method can lead to distinct contact maps even when the same sets of experimental measurements were used for ensemble-fitting, comparative analyses from these initial ensembles reveal commonly recurring structural features in their ensemble-averaged contact map. Specifically, nonlocal or long-range transient interactions were found consistently between the N-terminal segments and the central region, sufficient to mediate the conformational ensemble and regulate how the NTD interacts with its coactivator proteins.


Assuntos
Receptor alfa de Estrogênio , Proteínas Intrinsicamente Desordenadas , Espalhamento a Baixo Ângulo , Difração de Raios X , Conformação Proteica , Flúor , Proteínas Intrinsicamente Desordenadas/química , Solventes
2.
Cancer Res ; 80(21): 4815-4827, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32907836

RESUMO

PIK3CA encodes the p110α catalytic subunit of PI3K and is frequently mutated in human cancers, including ∼30% of colorectal cancer. Oncogenic mutations in PIK3CA render colorectal cancers more dependent on glutamine. Here we report that the glutaminase inhibitor CB-839 preferentially inhibits xenograft growth of PIK3CA-mutant, but not wild-type (WT), colorectal cancers. Moreover, the combination of CB-839 and 5-fluorouracil (5-FU) induces PIK3CA-mutant tumor regression in xenograft models. CB-839 treatment increased reactive oxygen species and caused nuclear translocation of Nrf2, which in turn upregulated mRNA expression of uridine phosphorylase 1 (UPP1). UPP1 facilitated the conversion of 5-FU to its active compound, thereby enhancing the inhibition of thymidylate synthase. Consistently, knockout of UPP1 abrogated the tumor inhibitory effect of combined CB-839 and 5-FU administration. A phase I clinical trial showed that the combination of CB-839 and capecitabine, a prodrug of 5-FU, was well tolerated at biologically-active doses. Although not designed to test efficacy, an exploratory analysis of the phase I data showed a trend that PIK3CA-mutant patients with colorectal cancer might derive greater benefit from this treatment strategy as compared with PIK3CA WT patients with colorectal cancer. These results effectively demonstrate that targeting glutamine metabolism may be an effective approach for treating patients with PIK3CA-mutant colorectal cancers and warrants further clinical evaluation. SIGNIFICANCE: Preclinical and clinical trial data suggest that the combination of CB-839 with capecitabine could serve as an effective treatment for PIK3CA-mutant colorectal cancers.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Benzenoacetamidas/administração & dosagem , Classe I de Fosfatidilinositol 3-Quinases/genética , Neoplasias Colorretais/tratamento farmacológico , Fluoruracila/administração & dosagem , Tiadiazóis/administração & dosagem , Adulto , Animais , Benzenoacetamidas/efeitos adversos , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Relação Dose-Resposta a Droga , Feminino , Fluoruracila/efeitos adversos , Humanos , Masculino , Dose Máxima Tolerável , Camundongos , Pessoa de Meia-Idade , Tiadiazóis/efeitos adversos , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Structure ; 27(2): 229-240.e4, 2019 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-30581045

RESUMO

The N-terminal transactivation domain (NTD) of estrogen receptor alpha, a well-known member of the family of intrinsically disordered proteins, mediates the receptor's transactivation function. However, an accurate molecular dissection of NTD's structure-function relationships remains elusive. Here, we show that the NTD adopts a mostly disordered, unexpectedly compact conformation that undergoes structural expansion on chemical denaturation. By combining small-angle X-ray scattering, hydroxyl radical protein footprinting, and computational modeling, we derive the ensemble-structures of the NTD and determine its ensemble-contact map revealing metastable long-range contacts, e.g., between residues I33 and S118. We show that mutation at S118, a known phosphorylation site, promotes conformational changes and increases coactivator binding. We further demonstrate via fluorine-19 (19F) nuclear magnetic resonance that mutations near I33 alter 19F chemical shifts at S118, confirming the proposed I33-S118 contact in the ensemble of structural disorder. These findings extend our understanding of how specific contact metastability mediates critical functions of disordered proteins.


Assuntos
Receptor alfa de Estrogênio/química , Receptor alfa de Estrogênio/metabolismo , Mutação , Receptor alfa de Estrogênio/genética , Imagem por Ressonância Magnética de Flúor-19 , Humanos , Isoleucina/genética , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Domínios Proteicos , Espalhamento a Baixo Ângulo , Serina/genética , Ativação Transcricional , Difração de Raios X
4.
Clin Cancer Res ; 24(7): 1629-1643, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29326282

RESUMO

Purpose: (i) To investigate the expression of the E3 ligase, RNF126, in human invasive breast cancer and its links with breast cancer outcomes; and (ii) to test the hypothesis that RNF126 determines the efficacy of inhibitors targeting the cell-cycle checkpoint kinase, CHEK1.Experimental Design: A retrospective analysis by immunohistochemistry (IHC) compared RNF126 staining in 110 invasive breast cancer and 78 paired adjacent normal tissues with clinicopathologic data. Whether RNF126 controls CHEK1 expression was determined by chromatin immunoprecipitation and a CHEK1 promoter driven luciferase reporter. Staining for these two proteins by IHC using tissue microarrays was also conducted. Cell killing/replication stress induced by CHEK1 inhibition was evaluated in cells, with or without RNF126 knockdown, by MTT/colony formation, replication stress biomarker immunostaining and DNA fiber assays.Results: RNF126 protein expression was elevated in breast cancer tissue samples. RNF126 was associated with a poor clinical outcome after multivariate analysis and was an independent predictor. RNF126 promotes CHEK1 transcript expression. Critically, a strong correlation between RNF126 and CHEK1 proteins was identified in breast cancer tissue and cell lines. The inhibition of CHEK1 induced a greater cell killing and a higher level of replication stress in breast cancer cells expressing RNF126 compared to RNF126 depleted cells.Conclusions: RNF126 protein is highly expressed in invasive breast cancer tissue. The high expression of RNF126 is an independent predictor of a poor prognosis in invasive breast cancer and is considered a potential biomarker of a cancer's responsiveness to CHEK1 inhibitors. CHEK1 inhibition targets breast cancer cells expressing higher levels of RNF126 by enhancing replication stress. Clin Cancer Res; 24(7); 1629-43. ©2018 AACR.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias da Mama/genética , Quinase 1 do Ponto de Checagem/genética , Ubiquitina-Proteína Ligases/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Estudos de Coortes , Replicação do DNA/genética , Feminino , Humanos , Imuno-Histoquímica/métodos , Células MCF-7 , Prognóstico , Regiões Promotoras Genéticas/genética , Estudos Retrospectivos
5.
Oncotarget ; 7(23): 34688-702, 2016 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-27167194

RESUMO

Radiotherapy (RT) remains a standard therapeutic modality for breast cancer patients. However, intrinsic or acquired resistance limits the efficacy of RT. Here, we demonstrate that CHK1 inhibitor AZD7762 alone significantly inhibited the growth of radioresistant breast cancer cells (RBCC). Given the critical role of ATR/CHK1 signaling in suppressing oncogene-induced replication stress (RS), we hypothesize that CHK1 inhibition leads to the specific killing for RBCC due to its abrogation in the suppression of RS induced by oncogenes. In agreement, the expression of oncogenes c-Myc/CDC25A/c-Src/H-ras/E2F1 and DNA damage response (DDR) proteins ATR/CHK1/BRCA1/CtIP were elevated in RBCC. AZD7762 exposure led to significantly higher levels of RS in RBCC, compared to the parental cells. The mechanisms by which CHK1 inhibition led to specific increase of RS in RBCC were related to the interruptions in the replication fork dynamics and the homologous recombination (HR). In summary, RBCC activate oncogenic pathways and thus depend upon mechanisms controlled by CHK1 signaling to maintain RS under control for survival. Our study provided the first example where upregulating RS by CHK1 inhibitor contributes to the specific killing of RBCC, and highlight the importance of the CHK1 as a potential target for treatment of radioresistant cancer cells.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/radioterapia , Quinase 1 do Ponto de Checagem/antagonistas & inibidores , Tolerância a Radiação/efeitos dos fármacos , Radiossensibilizantes/farmacologia , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Reparo do DNA/genética , Recombinação Homóloga/genética , Humanos , Células MCF-7 , Compostos de Fenilureia/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Pirazinas/farmacologia , Tiofenos/farmacologia , Ureia/análogos & derivados , Ureia/farmacologia
6.
Cell Rep ; 15(5): 1111-1122, 2016 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-27117404

RESUMO

Fbxo30 is an orphan member of the F-box protein family with no known substrate or function. Here we report that, while Fbxo30(-/-) mice exhibit normal development, growth, lifespan, and fertility, the females fail to nurture their offspring as a result of defective mammopoiesis. Mass spectrometry analysis of Fbxo30-associated proteins revealed that Fbxo30 specifically interacts with the bipolar spindle kinesin EG5 (encoded by Kif11). As a result, Fbxo30 targets Eg5 for ubiquitinylation and controls its oscillation during the cell cycle. Correlated with EG5 dysregulation, Fbxo30(-/-) mammary epithelial cells exhibit multiple defects in centrosome homeostasis, mitotic spindle formation, and proliferation. Effects on proliferation, centrosome homeostasis, and mammopoiesis in the Fbxo30(-/-) mice were rescued through normalization of Eg5 activity using shRNA and/or an EG5 inhibitor. Our data reveal the Fbxo30-Eg5 interaction as a critical checkpoint in mammopoiesis and a critical role for ubiquitinylation-regulated Eg5 oscillation in the cell cycle.


Assuntos
Proteínas F-Box/metabolismo , Cinesinas/metabolismo , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/metabolismo , Mitose , Animais , Contagem de Células , Diferenciação Celular , Linhagem da Célula/genética , Proliferação de Células , Instabilidade Cromossômica , Feminino , Marcação de Genes , Células HCT116 , Humanos , Camundongos , Mutação/genética , Fuso Acromático/metabolismo , Células-Tronco/citologia , Ubiquitina-Proteína Ligases/metabolismo
7.
Mol Cancer Res ; 13(6): 969-81, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25753001

RESUMO

UNLABELLED: Aberrant activation of the Wnt signaling pathway is an important step in the initiation and progression of tumor development in diverse cancers. The central effector of canonical Wnt signaling, ß-catenin (CTNNB1), is a multifunctional protein, and has been extensively studied with respect to its roles in cell-cell adhesion and in regulation of Wnt-driven transcription. Here, a novel mass spectrometry-based proteomics technique in colorectal cancer cells expressing stabilized ß-catenin, was used to identify a protein-protein interaction between ß-catenin and DNA methyltransferase I (Dnmt1) protein, the primary regulator of DNA methylation patterns in mammalian cells. Dnmt1 and ß-catenin strongly colocalized in the nuclei of colorectal cancer cells, and the interaction is mediated by the central domain of the Dnmt1 protein. Dnmt1 protein abundance is dependent upon the levels of ß-catenin, and is increased in cells expressing stabilized mutant ß-catenin. Conversely, the Dnmt1 regulates the levels of nuclear ß-catenin and ß-catenin/TCF-driven transcription. In addition, lysine-specific demethylase 1 (LSD1/KDM1A), a regulator of DNMT1 stability, was identified as a component of the Dnmt1-ß-catenin protein complex and perturbation of the Dnmt1-ß-catenin interaction altered DNA methylation. In summary, a functional protein-protein interaction was identified between two critically important oncoproteins, in turn revealing a link between Wnt signaling and downstream nuclear functions mediated by Dnmt1. IMPLICATIONS: Two critical oncoproteins, Dnmt1 and ß-catenin, mutually regulate one each other's levels and activities in colorectal cancer cells.


Assuntos
Neoplasias Colorretais/metabolismo , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , Via de Sinalização Wnt , beta Catenina/metabolismo , Linhagem Celular Tumoral , Ilhas de CpG , DNA (Citosina-5-)-Metiltransferase 1 , Histona Desmetilases/metabolismo , Humanos , Ligação Proteica , Fatores de Transcrição TCF/metabolismo
8.
Nucleic Acids Res ; 43(3): 1659-70, 2015 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-25586219

RESUMO

Alternative non-homologous end joining (alt-NHEJ) was originally identified as a backup repair mechanism in the absence of classical NHEJ (c-NHEJ) factors but recent studies have demonstrated that alt-NHEJ is active even when c-NHEJ as well as homologous recombination is available. The functions of 53BP1 in NHEJ processes are not well understood. Here, we report that 53BP1 promotes DNA double-strand break (DSB) repair and genomic stability not only in c-NHEJ-proficient but also -deficient human G1-phase cells. Using an array of repair substrates we show that these effects of 53BP1 are correlated with a promotion of microhomology-mediated end-joining (MMEJ), a subtype of alt-NHEJ, in G1-phase. Consistent with a specific role in MMEJ we confirm that 53BP1 status does not affect c-NHEJ. 53BP1 supports sequence deletion during MMEJ consistent with a putative role in facilitating end-resection. Interestingly, promotion of MMEJ by 53BP1 in G1-phase cells is only observed in the presence of functional BRCA1. Depletion of both 53BP1 and BRCA1 increases repair needing microhomology usage and augments loss of DNA sequence, suggesting that MMEJ is a highly regulated DSB repair process. Together, these findings significantly expand our understanding of the cell-cycle-dependent roles of 53BP1 in DSB repair.


Assuntos
Reparo do DNA por Junção de Extremidades , Fase G1 , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Sequência de Bases , Western Blotting , Linhagem Celular , Ensaio Cometa , Primers do DNA , Imunofluorescência , Instabilidade Genômica , Humanos , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
9.
Cancer Res ; 73(7): 2170-80, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23319807

RESUMO

FOXP3 is an X-linked tumor suppressor gene and a master regulator in T regulatory cell function. This gene has been found to be mutated frequently in breast and prostate cancers and to inhibit tumor cell growth, but its functional significance in DNA repair has not been studied. We found that FOXP3 silencing stimulates homologous recombination-mediated DNA repair and also repair of γ-irradiation-induced DNA damage. Expression profiling and chromatin-immunoprecipitation analyses revealed that FOXP3 regulated the BRCA1-mediated DNA repair program. Among 48 FOXP3-regulated DNA repair genes, BRCA1 and 12 others were direct targets of FOXP3 transcriptional control. Site-specific interaction of FOXP3 with the BRCA1 promoter repressed its transcription. Somatic FOXP3 mutants identified in breast cancer samples had reduced BRCA1 repressor activity, whereas FOXP3 silencing and knock-in of a prostate cancer-derived somatic FOXP3 mutant increased the radioresistance of cancer cells. Together our findings provide a missing link between FOXP3 function and DNA repair programs.


Assuntos
Proteína BRCA1/genética , Neoplasias da Mama/patologia , Fatores de Transcrição Forkhead/fisiologia , Raios gama , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Tolerância a Radiação/genética , Transcrição Gênica/efeitos da radiação , Animais , Apoptose , Proteína BRCA1/metabolismo , Western Blotting , Neoplasias da Mama/metabolismo , Neoplasias da Mama/radioterapia , Proliferação de Células , Imunoprecipitação da Cromatina , Ensaio Cometa , Dano ao DNA/efeitos da radiação , Reparo do DNA/efeitos da radiação , Feminino , Recombinação Homóloga , Humanos , Técnicas Imunoenzimáticas , Camundongos , Camundongos Endogâmicos BALB C , Mutagênese Sítio-Dirigida , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
J Proteome Res ; 11(12): 5630-41, 2012 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-23106643

RESUMO

Affinity-purification mass spectrometry (AP-MS) is the preeminent technique for identification of eukaryotic protein complexes in vivo. AP-MS workflows typically express epitope-tagged bait proteins, immunopurify, and then identify associated protein complexes using mass spectrometry. However, challenges of existing strategies include the construction of expression vectors for large open reading frames and the possibility that overexpression of bait proteins may result in expression of nonphysiological levels of the bait protein with concomitant perturbation of endogenous protein complexes. To address these issues, we use human cell lines with epitope-tagged endogenous genes as AP-MS substrates to develop a platform that we call "knock-in AP-MS", thereby avoiding the challenges of expression vector construction and ensuring that expression of tagged proteins is driven by endogenous regulatory mechanisms. Using three different bait genes (MRE11A, DNMT1 and APC), we show that cell lines expressing epitope-tagged endogenous genes make good substrates for sensitive and reproducible identification of protein interactions using AP-MS. In particular, we identify novel interactors of the important oncoprotein Adenomatous Polyposis Coli (APC), including an interaction with Flightless-1 homologue (FLII) that is enriched in nuclear fractions.


Assuntos
Cromatografia de Afinidade/métodos , Neoplasias do Colo/patologia , Mapeamento de Epitopos/métodos , Genoma Humano , Espectrometria de Massas/métodos , Complexos Multiproteicos/isolamento & purificação , Proteínas Adaptadoras de Transdução de Sinal/isolamento & purificação , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Núcleo Celular/metabolismo , Núcleo Celular/patologia , Neoplasias do Colo/metabolismo , Citoplasma/metabolismo , Citoplasma/patologia , Dependovirus , Vetores Genéticos , Células HCT116 , Células HEK293 , Humanos , Proteínas dos Microfilamentos/isolamento & purificação , Proteínas dos Microfilamentos/metabolismo , Complexos Multiproteicos/metabolismo , Proteínas de Neoplasias/isolamento & purificação , Proteínas de Neoplasias/metabolismo , Mapeamento de Interação de Proteínas/métodos , Proteômica/métodos , Receptores Citoplasmáticos e Nucleares/isolamento & purificação , Receptores Citoplasmáticos e Nucleares/metabolismo , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Transativadores , Transfecção
11.
Mol Cell ; 44(5): 770-84, 2011 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-22152480

RESUMO

Both H4K16 acetylation and H3K4 trimethylation are required for gene activation. However, it is still largely unclear how these modifications are orchestrated by transcriptional factors. Here, we analyzed the mechanism of the transcriptional activation by FOXP3, an X-linked suppressor of autoimmune diseases and cancers. FOXP3 binds near transcriptional start sites of its target genes. By recruiting MOF and displacing histone H3K4 demethylase PLU-1, FOXP3 increases both H4K16 acetylation and H3K4 trimethylation at the FOXP3-associated chromatins of multiple FOXP3-activated genes. RNAi-mediated silencing of MOF reduced both gene activation and tumor suppression by FOXP3, while both somatic mutations in clinical cancer samples and targeted mutation of FOXP3 in mouse prostate epithelial cells disrupted nuclear localization of MOF. Our data demonstrate a pull-push model in which a single transcription factor orchestrates two epigenetic alterations necessary for gene activation and provide a mechanism for somatic inactivation of the FOXP3 protein function in cancer cells.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Histona Acetiltransferases/metabolismo , Histonas/metabolismo , Histona Desmetilases com o Domínio Jumonji/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Acetilação , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Feminino , Fatores de Transcrição Forkhead/genética , Regulação da Expressão Gênica , Células HEK293 , Humanos , Metilação , Mutação
12.
Mol Med Rep ; 4(6): 1131-7, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21874239

RESUMO

Previously, we characterized a zinc finger protein gene HZF1 (ZNF16) and demonstrated that it played a significant role in the erythroid and megakaryocytic differentiation of K562 cells by knockdown of the gene. In this study, we examined the effect of HZF1 on the proliferation and apoptosis of K562 cells and identified the possible mechanism for this effect. By lentivirus-mediated gene transfer, we obtained stable K562 transductants with HZF1 overexpression (K562/WPXL-HZF1) and stable control transductants (K562/WPXL). Significantly rapid cell amplification was observed in K562/WPXL-HZF1 cells compared to K562/WPXL cells. The cell cycles of the two transductants were analyzed and the results demonstrated that HZF1 overexpression promoted the S to G2/M phase transition. Additionally, we found that the overexpression of HZF1 slightly inhibits the apoptosis of K562 cells induced by sodium arsenate. Furthermore, using a yeast two-hybrid (Y2H) system we identified the HZF1-interacting proteins and screened 29 potential binding partners of HZF1. Using a co-immunoprecipitation (Co-IP) assay, we confirmed the interaction between HZF1 and the inhibitor of cyclin-dependent kinase (CDK) interacting with cyclin A1 (INCA1), and proved that this interaction leads to the inhibition of INCA1 function, which rescued the activity of CDK2 inhibited by INCA1. In conclusion, our results identified novel functions of the HZF1 gene and revealed a possible mechanism through which HZF1 affects K562 cell proliferation.


Assuntos
Proteínas de Transporte/antagonistas & inibidores , Proteínas de Ligação a DNA/metabolismo , Fosfoproteínas/antagonistas & inibidores , Apoptose , Arseniatos/toxicidade , Proteínas de Transporte/metabolismo , Divisão Celular , Proliferação de Células , Quinase 2 Dependente de Ciclina/metabolismo , Proteínas de Ligação a DNA/genética , Fase G2 , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Humanos , Imunoprecipitação , Células K562 , Lentivirus/genética , Fosfoproteínas/metabolismo , Ligação Proteica
13.
Sci Signal ; 3(146): ra80, 2010 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-21045206

RESUMO

DNA methyltransferase 1 (DNMT1) is the primary enzyme that maintains DNA methylation. We describe a previously unknown mode of regulation of DNMT1 protein stability through the coordinated action of an array of DNMT1-associated proteins. DNMT1 was destabilized by acetylation by the acetyltransferase Tip60, which triggered ubiquitination by the E3 ligase UHRF1, thereby targeting DNMT1 for proteasomal degradation. In contrast, DNMT1 was stabilized by histone deacetylase 1 (HDAC1) and the deubiquitinase HAUSP (herpes virus-associated ubiquitin-specific protease). Analysis of the abundance of DNMT1 and Tip60, as well as the association between HAUSP and DNMT1, suggested that during the cell cycle the initiation of DNMT1 degradation was coordinated with the end of DNA replication and the need for DNMT activity. In human colon cancers, the abundance of DNMT1 correlated with that of HAUSP. HAUSP knockdown rendered colon cancer cells more sensitive to killing by HDAC inhibitors both in tissue culture and in tumor xenograft models. Thus, these studies provide a mechanism-based rationale for the development of HDAC and HAUSP inhibitors for combined use in cancer therapy.


Assuntos
DNA (Citosina-5-)-Metiltransferases/metabolismo , Acetilação , Animais , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Ciclo Celular/fisiologia , Linhagem Celular , Neoplasias do Colo/enzimologia , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/biossíntese , Estabilidade Enzimática , Células HCT116 , Células HEK293 , Histona Acetiltransferases/metabolismo , Histona Desacetilase 1/metabolismo , Humanos , Lisina Acetiltransferase 5 , Camundongos , Camundongos Nus , Antígeno Nuclear de Célula em Proliferação/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina Tiolesterase/metabolismo , Ubiquitina-Proteína Ligases , Peptidase 7 Específica de Ubiquitina , Ubiquitinação
14.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 29(6): 772-6, 2007 Dec.
Artigo em Chinês | MEDLINE | ID: mdl-18595257

RESUMO

OBJECTIVE: To express human HZF1 fusion protein in E. coli and to obtain an anti-HZF1 antibody. METHODS: A DNA fragment encoding non-zinc finger region of HZF1 protein was inserted into pET30a vector to get the recombination expression plasmid pET30a-HZF1. E. coli was transformed with pET30a-HZF1 and the selected clones were cultured with isopropy-beta-D-thiogalactoside induction. The proteins were prepared from the culture and the fusion protein was purified by Ni column. Rabbits were immunized and reinforced three times with the purified fusion protein. The antiserum was collected and the titer and the specificity of the antibody were checked by ELISA and Western blot. RESULTS: Antibody against HZF1 was obtained and its titer was more than 1:100 000, as proven by ELISA. Western blot analysis showed specific reaction between this antibody and HZF1 fusion protein or the endogenetic HZF1 protein in hemin-induced K562 cells. CONCLUSIONS: The specific antibody against HZF1 is obtained. The antibody may have potential application in farther HZF1 function study and HZF1 determination in tissues and cells.


Assuntos
Proteínas de Ligação a DNA/imunologia , Escherichia coli/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Escherichia coli/metabolismo , Técnicas de Transferência de Genes , Soros Imunes/imunologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transformação Bacteriana
15.
Biochem Biophys Res Commun ; 349(1): 59-68, 2006 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-16934749

RESUMO

MicroRNAs (miRNAs) play an important role in diverse physiological and developmental processes by negatively regulating expression of target genes at the post-transcriptional level. Here, we globally analyzed the genomic organization of all registered 326 human miRNA genes in miRNA registry 7.1 and found that 148 human miRNA genes appeared in a total of 51 clusters. Alignment of the miRNA sequences in different clusters revealed a significant number of miRNA paralogs among the clusters, implying an evolution process targeting the potentially conserved roles of these molecules. Then we performed Northern blot analysis for expression profiling of all clustered miRNAs in several human leukemia cell lines. Consistent expression of the miRNAs in a single cluster was revealed in 39 clusters, while inconsistent expression of members in a single cluster was detected in the other 12 clusters. Meanwhile, we identified several hematopoietic lineage-specific or -enriched miRNA clusters (e.g., the mir-29c, mir-302, mir-98, mir-29a, and let-7a-1 clusters) and individual miRNAs (e.g., mir-181c, mir-181d, mir-191, and mir-136). These findings may suggest vital roles of these miRNA clusters or miRNAs in human hematopoiesis and oncogenesis, and provide clues for understanding the function and mechanism of miRNAs in various biological processes.


Assuntos
Regulação Neoplásica da Expressão Gênica , Leucemia/genética , MicroRNAs , Família Multigênica , Sequência de Bases , Linhagem Celular Tumoral , Linhagem da Célula , Perfilação da Expressão Gênica , Genoma , Células-Tronco Hematopoéticas , Humanos , Dados de Sequência Molecular , Hibridização de Ácido Nucleico
16.
Mol Cell Biochem ; 292(1-2): 155-67, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16786195

RESUMO

K562 cell line has been used as a model of common progenitor of erythroblasts and magakaryocytes and can be differentiated into erythroid and megakaryocytic lineages by hemin and phorbol myristate acetate (PMA) respectively. We analyzed mRNA expression in un-induced, hemin-induced and PMA-induced K562 cells by differential display reverse transcription polymerase chain reaction (DDRT-PCR) method. 314 differential expression sequence tags (ESTs) were obtained. Among them, 201 ESTs displayed up-regulation and 85 ESTs down-regulation after hemin induction, 186 ESTs showed up-regulation and 72 ESTs down-regulation after PMA induction. The differentially expressed genes included those encoding transcription factors, signaling factors, apoptosis-associated factors and others. 45 of these ESTs stand for genes whose open reading frames were found but whose functions remain unknown. 4 ESTs represent possibly new genes. Furthermore we compared differences of gene expression during hemin-induced erythroid differentiation and PMA-induced megakaryocytic differentiation and found that the expressional changes of some transcription factors and metabolism proteins are the common but the expressional changes of some signal pathways in these two differentiation processes are different. These results suggested that erythroid differentiation and megakaryocytic differentiation are associated in activation and repression of different signal pathways.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Células Eritroides/citologia , Perfilação da Expressão Gênica , Hemina/farmacologia , Megacariócitos/citologia , Acetato de Tetradecanoilforbol/farmacologia , Northern Blotting , Células Eritroides/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Globinas/genética , Humanos , Células K562 , Megacariócitos/efeitos dos fármacos , Ploidias , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
17.
J Biol Chem ; 280(9): 7452-9, 2005 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-15613485

RESUMO

The T to C substitution at position -175 of the gamma-globin gene has been identified in some individuals with non-deletion hereditary persistence of fetal hemoglobin (HPFH). In this study, the HPFH phenotype was reestablished in transgenic mice carrying the mu'LCRAgamma(-175)psibetadeltabeta construct, which contained a 3.1-kb mu'LCR cassette linked to a 29-kb fragment from the Agamma-to beta-globin gene with the natural chromosome arrangement but with the -175 mutation, which provided evidence for this single mutation as the cause of this form of HPFH. The HPFH phenotype was also reproduced in transgenic mice carrying the mu'LCRAgamma(-173)psibetadeltabeta construct, in which the -175 T to C Agamma gene was substituted with the -173 T to C Agamma gene. In vitro experiments proved that the -175 mutation significantly reduced binding of Oct-1 but not GATA-1, whereas the -173 mutation dramatically decreased binding of GATA-1 but not Oct-1. These results suggest that abrogation of either GATA-1 or Oct-1 binding to this promoter region may result in the HPFH phenotype. An in vivo footprinting assay revealed that either the -175 mutation or the -173 mutation significantly decreased overall protein binding to this promoter region in adult erythrocytes of transgenic mice. We hypothesize that a multiprotein complex containing GATA-1, Oct-1, and other protein factors may contribute to the formation of a repressive chromatin structure that silences gamma-globin gene expression in normal adult erythrocytes. Both the -173 and -175 T to C substitutions may disrupt the complex assembly and result in the reactivation of the gamma-globin gene in adult erythrocytes.


Assuntos
Proteínas de Ligação a DNA/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Globinas/genética , Fatores de Transcrição/biossíntese , Animais , Sequência de Bases , Sítios de Ligação , Linhagem Celular Tumoral , Cromatina/genética , Imunoprecipitação da Cromatina , Cosmídeos/metabolismo , DNA/química , DNA/metabolismo , Proteínas de Ligação a DNA/química , Eritrócitos/metabolismo , Fatores de Ligação de DNA Eritroide Específicos , Fator de Transcrição GATA1 , Deleção de Genes , Regulação da Expressão Gênica , Globinas/biossíntese , Globinas/metabolismo , Células HeLa , Humanos , Células K562 , Camundongos , Camundongos Transgênicos , Modelos Genéticos , Dados de Sequência Molecular , Mutação , Fator 1 de Transcrição de Octâmero , Fenótipo , Plasmídeos/metabolismo , Mutação Puntual , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas , Ligação Proteica , Conformação Proteica , RNA/metabolismo , RNA Mensageiro/metabolismo , Fatores de Tempo , Fatores de Transcrição/química
18.
Yi Chuan ; 24(3): 329-31, 2002 May.
Artigo em Chinês | MEDLINE | ID: mdl-16126693

RESUMO

Most of the important functionally proteins contain the corresponding function domains that consist of conserved amino acid sequences. The study provided a method to identify novel genes that encode proteins containing important functionally domains with conserved sequences. First, primers were designed according to the sequence of the cDNA library vector and the ESTs that have been obtained by reverse PCR and degenerate primers encoding Zinc finger domain. The cDNA library DNA was used as template for PCR amplification. The amplified fragment that contains nonhomologous sequences of the cDNA was inserted into pGEM-T easy vector. The fragment was recovered and used as a probe for screening the cDNA library. Several cDNAs with full length that encode proteins with Zinc finger domain and represent the original ESTs have been successfully cloned from a human bone marrow cDNA library. This strategy can also be used in screening genes that encode proteins containing differential function domains with conserved sequences.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...