Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 167
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
1.
Nucleic Acids Res ; 52(5): 2711-2723, 2024 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-38281192

RESUMEN

Class IIa Histone deacetylases (HDACs), including HDAC4, 5, 7 and 9, play key roles in multiple important developmental and differentiation processes. Recent studies have shown that class IIa HDACs exert their transcriptional repressive function by interacting with tissue-specific transcription factors, such as members of the myocyte enhancer factor 2 (MEF2) family of transcription factors. However, the molecular mechanism is not well understood. In this study, we determined the crystal structure of an HDAC4-MEF2A-DNA complex. This complex adopts a dumbbell-shaped overall architecture, with a 2:4:2 stoichiometry of HDAC4, MEF2A and DNA molecules. In the complex, two HDAC4 molecules form a dimer through the interaction of their glutamine-rich domain (GRD) to form the stem of the 'dumbbell'; while two MEF2A dimers and their cognate DNA molecules are bridged by the HDAC4 dimer. Our structural observations were then validated using biochemical and mutagenesis assays. Further cell-based luciferase reporter gene assays revealed that the dimerization of HDAC4 is crucial in its ability to repress the transcriptional activities of MEF2 proteins. Taken together, our findings not only provide the structural basis for the assembly of the HDAC4-MEF2A-DNA complex but also shed light on the molecular mechanism of HDAC4-mediated long-range gene regulation.


Asunto(s)
ADN , Histona Desacetilasas , Factores de Transcripción MEF2 , Proteínas Represoras , ADN/química , ADN/metabolismo , Regulación de la Expresión Génica , Genes Reporteros , Factores de Transcripción MEF2/química , Factores de Transcripción MEF2/metabolismo , Factores Reguladores Miogénicos/química , Factores Reguladores Miogénicos/genética , Factores Reguladores Miogénicos/metabolismo , Proteínas Represoras/química , Proteínas Represoras/metabolismo , Humanos , Histona Desacetilasas/química , Histona Desacetilasas/metabolismo
2.
Mol Cell Proteomics ; 22(6): 100567, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37172717

RESUMEN

Nasopharyngeal carcinoma (NPC), a malignant tumor distinctly characterized by ethnic and geographic distribution, is highly prevalent in Southern China and Southeast Asia. However, the molecular mechanisms of NPC have not been fully revealed at the proteomic level. In this study, 30 primary NPC samples and 22 normal nasopharyngeal epithelial tissues were collected for proteomics analysis, and a relatively complete proteomics landscape of NPC was depicted for the first time. By combining differential expression analysis, differential co-expression analysis, and network analysis, potential biomarkers and therapeutic targets were identified. Some identified targets were verified by biological experiments. We found that 17-AAG, a specific inhibitor of the identified target heat shock protein 90 (HSP90), could be a potential therapeutic drug for NPC. Finally, consensus clustering identified two NPC subtypes with specific molecular features. The subtypes and the related molecules were verified by an independent data set and may have different progression-free survival. The results of this study provide a comprehensive understanding of the proteomics molecular signatures of NPC and provide new perspectives and inspiration for prognostic determination and treatment of NPC.


Asunto(s)
Carcinoma , Neoplasias Nasofaríngeas , Humanos , Carcinoma Nasofaríngeo , Carcinoma/metabolismo , Neoplasias Nasofaríngeas/metabolismo , Proteómica/métodos , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo
3.
Nucleic Acids Res ; 49(6): 3573-3583, 2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33577686

RESUMEN

Forkhead transcription factors bind a canonical consensus DNA motif, RYAAAYA (R = A/G, Y = C/T), as a monomer. However, the molecular mechanisms by which forkhead transcription factors bind DNA as a dimer are not well understood. In this study, we show that FOXO1 recognizes a palindromic DNA element DIV2, and mediates transcriptional regulation. The crystal structure of FOXO1/DIV2 reveals that the FOXO1 DNA binding domain (DBD) binds the DIV2 site as a homodimer. The wing1 region of FOXO1 mediates the dimerization, which enhances FOXO1 DNA binding affinity and complex stability. Further biochemical assays show that FOXO3, FOXM1 and FOXI1 also bind the DIV2 site as homodimer, while FOXC2 can only bind this site as a monomer. Our structural, biochemical and bioinformatics analyses not only provide a novel mechanism by which FOXO1 binds DNA as a homodimer, but also shed light on the target selection of forkhead transcription factors.


Asunto(s)
ADN/metabolismo , Proteína Forkhead Box O1/química , Proteína Forkhead Box O1/metabolismo , ADN/química , Factores de Transcripción Forkhead/química , Factores de Transcripción Forkhead/metabolismo , Células HEK293 , Humanos , Secuencias Invertidas Repetidas , Modelos Moleculares , Unión Proteica , Multimerización de Proteína , Transcripción Genética
4.
Biochem Biophys Res Commun ; 605: 9-15, 2022 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-35306364

RESUMEN

Fumarates (fumaric acid esters), primarily dimethyl fumarate (DMF) and monoethyl fumarate (MEF) and its salts, are orally administered systemic agents used for the treatment of psoriasis and multiple sclerosis. It is widely believed that the pharmaceutical activities of fumarates are exerted through the Keap1-Nrf2 pathway. Although it has been revealed that DMF and MEF differentially modify specific Keap1 cysteine residues and result in the differential activation of Nrf2, how the modification of DMF and MEF impacts the biochemical properties of Keap1 has not been well characterized. Here, we found that both DMF and MEF can only modify the BTB domain of Keap1 and that only C151 is accessible for covalent binding in vitro. Dynamic fluorescence scanning (DSF) assays showed that the modification of DMF to Keap1 BTB increased its thermal stability, while the modification of MEF dramatically decreased its thermal stability. Further crystal structures revealed no significant conformational variation between the DMF-modified and MEF-modified BTBs. Overall, our biochemical and structural study provides a better understanding of the covalent modification of fumarates to Keap1 and may suggest fundamentally different mechanisms adopted by fumarates in regulating the Keap1-Nrf2 pathway.


Asunto(s)
Dimetilfumarato , Factor 2 Relacionado con NF-E2 , Dimetilfumarato/farmacología , Fumaratos/química , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Unión Proteica
5.
Biochem Biophys Res Commun ; 598: 15-19, 2022 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-35151199

RESUMEN

Ponatinib is a multi-target tyrosine kinase inhibitor that targets ABL, SRC, FGFR, and so on. It was designed to overcome the resistance of BCR-ABL mutation to imatinib, especially the gatekeeper mutation ABLT315I. The molecular mechanism by which ponatinib overcomes mutations of BCR-ABL and some other targets has been explained, but little information is known about the characteristics of ponatinib binding to SRC. Here, we showed that ponatinib inhibited wild type SRC kinase but failed to inhibit SRC gatekeeper mutants in both biochemical and cellular assays. We determined the crystal structure of ponatinib in complex with the SRC kinase domain. In addition, by structural analysis, we provided a possible explanation for why ponatinib showed different effects on SRC and other kinases with gatekeeper mutations. The resistance mechanism of SRC gatekeeper mutations to ponatinib may provide meaningful information for designing inhibitors against SRC family kinases in the future.


Asunto(s)
Imidazoles/química , Imidazoles/farmacología , Inhibidores de Proteínas Quinasas/química , Piridazinas/química , Piridazinas/farmacología , Familia-src Quinasas/química , Sitios de Unión , Cristalografía por Rayos X , Humanos , Imidazoles/metabolismo , Modelos Moleculares , Mutación , Conformación Proteica , Dominios Proteicos , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-hck/química , Proteínas Proto-Oncogénicas c-hck/metabolismo , Piridazinas/metabolismo , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
6.
World J Surg Oncol ; 20(1): 347, 2022 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-36258216

RESUMEN

BACKGROUND: Gelsolin-like capping actin protein (CapG) modulates actin dynamics and actin-based motility with a debatable role in tumorigenic progression. The motility-associated functions and potential molecular mechanisms of CapG in nasopharyngeal carcinoma (NPC) remain unclear. METHODS: CapG expression was detected by immunohistochemistry in a cohort of NPC tissue specimens and by Western blotting assay in a variety of NPC cell lines. Loss of function and gain of function of CapG in scratch wound-healing and transwell assays were performed. Inactivation of Rac1 and ROCK with the specific small molecular inhibitors was applied to evaluate CapG's role in NPC cell motility. GTP-bound Rac1 and phosphorylated-myosin light chain 2 (p-MLC2) were measured in the ectopic CapG overexpressing cells. Finally, CapG-related gene set enrichment analysis was conducted to figure out the significant CapG-associated pathways in NPC. RESULTS: CapG disclosed increased level in the poorly differentiated NPC tissues and highly metastatic cells. Knockdown of CapG reduced NPC cell migration and invasion in vitro, while ectopic CapG overexpression showed the opposite effect. Ectopic overexpression of CapG compensated for the cell motility loss caused by simultaneous inactivation of ROCK and Rac1 or inactivation of ROCK alone. GTP-bound Rac1 weakened, and p-MLC2 increased in the CapG overexpressing cells. Bioinformatics analysis validated a positive correlation of CapG with Rho motility signaling, while Rac1 motility pathway showed no significant relationship. CONCLUSIONS: The present findings highlight the contribution of CapG to NPC cell motility independent of ROCK and Rac1. CapG promotes NPC cell motility at least partly through MLC2 phosphorylation and contradicts with Rac1 activation.


Asunto(s)
Actinas , Neoplasias Nasofaríngeas , Humanos , Actinas/metabolismo , Carcinoma Nasofaríngeo/genética , Gelsolina/análisis , Gelsolina/genética , Gelsolina/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Neoplasias Nasofaríngeas/genética , Guanosina Trifosfato , Regulación Neoplásica de la Expresión Génica , Proteínas de Microfilamentos/metabolismo , Proteínas Nucleares/genética
7.
Bioorg Med Chem Lett ; 34: 127757, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33359446

RESUMEN

Ibrutinib is a BTK-targeted irreversible inhibitor. In this study, we demonstrate that ibrutinib potently inhibits SRC activity in a non-covalent manner via mass spectrometry and crystallography. The S345C mutation renders SRC to bind covalently with ibrutinib, and restores the potency of ibrutinib against the gatekeeper mutant. The co-crystal structure of ibrutinib/SRC shows Ser345 of SRC did not form covalent bond with ibrutinib, leading to a decrease of potency and loss of the ability to overcome the gatekeeper mutation of SRC. The X-ray crystallographic studies also provide structural insight into why ibrutinib behaves differently against gatekeeper mutants of different kinases.


Asunto(s)
Adenina/análogos & derivados , Piperidinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Familia-src Quinasas/antagonistas & inhibidores , Adenina/química , Adenina/farmacología , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Humanos , Modelos Moleculares , Estructura Molecular , Piperidinas/química , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad , Familia-src Quinasas/metabolismo
8.
Nucleic Acids Res ; 47(7): 3752-3764, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-30722065

RESUMEN

The FOXC family of transcription factors (FOXC1 and FOXC2) plays essential roles in the regulation of embryonic, ocular, and cardiac development. Mutations and abnormal expression of FOXC proteins are implicated in genetic diseases as well as cancer. In this study, we determined two crystal structures of the DNA-binding domain (DBD) of human FOXC2 protein, in complex with different DNA sites. The FOXC2-DBD adopts the winged-helix fold with helix H3 contributing to all the base specific contacts, while the N-terminus, wing 1, and the C-terminus of FOXC2-DBD all make additional contacts with the phosphate groups of DNA. Our structural, biochemical, and bioinformatics analyses allow us to revise the previously proposed DNA recognition mechanism and provide a model of DNA binding for the FOXC proteins. In addition, our structural analysis and accompanying biochemical assays provide a molecular basis for understanding disease-causing mutations in FOXC1 and FOXC2.


Asunto(s)
Proteínas de Unión al ADN/química , ADN/química , Factores de Transcripción Forkhead/química , ADN/genética , Proteínas de Unión al ADN/genética , Factores de Transcripción Forkhead/genética , Regulación de la Expresión Génica/genética , Humanos , Mutación , Conformación de Ácido Nucleico , Dominios Proteicos/genética
9.
J Biol Chem ; 294(51): 19795-19803, 2019 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-31723028

RESUMEN

Proteins of nuclear receptor subfamily 4 group A (NR4A), including NR4A1/NGFI-B, NR4A2/Nurr1, and NR4A3/NOR-1, are nuclear transcription factors that play important roles in metabolism, apoptosis, and proliferation. NR4A proteins recognize DNA response elements as monomers or dimers to regulate the transcription of a variety of genes involved in multiple biological processes. In this study, we determined two crystal structures of the NR4A2 DNA-binding domain (NR4A2-DBD) bound to two Nur-responsive elements: an inverted repeat and an everted repeat at 2.6-2.8 Å resolution. The structures revealed that two NR4A2-DBD molecules bind independently to the everted repeat, whereas two other NR4A2-DBD molecules form a novel dimer interface on the inverted repeat. Moreover, substitution of the interfacial residue valine 298 to lysine as well as mutation of DNA bases involved in the interactions abolished the dimerization. Overall, our structural, biochemical, and bioinformatics analyses provide a molecular basis for the binding of the NR4A2 protein dimers to NurREs and advance our understanding of the dimerization specificity of nuclear receptors.


Asunto(s)
ADN/química , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/química , Receptores X Retinoide/química , Secuencias de Aminoácidos , Apoptosis , Núcleo Celular/química , Proliferación Celular , Dimerización , Escherichia coli , Humanos , Lisina/química , Mutación , Unión Proteica , Mapeo de Interacción de Proteínas , Transcripción Genética , Valina/química
10.
Biochem Biophys Res Commun ; 523(1): 1-5, 2020 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-31822342

RESUMEN

The nuclear receptor NR4A subfamily (NR4A1/NGFI-B, NR4A2/Nurr1 and NR4A3/NOR-1) can recognize different classes of DNA response elements either as a monomer, homodimer, or heterodimer. In this study, we determined the structure of the NR4A1 DNA-binding domain (NR4A1-DBD) bound to natural Nur-responsive elements (NurREs) in the promoter region of the pituitary proopiomelanocortin (POMC) gene (NurREPOMC) at 3.12 Å resolution. The NR4A1-DBD molecules bound independently to this element in our structure. The N-terminal helix H1 forms specific contacts with major groove, and C-terminal extension interact extensively with minor groove. Moreover our modelling structure of NR4A1 large fragment complexed with NurREPOMC indicated that ligand binding domain of NR4A might form dimer interactions to help recognize DNA. Overall, our analyses provide a molecular basis for DNA binding of NR4A proteins as a homodimer and novel insight into the molecular functions of NR4A modulation of gene expression.


Asunto(s)
Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/química , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Hipófisis/metabolismo , Proopiomelanocortina/genética , Regiones Promotoras Genéticas/genética , Humanos , Modelos Moleculares , Conformación Proteica
11.
Expert Rev Proteomics ; 16(6): 475-485, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31056975

RESUMEN

Introduction: Nasopharyngeal carcinoma (NPC) is a distinct head and neck squamous cell carcinoma in its etiological association of Epstein-Barr virus (EBV) infection, hidden anatomical location, remarkable racial and geographical distribution, and high incidence of locoregional recurrence or metastasis. Thanks to the advancements in proteomics in recent decades, more understanding of the disease etiology, carcinogenesis, and progression has been gained, potentially deciphering the molecular characteristics of the malignancy. Areas covered: In this review, we provide an overview of the proteomic aberrations that are likely involved or drive NPC development and progression, focusing on the contributions of major EBV-encoded factors, intercommunication with environment, protein features of high metastasis and therapy resistance, and protein-protein interactions that allow NPC cells to evade immune recognition and elimination. Finally, multistep carcinogenesis and subtypes of NPC from a proteomic perspective are inquired. Expert commentary: Proteomic studies have covered various aspects involved in NPC pathogenesis, yet much remains to be uncovered. Coherent study designs, optimal conditions for obtaining high-quality data, and compelling interpretation are critical in ensuring the emergence of good science out of NPC proteomics. NPC proteogenomics and proteoform analysis are two promising fields to promote the application of the proteomic findings from bench to bedside.


Asunto(s)
Carcinoma Nasofaríngeo/metabolismo , Carcinoma Nasofaríngeo/patología , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/patología , Proteómica/métodos , Animales , Infecciones por Virus de Epstein-Barr/metabolismo , Infecciones por Virus de Epstein-Barr/patología , Humanos
12.
J Neurooncol ; 141(1): 19-30, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30539341

RESUMEN

BACKGROUND: Glioblastoma (GBM) is the most common malignant tumor originating in the brain parenchyma. The invasive and infiltrative properties of glioblastoma result in poor clinical prognosis to conventional therapies. Emerging reports on microRNAs as important regulators during the process of EMT provide new insights into treating glioblastoma through new targets. However, underlying molecular mechanism of the regulation of miR-101-3p in glioblastoma remains unclear. METHODS: Level of miR-101-3p was determined in GBM cell lines by qRT-PCR. MTT, colony formation and transwell assays were utilized to evaluate functions of overexpression of miR-101-3p/knock down of TRIM44 on proliferation, migration and invasion in GBM cells. Direct interaction between miR-101-3p and TRIM44 was validated using dual luciferase reporter system and impacts of overexpression of miR-101-3p/knock down of TRIM44 on regulation of EMT markers were assessed by Western blotting. RESULTS: MiR-101-3p was validated to be repressed expressed in glioblastoma cancer cell lines. Both overexpression of miR-101-3p and knock down of TRIM44 attenuated proliferation, migration and invasion of glioblastoma cell lines in vitro. TRIM44 was shown to promote EMT in GBM progress and reverse inhibitory function of miR-101-3p. MiR-101-3p was found to suppress the expression of TRIM44 via directly targeting its 3'UTR. CONCLUSIONS: Our findings suggested miR-101-3p regulated proliferation and migration of glioblastoma cells through attenuating TRIM44 induced EMT via direct targeting 3'UTR of TRIM44, which provided preliminary study of potential therapeutic target in future GBM treatment.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Proteínas Portadoras/metabolismo , Proliferación Celular , Glioblastoma/metabolismo , MicroARNs/metabolismo , Metástasis de la Neoplasia , Línea Celular Tumoral , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular , Proteínas de Motivos Tripartitos
13.
Bioinformatics ; 33(8): 1187-1196, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28096083

RESUMEN

Motivation: : Exploring the potential curative effects of drugs is crucial for effective drug development. Previous studies have indicated that integration of multiple types of information could be conducive to discovering novel indications of drugs. However, how to efficiently identify the mechanism behind drug-disease associations while integrating data from different sources remains a challenging problem. Results: : In this research, we present a novel method for indication prediction of both new drugs and approved drugs. This method is based on Laplacian regularized sparse subspace learning (LRSSL), which integrates drug chemical information, drug target domain information and target annotation information. Experimental results show that the proposed method outperforms several recent approaches for predicting drug-disease associations. Some drug therapeutic effects predicted by the method could be validated by database records or literatures. Moreover, with L1-norm constraint, important drug features have been extracted from multiple drug feature profiles. Case studies suggest that the extracted drug features could be beneficial to interpretation of the predicted results. Availability and Implementation: https://github.com/LiangXujun/LRSSL. Contact: proteomics@csu.edu.cn. Supplementary information: Supplementary data are available at Bioinformatics online.


Asunto(s)
Algoritmos , Enfermedad , Preparaciones Farmacéuticas/metabolismo , Estadística como Asunto , Colitis Ulcerosa/tratamiento farmacológico , Reposicionamiento de Medicamentos , Humanos
15.
Biochemistry ; 56(29): 3745-3753, 2017 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-28644006

RESUMEN

FOXA2, a member of the forkhead family of transcription factors, plays essential roles in liver development and bile acid homeostasis. In this study, we report a 2.8 Å co-crystal structure of the FOXA2 DNA-binding domain (FOXA2-DBD) bound to a DNA duplex containing a forkhead consensus binding site (GTAAACA). The FOXA2-DBD adopts the canonical winged-helix fold, with helix H3 and wing 1 regions mainly mediating the DNA recognition. Although the wing 2 region was not defined in the structure, isothermal titration calorimetry assays suggested that this region was required for optimal DNA binding. Structure comparison with the FOXA3-DBD bound to DNA revealed more major groove contacts and fewer minor groove contacts in the FOXA2 structure than in the FOXA3 structure. Structure comparison with the FOXO1-DBD bound to DNA showed that different forkhead proteins could induce different DNA conformations upon binding to identical DNA sequences. Our findings provide the structural basis for FOXA2 protein binding to a consensus forkhead site and elucidate how members of the forkhead protein family bind different DNA sites.


Asunto(s)
ADN/química , Factor Nuclear 3-beta del Hepatocito/química , Motivos de Nucleótidos , Cristalografía por Rayos X , ADN/metabolismo , Factor Nuclear 3-beta del Hepatocito/metabolismo , Factor Nuclear 3-gamma del Hepatocito/química , Factor Nuclear 3-gamma del Hepatocito/metabolismo , Humanos , Unión Proteica , Dominios Proteicos , Homología Estructural de Proteína
16.
Acta Biochim Biophys Sin (Shanghai) ; 49(9): 792-799, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28910978

RESUMEN

FOXP3, a lineage-specific forkhead (FKH) transcription factor, plays essential roles in the development and function of regulatory T cells. However, the DNA-binding properties of FOXP3 are not well understood. In this study, FOXP3 fragments containing different domains were purified, and their DNA-binding properties were investigated using electrophoretic mobility shift assay and isothermal titration calorimetry (ITC). Both the FKH and leucine-zipper domains were required for optimal DNA binding for FOXP3. FOXP3 protein not only binds with DNA sequences containing one FKH consensus sequence, but also binds with DNA sequences with two direct repeats of consensus sequences separated by three-nucleotides (DRE3). Our results shed lights on the mechanisms by which FOXP3 recognizes cognate DNA elements, and would facilitate further structural and functional studies of FOXP3.


Asunto(s)
ADN/química , Factores de Transcripción Forkhead/química , Conformación de Ácido Nucleico , Dominios Proteicos , Secuencia de Aminoácidos , Secuencia de Bases , Sitios de Unión/genética , ADN/genética , ADN/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Humanos , Modelos Moleculares , Unión Proteica
17.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 42(1): 1-7, 2017 Jan 28.
Artículo en Zh | MEDLINE | ID: mdl-28216490

RESUMEN

OBJECTIVE: To express and purify FOXO1 DNA binding domain (DBD) and to evaluate its DNA binding properties.
 Methods: Gene sequence of FOXO1-DBD was optimized and the recombinant strains were induced at low temperature to obtain soluble protein of FOXO1-DBD. Then, FOXO1-DBD protein was purified through Ni affinity chromatography and cation exchange. Finally, the DNA binding properties of FOXO1-DBD was evaluated by electrophoretic mobility shift assay (EMSA).
 Results: Most of soluble proteins were obtained by optimizing its genes sequence and induction at 21 ℃. Over 95% purity of FOXO1-DBD protein was obtained through two-steps purification. Purified protein exhibited good binding property to the DNA binding motif (G/ATAAACA) of FOX family.
 Conclusion: An efficient expression and purification method for FOXO1-DBD is established, and the complexity of FOXO1 protein in recognition of DNA is verified.


Asunto(s)
Proteínas de Unión al ADN , ADN , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/aislamiento & purificación , Proteína Forkhead Box O1/metabolismo , Dominios Proteicos , ADN/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Datos de Secuencia Molecular , Unión Proteica , Proteínas Recombinantes
18.
Protein Expr Purif ; 128: 67-72, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27546451

RESUMEN

PTEN-induced putative kinase 1 (PINK1) is a Ser/Thr kinase that specifically localizes on the mitochondrial membrane. It cooperates with Parkin to regulate mitochondrial quality control. Mutations in PINK1 protein which account for 8-15% of Parkinson's disease (PD), are the second most common cause of early-onset Autosomal Recessive Parkinson's disease (AR-PD). The lack of methods for PINK1 heterologous expression and purification has slowed progress in the AR-PD research field. To pave the way for direct structural study of this important protein, in this study, we developed an efficient expression system of recombinant PINK1 kinase domain (rPINK1) using Pichia pastoris (P. pastoris). Our results showed that rPINK1 is best expressed in P. pastoris at 25 °C induction. Additionally, we determined that the optimal induction time was 72 h and the optimal induction methanol concentration was 1% for the expression of rPINK1 in P. pastoris. Subsequent purification by Ni affinity chromatography (Ni-NTA) and cation-exchange chromatography (Mono S) produced the protein with purity higher than 95%. The pure rPINK1 was active to phosphorylate ubiquitin in a substrate phosphorylation assay. Overall, these studies provide the first effective method for heterologous expression and purification of the rPINK1 with a high purity. These findings can help contribute to further researches on the interactions study and biochemical characterization of PINK1.


Asunto(s)
Expresión Génica , Pichia/metabolismo , Proteínas Quinasas , Cromatografía de Afinidad/métodos , Cromatografía por Intercambio Iónico/métodos , Humanos , Pichia/genética , Dominios Proteicos , Proteínas Quinasas/biosíntesis , Proteínas Quinasas/química , Proteínas Quinasas/genética , Proteínas Quinasas/aislamiento & purificación , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/aislamiento & purificación
19.
Mol Cancer ; 14: 73, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25889562

RESUMEN

BACKGROUND: microRNAs are small noncoding RNAs that modulate a variety of cellular processes by regulating multiple targets, which can promote or inhibit the development of malignant behaviors. Accumulating evidence suggests that miR-675-5p plays important roles in human carcinogenesis. However, its precise biological role remains largely elusive. This study examined the role of miR-675-5p in non- small cell lung cancer (NSCLC). METHODS: The expression of miR-675-5p was analyzed by real-time quantitative PCR (qRT-PCR). The effect of miR-675-5p on proliferation was evaluated through MTT and colony formation assays, and cell migration and invasion were evaluated through transwell assays. The expression of target proteins and downstream molecules was analyzed by western blotting and immunohistochemical staining. The luciferase reporter assay was used to assess the target genes of miR-675-5p in non-small cell lung cancer cells. RESULTS: The expression levels of miR-675-5p in NSCLC tissues were significantly reduced compared to those in adjacent non-cancerous tissues (P < 0.001). The expression of miR-675-5p in patients with non-small cell lung cancer had a negative correlation with lymph node metastasis (P < 0.01) and TNM stage (P < 0.05). Down-regulation of miR-675-5p promoted cell growth, proliferation, colony formation, invasion and migration, and promoted the tumorigenicity graft growth of nude mice in vivo (P < 0.01); whereas up-regulation of miR-675-5p had the contrary effects. The luciferase reporter assay showed that GPR55 was a direct target gene of miR-675-5p. Overexpression of miR-675-5p can lead to the down-regulation of GPR55 and its signaling pathway, whereas the effect can be reversed by down-regulation of miR-675-5p expression. CONCLUSIONS: miR-675-5p functions as a novel tumor suppressor in NSCLC and the anti-oncogenic activity may involve its inhibition of the target gene GPR55. These findings suggest the possibility for miR-675-5p as a therapeutic target in NSCLC.


Asunto(s)
Carcinogénesis/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Regulación hacia Abajo/genética , Neoplasias Pulmonares/genética , MicroARNs/genética , Receptores Acoplados a Proteínas G/genética , Animales , Carcinogénesis/patología , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Progresión de la Enfermedad , Femenino , Humanos , Neoplasias Pulmonares/patología , Metástasis Linfática/genética , Metástasis Linfática/patología , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Receptores de Cannabinoides , Transducción de Señal/genética , Regulación hacia Arriba/genética
20.
Nucleic Acids Res ; 41(17): 8368-76, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23836939

RESUMEN

The p53 core domain binds to response elements (REs) that contain two continuous half-sites as a cooperative tetramer, but how p53 recognizes discontinuous REs is not well understood. Here we describe the crystal structure of the p53 core domain bound to a naturally occurring RE located at the promoter of the Bcl-2-associated X protein (BAX) gene, which contains a one base-pair insertion between the two half-sites. Surprisingly, p53 forms a tetramer on the BAX-RE that is nearly identical to what has been reported on other REs with a 0-bp spacer. Each p53 dimer of the tetramer binds in register to a half-site and maintains the same protein-DNA interactions as previously observed, and the two dimers retain all the protein-protein contacts without undergoing rotation or translation. To accommodate the additional base pair, the DNA is deformed and partially disordered around the spacer region, resulting in an apparent unwinding and compression, such that the interactions between the dimers are maintained. Furthermore, DNA deformation within the p53-bound BAX-RE is confirmed in solution by site-directed spin labeling measurements. Our results provide a structural insight into the mechanism by which p53 binds to discontinuous sites with one base-pair spacer.


Asunto(s)
ADN/química , Elementos de Respuesta , Proteína p53 Supresora de Tumor/química , Proteína X Asociada a bcl-2/genética , Emparejamiento Base , Sitios de Unión , Modelos Moleculares , Conformación de Ácido Nucleico , Proteína p53 Supresora de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA