Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Aging (Albany NY) ; 13(10): 13739-13763, 2021 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-34023818

RESUMEN

ETS1 - an evolutionarily conserved transcription factor involved in the regulation of a number of cellular processes - is overexpressed in several malignancies, including ovarian cancer. Most studies on ETS1 expression have been focused on the transcriptional and RNA levels, with post-translational control mechanisms remaining relatively unexplored in the pathogenesis of malignancies. Here, we show that ETS1 forms a complex with glycogen synthase kinase-3ß (GSK3ß). Specifically, GSK3ß-mediated phosphorylation of ETS1 at threonine 265 and serine 269 promoted protein stability, induced the transcriptional activation of matrix metalloproteinase (MMP)-9, and increased cell migration. In vivo experiments revealed that a GSK3ß inhibitor was able to suppress both endogenous ETS1 expression and induction of MMP-9 expression. Upon generation of a specific antibody against phosphorylated ETS1, we demonstrated that phospho-ETS1 immunohistochemical expression in ovarian cancer specimens was correlated with that of MMP-9. Notably, the cumulative overall survival of patients with low phospho-ETS1 histoscores was significantly longer than that of those showing higher scores. We conclude that the GSK3ß/ETS1/MMP-9 axis may regulate the biological aggressiveness of ovarian cancer and can serve as a prognostic factor in patients with this malignancy.


Asunto(s)
Progresión de la Enfermedad , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Proteína Proto-Oncogénica c-ets-1/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Movimiento Celular/genética , Femenino , Humanos , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Endogámicos C57BL , Modelos Biológicos , Mutación/genética , Estadificación de Neoplasias , Fosforilación , Unión Proteica , Estabilidad Proteica , Proteína Proto-Oncogénica c-ets-1/química , Proteína Proto-Oncogénica c-ets-1/genética , Serina/metabolismo , Especificidad por Sustrato , Treonina/metabolismo
3.
J Biol Chem ; 294(25): 9666-9678, 2019 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-31048376

RESUMEN

Functional evidence increasingly implicates low-affinity DNA recognition by transcription factors as a general mechanism for the spatiotemporal control of developmental genes. Although the DNA sequence requirements for affinity are well-defined, the dynamic mechanisms that execute cognate recognition are much less resolved. To address this gap, here we examined ETS1, a paradigm developmental transcription factor, as a model for which cognate discrimination remains enigmatic. Using molecular dynamics simulations, we interrogated the DNA-binding domain of murine ETS1 alone and when bound to high-and low-affinity cognate sites or to nonspecific DNA. The results of our analyses revealed collective backbone and side-chain motions that distinguished cognate versus nonspecific as well as high- versus low-affinity cognate DNA binding. Combined with binding experiments with site-directed ETS1 mutants, the molecular dynamics data disclosed a triad of residues that respond specifically to low-affinity cognate DNA. We found that a DNA-contacting residue (Gln-336) specifically recognizes low-affinity DNA and triggers the loss of a distal salt bridge (Glu-343/Arg-378) via a large side-chain motion that compromises the hydrophobic packing of two core helices. As an intact Glu-343/Arg-378 bridge is the default state in unbound ETS1 and maintained in high-affinity and nonspecific complexes, the low-affinity complex represents a unique conformational adaptation to the suboptimization of developmental enhancers.


Asunto(s)
ADN/química , ADN/metabolismo , Proteína Proto-Oncogénica c-ets-1/química , Proteína Proto-Oncogénica c-ets-1/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Ratones , Modelos Moleculares , Simulación de Dinámica Molecular , Unión Proteica , Conformación Proteica
4.
Genes (Basel) ; 10(3)2019 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-30857266

RESUMEN

The transcription factor Ets-1 (ETS proto-oncogene 1) shows low expression levels except in specific biological processes like haematopoiesis or angiogenesis. Elevated levels of expression are observed in tumor progression, resulting in Ets-1 being named an oncoprotein. It has recently been shown that Ets-1 interacts with two DNA repair enzymes, PARP-1 (poly(ADP-ribose) polymerase 1) and DNA-PK (DNA-dependent protein kinase), through two different domains and that these interactions play a role in cancer. Considering that Ets-1 can bind to distinctly different domains of two DNA repair enzymes, we hypothesized that the interaction can be transposed onto homologs of the respective domains. We have searched for sequence and structure homologs of the interacting ETS(Ets-1), BRCT(PARP-1) and SAP(DNA-PK) domains, and have identified several candidate binding pairs that are currently not annotated as such. Many of the Ets-1 partners are associated to DNA repair mechanisms. We have applied protein-protein docking to establish putative interaction poses and investigated these using centrality analyses at the protein residue level. Most of the identified poses are virtually similar to our recently established interaction model for Ets-1/PARP-1 and Ets-1/DNA-PK. Our work illustrates the potentially high number of interactors of Ets-1, in particular involved in DNA repair mechanisms, which shows the oncoprotein as a potential important regulator of the mechanism.


Asunto(s)
Reparación del ADN , Mapas de Interacción de Proteínas , Proteína Proto-Oncogénica c-ets-1/metabolismo , Sitios de Unión , Proteína Quinasa Activada por ADN/química , Proteína Quinasa Activada por ADN/metabolismo , Humanos , Simulación del Acoplamiento Molecular , Poli(ADP-Ribosa) Polimerasa-1/química , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Unión Proteica , Proto-Oncogenes Mas , Proteína Proto-Oncogénica c-ets-1/química
5.
Mol Carcinog ; 58(6): 1068-1081, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30790340

RESUMEN

Epithelial-mesenchymal transition (EMT) is an embryonic program that is reactivated in cancer and regulates the invasion and metastasis of tumor cells. Zinc finger E-box binding homeobox 2 (ZEB2) induces EMT by upregulating matrix metalloproteinases (MMP), yet MMP genes lack ZEB2 binding motif in their promoters. Recently, expression of MMPs was associated to the activation of ETS1 transcription factor; however, a link between ZEB2 and ETS proto-oncogene 1, transcription factor (ETS1) remains to be elucidated. Hence, we investigated the transcriptional regulation of ETS1 by ZEB2 after our initial observation that ZEB2 and ETS1 are coexpressed in hepatocellular carcinoma cells (HCCs). Chromatin immunoprecipitation and luciferase reporter assays clearly showed that ZEB2 binds to E-box sequences on the promoter of ETS1. Elevated expression of ETS1 was found in DLD-ZEB2 and A431-ZEB2 inducible systems, and knockdown of ZEB2 caused an explicit downregulation of ETS1 in shZEB2-SNU398 and shZEB2-SK-HEP-1 cells. Repression of ETS1 expression in ZEB2-induced conditions substantially impaired the migration and invasive capacities of DLD1 cells. Mechanistically, knockdown of ETS1 in ZEB2-expressing cells resulted in the downregulation of established ZEB2 targets TWIST and MMP9. Correlation analyses in HCC lines, cancer complementary DNA arrays, and The Cancer Genome Atlas RNA-sequencing data set revealed that ZEB2 and ETS1 are coexpressed, and their expressions in human tumors show a highly significant positive correlation. Our results demonstrated that ZEB2 acts as an upstream regulator of ETS1 and, in turn, ETS1 maintains ZEB2-induced EMT. These findings add another level of complexity to the understanding of ZEB2 in the invasion and metastasis of cancer cells, and put ZEB2/ETS1 axis as a novel therapeutic target in human malignancies.


Asunto(s)
Neoplasias/metabolismo , Proteína Proto-Oncogénica c-ets-1/genética , Proteína Proto-Oncogénica c-ets-1/metabolismo , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc/genética , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc/metabolismo , Sitios de Unión , Línea Celular Tumoral , Movimiento Celular , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células Hep G2 , Humanos , Metaloproteinasa 9 de la Matriz/metabolismo , Neoplasias/genética , Proteínas Nucleares/metabolismo , Proto-Oncogenes Mas , Proteína Proto-Oncogénica c-ets-1/química , Proteína 1 Relacionada con Twist/metabolismo
6.
Nucleic Acids Res ; 46(20): 10577-10588, 2018 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-30295801

RESUMEN

Hydration of interfaces is a major determinant of target specificity in protein/DNA interactions. Interfacial hydration is a highly variable feature in DNA recognition by ETS transcription factors and functionally relates to cellular responses to osmotic stress. To understand how hydration is mediated in the conserved ETS/DNA binding interface, secondary structures comprising the DNA contact surface of the strongly hydrated ETS member PU.1 were substituted, one at a time, with corresponding elements from its sparsely hydrated relative Ets-1. The resultant PU.1/Ets-1 chimeras exhibited variably reduced sensitivity to osmotic pressure, indicative of a distributed pattern of interfacial hydration in wildt-ype PU.1. With the exception of the recognition helix H3, the chimeras retained substantially high affinities. Ets-1 residues could therefore offset the loss of favorable hydration contributions in PU.1 via low-water interactions, but at the cost of decreased selectivity at base positions flanking the 5'-GGA-3' core consensus. Substitutions within H3 alone, which contacts the core consensus, impaired binding affinity and PU.1 transactivation in accordance with the evolutionary separation of the chimeric residues involved. The combined biophysical, bioinformatics and functional data therefore supports hydration as an evolved specificity determinant that endows PU.1 with more stringent sequence selection over its ancestral relative Ets-1.


Asunto(s)
ADN/química , Proteína Proto-Oncogénica c-ets-1/química , Proteínas Proto-Oncogénicas/química , Transactivadores/química , Animales , Sitios de Unión , Clonación Molecular , Biología Computacional , Cristalización , Genes Reporteros , Células HEK293 , Humanos , Ratones , Modelos Moleculares , Simulación de Dinámica Molecular , Conformación de Ácido Nucleico , Ósmosis , Unión Proteica , Proteínas Recombinantes de Fusión/química , Termodinámica , Agua/química
7.
Proteins ; 86(10): 1055-1063, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30019773

RESUMEN

The Ets-1 oncoprotein is a transcription factor that promotes target gene expression in specific biological processes. Typically, Ets-1 activity is low in healthy cells, but elevated levels of expression have been found in cancerous cells, specifically related to tumor progression. Like the vast majority of the cellular effectors, Ets-1 does not act alone but in association with partners. Given the important role that is attributed to Ets-1 in major human diseases, it is crucial to identify its partners and characterize their interactions. In this context, two DNA-repair enzymes, PARP-1 and DNA-PK, have been identified recently as interaction partners of Ets-1. We here identify their binding mode by means of protein docking. The results identify the interacting surface between Ets-1 and the two DNA-repair enzymes centered on the α-helix H1 of the ETS domain, leaving α-helix H3 available to bind DNA. The models highlight a hydrophobic patch on Ets-1 at the center of the interaction interface that includes three tryptophans (Trp338, Trp356, and Trp361). We rationalize the binding mode using a series of computational analyses, including alanine scanning, molecular dynamics simulation, and residue centrality analysis. Our study constitutes a first but important step in the characterization, at the molecular level, of the interaction between an oncoprotein and DNA-repair enzymes.


Asunto(s)
Enzimas Reparadoras del ADN/metabolismo , Mapas de Interacción de Proteínas , Proteína Proto-Oncogénica c-ets-1/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Enzimas Reparadoras del ADN/química , Proteína Quinasa Activada por ADN/química , Proteína Quinasa Activada por ADN/metabolismo , Humanos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Poli(ADP-Ribosa) Polimerasa-1/química , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Unión Proteica , Conformación Proteica , Conformación Proteica en Hélice alfa , Proteína Proto-Oncogénica c-ets-1/química , Alineación de Secuencia
8.
Biosci Biotechnol Biochem ; 82(10): 1753-1759, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29912634

RESUMEN

The Ets-1 transcription factor plays an important role in various physiological and pathological processes. These diverse roles of Ets-1 are likely to depend on its interaction proteins. We have previously showed that Ets-1 interacted with DNA-dependent protein kinase (DNA-PK) complex including its regulatory subunits, Ku70 and Ku86 and with poly (ADP-ribose) polymerase-1 (PARP-1). In this study, the binding domains for the interaction between Ets-1 and these proteins were reported. We demonstrated that the interaction of Ets-1 with DNA-PK was mediated through the Ku70 subunit and was mapped to the C-terminal region of Ets-1 and the C-terminal part of Ku70 including SAP domain. The interactive domains between Ets-1 and PARP-1 have been mapped to the C-terminal region of Ets-1 and the BRCA1 carboxy-terminal (BRCT) domain of PARP-1. The results presented in this study may advance our understanding of the functional link between Ets-1 and its interaction partners, DNA-PK and PARP-1.


Asunto(s)
Autoantígeno Ku/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Proteína Proto-Oncogénica c-ets-1/metabolismo , Sitios de Unión , Humanos , Unión Proteica , Proteína Proto-Oncogénica c-ets-1/química
9.
Nucleic Acids Res ; 46(5): 2243-2251, 2018 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-29309620

RESUMEN

Multi-modal interactions are frequently observed in intrinsically disordered regions (IDRs) of proteins upon binding to their partners. In many cases, post-translational modifications in IDRs are accompanied by coupled folding and binding. From both molecular simulations and biochemical experiments with mutational studies, we show that the IDR including a Ser rich region (SRR) of the transcription factor Ets1, just before the DNA-binding core domain, undergoes multi-modal interactions when the SRR is not phosphorylated. In the phosphorylated state, the SRR forms a few specific complex structures with the Ets1 core, covering the recognition helix in the core and drastically reducing the DNA binding affinities as the auto-inhibitory state. The binding kinetics of mutated Ets1 indicates that aromatic residues in the SRR can be substituted with other hydrophobic residues for the interactions with the Ets1 core.


Asunto(s)
Proteínas Intrínsecamente Desordenadas/química , Simulación de Dinámica Molecular , Pliegue de Proteína , Procesamiento Proteico-Postraduccional , Proteína Proto-Oncogénica c-ets-1/química , Sustitución de Aminoácidos , Proteínas Intrínsecamente Desordenadas/metabolismo , Cinética , Fosforilación , Unión Proteica , Proteína Proto-Oncogénica c-ets-1/metabolismo , Serina/química , Serina/genética , Serina/metabolismo
10.
Biochemistry ; 55(29): 4105-18, 2016 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-27362745

RESUMEN

The affinity of the Ets-1 transcription factor for DNA is autoinhibited by an intrinsically disordered serine-rich region (SRR) and a helical inhibitory module (IM) appended to its winged helix-turn-helix ETS domain. Using NMR spectroscopy, we investigated how Ets-1 recognizes specific versus nonspecific DNA, with a focus on the roles of protein dynamics and autoinhibition in these processes. Upon binding either DNA, the two marginally stable N-terminal helices of the IM predominantly unfold, but still sample partially ordered conformations. Also, on the basis of amide chemical shift perturbation mapping, Ets-1 associates with both specific and nonspecific DNA through the same canonical ETS domain interface. These interactions are structurally independent of the SRR, and thus autoinhibition does not impart DNA-binding specificity. However, relative to the pronounced NMR spectroscopic changes in Ets-1 resulting from specific DNA binding, the spectra of the nonspecific DNA complexes showed conformational exchange broadening and lacked several diagnostic amide and indole signals attributable to hydrogen bonding interactions seen in reported X-ray crystallographic structures of this transcription factor with its cognate DNA sequences. Such differences are highlighted by the chemical shift and relaxation properties of several interfacial lysine and arginine side chains. Collectively, these data support a general model in which Ets-1 interacts with nonspecific DNA via dynamic electrostatic interactions, whereas hydrogen bonding drives the formation of well-ordered complexes with specific DNA.


Asunto(s)
Proteína Proto-Oncogénica c-ets-1/química , Proteína Proto-Oncogénica c-ets-1/metabolismo , Animales , Arginina/química , Secuencia de Bases , ADN/química , ADN/metabolismo , Lisina/química , Ratones , Modelos Moleculares , Simulación de Dinámica Molecular , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , Conformación Proteica , Estructura Secundaria de Proteína , Proteína Proto-Oncogénica c-ets-1/antagonistas & inhibidores , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Electricidad Estática
11.
Mol Carcinog ; 55(2): 131-47, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25630658

RESUMEN

During cancer progression, the genome instability incurred rearrangement could possibly turn some of the tumor suppressor micro-RNAs into pro-oncogenic ones. We aimed to investigate miR-204 in the context of prostate cancer progression using a cell line model of different levels of genome instability (LNCaP, PC3, VCaP and NCI H660), as demonstrated by the availability of ERG fusion. We studied the effect of miR-204 modulation on master transcription factors important for lineage development, cell differentiation and prostate cancer bone marrow metastasis. We followed c-MYB, ETS1 and RUNX2 transcript and protein expression and the miR-204 affected global proteome. We further investigated if these transcription factors exert an effect on miR-204 expression (qPCR, luciferase reporter assay) by silencing them using esiRNA. We found dualistic miR-204 effects, either acting as a tumor suppressor on c-MYB, or as an oncomiR on ETS1. RUNX2 and ETS1 regulation by miR-204 was ERG fusion dependent, demonstrating regulatory circuitry disruption in advanced metastatic models. miR-204 also differentially affected mRNA splicing and protein stability. miR-204 levels were found dependent on cancer hypermethylation and supported by positive feedback induced by all three transcription factors. In this regulatory circuitry among miR-204, c-MYB, RUNX2 and ETS1, the c-MYB was found to induce all three other members, but its expression was differentially affected by the methylation status in lymph node vs. bone metastasis. We demonstrate that not only tumor suppressor micro-RNA loss, but also significant genome rearrangement-driven regulatory loop perturbations play a role in the advanced cancer progression, conferring better pro-survival and metastatic potential.


Asunto(s)
MicroARNs/genética , Neoplasias de la Próstata/genética , Proteoma/genética , Proteoma/metabolismo , Empalme Alternativo , Línea Celular Tumoral , Subunidad alfa 1 del Factor de Unión al Sitio Principal/química , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Humanos , Técnicas In Vitro , Masculino , Metástasis de la Neoplasia , Proteínas de Fusión Oncogénica/genética , Neoplasias de la Próstata/metabolismo , Estabilidad Proteica , Proteoma/química , Proteína Proto-Oncogénica c-ets-1/química , Proteína Proto-Oncogénica c-ets-1/genética , Proteína Proto-Oncogénica c-ets-1/metabolismo , Proteínas Proto-Oncogénicas c-myb/química , Proteínas Proto-Oncogénicas c-myb/genética , Proteínas Proto-Oncogénicas c-myb/metabolismo , Transactivadores/genética , Regulador Transcripcional ERG
12.
Semin Cancer Biol ; 35: 20-38, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26392377

RESUMEN

Ets1 belongs to the large family of the ETS domain family of transcription factors and is involved in cancer progression. In most carcinomas, Ets1 expression is linked to poor survival. In breast cancer, Ets1 is primarily expressed in the triple-negative subtype, which is associated with unfavorable prognosis. Ets1 contributes to the acquisition of cancer cell invasiveness, to EMT (epithelial-to-mesenchymal transition), to the development of drug resistance and neo-angiogenesis. The aim of this review is to summarize the current knowledge on the functions of Ets1 in carcinoma progression and on the mechanisms that regulate Ets1 activity in cancer.


Asunto(s)
Carcinoma/genética , Carcinoma/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteína Proto-Oncogénica c-ets-1/metabolismo , Animales , Carcinoma/tratamiento farmacológico , Carcinoma/mortalidad , Carcinoma/patología , Progresión de la Enfermedad , Resistencia a Antineoplásicos/genética , Transición Epitelial-Mesenquimal/genética , Humanos , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Fosforilación , Pronóstico , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Procesamiento Proteico-Postraduccional , Proteína Proto-Oncogénica c-ets-1/química , Proteína Proto-Oncogénica c-ets-1/genética , Interferencia de ARN , Transducción de Señal
13.
J Biol Chem ; 290(36): 22101-10, 2015 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-26195629

RESUMEN

ETS1 is the archetype of the ETS transcription factor (TF) family. ETS TFs share a DNA-binding domain, the ETS domain. All ETS TFs recognize a core GGA(A/T) binding site, and thus ETS TFs are found to redundantly regulate the same genes. However, each ETS TF has unique targets as well. One prevailing hypotheses explaining this duality is that protein-protein interactions, including homodimerization, allow each ETS TF to display distinct behavior. The behavior of ETS1 is further regulated by autoinhibition. Autoinhibition apparently modulates ETS1 DNA binding affinity, but the mechanism of this inhibition is not completely understood. We sought to characterize the relationship between DNA binding and ETS1 homodimer formation. We find that ETS1 interrogates DNA and forms dimers even when the DNA does not contain an ETS recognition sequence. Mutational studies also link nonspecific DNA backbone contacts with dimer formation, in addition to providing a new role for the recognition helix of ETS1 in maintaining the autoinhibited state. Finally, in showing that residues in the DNA recognition helix affect autoinhibition, we define a new function of ETS1 autoinhibition: maintenance of a monomeric state in the absence of DNA. The conservation of relevant amino acid residues across all ETS TFs indicates that the mechanisms of nonspecific DNA interrogation and protein oligomer formation elucidated here may be common to all ETS proteins that autoinhibit.


Asunto(s)
ADN/química , Multimerización de Proteína , Proteína Proto-Oncogénica c-ets-1/química , Factores de Transcripción/química , Sitios de Unión/genética , Unión Competitiva , Dicroismo Circular , ADN/metabolismo , Huella de ADN/métodos , Desoxirribonucleasa I/metabolismo , Humanos , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutación , Oligonucleótidos/química , Oligonucleótidos/metabolismo , Unión Proteica , Proteína Proto-Oncogénica c-ets-1/genética , Proteína Proto-Oncogénica c-ets-1/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
14.
Dev Biol ; 407(2): 300-12, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25912689

RESUMEN

Melanocytes, the pigment-producing cells, arise from multipotent neural crest (NC) cells during embryogenesis. Many genes required for melanocyte development were identified using mouse pigmentation mutants. The variable spotting mouse pigmentation mutant arose spontaneously at the Jackson Laboratory. We identified a G-to-A nucleotide transition in exon 3 of the Ets1 gene in variable spotting, which results in a missense G102E mutation. Homozygous variable spotting mice exhibit sporadic white spotting. Similarly, mice carrying a targeted deletion of Ets1 exhibit hypopigmentation; nevertheless, the function of Ets1 in melanocyte development is unknown. The transcription factor Ets1 is widely expressed in developing organs and tissues, including the NC. In the chick, Ets1 is required for the expression of Sox10, a transcription factor critical for the development of various NC derivatives, including melanocytes. We show that Ets1 is required early for murine NC cell and melanocyte precursor survival in vivo. Given the importance of Ets1 for Sox10 expression in the chick, we investigated a potential genetic interaction between these genes by comparing the hypopigmentation phenotypes of single and double heterozygous mice. The incidence of hypopigmentation in double heterozygotes was significantly greater than in single heterozygotes. The area of hypopigmentation in double heterozygotes was significantly larger than would be expected from the addition of the areas of hypopigmentation of single heterozygotes, suggesting that Ets1 and Sox10 interact synergistically in melanocyte development. Since Sox10 is also essential for enteric ganglia development, we examined the distal colons of Ets1 null mutants and found a significant decrease in enteric innervation, which was exacerbated by Sox10 heterozygosity. At the molecular level, Ets1 was found to activate an enhancer critical for Sox10 expression in NC-derived structures. Furthermore, enhancer activation was significantly inhibited by the variable spotting mutation. Together, these results suggest that Ets1 and Sox10 interact to promote proper melanocyte and enteric ganglia development from the NC.


Asunto(s)
Melanocitos/citología , Melanocitos/metabolismo , Proteína Proto-Oncogénica c-ets-1/metabolismo , Factores de Transcripción SOXE/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Tipificación del Cuerpo , Recuento de Células , Línea Celular Tumoral , Linaje de la Célula , Supervivencia Celular , Embrión de Mamíferos/metabolismo , Elementos de Facilitación Genéticos/genética , Epigénesis Genética , Ganglios/embriología , Ganglios/metabolismo , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones Transgénicos , Datos de Secuencia Molecular , Mutación Missense/genética , Cresta Neural/citología , Unión Proteica , Proteína Proto-Oncogénica c-ets-1/química , Proteína Proto-Oncogénica c-ets-1/genética , Activación Transcripcional/genética
15.
Nucleic Acids Res ; 43(8): 4322-31, 2015 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-25824951

RESUMEN

The ETS family of transcription factors exemplifies current uncertainty in how eukaryotic genetic regulators with overlapping DNA sequence preferences achieve target site specificity. PU.1 and Ets-1 represent archetypes for studying site discrimination by ETS proteins because their DNA-binding domains are the most divergent in sequence, yet they share remarkably superimposable DNA-bound structures. To gain insight into the contrasting thermodynamics and kinetics of DNA recognition by these two proteins, we investigated the structure and dynamics of site discrimination by their DNA-binding domains. Electrophoretic mobilities of complexes formed by the two homologs with circularly permuted binding sites showed significant dynamic differences only for DNA complexes of PU.1. Free solution measurements by dynamic light scattering showed PU.1 to be more dynamic than Ets-1; moreover, dynamic changes are strongly coupled to site discrimination by PU.1, but not Ets-1. Interrogation of the protein/DNA interface by DNA footprinting showed similar accessibility to dimethyl sulfate for PU.1/DNA and Ets-1/DNA complexes, indicating that the dynamics of PU.1/DNA complexes reside primarily outside that interface. An information-based analysis of the two homologs' binding motifs suggests a role for dynamic coupling in PU.1's ability to enforce a more stringent sequence preference than Ets-1 and its proximal sequence homologs.


Asunto(s)
ADN/química , Proteína Proto-Oncogénica c-ets-1/química , Proteínas Proto-Oncogénicas/química , Transactivadores/química , Sitios de Unión , ADN/metabolismo , Modelos Moleculares , Unión Proteica , Estructura Terciaria de Proteína , Proteína Proto-Oncogénica c-ets-1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo
16.
J Mol Biol ; 427(8): 1655-69, 2015 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-25083921

RESUMEN

Cooperative assemblies of transcription factors (TFs) on target gene enhancers coordinate cell proliferation, fate specification, and differentiation through precise and complicated transcriptional mechanisms. Chemical modifications, such as phosphorylation, of TFs induced by cell signaling further modulate the dynamic cooperativity of TFs. In this study, we found that various Ets1-containing TF-DNA complexes respond differently to calcium-induced phosphorylation of Ets1, which is known to inhibit Ets1-DNA binding. Crystallographic analysis of a complex comprising Ets1, Runx1, and CBFß at the TCRα enhancer revealed that Ets1 acquires robust binding stability in the Runx1 and DNA-complexed state, via allosteric mechanisms. This allows phosphorylated Ets1 to be retained at the TCRα enhancer with Runx1, in contrast to other Ets1 target gene enhancers including mb-1 and stromelysin-1. This study provides a structure-based model for cell-signaling-dependent regulation of target genes, mediated via chemical modification of TFs.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Subunidad beta del Factor de Unión al Sitio Principal/metabolismo , ADN/metabolismo , Proteína Proto-Oncogénica c-ets-1/metabolismo , Animales , Secuencia de Bases , Línea Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/química , Subunidad beta del Factor de Unión al Sitio Principal/química , Cristalografía por Rayos X , ADN/química , Humanos , Ratones , Modelos Moleculares , Fosforilación , Unión Proteica , Conformación Proteica , Proteína Proto-Oncogénica c-ets-1/química , Activación Transcripcional
17.
PLoS One ; 9(11): e112419, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25380315

RESUMEN

The dynamic cross correlation (DCC) analysis is a popular method for analyzing the trajectories of molecular dynamics (MD) simulations. However, it is difficult to detect correlative motions that appear transiently in only a part of the trajectory, such as atomic contacts between the side-chains of amino acids, which may rapidly flip. In order to capture these multi-modal behaviors of atoms, which often play essential roles, particularly at the interfaces of macromolecules, we have developed the "multi-modal DCC (mDCC)" analysis. The mDCC is an extension of the DCC and it takes advantage of a Bayesian-based pattern recognition technique. We performed MD simulations for molecular systems modeled from the (Ets1)2-DNA complex and analyzed their results with the mDCC method. Ets1 is an essential transcription factor for a variety of physiological processes, such as immunity and cancer development. Although many structural and biochemical studies have so far been performed, its DNA binding properties are still not well characterized. In particular, it is not straightforward to understand the molecular mechanisms how the cooperative binding of two Ets1 molecules facilitates their recognition of Stromelysin-1 gene regulatory elements. A correlation network was constructed among the essential atomic contacts, and the two major pathways by which the two Ets1 molecules communicate were identified. One is a pathway via direct protein-protein interactions and the other is that via the bound DNA intervening two recognition helices. These two pathways intersected at the particular cytosine bases (C110/C11), interacting with the H1, H2, and H3 helices. Furthermore, the mDCC analysis showed that both pathways included the transient interactions at their intermolecular interfaces of Tyr396-C11 and Ala327-Asn380 in multi-modal motions of the amino acid side chains and the nucleotide backbone. Thus, the current mDCC approach is a powerful tool to reveal these complicated behaviors and scrutinize intermolecular communications in a molecular system.


Asunto(s)
ADN/química , Simulación de Dinámica Molecular , Reconocimiento de Normas Patrones Automatizadas/métodos , Proteína Proto-Oncogénica c-ets-1/química , Teorema de Bayes , Sitios de Unión/genética , Simulación por Computador , ADN/metabolismo , Cinética , Mutación , Conformación de Ácido Nucleico , Unión Proteica , Mapas de Interacción de Proteínas , Multimerización de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteína Proto-Oncogénica c-ets-1/genética , Proteína Proto-Oncogénica c-ets-1/metabolismo , Reproducibilidad de los Resultados
18.
Acta Crystallogr F Struct Biol Commun ; 70(Pt 10): 1380-4, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25286944

RESUMEN

Gene transcription is regulated in part through the assembly of multiple transcription factors (TFs) on gene enhancers. To enable examination of the mechanism underlying the formation of these complexes and their response to a phosphorylation signal, two kinds of higher-order TF-DNA assemblies were crystallized composed of an unmodified or phosphorylated Ets1 fragment, a Runx1(L94K) fragment and a CBFß fragment on the T-cell receptor (TCR) α gene enhancer. Within these complexes, the Ets1 and Runx1 fragments contain intrinsically disordered regulatory regions as well as their DNA-binding domains. Crystals of the complex containing unmodified Ets1 belonged to space group P212121, with unit-cell parameters a = 78.7, b = 102.1, c = 195.0 Å, and diffracted X-rays to a resolution of 2.35 Å, and those containing phosphorylated Ets1 belonged to the same space group, with unit-cell parameters a = 78.6, b = 101.7, c = 194.7 Å, and diffracted X-rays to a similar resolution. To facilitate crystallization, a Runx1 residue involved in a hydrophobic patch that was predicted to be engaged in crystal packing based on the previously reported structures of Runx1-containing crystals was mutated.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/química , Elementos de Facilitación Genéticos , Genes Codificadores de la Cadena alfa de los Receptores de Linfocito T , Proteína Proto-Oncogénica c-ets-1/química , Secuencia de Aminoácidos , Animales , Cristalización , ADN/química , Humanos , Sustancias Macromoleculares/química , Ratones , Datos de Secuencia Molecular , Unión Proteica
19.
Proc Natl Acad Sci U S A ; 111(30): 11019-24, 2014 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-25024220

RESUMEN

The E26 transformation-specific (Ets-1) transcription factor is autoinhibited by a conformationally disordered serine-rich region (SRR) that transiently interacts with its DNA-binding ETS domain. In response to calcium signaling, autoinhibition is reinforced by calmodulin-dependent kinase II phosphorylation of serines within the SRR. Using mutagenesis and quantitative DNA-binding measurements, we demonstrate that phosphorylation-enhanced autoinhibition requires the presence of phenylalanine or tyrosine (ϕ) residues adjacent to the SRR phosphoacceptor serines. The introduction of additional phosphorylated Ser-ϕ-Asp, but not Ser-Ala-Asp, repeats within the SRR dramatically reinforces autoinhibition. NMR spectroscopic studies of phosphorylated and mutated SRR variants, both within their native context and as separate trans-acting peptides, confirmed that the aromatic residues and phosphoserines contribute to the formation of a dynamic complex with the ETS domain. Complementary NMR studies also identified the SRR-interacting surface of the ETS domain, which encompasses its positively charged DNA-recognition interface and an adjacent region of neutral polar and nonpolar residues. Collectively, these studies highlight the role of aromatic residues and their synergy with phosphoserines in an intrinsically disordered regulatory sequence that integrates cellular signaling and gene expression.


Asunto(s)
Fosfoserina/química , Proteína Proto-Oncogénica c-ets-1/química , Humanos , Resonancia Magnética Nuclear Biomolecular , Péptidos , Fosfoserina/metabolismo , Estructura Terciaria de Proteína , Proteína Proto-Oncogénica c-ets-1/genética , Proteína Proto-Oncogénica c-ets-1/metabolismo , Secuencias Repetitivas de Aminoácido
20.
Structure ; 22(8): 1196-1203, 2014 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-25017730

RESUMEN

Functions of many proteins are affected by posttranslational modifications of intrinsically disordered (ID) regions, yet little is known about the underlying molecular mechanisms. By combining molecular dynamics simulations and protein docking, we demonstrate that the addition of phosphates to an ID segment adjacent to the PNT domain of Ets1 directs conformational sampling toward substates that are most compatible with high-affinity binding of the TAZ1 domain of its coactivator CBP. The phosphate charges disrupt salt bridges and thereby open a hydrophobic cleft and expose hydrophobic residues at the ID N terminus. The structure of the PNT-TAZ1 complex that we determined shows that PNT binds to TAZ1 via these hydrophobic regions in a similar manner to how it interacts with other partners. Our calculations reveal a dual effect of phosphorylation in that it changes the dynamics of PNT so that it becomes more compatible for TAZ1 binding and increases complementarity with this binding partner.


Asunto(s)
Proteínas Intrínsecamente Desordenadas/química , Proteínas Intrínsecamente Desordenadas/metabolismo , Modelos Moleculares , Proteína Proto-Oncogénica c-ets-1/química , Proteína Proto-Oncogénica c-ets-1/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Simulación de Dinámica Molecular , Fragmentos de Péptidos/metabolismo , Fosforilación , Unión Proteica , Conformación Proteica , Sialoglicoproteínas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...