Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Anal Biochem ; 629: 114298, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34252439

ABSTRACT

To extend the value of biosensor-SPR in the characterization of DNA recognition by nucleoproteins, we report a comparative analysis of DNA-facilitated target search by two ETS-family transcription factors: Elk1 and ETV6. ETS domains represent an attractive system for developing biosensor-based techniques due to a broad range of physicochemical properties encoded within a highly conserved DNA-binding motif. Building on a biosensor approach in which the protein is quantitatively sequestered and presented to immobilized cognate DNA as nonspecific complexes, we assessed the impact of intrinsic cognate and nonspecific affinities on long-range (intersegmental) target search. The equilibrium constants of DNA-facilitated binding were sensitive to the intrinsic binding properties of the proteins such that their relative specificity for cognate DNA were reinforced when binding occurred by transfer vs. without nonspecific DNA. Direct measurement of association and dissociation kinetics revealed ionic features of the activated complex that evidenced DNA-facilitated dissociation, even though Elk1 and ETV6 harbor only a single DNA-binding surface. At salt concentrations that masked the effects of nonspecific pre-binding at equilibrium, the dissociation kinetics of cognate binding were nevertheless distinct from conditions under which nonspecific DNA was absent. These results further strengthen the significance of long-range DNA-facilitated translocation in the physiologic environment.


Subject(s)
DNA/analysis , Nucleoproteins/chemistry , Binding Sites , Biosensing Techniques , Escherichia coli/genetics , Nucleoproteins/genetics , Protein Binding , Proto-Oncogene Proteins c-ets/chemistry , Repressor Proteins/chemistry , Surface Plasmon Resonance , ets-Domain Protein Elk-1/chemistry , ETS Translocation Variant 6 Protein
2.
Phys Chem Chem Phys ; 23(24): 13490-13502, 2021 Jun 23.
Article in English | MEDLINE | ID: mdl-34120158

ABSTRACT

Electrostatic protein/DNA interactions arise from the neutralization of the DNA phosphodiester backbone as well as coupled exchanges by charged protein residues as salt bridges or with mobile ions. Much focus has been and continues to be paid to interfacial ion pairs with DNA. The role of extra-interfacial ionic interactions, particularly as dynamic drivers of DNA sequence selectivity, remain poorly known. The ETS family of transcription factors represents an attractive model for addressing this knowledge gap given their diverse ionic composition in primary structures that fold to a tightly conserved DNA-binding motif. To probe the importance of extra-interfacial salt bridges in DNA recognition, we compared the salt-dependent binding by Elk1 with ETV6, two ETS homologs differing markedly in ionic composition. While both proteins exhibit salt-dependent binding with cognate DNA that corresponds to interfacial phosphate contacts, their nonspecific binding diverges from cognate binding as well as each other. Molecular dynamics simulations in explicit solvent, which generated ionic interactions in agreement with the experimental binding data, revealed distinct salt-bridge dynamics in the nonspecific complexes formed by the two proteins. Impaired DNA contact by ETV6 resulted in fewer backbone contacts in the nonspecific complex, while Elk1 exhibited a redistribution of extra-interfacial salt bridges via residues that are non-conserved between the two ETS relatives. Thus, primary structure variation in ionic residues can encode highly differentiated specificity mechanisms in a highly conserved DNA-binding motif.


Subject(s)
DNA/chemistry , Molecular Dynamics Simulation , Proto-Oncogene Proteins c-ets/chemistry , Repressor Proteins/chemistry , ets-Domain Protein Elk-1/chemistry , Density Functional Theory , Humans , ETS Translocation Variant 6 Protein
3.
Int J Hematol ; 114(2): 297-300, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33768492

ABSTRACT

ETV6-related thrombocytopenia is an autosomal dominant thrombocytopenia, characterized by a bleeding tendency and predisposition to hematological malignancies. The similarity in symptoms makes differentiating immune and congenital thrombocytopenia challenging. We report a 5-year-old girl who presented with chronic thrombocytopenia associated with repetitive and long-lasting epistaxis, leading to blood transfusion for severe anemia. Blood tests showed thrombocytopenia (52 × 103/µL) with normal-sized platelets and transiently low von Willebrand factor (VWF) levels (VWF:RCo 13%, VWF:Ag 50%); therefore, von Willebrand disease type 2 was initially suspected. Repetition of the blood tests revealed normal levels of VWF. Exome and Sanger sequencing identified a germline ETV6 heterozygous variant, c.641C > T:p.(P214L). No additional pathogenic variants were found, including VWF, in the gene panel testing of the 53 known target causative genes for thrombocytopenia. High-throughput exome sequencing for chronic thrombocytopenia can be utilized to differentially diagnose ETV6-related thrombocytopenia from chronic/intractable immune thrombocytopenia and to effectively monitor malignancy.


Subject(s)
Proto-Oncogene Proteins c-ets/genetics , Repressor Proteins/genetics , Thrombocytopenia/blood , Thrombocytopenia/genetics , von Willebrand Factor , Biomarkers , Child, Preschool , Disease Susceptibility , Erythrocyte Indices , Female , Genotype , Hematologic Tests , Hemorrhage/diagnosis , Hemorrhage/etiology , Humans , Mutation , Platelet Count , Proto-Oncogene Proteins c-ets/chemistry , Proto-Oncogene Proteins c-ets/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Thrombocytopenia/diagnosis , Thrombocytopenia/therapy , ETS Translocation Variant 6 Protein
4.
J Biol Chem ; 296: 100284, 2021.
Article in English | MEDLINE | ID: mdl-33450226

ABSTRACT

ETV6 is an E26 transformation specific family transcriptional repressor that self-associates by its PNT domain to facilitate cooperative DNA binding. Chromosomal translocations frequently generate constitutively active oncoproteins with the ETV6 PNT domain fused to the kinase domain of one of many protein tyrosine kinases. Although an attractive target for therapeutic intervention, the propensity of the ETV6 PNT domain to polymerize via the tight head-to-tail association of two relatively flat interfaces makes it challenging to identify suitable small molecule inhibitors of this protein-protein interaction. Herein, we provide a comprehensive biophysical characterization of the ETV6 PNT domain interaction interfaces to aid future drug discovery efforts and help define the mechanisms by which its self-association mediates transcriptional repression. Using NMR spectroscopy, X-ray crystallography, and molecular dynamics simulations, along with amide hydrogen exchange measurements, we demonstrate that monomeric PNT domain variants adopt very stable helical bundle folds that do not change in conformation upon self-association into heterodimer models of the ETV6 polymer. Surface plasmon resonance-monitored alanine scanning mutagenesis studies identified hot spot regions within the self-association interfaces. These regions include both central hydrophobic residues and flanking salt-bridging residues. Collectively, these studies indicate that small molecules targeted to these hydrophobic or charged regions within the relatively rigid interfaces could potentially serve as orthosteric inhibitors of ETV6 PNT domain polymerization.


Subject(s)
Alanine/chemistry , Aspartic Acid/chemistry , Glutamic Acid/chemistry , Proto-Oncogene Proteins c-ets/chemistry , Repressor Proteins/chemistry , Transcription, Genetic , Valine/chemistry , Alanine/metabolism , Amino Acid Substitution , Aspartic Acid/metabolism , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Glutamic Acid/metabolism , Humans , Models, Molecular , Mutagenesis, Site-Directed , Mutation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Thermodynamics , Valine/metabolism , ETS Translocation Variant 6 Protein
5.
SLAS Discov ; 26(5): 698-711, 2021 06.
Article in English | MEDLINE | ID: mdl-33345679

ABSTRACT

ETV6 is an ETS family transcriptional repressor for which head-to-tail polymerization of its PNT domain facilitates cooperative binding to DNA by its ETS domain. Chromosomal translocations frequently fuse the ETV6 PNT domain to one of several protein tyrosine kinases. The resulting chimeric oncoproteins undergo ligand-independent self-association, autophosphorylation, and aberrant stimulation of downstream signaling pathways, leading to a variety of cancers. Currently, no small-molecule inhibitors of ETV6 PNT domain polymerization are known and no assays targeting PNT domain polymerization have been described. In this study, we developed complementary experimental and computational approaches for identifying such inhibitory compounds. One mammalian cellular approach utilized a mutant PNT domain heterodimer system covalently attached to split Gaussia luciferase fragments. In this protein-fragment complementation assay, inhibition of PNT domain heterodimerization reduces luminescence. A yeast assay took advantage of activation of the reporter HIS3 gene upon heterodimerization of mutant PNT domains fused to DNA-binding and transactivation domains. In this two-hybrid screen, inhibition of PNT domain heterodimerization prevents cell growth in medium lacking histidine. The Bristol University Docking Engine (BUDE) was used to identify virtual ligands from the ZINC8 library predicted to bind the PNT domain polymerization interfaces. More than 75 hits from these three assays were tested by nuclear magnetic resonance spectroscopy for binding to the purified ETV6 PNT domain. Although none were found to bind, the lessons learned from this study may facilitate future approaches for developing therapeutics that act against ETV6 oncoproteins by disrupting PNT domain polymerization.


Subject(s)
Drug Discovery/methods , Drug Screening Assays, Antitumor/methods , Protein Interaction Domains and Motifs/drug effects , Protein Multimerization/drug effects , Proto-Oncogene Proteins c-ets/antagonists & inhibitors , Proto-Oncogene Proteins c-ets/chemistry , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/chemistry , Biological Assay/methods , Genes, Reporter , Humans , Protein Binding , Structure-Activity Relationship , ETS Translocation Variant 6 Protein
6.
Med Res Rev ; 40(1): 413-430, 2020 01.
Article in English | MEDLINE | ID: mdl-30927317

ABSTRACT

The ETS family of proteins consists of 28 transcription factors, many of which have been implicated in development and progression of a variety of cancers. While one family member, ERG, has been rigorously studied in the context of prostate cancer where it plays a critical role, other ETS factors keep emerging as potential hallmark oncodrivers. In recent years, numerous studies have reported initial discoveries of small molecule inhibitors of ETS proteins and opened novel avenues for ETS-directed cancer therapies. This review summarizes the state of the art data on therapeutic targeting of ETS family members and highlights the corresponding drug discovery strategies.


Subject(s)
Drug Delivery Systems , Neoplasms/drug therapy , Proto-Oncogene Proteins c-ets/metabolism , Amino Acid Sequence , Animals , Humans , Proto-Oncogene Proteins c-ets/chemistry , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use
7.
Mol Carcinog ; 58(6): 922-932, 2019 06.
Article in English | MEDLINE | ID: mdl-30676667

ABSTRACT

Epithelial specific ETS-1 (ESE-1) belongs to the E26 transformation-specific transcription factor superfamily and is of great interest as a potential target for managing several types of cancer. Despite its clinical significance, the documented effects of ESE-1 on cancer development and progression are contradictory and its underlying biological mechanism of action remains elusive. The objectives of this study are to investigate whether ESE-1 is a tumor suppressor and to identify dietary anti-cancer compound to activate ESE-1 expression in human colon cancer model. ESE-1 knockout and xenograft mouse models were used to examine the effect of ESE-1 in colon tumorigenesis. Stable human colon cancer cell lines were used for in vitro mechanistic studies. ESE-1 knockout in mice increased azoxymethane (AOM)-induced and dextran sulfate sodium (DSS)-promoted formation of aberrant crypt foci (ACF). Conversely, overexpression of ESE-1 suppressed tumorigenicity in a xenograft mouse study, and repressed anchorage-independent growth and migration/invasion in human colon cancer cells. Full length ESE-1 localized abundantly in the nucleus, and internal deletion of nuclear localization sequence 2 (NLS2) reduced nuclear ESE-1. Three lysine residues (318 KKK320 ) in the NLS2 determine its nuclear localization. We identified epigallocatechin-3-gallate (EGCG) that acts as a transcriptional activator of ESE-1 in human colon cancer cells. These findings propose a novel and promising molecular target of dietary anti-cancer compounds for prevention of colon cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Catechin/analogs & derivatives , Colonic Neoplasms/drug therapy , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Azoxymethane/adverse effects , Caco-2 Cells , Catechin/administration & dosage , Catechin/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Nucleus/metabolism , Colonic Neoplasms/chemically induced , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , DNA-Binding Proteins/chemistry , Dextran Sulfate/adverse effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , HCT116 Cells , HT29 Cells , Humans , Mice , Nuclear Localization Signals , Proto-Oncogene Proteins c-ets/chemistry , Transcription Factors/chemistry , Xenograft Model Antitumor Assays
8.
J Biol Chem ; 292(32): 13187-13196, 2017 08 11.
Article in English | MEDLINE | ID: mdl-28592487

ABSTRACT

To find their DNA target sites in complex solution environments containing excess heterogeneous DNA, sequence-specific DNA-binding proteins execute various translocation mechanisms known collectively as facilitated diffusion. For proteins harboring a single DNA contact surface, long-range translocation occurs by jumping between widely spaced DNA segments. We have configured biosensor-based surface plasmon resonance to directly measure the affinity and kinetics of this intersegmental jumping by the ETS-family transcription factor ETS variant 6 (ETV6). To isolate intersegmental target binding in a functionally defined manner, we pre-equilibrated ETV6 with excess salmon sperm DNA, a heterogeneous polymer, before exposing the nonspecifically bound protein to immobilized oligomeric DNA harboring a high-affinity ETV6 site. In this way, the mechanism of ETV6-target association could be toggled electrostatically through varying NaCl concentration in the bulk solution. Direct measurements of association and dissociation kinetics of the site-specific complex indicated that 1) freely diffusive binding by ETV6 proceeds through a nonspecific-like intermediate, 2) intersegmental jumping is rate-limited by dissociation from the nonspecific polymer, and 3) dissociation of the specific complex is independent of the history of complex formation. These results show that target searches by proteins with an ETS domain, such as ETV6, whose single DNA-binding domain cannot contact both source and destination sites simultaneously, are nonetheless strongly modulated by intersegmental jumping in heterogeneous site environments. Our findings establish biosensors as a general technique for directly and specifically measuring target site search by DNA-binding proteins via intersegmental translocation.


Subject(s)
DNA/metabolism , Models, Molecular , Proto-Oncogene Proteins c-ets/metabolism , Repressor Proteins/metabolism , Animals , Binding Sites , Biosensing Techniques , DNA/chemistry , DNA, Single-Stranded/chemistry , DNA, Single-Stranded/metabolism , ETS Motif , Facilitated Diffusion , Humans , Kinetics , Nucleotide Motifs , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Polydeoxyribonucleotides/chemistry , Polydeoxyribonucleotides/metabolism , Protein Conformation , Protein Interaction Domains and Motifs , Proto-Oncogene Proteins c-ets/chemistry , Proto-Oncogene Proteins c-ets/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Repressor Proteins/chemistry , Repressor Proteins/genetics , Response Elements , Salmon , Static Electricity , Surface Plasmon Resonance , Thermodynamics , ETS Translocation Variant 6 Protein
9.
J Phys Chem B ; 121(13): 2748-2758, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28296403

ABSTRACT

The ETS family of transcription factors is a functionally heterogeneous group of gene regulators that share a structurally conserved, eponymous DNA-binding domain. Unlike other ETS homologues, such as Ets-1, DNA recognition by PU.1 is highly sensitive to its osmotic environment due to excess interfacial hydration in the complex. To investigate interfacial hydration in the two homologues, we mutated a conserved tyrosine residue, which is exclusively engaged in coordinating a well-defined water contact between the protein and DNA among ETS proteins, to phenylalanine. The loss of this water-mediated contact blunted the osmotic sensitivity of PU.1/DNA binding, but did not alter binding under normo-osmotic conditions, suggesting that PU.1 has evolved to maximize osmotic sensitivity. The homologous mutation in Ets-1, which was minimally sensitive to osmotic stress due to a sparsely hydrated interface, reduced DNA-binding affinity at normal osmolality but the complex became stabilized by osmotic stress. Molecular dynamics simulations of wildtype and mutant PU.1 and Ets-1 in their free and DNA-bound states, which recapitulated experimental features of the proteins, showed that abrogation of this tyrosine-mediated water contact perturbed the Ets-1/DNA complex not through disruption of interfacial hydration, but by inhibiting local dynamics induced specifically in the bound state. Thus, a configurationally identical water-mediated contact plays mechanistically distinct roles in mediating DNA recognition by structurally homologous ETS transcription factors.


Subject(s)
DNA/chemistry , Proto-Oncogene Proteins c-ets/chemistry , Humans , Molecular Dynamics Simulation , Water/chemistry
10.
Sci Rep ; 6: 34725, 2016 10 07.
Article in English | MEDLINE | ID: mdl-27713480

ABSTRACT

Tumorigenesis is driven by genetic alterations that perturb the signaling networks regulating proliferation or cell death. In order to block tumor growth, one has to precisely know how these signaling pathways function and interplay. Here, we identified the transcription factor Ets21C as a pivotal regulator of tumor growth and propose a new model of how Ets21C could affect this process. We demonstrate that a depletion of Ets21C strongly suppressed tumor growth while ectopic expression of Ets21C further increased tumor size. We confirm that Ets21C expression is regulated by the JNK pathway and show that Ets21C acts via a positive feed-forward mechanism to induce a specific set of target genes that is critical for tumor growth. These genes are known downstream targets of the JNK pathway and we demonstrate that their expression not only depends on the transcription factor AP-1, but also on Ets21C suggesting a cooperative transcriptional activation mechanism. Taken together we show that Ets21C is a crucial player in regulating the transcriptional program of the JNK pathway and enhances our understanding of the mechanisms that govern neoplastic growth.


Subject(s)
Carcinogenesis/pathology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/metabolism , Transcription Factor AP-1/metabolism , Animals , Binding Sites , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Line , Cell Proliferation , Drosophila Proteins/chemistry , Drosophila melanogaster , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , MAP Kinase Signaling System , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins c-ets/chemistry , Transcription Factor AP-1/genetics , Transcriptional Activation , Tumor Burden
11.
Nucleic Acids Res ; 44(18): 8671-8681, 2016 Oct 14.
Article in English | MEDLINE | ID: mdl-27270080

ABSTRACT

Transactivation by the ETS family of transcription factors, whose members share structurally conserved DNA-binding domains, is variably sensitive to methylation of their target genes. The mechanism by which DNA methylation controls ETS proteins remains poorly understood. Uncertainly also pervades the effects of hemi-methylated DNA, which occurs following DNA replication and in response to hypomethylating agents, on site recognition by ETS proteins. To address these questions, we measured the affinities of two sequence-divergent ETS homologs, PU.1 and Ets-1, to DNA sites harboring a hemi- and fully methylated CpG dinucleotide. While the two proteins bound unmethylated DNA with indistinguishable affinity, their affinities to methylated DNA are markedly heterogeneous and exhibit major energetic coupling between the two CpG methylcytosines. Analysis of simulated DNA and existing co-crystal structures revealed that hemi-methylation induced non-local backbone and groove geometries that were not conserved in the fully methylated state. Indirect readout of these perturbations was differentially achieved by the two ETS homologs, with the distinctive interfacial hydration in PU.1/DNA binding moderating the inhibitory effects of DNA methylation on binding. This data established a biophysical basis for the pioneering properties associated with PU.1, which robustly bound fully methylated DNA, but not Ets-1, which was substantially inhibited.


Subject(s)
DNA Methylation/genetics , DNA/metabolism , Proto-Oncogene Proteins c-ets/chemistry , Proto-Oncogene Proteins c-ets/metabolism , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Trans-Activators/chemistry , Trans-Activators/metabolism , 5-Methylcytosine/metabolism , Animals , Binding Sites , CpG Islands/genetics , DNA/chemistry , Mice , Nucleic Acid Conformation , Principal Component Analysis , Protein Binding/genetics , Protein Domains , Thermodynamics
12.
Biochem J ; 473(8): 1047-61, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26920025

ABSTRACT

Oestrogen receptor-α (ERα) is a ligand-dependent transcription factor that primarily mediates oestrogen (E2)-dependent gene transcription required for mammary gland development. Coregulators critically regulate ERα transcription functions by directly interacting with it. In the present study, we report that ELF3, an epithelial-specific ETS transcription factor, acts as a transcriptional repressor of ERα. Co-immunoprecipitation (Co-IP) analysis demonstrated that ELF3 strongly binds to ERα in the absence of E2, but ELF3 dissociation occurs upon E2 treatment in a dose- and time-dependent manner suggesting that E2 negatively influences such interaction. Domain mapping studies further revealed that the ETS (E-twenty six) domain of ELF3 interacts with the DNA binding domain of ERα. Accordingly, ELF3 inhibited ERα's DNA binding activity by preventing receptor dimerization, partly explaining the mechanism by which ELF3 represses ERα transcriptional activity. Ectopic expression of ELF3 decreases ERα transcriptional activity as demonstrated by oestrogen response elements (ERE)-luciferase reporter assay or by endogenous ERα target genes. Conversely ELF3 knockdown increases ERα transcriptional activity. Consistent with these results, ELF3 ectopic expression decreases E2-dependent MCF7 cell proliferation whereas ELF3 knockdown increases it. We also found that E2 induces ELF3 expression in MCF7 cells suggesting a negative feedback regulation of ERα signalling in breast cancer cells. A small peptide sequence of ELF3 derived through functional interaction between ERα and ELF3 could inhibit DNA binding activity of ERα and breast cancer cell growth. These findings demonstrate that ELF3 is a novel transcriptional repressor of ERα in breast cancer cells. Peptide interaction studies further represent a novel therapeutic option in breast cancer therapy.


Subject(s)
Breast Neoplasms/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/metabolism , Proto-Oncogene Proteins c-ets/chemistry , Proto-Oncogene Proteins c-ets/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism , Transcription, Genetic/physiology , Amino Acid Sequence , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/physiology , DNA-Binding Proteins/genetics , Dose-Response Relationship, Drug , Estrogen Receptor alpha/genetics , Female , HeLa Cells , Humans , MCF-7 Cells , Molecular Sequence Data , Protein Structure, Secondary , Proto-Oncogene Proteins c-ets/genetics , Tamoxifen/metabolism , Tamoxifen/pharmacology , Transcription Factors/genetics , Transcription, Genetic/drug effects
13.
J Mol Biol ; 428(8): 1515-30, 2016 Apr 24.
Article in English | MEDLINE | ID: mdl-26920109

ABSTRACT

The ETS transcriptional repressor ETV6 (or TEL) is autoinhibited by an α-helix that sterically blocks its DNA-binding ETS domain. The inhibitory helix is marginally stable and unfolds when ETV6 binds to either specific or non-specific DNA. Using NMR spectroscopy, we show that folding of the inhibitory helix requires a buried charge-dipole interaction with helix H1 of the ETS domain. This interaction also contributes directly to autoinhibition by precluding a highly conserved dipole-enhanced hydrogen bond between the phosphodiester backbone of bound DNA and the N terminus of helix H1. To probe further the thermodynamic basis of autoinhibition, ETV6 variants were generated with amino acid substitutions introduced along the solvent exposed surface of the inhibitory helix. These changes were designed to increase the intrinsic helical propensity of the inhibitory helix without perturbing its packing interactions with the ETS domain. NMR-monitored amide hydrogen exchange measurements confirmed that the stability of the folded inhibitory helix increases progressively with added helix-promoting substitutions. This also results in progressively reinforced autoinhibition and decreased DNA-binding affinity. Surprisingly, locking the inhibitory helix onto the ETS domain by a disulfide bridge severely impairs, but does not abolish DNA binding. Weak interactions still occur via an interface displaced from the canonical ETS domain DNA-binding surface. Collectively, these studies establish a direct thermodynamic linkage between inhibitory helix stability and ETV6 autoinhibition, and demonstrate that helix unfolding does not strictly precede DNA binding. Modulating inhibitory helix stability provides a potential route for the in vivo regulation of ETV6 activity.


Subject(s)
Proto-Oncogene Proteins c-ets/chemistry , Proto-Oncogene Proteins c-ets/physiology , Repressor Proteins/chemistry , Repressor Proteins/physiology , Amides/chemistry , Amino Acids/chemistry , Animals , DNA/chemistry , Disulfides/chemistry , Hydrogen/chemistry , Hydrogen-Ion Concentration , Kinetics , Magnetic Resonance Spectroscopy , Mice , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Thermodynamics , ETS Translocation Variant 6 Protein
14.
Oncotarget ; 7(12): 13865-79, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26885618

ABSTRACT

The epithelium-specific Ets transcription factor, SPDEF, plays a critical role in metastasis of prostate and breast cancer cells. While enhanced SPDEF expression blocks migration and invasion, knockdown of SPDEF expression enhances migration, invasion, and metastasis of cancer cells. SPDEF expression and activation is tightly regulated in cancer cells; however, the precise mechanism of SPDEF regulation has not been explored in detail. In this study we provide evidence that the cell cycle kinase CDK11p58, a protein involved in G2/M transition and degradation of several transcription factors, directly interacts with and phosphorylates SPDEF on serine residues, leading to subsequent ubiquitination and degradation of SPDEF through the proteasome pathway. As a consequence of CDK11p58 mediated degradation of SPDEF, this loss of SPDEF protein results in increased prostate cancer cell migration and invasion. In contrast, knockdown of CDK11p58 protein expression by interfering RNA or SPDEF overexpression inhibit migration and invasion of cancer cells. We demonstrate that CDK11p58 mediated degradation of SPDEF is attenuated by Growth Arrest and DNA damage-inducible 45 (GADD45) α and , two proteins inducing G2/M cell cycle arrest. We show that GADD45 α and γ, directly interact with CDK11p58 and thereby inhibit CDK11p58 activity, and consequentially SPDEF phosphorylation and degradation, ultimately reducing prostate cancer cell migration and invasion. Our findings provide new mechanistic insights into the complex regulation of SPDEF activity linked to cancer metastasis and characterize a previously unidentified SPDEF/CDK11p58/GADD45α/γ pathway that controls SPDEF protein stability and SPDEF-mediated effects on cancer cell migration and invasion.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Movement , Cyclin D3/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-ets/chemistry , Proto-Oncogene Proteins c-ets/metabolism , Apoptosis , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Male , Phosphorylation , Prostatic Neoplasms/metabolism , Protein Interaction Maps , Protein Stability , Proteolysis , Tumor Cells, Cultured
15.
PLoS Genet ; 11(6): e1005262, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26102509

ABSTRACT

Somatic mutations affecting ETV6 often occur in acute lymphoblastic leukemia (ALL), the most common childhood malignancy. The genetic factors that predispose to ALL remain poorly understood. Here we identify a novel germline ETV6 p. L349P mutation in a kindred affected by thrombocytopenia and ALL. A second ETV6 p. N385fs mutation was identified in an unrelated kindred characterized by thrombocytopenia, ALL and secondary myelodysplasia/acute myeloid leukemia. Leukemic cells from the proband in the second kindred showed deletion of wild type ETV6 with retention of the ETV6 p. N385fs. Enforced expression of the ETV6 mutants revealed normal transcript and protein levels, but impaired nuclear localization. Accordingly, these mutants exhibited significantly reduced ability to regulate the transcription of ETV6 target genes. Our findings highlight a novel role for ETV6 in leukemia predisposition.


Subject(s)
Germ-Line Mutation , Mutation, Missense , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins c-ets/genetics , Repressor Proteins/genetics , Thrombocytopenia/genetics , Amino Acid Sequence , Case-Control Studies , HeLa Cells , Humans , Molecular Sequence Data , Proto-Oncogene Proteins c-ets/chemistry , Proto-Oncogene Proteins c-ets/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , ETS Translocation Variant 6 Protein
16.
J Biol Chem ; 290(22): 13692-709, 2015 May 29.
Article in English | MEDLINE | ID: mdl-25866208

ABSTRACT

Ets transcription factors, which share the conserved Ets DNA-binding domain, number nearly 30 members in humans and are particularly involved in developmental processes. Their deregulation following changes in expression, transcriptional activity, or by chromosomal translocation plays a critical role in carcinogenesis. Ets DNA binding, selectivity, and regulation have been extensively studied; however, questions still arise regarding binding specificity outside the core GGA recognition sequence and the mode of action of Ets post-translational modifications. Here, we report the crystal structures of Etv1, Etv4, Etv5, and Fev, alone and in complex with DNA. We identify previously unrecognized features of the protein-DNA interface. Interactions with the DNA backbone account for most of the binding affinity. We describe a highly coordinated network of water molecules acting in base selection upstream of the GGAA core and the structural features that may account for discrimination against methylated cytidine residues. Unexpectedly, all proteins crystallized as disulfide-linked dimers, exhibiting a novel interface (distant to the DNA recognition helix). Homodimers of Etv1, Etv4, and Etv5 could be reduced to monomers, leading to a 40-200-fold increase in DNA binding affinity. Hence, we present the first indication of a redox-dependent regulatory mechanism that may control the activity of this subset of oncogenic Ets transcription factors.


Subject(s)
Adenovirus E1A Proteins/chemistry , DNA-Binding Proteins/chemistry , Disulfides/chemistry , Nuclear Proteins/chemistry , Proto-Oncogene Proteins c-ets/chemistry , Proto-Oncogene Proteins/chemistry , Transcription Factors/chemistry , Amino Acid Sequence , Chromatography, Liquid , DNA/chemistry , Gene Expression Regulation , Humans , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Neoplasms/metabolism , Oxidation-Reduction , Oxygen/chemistry , Protein Binding , Protein Conformation , Protein Multimerization , Recombinant Proteins/chemistry , Sequence Homology, Amino Acid , Spectrometry, Mass, Electrospray Ionization
17.
PLoS One ; 9(8): e104391, 2014.
Article in English | MEDLINE | ID: mdl-25148142

ABSTRACT

Nonsense-mediated mRNA decay (NMD), which is best known for degrading mRNAs with premature termination codons (PTCs), is thought to be triggered by aberrant translation termination at stop codons located in an environment of the mRNP that is devoid of signals necessary for proper termination. In mammals, the cytoplasmic poly(A)-binding protein 1 (PABPC1) has been reported to promote correct termination and therewith antagonize NMD by interacting with the eukaryotic release factors 1 (eRF1) and 3 (eRF3). Using tethering assays in which proteins of interest are recruited as MS2 fusions to a NMD reporter transcript, we show that the three N-terminal RNA recognition motifs (RRMs) of PABPC1 are sufficient to antagonize NMD, while the eRF3-interacting C-terminal domain is dispensable. The RRM1-3 portion of PABPC1 interacts with eukaryotic initiation factor 4G (eIF4G) and tethering of eIF4G to the NMD reporter also suppresses NMD. We identified the interactions of the eIF4G N-terminus with PABPC1 and the eIF4G core domain with eIF3 as two genetically separable features that independently enable tethered eIF4G to inhibit NMD. Collectively, our results reveal a function of PABPC1, eIF4G and eIF3 in translation termination and NMD suppression, and they provide additional evidence for a tight coupling between translation termination and initiation.


Subject(s)
Eukaryotic Initiation Factor-4G/metabolism , Gene Expression Regulation , Nonsense Mediated mRNA Decay , Codon, Nonsense/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Eukaryotic Initiation Factor-4G/chemistry , Humans , Poly(A)-Binding Protein I/chemistry , Poly(A)-Binding Protein I/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Subunits , Proto-Oncogene Proteins c-ets/chemistry , Proto-Oncogene Proteins c-ets/metabolism , Ribonucleoside Diphosphate Reductase/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism
19.
Cell Rep ; 7(1): 19-26, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-24656813

ABSTRACT

The R2TP cochaperone complex plays a critical role in the assembly of multisubunit machines, including small nucleolar ribonucleoproteins (snoRNPs), RNA polymerase II, and the mTORC1 and SMG1 kinase complexes, but the molecular basis of substrate recognition remains unclear. Here, we describe a phosphopeptide binding domain (PIH-N) in the PIH1D1 subunit of the R2TP complex that preferentially binds to highly acidic phosphorylated proteins. A cocrystal structure of a PIH-N domain/TEL2 phosphopeptide complex reveals a highly specific phosphopeptide recognition mechanism in which Lys57 and 64 in PIH1D1, along with a conserved DpSDD phosphopeptide motif within TEL2, are essential and sufficient for binding. Proteomic analysis of PIH1D1 interactors identified R2TP complex substrates that are recruited by the PIH-N domain in a sequence-specific and phosphorylation-dependent manner suggestive of a common mechanism of substrate recognition. We propose that protein complexes assembled by the R2TP complex are defined by phosphorylation of a specific motif and recognition by the PIH1D1 subunit.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Crystallography, X-Ray/methods , Molecular Chaperones/metabolism , Amino Acid Sequence , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/genetics , Cell Line, Tumor , HEK293 Cells , HSP90 Heat-Shock Proteins/chemistry , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Humans , Models, Molecular , Molecular Chaperones/genetics , Molecular Sequence Data , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Phosphorylation , Protein Structure, Tertiary , Proto-Oncogene Proteins c-ets/chemistry , Proto-Oncogene Proteins c-ets/metabolism , Sequence Homology, Amino Acid , Substrate Specificity
20.
Biochem Soc Trans ; 42(1): 130-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24450640

ABSTRACT

The Ets family of eukaryotic transcription factors is based around the conserved Ets DNA-binding domain. Although their DNA-binding selectivity is biochemically and structurally well characterized, structures of homodimeric and ternary complexes point to Ets domains functioning as versatile protein-interaction modules. In the present paper, we review the progress made over the last decade to elucidate the structural mechanisms involved in modulation of DNA binding and protein partner selection during dimerization. We see that Ets domains, although conserved around a core architecture, have evolved to utilize a variety of interaction surfaces and binding mechanisms, reflecting Ets domains as dynamic interfaces for both DNA and protein interaction. Furthermore, we discuss recent advances in drug development for inhibition of Ets factors, and the roles structural biology can play in their future.


Subject(s)
Neoplasms/genetics , Proto-Oncogene Proteins c-ets/chemistry , Animals , Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Models, Molecular , Molecular Targeted Therapy , Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/chemistry , Multiprotein Complexes/physiology , Neoplasms/drug therapy , Neoplasms/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Proto-Oncogene Proteins c-ets/antagonists & inhibitors , Proto-Oncogene Proteins c-ets/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...