Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 141
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 51(16): 8864-8879, 2023 09 08.
Article in English | MEDLINE | ID: mdl-37503845

ABSTRACT

Transcription factors, such as nuclear receptors achieve precise transcriptional regulation by means of a tight and reciprocal communication with DNA, where cooperativity gained by receptor dimerization is added to binding site sequence specificity to expand the range of DNA target gene sequences. To unravel the evolutionary steps in the emergence of DNA selection by steroid receptors (SRs) from monomeric to dimeric palindromic binding sites, we carried out crystallographic, biophysical and phylogenetic studies, focusing on the estrogen-related receptors (ERRs, NR3B) that represent closest relatives of SRs. Our results, showing the structure of the ERR DNA-binding domain bound to a palindromic response element (RE), unveil the molecular mechanisms of ERR dimerization which are imprinted in the protein itself with DNA acting as an allosteric driver by allowing the formation of a novel extended asymmetric dimerization region (KR-box). Phylogenetic analyses suggest that this dimerization asymmetry is an ancestral feature necessary for establishing a strong overall dimerization interface, which was progressively modified in other SRs in the course of evolution.


Subject(s)
DNA , Transcription Factors , Transcription Factors/metabolism , Dimerization , Phylogeny , DNA/genetics , DNA/metabolism , Binding Sites , Receptors, Estrogen/genetics
2.
Front Endocrinol (Lausanne) ; 14: 1197063, 2023.
Article in English | MEDLINE | ID: mdl-37404310

ABSTRACT

Hepatocyte Nuclear Factor 4 (HNF4) is a transcription factor (TF) belonging to the nuclear receptor (NR) family that is expressed in liver, kidney, intestine and pancreas. It is a master regulator of liver-specific gene expression, in particular those genes involved in lipid transport and glucose metabolism and is crucial for the cellular differentiation during development. Dysregulation of HNF4 is linked to human diseases, such as type I diabetes (MODY1) and hemophilia. Here, we review the structures of the isolated HNF4 DNA binding domain (DBD) and ligand binding domain (LBD) and that of the multidomain receptor and compare them with the structures of other NRs. We will further discuss the biology of the HNF4α receptors from a structural perspective, in particular the effect of pathological mutations and of functionally critical post-translational modifications on the structure-function of the receptor.


Subject(s)
DNA-Binding Proteins , Hepatocyte Nuclear Factor 4 , Humans , DNA-Binding Proteins/genetics , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Gene Expression Regulation , Biology
3.
BMC Biol ; 20(1): 217, 2022 10 05.
Article in English | MEDLINE | ID: mdl-36199108

ABSTRACT

BACKGROUND: Nuclear receptors are transcription factors of central importance in human biology and associated diseases. Much of the knowledge related to their major functions, such as ligand and DNA binding or dimerization, derives from functional studies undertaken in classical model animals. It has become evident, however, that a deeper understanding of these molecular functions requires uncovering how these characteristics originated and diversified during evolution, by looking at more species. In particular, the comprehension of how dimerization evolved from ancestral homodimers to a more sophisticated state of heterodimers has been missing, due to a too narrow phylogenetic sampling. Here, we experimentally and phylogenetically define the evolutionary trajectory of nuclear receptor dimerization by analyzing a novel NR7 subgroup, present in various metazoan groups, including cnidarians, annelids, mollusks, sea urchins, and amphioxus, but lost in vertebrates, arthropods, and nematodes. RESULTS: We focused on NR7 of the cephalochordate amphioxus B. lanceolatum. We present a complementary set of functional, structural, and evolutionary analyses that establish that NR7 lies at a pivotal point in the evolutionary trajectory from homodimerizing to heterodimerizing nuclear receptors. The crystal structure of the NR7 ligand-binding domain suggests that the isolated domain is not capable of dimerizing with the ubiquitous dimerization partner RXR. In contrast, the full-length NR7 dimerizes with RXR in a DNA-dependent manner and acts as a constitutively active receptor. The phylogenetic and sequence analyses position NR7 at a pivotal point, just between the basal class I nuclear receptors that form monomers or homodimers on DNA and the derived class II nuclear receptors that exhibit the classical DNA-independent RXR heterodimers. CONCLUSIONS: Our data suggest that NR7 represents the "missing link" in the transition between class I and class II nuclear receptors and that the DNA independency of heterodimer formation is a feature that was acquired during evolution. Our studies define a novel paradigm of nuclear receptor dimerization that evolved from DNA-dependent to DNA-independent requirements. This new concept emphasizes the importance of DNA in the dimerization of nuclear receptors, such as the glucocorticoid receptor and other members of this pharmacologically important oxosteroid receptor subfamily. Our studies further underline the importance of studying emerging model organisms for supporting cutting-edge research.


Subject(s)
Receptors, Glucocorticoid , Receptors, Retinoic Acid , Animals , DNA , Dimerization , Humans , Ketosteroids , Ligands , Phylogeny , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Glucocorticoid/genetics , Receptors, Retinoic Acid/chemistry , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Retinoid X Receptors/chemistry , Retinoid X Receptors/genetics , Retinoid X Receptors/metabolism
4.
PLoS Genet ; 17(4): e1009492, 2021 04.
Article in English | MEDLINE | ID: mdl-33882063

ABSTRACT

Nuclear receptors are ligand-activated transcription factors that modulate gene regulatory networks from embryonic development to adult physiology and thus represent major targets for clinical interventions in many diseases. Most nuclear receptors function either as homodimers or as heterodimers. The dimerization is crucial for gene regulation by nuclear receptors, by extending the repertoire of binding sites in the promoters or the enhancers of target genes via combinatorial interactions. Here, we focused our attention on an unusual structural variation of the α-helix, called π-turn that is present in helix H7 of the ligand-binding domain of RXR and HNF4. By tracing back the complex evolutionary history of the π-turn, we demonstrate that it was present ancestrally and then independently lost in several nuclear receptor lineages. Importantly, the evolutionary history of the π-turn motif is parallel to the evolutionary diversification of the nuclear receptor dimerization ability from ancestral homodimers to derived heterodimers. We then carried out structural and biophysical analyses, in particular through point mutation studies of key RXR signature residues and showed that this motif plays a critical role in the network of interactions stabilizing homodimers. We further showed that the π-turn was instrumental in allowing a flexible heterodimeric interface of RXR in order to accommodate multiple interfaces with numerous partners and critical for the emergence of high affinity receptors. Altogether, our work allows to identify a functional role for the π-turn in oligomerization of nuclear receptors and reveals how this motif is linked to the emergence of a critical biological function. We conclude that the π-turn can be viewed as a structural exaptation that has contributed to enlarging the functional repertoire of nuclear receptors.


Subject(s)
Embryonic Development/genetics , Receptors, Cytoplasmic and Nuclear/ultrastructure , Retinoid X Receptors/genetics , Transcription Factors/ultrastructure , Amino Acid Sequence/genetics , Binding Sites/genetics , Dimerization , Gene Expression Regulation, Developmental/genetics , Gene Regulatory Networks/genetics , Humans , Ligands , Promoter Regions, Genetic/genetics , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/genetics , Retinoid X Receptors/ultrastructure , Transcription Factors/chemistry , Transcription Factors/genetics
5.
J Pestic Sci ; 46(1): 88-100, 2021 Feb 20.
Article in English | MEDLINE | ID: mdl-33746550

ABSTRACT

The ecdysone receptor (EcR) possesses the remarkable capacity to adapt structurally to different types of ligands. EcR binds ecdysteroids, including 20-hydroxyecdysone (20E), as well as nonsteroidal synthetic agonists such as insecticidal dibenzoylhydrazines (DBHs). Here, we report the crystal structures of the ligand-binding domains of Heliothis virescens EcR/USP bound to the DBH agonist BYI09181 and to the imidazole-type compound BYI08346. The region delineated by helices H7 and H10 opens up to tightly fit a phenyl ring of the ligands to an extent that depends on the bulkiness of ring substituent. In the structure of 20E-bound EcR, this part of the ligand-binding pocket (LBP) contains a channel filled by water molecules that form an intricate hydrogen bond network between 20E and LBP. The water channel present in the nuclear receptor bound to its natural hormone acts as a critical molecular adaptation spring used to accommodate synthetic agonists inside its binding cavity.

6.
Nucleic Acids Res ; 48(6): 3277-3285, 2020 04 06.
Article in English | MEDLINE | ID: mdl-31965182

ABSTRACT

The partition of aminoacyl-tRNA synthetases (aaRSs) into two classes of equal size and the correlated amino acid distribution is a puzzling still unexplained observation. We propose that the time scale of the amino-acid synthesis, assumed to be proportional to the number of reaction steps (NE) involved in the biosynthesis pathway, is one of the parameters that controlled the timescale of aaRSs appearance. Because all pathways are branched at fructose-6-phosphate on the metabolic pathway, this product is defined as the common origin for the NE comparison. For each amino-acid, the NE value, counted from the origin to the final product, provides a timescale for the pathways to be established. An archeological approach based on NE reveals that aaRSs of the two classes are generated in pair along this timescale. The results support the coevolution theory for the origin of the genetic code with an earlier appearance of class II aaRSs.


Subject(s)
Amino Acids/biosynthesis , Amino Acyl-tRNA Synthetases/genetics , Biosynthetic Pathways/genetics , Evolution, Molecular , Amino Acids/genetics , Fructosephosphates/genetics , Fructosephosphates/metabolism , Genetic Code/genetics
7.
Acta Crystallogr F Struct Biol Commun ; 75(Pt 2): 98-104, 2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30713160

ABSTRACT

The retinoic X receptor (RXR) plays a crucial role in the superfamily of nuclear receptors (NRs) by acting as an obligatory partner of several nuclear receptors; its role as a transcription factor is thus critical in many signalling pathways, such as metabolism, cell development, differentiation and cellular death. The first published structure of the apo ligand-binding domain (LBD) of RXRα, which is still used as a reference today, contained inaccuracies. In the present work, these inaccuracies were corrected using modern crystallographic tools. The most important correction concerns the presence of a π-bulge in helix H7, which was originally built as a regular α-helix. The presence of several CHAPS molecules, which are visible for the first time in the electron-density map and which stabilize the H1-H3 loop, which contains helix H2, are also revealed. The apo RXR structure has played an essential role in deciphering the molecular mode of action of NR ligands and is still used in numerous biophysical studies. This refined structure should be used preferentially in the future in interpreting experiments as well as for modelling and structural dynamics studies of the apo RXRα LBD.


Subject(s)
Apoproteins/chemistry , Apoproteins/metabolism , Retinoid X Receptor alpha/chemistry , Retinoid X Receptor alpha/metabolism , Amino Acid Sequence , Binding Sites , Crystallization , Crystallography, X-Ray , Humans , Ligands , Models, Molecular , Protein Binding , Protein Conformation , Protein Domains
8.
Article in English | MEDLINE | ID: mdl-28676789

ABSTRACT

Most nuclear receptors (NRs) bind DNA as dimers, either as hetero- or as homodimers on DNA sequences organized as two half-sites with specific orientation and spacing. The dimerization of NRs on their cognate response elements (REs) involves specific protein-DNA and protein-protein interactions. The estrogen-related receptor (ERR) belongs to the steroid hormone nuclear receptor (SHR) family and shares strong similarity in its DNA-binding domain (DBD) with that of the estrogen receptor (ER). In vitro, ERR binds with high affinity inverted repeat REs with a 3-bps spacing (IR3), but in vivo, it preferentially binds to single half-site REs extended at the 5'-end by 3 bp [estrogen-related response element (ERREs)], thus explaining why ERR was often inferred as a purely monomeric receptor. Since its C-terminal ligand-binding domain is known to homodimerize with a strong dimer interface, we investigated the binding behavior of the isolated DBDs to different REs using electrophoretic migration, multi-angle static laser light scattering (MALLS), non-denaturing mass spectrometry, and nuclear magnetic resonance. In contrast to ER DBD, ERR DBD binds as a monomer to EREs (IR3), such as the tff1 ERE-IR3, but we identified a DNA sequence composed of an extended half-site embedded within an IR3 element (embedded ERRE/IR3), where stable dimer binding is observed. Using a series of chimera and mutant DNA sequences of ERREs and IR3 REs, we have found the key determinants for the binding of ERR DBD as a dimer. Our results suggest that the sequence-directed DNA shape is more important than the exact nucleotide sequence for the binding of ERR DBD to DNA as a dimer. Our work underlines the importance of the shape-driven DNA readout mechanisms based on minor groove recognition and electrostatic potential. These conclusions may apply not only to ERR but also to other members of the SHR family, such as androgen or glucocorticoid, for which a strong well-conserved half-site is followed by a weaker one with degenerated sequence.

9.
Sci Adv ; 3(3): e1601778, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28435861

ABSTRACT

The origin of ancient ligand/receptor couples is often analyzed via reconstruction of ancient receptors and, when ligands are products of metabolic pathways, they are not supposed to evolve. However, because metabolic pathways are inherited by descent with modification, their structure can be compared using cladistic analysis. Using this approach, we studied the evolution of steroid hormones. We show that side-chain cleavage is common to most vertebrate steroids, whereas aromatization was co-opted for estrogen synthesis from a more ancient pathway. The ancestral products of aromatic activity were aromatized steroids with a side chain, which we named "paraestrols." We synthesized paraestrol A and show that it effectively binds and activates the ancestral steroid receptor. Our study opens the way to comparative studies of biologically active small molecules.


Subject(s)
Estrogens/genetics , Evolution, Molecular , Models, Genetic , Receptors, Estrogen/genetics , Animals
10.
J Biol Chem ; 291(28): 14430-46, 2016 Jul 08.
Article in English | MEDLINE | ID: mdl-27226617

ABSTRACT

Glycyl tRNA synthetase (GlyRS) provides a unique case among class II aminoacyl tRNA synthetases, with two clearly widespread types of enzymes: a dimeric (α2) species present in some bacteria, archaea, and eukaryotes; and a heterotetrameric form (α2ß2) present in most bacteria. Although the differences between both types of GlyRS at the anticodon binding domain level are evident, the extent and implications of the variations in the catalytic domain have not been described, and it is unclear whether the mechanism of amino acid recognition is also dissimilar. Here, we show that the α-subunit of the α2ß2 GlyRS from the bacterium Aquifex aeolicus is able to perform the first step of the aminoacylation reaction, which involves the activation of the amino acid with ATP. The crystal structure of the α-subunit in the complex with an analog of glycyl adenylate at 2.8 Å resolution presents a conformational arrangement that properly positions the cognate amino acid. This work shows that glycine is recognized by a subset of different residues in the two types of GlyRS. A structural and sequence analysis of class II catalytic domains shows that bacterial GlyRS is closely related to alanyl tRNA synthetase, which led us to define a new subclassification of these ancient enzymes and to propose an evolutionary path of α2ß2 GlyRS, convergent with α2 GlyRS and divergent from AlaRS, thus providing a possible explanation for the puzzling existence of two proteins sharing the same fold and function but not a common ancestor.


Subject(s)
Glycine-tRNA Ligase/chemistry , Phylogeny , Bacteria/enzymology , Crystallography, X-Ray , Models, Molecular , Protein Conformation
12.
Sci Rep ; 5: 8216, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25645674

ABSTRACT

Retinoid X receptors (RXRs) act as homodimers or heterodimerisation partners of class II nuclear receptors. RXR homo- and heterodimers bind direct repeats of the half-site (A/G)G(G/T)TCA separated by 1 nucleotide (DR1). We present a structural characterization of RXR-DNA binding domain (DBD) homodimers on several natural DR1s and an idealized symmetric DR1. Homodimers displayed asymmetric binding, with critical high-affinity interactions accounting for the 3' positioning of RXR in heterodimers on DR1s. Differing half-site and spacer DNA sequence induce changes in RXR-DBD homodimer conformation notably in the dimerization interface such that natural DR1s are bound with higher affinity than an idealized symmetric DR1. Subtle changes in the consensus DR1 DNA sequence therefore specify binding affinity through altered RXR-DBD-DNA contacts and changes in DBD conformation suggesting a general model whereby preferential half-site recognition determines polarity of heterodimer binding to response elements.


Subject(s)
Retinoid X Receptors/genetics , Base Sequence , Binding Sites , Calorimetry , Crystallography, X-Ray , Dimerization , Humans , Molecular Dynamics Simulation , Nucleic Acid Conformation , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , Response Elements/genetics , Retinoid X Receptors/chemistry , Retinoid X Receptors/metabolism
13.
Cell Rep ; 10(4): 516-26, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25620699

ABSTRACT

The bioactive form of vitamin D [1,25(OH)2D3] regulates mineral and bone homeostasis and exerts potent anti-inflammatory and antiproliferative properties through binding to the vitamin D receptor (VDR). The 3D structures of the VDR ligand-binding domain with 1,25(OH)2D3 or gemini analogs unveiled the molecular mechanism underlying ligand recognition. On the basis of structure-function correlations, we generated a point-mutated VDR (VDR(gem)) that is unresponsive to 1,25(OH)2D3, but the activity of which is efficiently induced by the gemini ligands. Moreover, we show that many VDR target genes are repressed by unliganded VDR(gem) and that mineral ion and bone homeostasis are more impaired in VDR(gem) mice than in VDR null mice, demonstrating that mutations abolishing VDR ligand binding result in more severe skeletal defects than VDR null mutations. As gemini ligands induce VDR(gem) transcriptional activity in mice and normalize their serum calcium levels, VDR(gem) is a powerful tool to further unravel both liganded and unliganded VDR signaling.


Subject(s)
Receptors, Calcitriol/chemistry , Receptors, Calcitriol/metabolism , Animals , Chromatin Immunoprecipitation , Crystallography, X-Ray , Fluorescence Polarization , Genotype , HEK293 Cells , Humans , MCF-7 Cells , Mice , Mice, Knockout , Mutation/genetics , Protein Binding/genetics , Receptors, Calcitriol/genetics , Spectrometry, Mass, Electrospray Ionization , Vitamin D/metabolism
14.
Nat Commun ; 5: 4139, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24942373

ABSTRACT

Nuclear receptors (NRs) regulate gene expression through DNA- and ligand-binding and thus represent crucial therapeutic targets. The ultraspiracle protein/ecdysone receptor (USP/EcR) complex binds to half-sites with a one base pair spaced inverted repeat (IR1), a palindromic DNA response element (RE) reminiscent of IRs observed for vertebrate steroid hormone receptors. Here we present the cryo electron microscopy structure of the USP/EcR complex bound to an IR1 RE which provides the first description of a full IR-bound NR complex. The structure reveals that even though the DNA is almost symmetric, the complex adopts a highly asymmetric architecture in which the ligand-binding domains (LBDs) are positioned 5' off-centred. Additional interactions of the USP LBD with the 5'-flanking sequence trigger transcription activity as monitored by transfection assays. The comparison with DR-bound NR complexes suggests that DNA is the major allosteric driver in inversely positioning the LBDs, which serve as the main binding-site for transcriptional regulators.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , DNA/genetics , Insect Proteins/chemistry , Insect Proteins/metabolism , Moths/metabolism , Receptors, Steroid/chemistry , Receptors, Steroid/metabolism , Animals , Crystallography, X-Ray , DNA/metabolism , DNA-Binding Proteins/genetics , Gene Expression Regulation , Insect Proteins/genetics , Inverted Repeat Sequences , Moths/chemistry , Moths/genetics , Protein Binding , Protein Structure, Tertiary , Receptors, Steroid/genetics , Response Elements
15.
Subcell Biochem ; 70: 21-36, 2014.
Article in English | MEDLINE | ID: mdl-24962879

ABSTRACT

Nuclear Retinoic Acid receptors (RARs) consist of three subtypes, α, ß, and γ, encoded by separate genes. They function as ligand-dependent transcriptional regulators, forming heterodimers with Retinoid X receptors (RXRs). RARs mediate the effects of retinoic acid (RA), the active metabolite of Vitamin A, and regulate many biological functions such as embryonic development, organogenesis, homeostasis, vision, immune functions, and reproduction. During the two last decades, a number of in-depth structure-function relationship studies have been performed, in particular with drug design perspectives in the therapeutics for cancer, dermatology, metabolic disease, and other human diseases. Recent structural results concerning integral receptors in diverse functional states, obtained using a combination of different methods, allow a better understanding of the mechanisms involved in molecular regulation. The structural data highlight the importance of DNA sequences for binding selectivity and the role of promoter response elements in the spatial organization of the protein domains into functional complexes.


Subject(s)
Receptors, Retinoic Acid/chemistry , Retinoid X Receptors/chemistry , Tretinoin/metabolism , Vitamin A/metabolism , Gene Expression Regulation , Humans , Ligands , Models, Molecular , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Multimerization , Protein Structure, Tertiary , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Response Elements , Retinoid X Receptors/genetics , Retinoid X Receptors/metabolism , Signal Transduction , Tretinoin/chemistry , Vitamin A/chemistry
16.
Biochemistry ; 52(39): 6844-55, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24000896

ABSTRACT

A recent renaissance in small-angle X-ray scattering (SAXS) made this technique a major tool for the low-resolution structural characterization of biological macromolecules in solution. The major limitation of existing methods for reconstructing 3D models from SAXS is imposed by the requirement of solute monodispersity. We present a novel approach that couples low-resolution 3D SAXS reconstruction with composition analysis of mixtures. The approach is applicable to polydisperse and difficult to purify systems, including weakly associated oligomers and transient complexes. Ab initio shape analysis is possible for symmetric homo-oligomers, whereas rigid body modeling is applied also to dissociating complexes when atomic structures of the individual subunits are available. In both approaches, the sample is considered as an equilibrium mixture of intact complexes/oligomers with their dissociation products or free subunits. The algorithms provide the 3D low-resolution model (for ab initio modeling, also the shape of the monomer) and the volume fractions of the bound and free state(s). The simultaneous fitting of multiple scattering data sets collected under different conditions allows one to restrain the modeling further. The possibilities of the approach are illustrated in simulated and experimental SAXS data from protein oligomers and multisubunit complexes including nucleoproteins. Using this approach, new structural insights are provided in the association behavior and conformations of estrogen-related receptors ERRα and ERRγ. The possibility of 3D modeling from the scattering by mixtures significantly widens the range of applicability of SAXS and opens novel avenues in the analysis of oligomeric mixtures and assembly/dissociation processes.


Subject(s)
Macromolecular Substances/chemistry , Receptors, Estrogen/chemistry , Humans , Models, Molecular , Protein Structure, Quaternary , Scattering, Small Angle , X-Ray Diffraction , ERRalpha Estrogen-Related Receptor
17.
PLoS One ; 8(7): e67810, 2013.
Article in English | MEDLINE | ID: mdl-23874451

ABSTRACT

BACKGROUND: PGC-1α is a crucial regulator of cellular metabolism and energy homeostasis that functionally acts together with the estrogen-related receptors (ERRα and ERRγ) in the regulation of mitochondrial and metabolic gene networks. Dimerization of the ERRs is a pre-requisite for interactions with PGC-1α and other coactivators, eventually leading to transactivation. It was suggested recently (Devarakonda et al) that PGC-1α binds in a strikingly different manner to ERRγ ligand-binding domains (LBDs) compared to its mode of binding to ERRα and other nuclear receptors (NRs), where it interacts directly with the two ERRγ homodimer subunits. METHODS/PRINCIPAL FINDINGS: Here, we show that PGC-1α receptor interacting domain (RID) binds in an almost identical manner to ERRα and ERRγ homodimers. Microscale thermophoresis demonstrated that the interactions between PGC-1α RID and ERR LBDs involve a single receptor subunit through high-affinity, ERR-specific L3 and low-affinity L2 interactions. NMR studies further defined the limits of PGC-1α RID that interacts with ERRs. Consistent with these findings, the solution structures of PGC-1α/ERRα LBDs and PGC-1α/ERRγ LBDs complexes share an identical architecture with an asymmetric binding of PGC-1α to homodimeric ERR. CONCLUSIONS/SIGNIFICANCE: These studies provide the molecular determinants for the specificity of interactions between PGC-1α and the ERRs, whereby negative cooperativity prevails in the binding of the coactivators to these receptors. Our work indicates that allosteric regulation may be a general mechanism controlling the binding of the coactivators to homodimers.


Subject(s)
Receptors, Estrogen/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Humans , Models, Molecular , Molecular Sequence Data , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Protein Binding , Protein Conformation , Protein Folding , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Subunits/chemistry , Protein Subunits/metabolism , Receptors, Estrogen/chemistry , Scattering, Small Angle , Transcription Factors/chemistry , X-Ray Diffraction , ERRalpha Estrogen-Related Receptor
18.
PLoS One ; 8(4): e60734, 2013.
Article in English | MEDLINE | ID: mdl-23593299

ABSTRACT

Integration of the HIV-1 cDNA into the human genome is catalyzed by the viral integrase (IN) protein. Several studies have shown the importance of cellular cofactors that interact with integrase and affect viral integration and infectivity. In this study, we produced a stable complex between HIV-1 integrase, viral U5 DNA, the cellular cofactor LEDGF/p75 and the integrase binding domain of INI1 (INI1-IBD), a subunit of the SWI/SNF chromatin remodeling factor. The stoichiometry of the IN/LEDGF/INI1-IBD/DNA complex components was found to be 4/2/2/2 by mass spectrometry and Fluorescence Correlation Spectroscopy. Functional assays showed that INI1-IBD inhibits the 3' processing reaction but does not interfere with specific viral DNA binding. Integration assays demonstrate that INI1-IBD decreases the amount of integration events but inhibits by-product formation such as donor/donor or linear full site integration molecules. Cryo-electron microscopy locates INI1-IBD within the cellular DNA binding site of the IN/LEDGF complex, constraining the highly flexible integrase in a stable conformation. Taken together, our results suggest that INI1 could stabilize the PIC in the host cell, by maintaining integrase in a stable constrained conformation which prevents non-specific interactions and auto integration on the route to its integration site within nucleosomes, while LEDGF organizes and stabilizes an active integrase tetramer suitable for specific vDNA integration. Moreover, our results provide the basis for a novel type of integrase inhibitor (conformational inhibitor) representing a potential new strategy for use in human therapy.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , HIV Integrase/metabolism , HIV-1/physiology , Models, Molecular , Multiprotein Complexes/metabolism , Transcription Factors/metabolism , Virus Integration/physiology , Cryoelectron Microscopy , Fluorescence Polarization , HIV-1/enzymology , Humans , Mass Spectrometry , Protein Conformation , SMARCB1 Protein , Spectrometry, Fluorescence
19.
J Mol Biol ; 425(13): 2317-29, 2013 Jul 10.
Article in English | MEDLINE | ID: mdl-23499886

ABSTRACT

Nuclear receptors control a large number of physiological events through the regulation of gene transcription. Their activation function involves and is controlled by ligands and multiple cofactors (repressors, activators and bridging proteins). Further post-translational modifications resulting from the crosstalk between different signaling pathways provide additional regulations. The numerous players involved allow the sophisticated fine-tuning of transcriptional regulation. The present review describes how allosteric mechanisms are used in the control of the sequential and ordered binding of nuclear receptors and the various protein effectors to target DNA.


Subject(s)
Allosteric Regulation , Gene Expression Regulation , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription, Genetic , Crystallography, X-Ray , Models, Biological , Models, Molecular , Molecular Dynamics Simulation , Protein Conformation , Signal Transduction
20.
Cell ; 152(1-2): 132-43, 2013 Jan 17.
Article in English | MEDLINE | ID: mdl-23332751

ABSTRACT

The sequence-specific transcription factor NF-Y binds the CCAAT box, one of the sequence elements most frequently found in eukaryotic promoters. NF-Y is composed of the NF-YA and NF-YB/NF-YC subunits, the latter two hosting histone-fold domains (HFDs). The crystal structure of NF-Y bound to a 25 bp CCAAT oligonucleotide shows that the HFD dimer binds to the DNA sugar-phosphate backbone, mimicking the nucleosome H2A/H2B-DNA assembly. NF-YA both binds to NF-YB/NF-YC and inserts an α helix deeply into the DNA minor groove, providing sequence-specific contacts to the CCAAT box. Structural considerations and mutational data indicate that NF-YB ubiquitination at Lys138 precedes and is equivalent to H2B Lys120 monoubiquitination, important in transcriptional activation. Thus, NF-Y is a sequence-specific transcription factor with nucleosome-like properties of nonspecific DNA binding and helps establish permissive chromatin modifications at CCAAT promoters. Our findings suggest that other HFD-containing proteins may function in similar ways.


Subject(s)
CCAAT-Binding Factor/chemistry , Amino Acid Sequence , Animals , CCAAT-Binding Factor/metabolism , Crystallography, X-Ray , DNA/chemistry , DNA/genetics , DNA/metabolism , HSP72 Heat-Shock Proteins/genetics , Histones/chemistry , Humans , Molecular Sequence Data , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Oligonucleotides/chemistry , Oligonucleotides/genetics , Oligonucleotides/metabolism , Promoter Regions, Genetic , Protein Structure, Tertiary , Sequence Alignment , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...