Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 37
1.
Int J Mol Sci ; 24(4)2023 Feb 10.
Article En | MEDLINE | ID: mdl-36835018

Investigation of RNA- and DNA-binding proteins to a defined regulatory sequence, such as an AU-rich RNA and a DNA enhancer element, is important for understanding gene regulation through their interactions. For in vitro binding studies, an electrophoretic mobility shift assay (EMSA) was widely used in the past. In line with the trend toward using non-radioactive materials in various bioassays, end-labeled biotinylated RNA and DNA oligonucleotides can be more practical probes to study protein-RNA and protein-DNA interactions; thereby, the binding complexes can be pulled down with streptavidin-conjugated resins and identified by Western blotting. However, setting up RNA and DNA pull-down assays with biotinylated probes in optimum protein binding conditions remains challenging. Here, we demonstrate the step-by step optimization of pull-down for IRP (iron-responsive-element-binding protein) with a 5'-biotinylated stem-loop IRE (iron-responsive element) RNA, HuR, and AUF1 with an AU-rich RNA element and Nrf2 binding to an antioxidant-responsive element (ARE) enhancer in the human ferritin H gene. This study was designed to address key technical questions in RNA and DNA pull-down assays: (1) how much RNA and DNA probes we should use; (2) what binding buffer and cell lysis buffer we can use; (3) how to verify the specific interaction; (4) what streptavidin resin (agarose or magnetic beads) works; and (5) what Western blotting results we can expect from varying to optimum conditions. We anticipate that our optimized pull-down conditions can be applicable to other RNA- and DNA-binding proteins along with emerging non-coding small RNA-binding proteins for their in vitro characterization.


Biotinylation , Carrier Proteins , Chromatography, Affinity , DNA , RNA , Humans , Carrier Proteins/chemistry , DNA/chemistry , Iron/metabolism , Iron Regulatory Protein 1/chemistry , Iron Regulatory Protein 2/chemistry , NF-E2-Related Factor 2/chemistry , RNA/chemistry , Streptavidin/metabolism , Chromatography, Affinity/methods
2.
Genes (Basel) ; 12(9)2021 08 30.
Article En | MEDLINE | ID: mdl-34573347

Iron responsive elements (IREs) are mRNA stem-loop targets for translational control by the two iron regulatory proteins IRP1 and IRP2. They are found in the untranslated regions (UTRs) of genes that code for proteins involved in iron metabolism. There are ten "classic" IRE types that define the conserved secondary and tertiary structure elements necessary for proper IRP binding, and there are 83 published "IRE-like" sequences, most of which depart from the established IRE model. Here are structurally-guided discussions regarding the essential features of an IRE and what is important for IRE family membership.


Iron , RNA, Messenger/chemistry , Response Elements , Animals , Humans , Iron/metabolism , Iron Regulatory Protein 1/chemistry , Iron Regulatory Protein 1/metabolism , Nucleic Acid Conformation , Response Elements/physiology
3.
Proc Natl Acad Sci U S A ; 115(39): E9085-E9094, 2018 09 25.
Article En | MEDLINE | ID: mdl-30201724

Maturation of iron-sulfur (Fe-S) proteins in eukaryotes requires complex machineries in mitochondria and cytosol. Initially, Fe-S clusters are assembled on dedicated scaffold proteins and then are trafficked to target apoproteins. Within the cytosolic Fe-S protein assembly (CIA) machinery, the conserved P-loop nucleoside triphosphatase Nbp35 performs a scaffold function. In yeast, Nbp35 cooperates with the related Cfd1, which is evolutionary less conserved and is absent in plants. Here, we investigated the potential scaffold function of human CFD1 (NUBP2) in CFD1-depleted HeLa cells by measuring Fe-S enzyme activities or 55Fe incorporation into Fe-S target proteins. We show that CFD1, in complex with NBP35 (NUBP1), performs a crucial role in the maturation of all tested cytosolic and nuclear Fe-S proteins, including essential ones involved in protein translation and DNA maintenance. CFD1 also matures iron regulatory protein 1 and thus is critical for cellular iron homeostasis. To better understand the scaffold function of CFD1-NBP35, we resolved the crystal structure of Chaetomium thermophilum holo-Cfd1 (ctCfd1) at 2.6-Å resolution as a model Cfd1 protein. Importantly, two ctCfd1 monomers coordinate a bridging [4Fe-4S] cluster via two conserved cysteine residues. The surface-exposed topology of the cluster is ideally suited for both de novo assembly and facile transfer to Fe-S apoproteins mediated by other CIA factors. ctCfd1 specifically interacted with ATP, which presumably associates with a pocket near the Cfd1 dimer interface formed by the conserved Walker motif. In contrast, ctNbp35 preferentially bound GTP, implying differential regulation of the two fungal scaffold components during Fe-S cluster assembly and/or release.


Apoproteins/chemistry , Chaetomium/chemistry , Fungal Proteins/chemistry , GTP-Binding Proteins/chemistry , Iron Regulatory Protein 1/chemistry , Iron-Sulfur Proteins/chemistry , Amino Acid Motifs , Apoproteins/genetics , Apoproteins/metabolism , Catalytic Domain , Chaetomium/genetics , Chaetomium/metabolism , Crystallography, X-Ray , Fungal Proteins/genetics , Fungal Proteins/metabolism , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , HeLa Cells , Humans , Iron Regulatory Protein 1/genetics , Iron Regulatory Protein 1/metabolism , Iron-Sulfur Proteins/genetics , Iron-Sulfur Proteins/metabolism
4.
Biochim Biophys Acta Gen Subj ; 1862(9): 1980-1987, 2018 09.
Article En | MEDLINE | ID: mdl-29842905

BACKGROUND: The CIA2A protein, in complex with CIAO1, has been proposed to be exclusively implicated in the maturation of cytosolic aconitase. However, how the CIA2A-CIAO1 complex generates active aconitase is still unknown and the available structural information has not provided any crucial insights into the molecular function of CIA2A. METHODS: In this work we have characterized the Fe/S cluster binding properties of CIA2A and of the CIA2A-CIAO1 complex via NMR, UV - vis absorption and EPR spectroscopies and we have investigated how the Fe/S cluster is transferred to inactive aconitase/IRP1 protein. RESULTS: We found that an heterotrimeric species formed by two molecules of CIA2A and one of CIAO1 can bind one [4Fe-4S] cluster and that residue Cys90 of CIA2A is one of the cluster ligand. The holo trimeric complex is able to transfer the [4Fe-4S] cluster to apo-IRP1 thus generating the active form of aconitase. CONCLUSIONS AND GENERAL SIGNIFICANCE: These findings, which highlight a functional role for CIA2A-CIAO1 complex in aconitase maturation, raises a broad interest and can have a high impact on the community studying metal trafficking and iron­sulfur protein biogenesis. The present study can provide solid bases for further investigation of the molecular mechanisms involving also other CIA machinery proteins.


Carrier Proteins/metabolism , Iron Regulatory Protein 1/metabolism , Iron/metabolism , Metallochaperones/metabolism , Sulfides/metabolism , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cytosol , Humans , Iron/chemistry , Iron Regulatory Protein 1/chemistry , Iron Regulatory Protein 1/genetics , Metallochaperones/chemistry , Metallochaperones/genetics , Metalloproteins , Mutagenesis, Site-Directed , Mutation , Protein Binding , Protein Conformation , Sulfides/chemistry
5.
Sci Rep ; 8(1): 5903, 2018 04 12.
Article En | MEDLINE | ID: mdl-29651044

Molecules of single proteins, echoforms, can be distributed between two (or more) subcellular locations, a phenomenon which we refer to as dual targeting or dual localization. The yeast aconitase gene ACO1 (778 amino acids), encodes a single translation product that is nonetheless dual localized to the cytosol and mitochondria by a reverse translocation mechanism. The solved crystal structure of aconitase isolated from porcine heart mitochondria shows that it has four domains. The first three tightly associated N-terminal domains are tethered to the larger C-terminal fourth domain (C-terminal amino acids 517-778). We have previously shown that the aconitase C terminal domain constitutes an independent dual targeting signal when fused to mitochondria-targeted passenger-proteins. We show that the aconitase N and C-terminal domains interact and that this interaction is important for efficient aconitase post translational import into mitochondria and for aconitase dual targeting (relative levels of aconitase echoforms). Our results suggest a "chaperone-like function" of the C terminal domain towards the N terminal domains which can be modulated by Ssa1/2 (cytosolic Hsp70).


Adenosine Triphosphatases/chemistry , Cytosol/metabolism , HSP70 Heat-Shock Proteins/chemistry , Iron Regulatory Protein 1/chemistry , Mitochondria/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Binding Sites , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Iron Regulatory Protein 1/genetics , Iron Regulatory Protein 1/metabolism , Models, Molecular , Mutation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Transport , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
6.
Methods Enzymol ; 595: 83-106, 2017.
Article En | MEDLINE | ID: mdl-28882209

MitoNEET is the first identified Fe-S protein anchored to mammalian outer mitochondrial membranes with the vast majority of the protein polypeptide located in the cytosol, including its [2Fe-2S] cluster-binding domain. The coordination of the cluster is unusual and involves three cysteines and one histidine. MitoNEET is capable of transferring its redox-active Fe-S cluster to a bacterial apo-ferredoxin in vitro even under aerobic conditions, unlike other Fe-S transfer proteins such as ISCU. This specificity suggests its possible involvement in Fe-S repair after oxidative and/or nitrosative stress. Recently, we identified cytosolic aconitase/iron regulatory protein 1 (IRP1) as the first physiological protein acceptor of the mitoNEET Fe-S cluster in an Fe-S repair process. This chapter describes methods to study in vitro mitoNEET Fe-S cluster transfer/repair to a bacterial ferredoxin used as a model aporeceptor and in a more comprehensive manner to cytosolic aconitase/IRP1 after a nitrosative stress using in vitro, in cellulo, and in vivo methods.


Aconitate Hydratase/metabolism , Iron Regulatory Protein 1/metabolism , Iron-Sulfur Proteins/metabolism , Iron/metabolism , Mitochondrial Proteins/metabolism , Aconitate Hydratase/chemistry , Animals , Cysteine/metabolism , Cytosol/enzymology , Escherichia coli , Ferredoxins/metabolism , Histidine/metabolism , Humans , Iron/chemistry , Iron Regulatory Protein 1/chemistry , Iron-Sulfur Proteins/chemistry , Mitochondrial Membranes/metabolism , Nitrosative Stress , Oxidation-Reduction
7.
J Biol Chem ; 292(38): 15976-15989, 2017 09 22.
Article En | MEDLINE | ID: mdl-28768766

Iron-regulatory protein 1 (IRP1) belongs to a family of RNA-binding proteins that modulate metazoan iron metabolism. Multiple mechanisms are employed to control the action of IRP1 in dictating changes in the uptake and metabolic fate of iron. Inactivation of IRP1 RNA binding by iron primarily involves insertion of a [4Fe-4S] cluster by the cytosolic iron-sulfur cluster assembly (CIA) system, converting it into cytosolic aconitase (c-acon), but can also involve iron-mediated degradation of IRP1 by the E3 ligase FBXL5 that also targets IRP2. How CIA and FBXL5 collaborate to maintain cellular iron homeostasis through IRP1 and other pathways is poorly understood. Because impaired Fe-S cluster biogenesis associates with human disease, we determined the importance of FBXL5 for regulating IRP1 when CIA is impaired. Suppression of FBXL5 expression coupled with induction of an IRP1 mutant (IRP13C>3S) that cannot insert the Fe-S cluster, or along with knockdown of the CIA factors NUBP2 or FAM96A, reduced cell viability. Iron supplementation reversed this growth defect and was associated with FBXL5-dependent polyubiquitination of IRP1. Phosphorylation of IRP1 at Ser-138 increased when CIA was inhibited and was required for iron rescue. Impaired CIA activity, as noted by reduced c-acon activity, was associated with enhanced FBXL5 expression and a concomitant reduction in IRP1 and IRP2 protein level and RNA-binding activity. Conversely, expression of either IRP induced FBXL5 protein level, demonstrating a negative feedback loop limiting excessive accumulation of iron-response element RNA-binding activity, whose disruption reduces cell growth. We conclude that a regulatory circuit involving FBXL5 and CIA acts through both IRPs to control iron metabolism and promote optimal cell growth.


F-Box Proteins/metabolism , Iron Regulatory Protein 1/metabolism , Iron/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , F-Box Proteins/genetics , Ferritins/metabolism , Gene Knockdown Techniques , HEK293 Cells , Humans , Iron Regulatory Protein 1/chemistry , Iron Regulatory Protein 2/metabolism , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Proteolysis , RNA/metabolism , Serine/metabolism , Sulfur/metabolism , Ubiquitin-Protein Ligase Complexes/deficiency , Ubiquitin-Protein Ligase Complexes/genetics , Ubiquitination
8.
J Biol Chem ; 292(31): 12744-12753, 2017 08 04.
Article En | MEDLINE | ID: mdl-28615439

Fe-S cofactors are composed of iron and inorganic sulfur in various stoichiometries. A complex assembly pathway conducts their initial synthesis and subsequent binding to recipient proteins. In this minireview, we discuss how discovery of the role of the mammalian cytosolic aconitase, known as iron regulatory protein 1 (IRP1), led to the characterization of the function of its Fe-S cluster in sensing and regulating cellular iron homeostasis. Moreover, we present an overview of recent studies that have provided insights into the mechanism of Fe-S cluster transfer to recipient Fe-S proteins.


Homeostasis , Iron Regulatory Protein 1/physiology , Iron/physiology , Models, Molecular , Animals , Apoenzymes/chemistry , Apoenzymes/metabolism , Carbon-Sulfur Lyases/biosynthesis , Carbon-Sulfur Lyases/chemistry , Carbon-Sulfur Lyases/physiology , Electron Transport , Gene Expression Regulation, Enzymologic , HSP70 Heat-Shock Proteins/biosynthesis , HSP70 Heat-Shock Proteins/chemistry , HSP70 Heat-Shock Proteins/physiology , Humans , Iron Regulatory Protein 1/biosynthesis , Iron Regulatory Protein 1/chemistry , Iron-Binding Proteins/biosynthesis , Iron-Binding Proteins/chemistry , Iron-Binding Proteins/physiology , Iron-Regulatory Proteins/biosynthesis , Iron-Regulatory Proteins/chemistry , Iron-Regulatory Proteins/physiology , Iron-Sulfur Proteins/biosynthesis , Iron-Sulfur Proteins/chemistry , Iron-Sulfur Proteins/physiology , Mitochondrial Proteins/biosynthesis , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/physiology , Molecular Chaperones/biosynthesis , Molecular Chaperones/chemistry , Molecular Chaperones/physiology , Protein Folding , Protein Interaction Domains and Motifs , Protein Multimerization , Response Elements , Succinate Dehydrogenase/biosynthesis , Succinate Dehydrogenase/chemistry , Succinate Dehydrogenase/physiology , Frataxin
9.
Biochem Biophys Res Commun ; 465(3): 620-4, 2015 Sep 25.
Article En | MEDLINE | ID: mdl-26296460

Glutaredoxin 3 (GLRX3) is a member of monothiol glutaredoxins with a CGFS active site that has been demonstrated to function in cellular iron sensing and trafficking via its bound iron-sulfur cluster. Human GLRX3 has been shown to form a dimer that binds two bridging [2Fe-2S] clusters with glutathione (GSH) as a ligand, assembling a compound 2GLRX3-2[2Fe-2S]-4GSH. Each iron of the iron-sulfur clusters is bound to the thiols of the cysteines, one of which is from the active site of GLRX3, the other from the noncovalently bound GSH. Here, we show that the recombinant human GLRX3 isolated anaerobically from Escherichia coli can incorporate [4Fe-4S] cluster in the absence of GSH, revealed by spectral and enzymatic analysis. [4Fe-4S] cluster-containing GLRX3 is competent for converting iron regulatory protein 1 (apo-IRP1) into aconitase within 30 min, via intact iron-sulfur cluster transfer. These in vitro studies suggest that human GLRX3 is important for cytosolic Fe-S protein maturation.


Aconitate Hydratase/chemical synthesis , Carrier Proteins/chemistry , Iron Regulatory Protein 1/chemistry , Iron-Sulfur Proteins/chemistry , Binding Sites , Humans , Protein Binding
10.
Exp Parasitol ; 157: 1-11, 2015 Oct.
Article En | MEDLINE | ID: mdl-26115940

During its parasitic life stages, the marine ectoparasitic copepod Lepeophtheirus salmonis ingests large amounts of host blood, which contains high amounts of iron. Iron is an essential micronutrient, but also toxic in high dosages, and blood-feeding parasites like the salmon louse must thus possess an efficient system to handle the excess iron. Iron regulatory protein 1 and 2 (IRP1 and IRP2) are known to play crucial roles in this process, by regulating several proteins involved in iron transport and storage, depending on the cellular iron concentration. To gain knowledge about the regulation of the iron metabolism in salmon lice, two IRP homologues (LsIRP1A and LsIRP1B) were identified by sequence and predicted structure similarity to known IRPs in other species. In situ hybridisation revealed that LsIRP1A and LsIRP1B mRNAs were expressed in the ovaries, oviducts and vitellogenic oocytes of adult females. Transcription levels of LsIRP1A and LsIRP1B mRNAs did not differ significantly between the different developmental stages of the salmon louse. Adults in the absence of blood as a feed source had decreased levels of LsIRP1A, but not LsIRP1B mRNA. RNA binding experiments indicated the presence of functioning IRP in salmon lice. In order to explore the biological functions of LsIRP1A and LsIRP1B, the mRNAs of both proteins were knocked down by RNA interference (RNAi) in preadult females. The knockdown was confirmed by qRT-PCR. LsIRP1B knockdown lice produced less offspring than control lice due to slightly shorter egg strings and had decreased levels of transcripts involved in egg development. Knockdown of both LsIRP1A and LsIRP1B caused increased expression of a salmon louse Ferritin (LsFer). These results confirm that salmon lice have two IRP1 homologues, LsIRP1A and LsIRP1B, and might suggest a function in cellular iron regulation in the reproductive organs and eggs.


Copepoda/chemistry , Ectoparasitic Infestations/veterinary , Fish Diseases/parasitology , Iron Regulatory Protein 1/physiology , Salmo salar/parasitology , Amino Acid Sequence , Animals , Copepoda/classification , Copepoda/metabolism , Ectoparasitic Infestations/parasitology , Female , Gene Expression Regulation , Humans , In Situ Hybridization , Iron/metabolism , Iron Regulatory Protein 1/chemistry , Iron Regulatory Protein 1/genetics , Male , Molecular Sequence Data , Phylogeny , RNA Interference , RNA, Messenger/analysis , RNA, Messenger/metabolism , Sequence Alignment
11.
J Biol Chem ; 289(41): 28070-86, 2014 Oct 10.
Article En | MEDLINE | ID: mdl-25012650

In eukaryotes, mitochondrial iron-sulfur cluster (ISC), export and cytosolic iron-sulfur cluster assembly (CIA) machineries carry out biogenesis of iron-sulfur (Fe-S) clusters, which are critical for multiple essential cellular pathways. However, little is known about their export out of mitochondria. Here we show that Fe-S assembly of mitoNEET, the first identified Fe-S protein anchored in the mitochondrial outer membrane, strictly depends on ISC machineries and not on the CIA or CIAPIN1. We identify a dedicated ISC/export pathway in which augmenter of liver regeneration, a mitochondrial Mia40-dependent protein, is specific to mitoNEET maturation. When inserted, the Fe-S cluster confers mitoNEET folding and stability in vitro and in vivo. The holo-form of mitoNEET is resistant to NO and H2O2 and is capable of repairing oxidatively damaged Fe-S of iron regulatory protein 1 (IRP1), a master regulator of cellular iron that has recently been involved in the mitochondrial iron supply. Therefore, our findings point to IRP1 as the missing link to explain the function of mitoNEET in the control of mitochondrial iron homeostasis.


Iron Regulatory Protein 1/chemistry , Iron/metabolism , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/chemistry , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/chemistry , Animals , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation , HeLa Cells , Hep G2 Cells , Homeostasis , Humans , Hydrogen Peroxide/chemistry , Iron Regulatory Protein 1/genetics , Iron Regulatory Protein 1/metabolism , Mice , Mice, Transgenic , Mitochondria/chemistry , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membranes/chemistry , Mitochondrial Precursor Protein Import Complex Proteins , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Nitric Oxide/chemistry , Oxidation-Reduction , Protein Folding , Protein Stability , Protein Structure, Tertiary , Protein Transport , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction
12.
Nucleic Acids Res ; 42(10): 6567-77, 2014 Jun.
Article En | MEDLINE | ID: mdl-24728987

Metal ion binding was previously shown to destabilize IRE-RNA/IRP1 equilibria and enhanced IRE-RNA/eIF4F equilibria. In order to understand the relative importance of kinetics and stability, we now report rapid rates of protein/RNA complex assembly and dissociation for two IRE-RNAs with IRP1, and quantitatively different metal ion response kinetics that coincide with the different iron responses in vivo. kon, for FRT IRE-RNA binding to IRP1 was eight times faster than ACO2 IRE-RNA. Mn(2+) decreased kon and increased koff for IRP1 binding to both FRT and ACO2 IRE-RNA, with a larger effect for FRT IRE-RNA. In order to further understand IRE-mRNA regulation in terms of kinetics and stability, eIF4F kinetics with FRT IRE-RNA were determined. kon for eIF4F binding to FRT IRE-RNA in the absence of metal ions was 5-times slower than the IRP1 binding to FRT IRE-RNA. Mn(2+) increased the association rate for eIF4F binding to FRT IRE-RNA, so that at 50 µM Mn(2+) eIF4F bound more than 3-times faster than IRP1. IRP1/IRE-RNA complex has a much shorter life-time than the eIF4F/IRE-RNA complex, which suggests that both rate of assembly and stability of the complexes are important, and that allows this regulatory system to respond rapidly to change in cellular iron.


Eukaryotic Initiation Factor-4F/metabolism , Iron Regulatory Protein 1/metabolism , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Regulatory Sequences, Ribonucleic Acid , Aconitate Hydratase/genetics , Aconitate Hydratase/metabolism , Animals , Ferritins/genetics , Ferritins/metabolism , Iron/metabolism , Iron Regulatory Protein 1/chemistry , Kinetics , Manganese/chemistry , Potassium/chemistry , Rabbits
13.
J Mol Biol ; 425(18): 3301-10, 2013 Sep 23.
Article En | MEDLINE | ID: mdl-23806658

IRP1 [iron regulatory protein (IRP) 1] is a bifunctional protein with mutually exclusive end-states. In one mode of operation, IRP1 binds iron-responsive element (IRE) stem-loops in messenger RNAs encoding proteins of iron metabolism to control their rate of translation. In its other mode, IRP1 serves as cytoplasmic aconitase to correlate iron availability with the energy and oxidative stress status of the cell. IRP1/IRE binding occurs through two separate interfaces, which together contribute about two-dozen hydrogen bonds. Five amino acids make base-specific contacts and are expected to contribute significantly to binding affinity and specificity of this protein:RNA interaction. In this mutagenesis study, each of the five base-specific amino acids was changed to alter binding at each site. Analysis of IRE binding affinity and translational repression activity of the resulting IRP1 mutants showed that four of the five contact points contribute uniquely to the overall binding affinity of the IRP1:IRE interaction, while one site was found to be unimportant. The stronger-than-expected effect on binding affinity of mutations at Lys379 and Ser681, residues that make contact with the conserved nucleotides G16 and C8, respectively, identified them as particularly critical for providing specificity and stability to IRP1:IRE complex formation. We also show that even though the base-specific RNA-binding residues are not part of the aconitase active site, their substitutions can affect the aconitase activity of holo-IRP1, positively or negatively.


Iron Regulatory Protein 1/metabolism , Iron/pharmacology , Nucleotides/metabolism , Response Elements/genetics , Animals , Binding Sites/genetics , Catalytic Domain/genetics , Down-Regulation/genetics , Gene Expression Regulation/drug effects , Iron Regulatory Protein 1/chemistry , Iron Regulatory Protein 1/genetics , Iron Regulatory Protein 1/physiology , Models, Molecular , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Nucleic Acid Conformation , Nucleotides/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rabbits , Saccharomyces cerevisiae , Substrate Specificity
14.
Gene ; 524(2): 341-6, 2013 Jul 25.
Article En | MEDLINE | ID: mdl-23590984

Iron is a cofactor for many proteins that are involved in essential metabolic processes. However, iron must be strictly regulated because it can react with oxygen to generate cytotoxic reactive oxygen intermediates. Iron regulatory protein 1 (IRP1) is a bi-functional protein that can act either as a post-transcriptional regulator of mRNAs containing iron responsive elements, or as a [4Fe-4S] cluster-containing cytosolic aconitase. Previous X-ray crystallography results show that IRP1 is in an open L-shape conformation when bound to IRE-RNAs, and in a globular conformation when it binds an iron-sulfur cluster. The structure of apo-IRP1 and the mechanism by which it transforms to either functional state is unknown. Therefore, small angle X-ray scattering was used to determine the low resolution solution structure of apo-IRP1 and to characterize its biophysical properties. These results show that apo-IRP1 has a radius of gyration (Rg) of 33.6±0.3Å, and a Dmax of 118±2Å. The ab initio and rigid-body modeling results show that apo-IRP1 is in an open conformation in solution, and the ensemble optimization results show that the molecules stay narrowly distributed about a Rg of 33-34Å. The open apo-IRP1 conformation seems optimal for subsequent conversion to either functional end state: RNA-binding, or cytosolic aconitase.


Apolipoproteins/chemistry , Iron Regulatory Protein 1/chemistry , RNA-Binding Proteins/chemistry , Animals , Crystallography, X-Ray , Cytosol/chemistry , Protein Binding , Protein Conformation , Protein Structure, Tertiary , RNA/chemistry , Rabbits , Saccharomyces cerevisiae/chemistry , Scattering, Small Angle , Solutions/chemistry , Structure-Activity Relationship
15.
FEBS Lett ; 586(1): 32-5, 2012 Jan 02.
Article En | MEDLINE | ID: mdl-22119729

Iron responsive elements (IREs) are short stem-loop structures found in several mRNAs encoding proteins involved in cellular iron metabolism. Iron regulatory proteins (IRPs) control iron homeostasis through differential binding to the IREs, accommodating any sequence or structural variations that the IREs may present. Here we report the structure of IRP1 in complex with transferrin receptor 1 B (TfR B) IRE, and compare it to the complex with ferritin H (Ftn H) IRE. The two IREs are bound to IRP1 through nearly identical protein-RNA contacts, although their stem conformations are significantly different. These results support the view that binding of different IREs with IRP1 depends both on protein and RNA conformational plasticity, adapting to RNA variation while retaining conserved protein-RNA contacts.


Iron Regulatory Protein 1/chemistry , Iron Regulatory Protein 1/metabolism , Iron , Receptors, Transferrin/chemistry , Receptors, Transferrin/metabolism , Response Elements , Animals , Crystallography, X-Ray , Nucleic Acid Conformation , Protein Conformation , Rabbits
16.
Acc Chem Res ; 44(12): 1320-8, 2011 Dec 20.
Article En | MEDLINE | ID: mdl-22026512

Messenger RNAs (mRNAs) are emerging as prime targets for small-molecule drugs. They afford an opportunity to assert control over an enormous range of biological processes: mRNAs regulate protein synthesis rates, have specific 3-D regulatory structures, and, in nucleated cells, are separated from DNA in space and time. All of the many steps between DNA copying (transcription) and ribosome binding (translation) represent potential control points. Messenger RNAs can fold into complex, 3-D shapes, such as tRNAs and rRNAs, providing an added dimension to the 2-D RNA structure (base pairing) targeted in many mRNA interference approaches. In this Account, we describe the structural and functional properties of the IRE (iron-responsive element) family, one of the few 3-D mRNA regulatory elements with known 3-D structure. This family of related base sequences regulates the mRNAs that encode proteins for iron metabolism. We begin by considering the IRE-RNA structure, which consists of a short (~30-nucleotide) RNA helix. Nature tuned the structure by combining a conserved AGU pseudotriloop, a closing C-G base pair, and a bulge C with various RNA helix base pairs. The result is a set of IRE-mRNAs with individual iron responses. The physiological iron signal is hexahydrated ferrous ion; in vivo iron responses vary over 10-fold depending on the individual IRE-RNA structure. We then discuss the interaction between the IRE-RNA structure and the proteins associated with it. IRE-RNA structures, which are usually noncoding, tightly bind specific proteins called IRPs. These repressor proteins are bound to IRE-RNA through C-bulge and AGU contacts that flip out a loop AG and a bulge C, bending the RNA helix. After binding, the exposed RNA surface then invites further interactions, such as with iron and other proteins. Binding of the IRE-RNA and the IRP also changes the IRP conformation. IRP binding stabilities vary 10-fold within the IRE family, reflecting individual IRE-RNA paired and unpaired bases. This variation contributes to the graded (hierarchical) iron responses in vivo. We also consider the mechanisms of IRE-mRNA control. The binding of Fe(2+) to IRE-RNA facilitates IRP release and the binding of eukaryotic initiation factors (eIFs), which are proteins that assemble mRNA, ribosomes, and tRNA for translation. IRE-RNAs are riboregulators for the inorganic metabolic signal, Fe(2+); they control protein synthesis rates by changing the distribution of the iron metabolic mRNAs between complexes with enhancing eIFs and inhibitory IRPs. The regulation of mRNA in the cytoplasm of eukaryotic cells is a burgeoning frontier in biomedicine. The evolutionarily refined IRE-RNAs, although absent in plants and bacteria, constitute a model system for 3-D mRNAs in all organisms. IRE-mRNAs have yielded "proof of principle" data for small-molecule targeting of mRNA structures, demonstrating tremendous potential for chemical manipulation of mRNA and protein synthesis in living systems.


Ferrous Compounds/chemistry , RNA, Messenger/metabolism , Base Pairing , Crystallography, X-Ray , Ferritins/metabolism , Iron Regulatory Protein 1/chemistry , Iron Regulatory Protein 1/metabolism , Kinetics , Nucleic Acid Conformation , Protein Binding , Protein Structure, Tertiary , RNA, Messenger/chemistry , Ribosomes/metabolism
17.
Biochem J ; 430(2): 315-24, 2010 Sep 01.
Article En | MEDLINE | ID: mdl-20569198

In its aconitase-inactive form, IRP-1 (iron regulatory protein-1)/cytosolic aconitase binds to the IRE (iron-responsive element) of several mRNAs to effect post-transcriptional regulation. We have shown previously that IRP-1 has ATPase activity and that binding of ATP suppresses the IRP-1/IRE interaction. In the present study, we characterize the binding activity further. Binding is observed with both [alpha-32P]ATP and [alpha-32P]ADP, but not with [gamma-32P]ATP. Recombinant IRP-1 binds approximately two molecules of ATP, and positive co-operativity is observed with a Hill coefficient of 1.67+/-0.36 (EC50=44 microM) commencing at 1 microM ATP. Similar characteristics are observed with both apoprotein and the aconitase form. On binding, ATP is hydrolysed to ADP, and similar binding parameters and co-operativity are seen with ADP, suggesting that ATP hydrolysis is not rate limiting in product formation. The non-hydrolysable analogue AMP-PNP (adenosine 5'-[beta,gamma-imido]triphosphate) does not induce co-operativity. Upon incubation of IRP-1 with increasing concentrations of ATP or ADP, the protein migrates more slowly on agarose gel electrophoresis, and there is a shift in the CD spectrum. In this new state, adenosine nucleotide binding is competed for by other nucleotides (CTP, GTP and AMP-PNP), although ATP and ADP, but not the other nucleotides, partially stabilize the protein against spontaneous loss of aconitase activity when incubated at 37 degrees C. A mutant IRP-1(C437S) lacking aconitase activity shows only one ATP-binding site and lacks co-operativity. It has increased IRE-binding capacity and lower ATPase activity (Km=75+/-17 nmol/min per mg of protein) compared with the wild-type protein (Km=147+/-48 nmol/min per mg of protein). Under normal cellular conditions, it is predicted that ATP/ADP will maintain IRP-1 in a non-IRE-binding state.


Adenosine Diphosphate/chemistry , Adenosine Triphosphate/chemistry , Iron Regulatory Protein 1/chemistry , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Humans , Hydrolysis , Iron/metabolism , Iron Regulatory Protein 1/genetics , Iron Regulatory Protein 1/metabolism , Kinetics , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
18.
Curr Opin Struct Biol ; 18(1): 106-11, 2008 Feb.
Article En | MEDLINE | ID: mdl-18261896

Iron homeostasis in animal cells is controlled post-transcriptionally by the iron regulatory proteins IRP1 and IRP2. IRP1 can assume two different functions in the cell, depending on conditions. During iron scarcity or oxidative stress, IRP1 binds to mRNA stem-loop structures called iron responsive elements (IREs) to modulate the translation of iron metabolism genes. In iron-rich conditions, IRP1 binds an iron-sulfur cluster to function as a cytosolic aconitase. This functional duality of IRP1 connects the translational control of iron metabolizing proteins to cellular iron levels. The recently determined structures of IRP1 in both functional states reveal the large-scale conformational changes required for these mutually exclusive roles, providing new insights into the mechanisms of IRP1 interconversion and ligand binding.


Iron Regulatory Protein 1/chemistry , Iron Regulatory Protein 1/metabolism , Iron/metabolism , Aconitate Hydratase/chemistry , Aconitate Hydratase/metabolism , Animals , Base Sequence , Binding Sites , Humans , Models, Biological , Models, Molecular , Molecular Sequence Data , Nucleic Acid Conformation , Protein Conformation , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism
19.
BMC Mol Biol ; 9: 15, 2008 Jan 28.
Article En | MEDLINE | ID: mdl-18226225

BACKGROUND: Iron regulatory protein 2 (IRP2), a post-transcriptional regulator of cellular iron metabolism, undergoes iron-dependent degradation via the ubiquitin-proteasome pathway. A stretch of 73 amino acids within the N-terminal domain 1 of the protein was reported to function as an iron sensor. However, mutants lacking this fragment remain sensitive to degradation in iron-replete cells. RESULTS: To identify elements within IRP2 involved in the control of its stability, we undertook a systematic mutagenesis approach. Truncated versions of IRP2 were expressed in H1299 cells and analyzed for their response to iron. Deletion mutants lacking the entire C-terminal domain 4 (amino acids 719-963) of IRP2 remained stable following iron treatments. Moreover, the replacement of domain 4 of IRP1 with the corresponding region of IRP2 sensitized the chimerical IRP11-3/IRP24 protein to iron-dependent degradation, while the reverse manipulation gave rise to a stable chimerical IRP21-3/IRP14 protein. The deletion of just 26 or 34 C-terminal amino acids stabilized IRP2 against iron. However, the fusion of C-terminal IRP2 fragments to luciferase failed to sensitize the indicator protein for degradation in iron-loaded cells. CONCLUSION: Our data suggest that the C-terminus of IRP2 contains elements that are necessary but not sufficient for iron-dependent degradation. The functionality of these elements depends upon the overall IRP structure.


Iron Regulatory Protein 2/chemistry , Iron/metabolism , Proteasome Endopeptidase Complex/metabolism , Cell Line, Tumor , Humans , Iron Regulatory Protein 1/chemistry , Iron Regulatory Protein 1/genetics , Iron Regulatory Protein 1/metabolism , Iron Regulatory Protein 2/genetics , Iron Regulatory Protein 2/metabolism , Mutation , Protein Structure, Tertiary , Ubiquitination
20.
Plant Cell Environ ; 31(1): 144-58, 2008 Jan.
Article En | MEDLINE | ID: mdl-18004982

The maize (Zea mays L.) stem pulvinus is a disc of tissue located apical to each node that functions to return a tipped stem to a more upright position via increased cell elongation on its lower side. We investigated the possibility that reactive oxygen species (ROS) and hydrogen peroxide (H2O2), in particular, are involved in the gravitropic response of the pulvinus prior to initiation of the growth response by employing the cytochemical stain 3,3'-diaminobenzidine (DAB). DAB polymers were found in the bundle sheath cells of gravistimulated pulvini in association with amyloplasts after 1 min of gravistimulation, and the signal spread throughout the cytosol of these cells by 30 min. Furthermore, treatment of maize stem explants containing pulvini with 1 mm H2O2 on their upper sides caused reversal of bending polarity. Similar, though less dramatic, results were obtained via application of 1 mm ascorbic acid to the lower side of the explants. In addition, we determined that a maize cytoplasmic aconitase/iron regulatory protein 1 (IRP1) homolog is up-regulated in the pulvinus bundle sheath cells after gravistimulation using suppressive subtractive hybridization PCR (SSH PCR), real-time RT-PCR and in situ hybridization. Although we do not yet know the role of the IRP1 homolog in the pulvinus, the protein is known to be a redox sensor in other systems. Collectively, our results point to an increase in ROS quite early in the gravitropic signalling pathway and its possible role in determining the direction of bending of the pulvini. We speculate that an ROS burst may serve to link the physical phenomenon of amyloplast sedimentation to the changes in cellular biochemistry and gene expression that facilitate directional growth.


Gene Expression Regulation, Plant/physiology , Gravitropism/physiology , Iron Regulatory Protein 1/metabolism , Reactive Oxygen Species/metabolism , Zea mays/growth & development , Zea mays/metabolism , Ascorbic Acid/pharmacology , Hydrogen Peroxide/pharmacology , Iron Regulatory Protein 1/chemistry , Iron Regulatory Protein 1/genetics , Plant Proteins/genetics , Plant Proteins/metabolism , Plant Stems/cytology , RNA, Messenger/metabolism , Resins, Plant/metabolism , Time Factors , Zea mays/drug effects
...