Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 175
Filter
1.
Nat Commun ; 12(1): 6964, 2021 11 29.
Article in English | MEDLINE | ID: mdl-34845214

ABSTRACT

Many proteins are molecular machines, whose function is dependent on multiple conformational changes that are initiated and tightly controlled through biochemical stimuli. Their mechanistic understanding calls for spectroscopy that can probe simultaneously such structural coordinates. Here we present two-colour fluorescence microscopy in combination with photoinduced electron transfer (PET) probes as a method that simultaneously detects two structural coordinates in single protein molecules, one colour per coordinate. This contrasts with the commonly applied resonance energy transfer (FRET) technique that requires two colours per coordinate. We demonstrate the technique by directly and simultaneously observing three critical structural changes within the Hsp90 molecular chaperone machinery. Our results reveal synchronicity of conformational motions at remote sites during ATPase-driven closure of the Hsp90 molecular clamp, providing evidence for a cooperativity mechanism in the chaperone's catalytic cycle. Single-molecule PET fluorescence microscopy opens up avenues in the multi-dimensional exploration of protein dynamics and allosteric mechanisms.


Subject(s)
HSP90 Heat-Shock Proteins/chemistry , Optical Imaging/methods , Saccharomyces cerevisiae/genetics , Single Molecule Imaging/methods , Adenylyl Imidodiphosphate/chemistry , Adenylyl Imidodiphosphate/metabolism , Cloning, Molecular , Color , Electron Transport , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Light , Models, Molecular , Photochemical Processes , Point Mutation , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/metabolism
2.
Biochem Biophys Res Commun ; 557: 187-191, 2021 06 11.
Article in English | MEDLINE | ID: mdl-33872987

ABSTRACT

Human ATP-binding cassette transporter 8 of subfamily B (hABCB8) is an ABC transporter that located in the inner membrane of mitochondria. The ABCB8 is involved in the maturation of Fe-S and protects the heart from oxidative stress. Here, we present the cryo-EM structure of human ABCB8 binding with AMPPNP in inward-facing conformation with resolution of 4.1 Å. hABCB8 shows an open-inward conformation when ATP is bound. Unexpectedly, cholesterol molecules were identified in the transmembrane domain of hABCB8. Our results provide structural basis for the transport mechanism of the ABC transporter in mitochondria.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 1/chemistry , Adenosine Triphosphate/chemistry , Adenylyl Imidodiphosphate/chemistry , ATP Binding Cassette Transporter, Subfamily G, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism , Binding Sites , Cholesterol/chemistry , Cryoelectron Microscopy , Gene Expression , Membrane Transport Proteins/chemistry , Mitochondria/chemistry , Mitochondria/metabolism , Models, Molecular , Protein Binding , Protein Conformation , Protein Domains , Recombinant Proteins
3.
Nat Commun ; 12(1): 828, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33547294

ABSTRACT

The co-chaperone p23 is a central part of the Hsp90 machinery. It stabilizes the closed conformation of Hsp90, inhibits its ATPase and is important for client maturation. Yet, how this is achieved has remained enigmatic. Here, we show that a tryptophan residue in the proximal region of the tail decelerates the ATPase by allosterically switching the conformation of the catalytic loop in Hsp90. We further show by NMR spectroscopy that the tail interacts with the Hsp90 client binding site via a conserved helix. This helical motif in the p23 tail also binds to the client protein glucocorticoid receptor (GR) in the free and Hsp90-bound form. In vivo experiments confirm the physiological importance of ATPase modulation and the role of the evolutionary conserved helical motif for GR activation in the cellular context.


Subject(s)
Adenylyl Imidodiphosphate/chemistry , HSP90 Heat-Shock Proteins/chemistry , Molecular Chaperones/chemistry , Prostaglandin-E Synthases/chemistry , Receptors, Glucocorticoid/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/chemistry , Adenylyl Imidodiphosphate/metabolism , Amino Acid Sequence , Binding Sites , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Humans , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Molecular Dynamics Simulation , Mutation , Nuclear Magnetic Resonance, Biomolecular , Prostaglandin-E Synthases/genetics , Prostaglandin-E Synthases/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid
4.
Protein Pept Lett ; 28(5): 481-488, 2021.
Article in English | MEDLINE | ID: mdl-33143608

ABSTRACT

BACKGROUND: Activation of mitogen-activated protein kinases (MAPKs) is regulated by a phosphorylation cascade comprising three kinases, MAPK kinase kinase (MAP3K), MAPK kinase (MAP2K), and MAPK. MAP2K1 and MAPK2K2, also known as MEK1 and MEK2, activate ERK1 and ERK2. The structure of the MAPK signaling cascade has been studied, but high-resolution structural studies of MAP2Ks have often focused on kinase domains or docking sites, but not on full-length proteins. OBJECTIVE: To understand the conformational dynamics of MEK1. METHODS: Full-length MEK1 was purified from Escherichia coli (BL21), and its conformational dynamics were analyzed using hydrogen/deuterium exchange mass spectrometry (HDX-MS). The effects of ATP binding were examined by co-incubating MEK1 and adenylyl-imidodiphosphate (AMP- PNP), a non-hydrolysable ATP analog. RESULTS: MEK1 exhibited mixed EX1/EX2 HDX kinetics within the N-terminal tail through ß1, αI, and the C-terminal helix. AMP-PNP binding was found to reduce conformational dynamics within the glycine-rich loop and regions near the DFG motif, along with the activation lip. CONCLUSION: We report for the first time that MEK1 has regions that slowly change its folded and unfolded states (mixed EX1/EX2 kinetics) and also report the conformational effects of ATP-binding to MEK1.


Subject(s)
Adenylyl Imidodiphosphate/chemistry , Hydrogen Deuterium Exchange-Mass Spectrometry , MAP Kinase Kinase 1/chemistry , Humans , Kinetics , Protein Domains , Protein Structure, Secondary , Recombinant Proteins
5.
Molecules ; 25(22)2020 Nov 12.
Article in English | MEDLINE | ID: mdl-33198135

ABSTRACT

Nucleoside triphosphates (NTPs) are used as chemical energy source in a variety of cell systems. Structural snapshots along the NTP hydrolysis reaction coordinate are typically obtained by adding stable, nonhydrolyzable adenosine triphosphate (ATP) -analogues to the proteins, with the goal to arrest a state that mimics as closely as possible a physiologically relevant state, e.g., the pre-hydrolytic, transition and post-hydrolytic states. We here present the lessons learned on two distinct ATPases on the best use and unexpected pitfalls observed for different analogues. The proteins investigated are the bacterial DnaB helicase from Helicobacter pylori and the multidrug ATP binding cassette (ABC) transporter BmrA from Bacillus subtilis, both belonging to the same division of P-loop fold NTPases. We review the magnetic-resonance strategies which can be of use to probe the binding of the ATP-mimics, and present carbon-13, phosphorus-31, and vanadium-51 solid-state nuclear magnetic resonance (NMR) spectra of the proteins or the bound molecules to unravel conformational and dynamic changes upon binding of the ATP-mimics. Electron paramagnetic resonance (EPR), and in particular W-band electron-electron double resonance (ELDOR)-detected NMR, is of complementary use to assess binding of vanadate. We discuss which analogues best mimic the different hydrolysis states for the DnaB helicase and the ABC transporter BmrA. These might be relevant also to structural and functional studies of other NTPases.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Adenosine Triphosphate/chemistry , Bacillus subtilis/enzymology , DnaB Helicases/metabolism , Helicobacter pylori/enzymology , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenylyl Imidodiphosphate/chemistry , Aluminum Compounds/chemistry , Bacterial Proteins/metabolism , Electron Spin Resonance Spectroscopy , Electrons , Fluorides/chemistry , Hydrolysis , Magnetic Resonance Spectroscopy , Protein Conformation
6.
Nucleic Acids Res ; 48(1): 200-211, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31665475

ABSTRACT

Escherichia coli replication initiator protein DnaA binds ATP with high affinity but the amount of ATP required to initiate replication greatly exceeds the amount required for binding. Previously, we showed that ATP-DnaA, not ADP-DnaA, undergoes a conformational change at the higher nucleotide concentration, which allows DnaA oligomerization at the replication origin but the association state remains unclear. Here, we used Small Angle X-ray Scattering (SAXS) to investigate oligomerization of DnaA in solution. Whereas ADP-DnaA was predominantly monomeric, AMP-PNP-DnaA (a non-hydrolysable ATP-analog bound-DnaA) was oligomeric, primarily dimeric. Functional studies using DnaA mutants revealed that DnaA(H136Q) is defective in initiating replication in vivo. The mutant retains high-affinity ATP binding, but was defective in producing replication-competent initiation complexes. Docking of ATP on a structure of E. coli DnaA, modeled upon the crystallographic structure of Aquifex aeolicus DnaA, predicts a hydrogen bond between ATP and imidazole ring of His136, which is disrupted when Gln is present at position 136. SAXS performed on AMP-PNP-DnaA (H136Q) indicates that the protein has lost its ability to form oligomers. These results show the importance of high ATP in DnaA oligomerization and its dependence on the His136 residue.


Subject(s)
Adenosine Diphosphate/chemistry , Adenosine Triphosphate/chemistry , Bacterial Proteins/chemistry , DNA Replication , DNA, Bacterial/genetics , DNA-Binding Proteins/chemistry , Escherichia coli/genetics , Gene Expression Regulation, Bacterial , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Adenylyl Imidodiphosphate/chemistry , Adenylyl Imidodiphosphate/metabolism , Aquifex , Bacteria/genetics , Bacteria/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Chromosomes, Bacterial/chemistry , Chromosomes, Bacterial/metabolism , Crystallography, X-Ray , DNA, Bacterial/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dimerization , Escherichia coli/metabolism , Hydrogen Bonding , Molecular Docking Simulation , Mutation , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Replication Origin , Thermodynamics
7.
Acta Crystallogr F Struct Biol Commun ; 75(Pt 9): 599-607, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31475927

ABSTRACT

Hygromycin B (HygB) is one of the aminoglycoside antibiotics, and it is widely used as a reagent in molecular-biology experiments. Two kinases are known to inactivate HygB through phosphorylation: aminoglycoside 7''-phosphotransferase-Ia [APH(7'')-Ia] from Streptomyces hygroscopicus and aminoglycoside 4-phosphotransferase-Ia [APH(4)-Ia] from Escherichia coli. They phosphorylate the hydroxyl groups at positions 7'' and 4 of the HygB molecule, respectively. Previously, the crystal structure of APH(4)-Ia was reported as a ternary complex with HygB and 5'-adenylyl-ß,γ-imidodiphosphate (AMP-PNP). To investigate the differences in the substrate-recognition mechanism between APH(7'')-Ia and APH(4)-Ia, the crystal structure of APH(7'')-Ia complexed with HygB is reported. The overall structure of APH(7'')-Ia is similar to those of other aminoglycoside phosphotransferases, including APH(4)-Ia, and consists of an N-terminal lobe (N-lobe) and a C-terminal lobe (C-lobe). The latter also comprises a core and a helical domain. Accordingly, the APH(7'')-Ia and APH(4)-Ia structures fit globally when the structures are superposed at three catalytically important conserved residues, His, Asp and Asn, in the Brenner motif, which is conserved in aminoglycoside phosphotransferases as well as in eukaryotic protein kinases. On the other hand, the phosphorylated hydroxyl groups of HygB in both structures come close to the Asp residue, and the HygB molecules in each structure lie in opposite directions. These molecules were held by the helical domain in the C-lobe, which exhibited structural differences between the two kinases. Furthermore, based on the crystal structures of APH(7'')-Ia and APH(4)-Ia, some mutated residues in their thermostable mutants reported previously were located at the same positions in the two enzymes.


Subject(s)
Anti-Bacterial Agents/chemistry , Hygromycin B/chemistry , Kanamycin Kinase/chemistry , Streptomyces/enzymology , Adenylyl Imidodiphosphate/chemistry , Amino Acid Motifs/genetics , Aminoglycosides/chemistry , Binding Sites , Catalysis , Crystallography, X-Ray , Escherichia coli/metabolism , Kanamycin Kinase/genetics , Kanamycin Kinase/metabolism , Phosphorylation , Protein Domains , Substrate Specificity
8.
Structure ; 27(4): 579-589.e5, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30744994

ABSTRACT

Despite sharing common features, previous studies have shown that gyrases from different species have been modified throughout evolution to modulate their properties. Here, we report two crystal structures of Mycobacterium tuberculosis DNA gyrase, an apo and AMPPNP-bound form at 2.6-Å and 3.3-Å resolution, respectively. These structures provide high-resolution structural data on the quaternary organization and interdomain connections of a gyrase (full-length GyrB-GyrA57)2 thus providing crucial inputs on this essential drug target. Together with small-angle X-ray scattering studies, they revealed an "extremely open" N-gate state, which persists even in the DNA-free gyrase-AMPPNP complex and an unexpected connection between the ATPase and cleavage core domains mediated by two Corynebacteriales-specific motifs, respectively the C-loop and DEEE-loop. We show that the C-loop participates in the stabilization of this open conformation, explaining why this gyrase has a lower ATPase activity. Our results image a conformational state which might be targeted for drug discovery.


Subject(s)
Adenosine Triphosphatases/genetics , Adenosine Triphosphate/chemistry , Apoproteins/chemistry , Corynebacterium/chemistry , DNA Gyrase/chemistry , Mycobacterium tuberculosis/chemistry , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Adenylyl Imidodiphosphate/chemistry , Adenylyl Imidodiphosphate/metabolism , Amino Acid Sequence , Apoproteins/genetics , Apoproteins/metabolism , Binding Sites , Cloning, Molecular , Corynebacterium/enzymology , Crystallography, X-Ray , DNA/chemistry , DNA/metabolism , DNA Gyrase/genetics , DNA Gyrase/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Kinetics , Models, Molecular , Mycobacterium tuberculosis/enzymology , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Structure, Quaternary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid
9.
Proc Natl Acad Sci U S A ; 116(10): 4265-4274, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30760599

ABSTRACT

We used cryo-electron microscopy (cryo-EM) to reconstruct actin filaments with bound AMPPNP (ß,γ-imidoadenosine 5'-triphosphate, an ATP analog, resolution 3.1 Å), ADP-Pi (ADP with inorganic phosphate, resolution 3.1 Å), or ADP (resolution 3.6 Å). Subunits in the three filaments have similar backbone conformations, so assembly rather than ATP hydrolysis or phosphate dissociation is responsible for their flattened conformation in filaments. Polymerization increases the rate of ATP hydrolysis by changing the positions of the side chains of Q137 and H161 in the active site. Flattening during assembly also promotes interactions along both the long-pitch and short-pitch helices. In particular, conformational changes in subdomain 3 open up multiple favorable interactions with the DNase-I binding loop in subdomain 2 of the adjacent subunit. Subunits at the barbed end of the filament are likely to be in this favorable conformation, while monomers are not. This difference explains why filaments grow faster at the barbed end than the pointed end. When phosphate dissociates from ADP-Pi-actin through a backdoor channel, the conformation of the C terminus changes so it distorts the DNase binding loop, which allows cofilin binding, and a network of interactions among S14, H73, G74, N111, R177, and G158 rearranges to open the phosphate release site.


Subject(s)
Actin Cytoskeleton/chemistry , Actins/chemistry , Adenylyl Imidodiphosphate/chemistry , Avian Proteins/chemistry , Polymerization , Actin Cytoskeleton/ultrastructure , Animals , Catalytic Domain , Chickens , Cryoelectron Microscopy
10.
Mol Cell ; 73(4): 738-748.e9, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30595437

ABSTRACT

A class of translation inhibitors, exemplified by the natural product rocaglamide A (RocA), isolated from Aglaia genus plants, exhibits antitumor activity by clamping eukaryotic translation initiation factor 4A (eIF4A) onto polypurine sequences in mRNAs. This unusual inhibitory mechanism raises the question of how the drug imposes sequence selectivity onto a general translation factor. Here, we determined the crystal structure of the human eIF4A1⋅ATP analog⋅RocA⋅polypurine RNA complex. RocA targets the "bi-molecular cavity" formed characteristically by eIF4A1 and a sharply bent pair of consecutive purines in the RNA. Natural amino acid substitutions found in Aglaia eIF4As changed the cavity shape, leading to RocA resistance. This study provides an example of an RNA-sequence-selective interfacial inhibitor fitting into the space shaped cooperatively by protein and RNA with specific sequences.


Subject(s)
Benzofurans/metabolism , Eukaryotic Initiation Factor-4A/metabolism , Protein Biosynthesis , Protein Synthesis Inhibitors/metabolism , RNA/metabolism , Ribosomes/metabolism , Adenylyl Imidodiphosphate/chemistry , Adenylyl Imidodiphosphate/metabolism , Aglaia/chemistry , Aglaia/genetics , Aglaia/metabolism , Amino Acid Substitution , Benzofurans/chemistry , Benzofurans/isolation & purification , Benzofurans/pharmacology , Binding Sites , Drug Resistance/genetics , Eukaryotic Initiation Factor-4A/chemistry , Eukaryotic Initiation Factor-4A/genetics , HEK293 Cells , Humans , Models, Molecular , Molecular Structure , Mutation , Plant Proteins/chemistry , Plant Proteins/genetics , Plant Proteins/metabolism , Protein Binding , Protein Biosynthesis/drug effects , Protein Biosynthesis/genetics , Protein Interaction Domains and Motifs , Protein Synthesis Inhibitors/chemistry , Protein Synthesis Inhibitors/isolation & purification , Protein Synthesis Inhibitors/pharmacology , RNA/chemistry , Ribosomes/chemistry , Ribosomes/drug effects , Ribosomes/genetics , Structure-Activity Relationship
11.
Protein Sci ; 28(3): 524-532, 2019 03.
Article in English | MEDLINE | ID: mdl-30461091

ABSTRACT

Vaccinia-related kinase 1 (VRK1), a serine/threonine mitotic kinase, is widely over-expressed in dividing cells and regarded as a cancer drug target primarily due to its function as an early response gene in cell proliferation. However, the mechanism of VRK1 phosphorylation and substrate activation is not well understood. More importantly even the molecular basis of VRK1 interaction with its cofactor, adenosine triphosphate (ATP), is unavailable to-date. As designing specific inhibitors remains to be the major challenge in kinase research, such a molecular understanding will enable us to design ATP-competitive specific inhibitors of VRK1. Here we report the molecular characterization of VRK1 in complex with AMP-PNP, a non-hydrolyzable ATP-analog, using NMR titration followed by the co-crystal structure determined upto 2.07 Å resolution. We also carried out the structural comparison of the AMP-PNP bound-form with its apo and inhibitor-bound counterparts, which has enabled us to present our rationale toward designing VRK1-specific inhibitors.


Subject(s)
Adenosine Triphosphate/metabolism , Adenylyl Imidodiphosphate/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenylyl Imidodiphosphate/chemistry , Crystallography, X-Ray , Enzyme Inhibitors/pharmacology , Humans , Hydrolysis , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/chemistry , Models, Molecular , Protein Conformation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/chemistry
12.
J Cell Biol ; 217(12): 4164-4183, 2018 12 03.
Article in English | MEDLINE | ID: mdl-30297389

ABSTRACT

Kinesin-1, the founding member of the kinesin superfamily of proteins, is known to use only a subset of microtubules for transport in living cells. This biased use of microtubules is proposed as the guidance cue for polarized transport in neurons, but the underlying mechanisms are still poorly understood. Here, we report that kinesin-1 binding changes the microtubule lattice and promotes further kinesin-1 binding. This high-affinity state requires the binding of kinesin-1 in the nucleotide-free state. Microtubules return to the initial low-affinity state by washing out the binding kinesin-1 or by the binding of non-hydrolyzable ATP analogue AMPPNP to kinesin-1. X-ray fiber diffraction, fluorescence speckle microscopy, and second-harmonic generation microscopy, as well as cryo-EM, collectively demonstrated that the binding of nucleotide-free kinesin-1 to GDP microtubules changes the conformation of the GDP microtubule to a conformation resembling the GTP microtubule.


Subject(s)
Kinesins , Microtubules , Adenylyl Imidodiphosphate/chemistry , Adenylyl Imidodiphosphate/pharmacology , Animals , Biological Transport, Active , Chlorocebus aethiops , Dogs , Guanosine Diphosphate/chemistry , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/chemistry , Guanosine Triphosphate/metabolism , HeLa Cells , Humans , Kinesins/chemistry , Kinesins/metabolism , Madin Darby Canine Kidney Cells , Microtubules/chemistry , Microtubules/metabolism , Vero Cells
13.
Acta Crystallogr F Struct Biol Commun ; 74(Pt 4): 205-213, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29633968

ABSTRACT

Recent studies suggest a link between infection by Zika virus (ZIKV) and the development of neurological complications. The lack of ZIKV-specific therapeutics has alarmed healthcare professionals worldwide. Here, crystal structures of apo and AMPPNP- and Mn2+-bound forms of the essential helicase of ZIKV refined to 1.78 and 1.3 Šresolution, respectively, are reported. The structures reveal a conserved trimodular topology of the helicase. ATP and Mn2+ are tethered between two RecA-like domains by conserved hydrogen-bonding interactions. The binding of ligands induces the movement of backbone Cα and side-chain atoms. Numerous solvent molecules are observed in the vicinity of the AMPPNP, suggesting a role in catalysis. These high-resolution structures could be useful for the design of inhibitors targeting the helicase of ZIKV for the treatment of infections caused by ZIKV.


Subject(s)
RNA Helicases/chemistry , RNA, Viral/chemistry , Viral Nonstructural Proteins/chemistry , Zika Virus/enzymology , Adenosine Triphosphate/metabolism , Adenylyl Imidodiphosphate/chemistry , Adenylyl Imidodiphosphate/metabolism , Amino Acid Sequence , Crystallization , Crystallography, X-Ray , Manganese/chemistry , Manganese/metabolism , Models, Molecular , Protein Conformation , Protein Domains , RNA Helicases/metabolism , RNA, Viral/metabolism , Sequence Homology , Viral Nonstructural Proteins/metabolism
14.
Nat Nanotechnol ; 13(5): 386-391, 2018 05.
Article in English | MEDLINE | ID: mdl-29531331

ABSTRACT

Kinesin-1 is a nanoscale molecular motor that walks towards the fast-growing (plus) ends of microtubules, hauling molecular cargo to specific reaction sites in cells. Kinesin-driven transport is central to the self-organization of eukaryotic cells and shows great promise as a tool for nano-engineering 1 . Recent work hints that kinesin may also play a role in modulating the stability of its microtubule track, both in vitro2,3 and in vivo 4 , but the results are conflicting5-7 and the mechanisms are unclear. Here, we report a new dimension to the kinesin-microtubule interaction, whereby strong-binding state (adenosine triphosphate (ATP)-bound and apo) kinesin-1 motor domains inhibit the shrinkage of guanosine diphosphate (GDP) microtubules by up to two orders of magnitude and expand their lattice spacing by ~1.6%. Our data reveal an unexpected mechanism by which the mechanochemical cycles of kinesin and tubulin interlock, and so allow motile kinesins to influence the structure, stability and mechanics of their microtubule track.


Subject(s)
Guanosine Diphosphate/chemistry , Kinesins/chemistry , Microtubules/chemistry , Mutation, Missense , Adenylyl Imidodiphosphate/chemistry , Amino Acid Substitution , Animals , Guanosine Diphosphate/metabolism , Kinesins/genetics , Kinesins/metabolism , Microtubules/genetics , Microtubules/metabolism , Swine
15.
Phys Chem Chem Phys ; 20(5): 3054-3062, 2018 Jan 31.
Article in English | MEDLINE | ID: mdl-28721412

ABSTRACT

Ligand binding to a protein can stabilize it significantly against unfolding. The variation of the folding free energy, ΔΔG0, due to ligand binding can be derived from a simple reaction scheme involving exclusive binding to the native state. One obtains the following expression: , where Kd is the ligand dissociation constant and L is its concentration, R is the universal gas constant and T is the temperature. This expression has been shown to correctly describe experimental results on multiple proteins. In the current work we studied the effect of ligand binding on the stability of the multi-domain protein adenylate kinase from E. coli (AKE). Unfolding experiments were conducted using single-molecule FRET spectroscopy, which allowed us to directly obtain the fraction of unfolded protein in a model-free way from FRET efficiency histograms. Surprisingly, it was found that the effect of two inhibitors (Ap5A and AMPPNP) and a substrate (AMP) on the stability of AKE was much smaller than expected based on Kd values obtained independently using microscale thermophoresis. To shed light on this issue, we measured the Kd for Ap5A over a range of chemical denaturant concentrations where the protein is still folded. It was found that Kd increases dramatically over this range, likely due to the population of folding intermediates, whose binding to the ligand is much weaker than that of the native state. We propose that binding to folding intermediates may dominate the effect of ligands on the stability of multi-domain proteins, and could therefore have a strong impact on protein homeostasis in vivo.


Subject(s)
Adenylate Kinase/metabolism , Escherichia coli Proteins/metabolism , Ligands , Adenylate Kinase/chemistry , Adenylate Kinase/genetics , Adenylyl Imidodiphosphate/chemistry , Adenylyl Imidodiphosphate/metabolism , Circular Dichroism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Escherichia coli/enzymology , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Fluorescence Resonance Energy Transfer , Kinetics , Protein Binding , Protein Denaturation , Protein Folding , Protein Stability , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Substrate Specificity , Thermodynamics
16.
J Biol Chem ; 292(35): 14680-14694, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28701465

ABSTRACT

Kinesin motors play central roles in establishing and maintaining the mitotic spindle during cell division. Unlike most other kinesins, Cin8, a kinesin-5 motor in Saccharomyces cerevisiae, can move bidirectionally along microtubules, switching directionality according to biochemical conditions, a behavior that remains largely unexplained. To this end, we used biochemical rate and equilibrium constant measurements as well as cryo-electron microscopy methodologies to investigate the microtubule interactions of the Cin8 motor domain. These experiments unexpectedly revealed that, whereas Cin8 ATPase kinetics fell within measured ranges for kinesins (especially kinesin-5 proteins), approximately four motors can bind each αß-tubulin dimer within the microtubule lattice. This result contrasted with those observations on other known kinesins, which can bind only a single "canonical" site per tubulin dimer. Competition assays with human kinesin-5 (Eg5) only partially abrogated this behavior, indicating that Cin8 binds microtubules not only at the canonical site, but also one or more separate ("noncanonical") sites. Moreover, we found that deleting the large, class-specific insert in the microtubule-binding loop 8 reverts Cin8 to one motor per αß-tubulin in the microtubule. The novel microtubule-binding mode of Cin8 identified here provides a potential explanation for Cin8 clustering along microtubules and potentially may contribute to the mechanism for direction reversal.


Subject(s)
Kinesins/metabolism , Microtubules/enzymology , Models, Molecular , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Tubulin/metabolism , Adenosine Diphosphate/chemistry , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Adenylyl Imidodiphosphate/chemistry , Adenylyl Imidodiphosphate/metabolism , Amino Acid Substitution , Binding Sites , Binding, Competitive , Biocatalysis , Cryoelectron Microscopy , Crystallography, X-Ray , Gene Deletion , Humans , Kinesins/chemistry , Kinesins/genetics , Microtubules/chemistry , Microtubules/metabolism , Mutation , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Conformation , Protein Interaction Domains and Motifs , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Tubulin/chemistry
17.
Genes Dev ; 31(5): 481-492, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28373206

ABSTRACT

Walker-box partition systems are ubiquitous in nature and mediate the segregation of bacterial and archaeal DNA. Well-studied plasmid Walker-box partition modules require ParA, centromere-DNA, and a centromere-binding protein, ParB. In these systems, ParA-ATP binds nucleoid DNA and uses it as a substratum to deliver ParB-attached cargo DNA, and ParB drives ParA dynamics, allowing ParA progression along the nucleoid. How ParA-ATP binds nonspecific DNA and is regulated by ParB is unclear. Also under debate is whether ParA polymerizes on DNA to mediate segregation. Here we describe structures of key ParA segregation complexes. The ParA-ß,γ-imidoadenosine 5'-triphosphate (AMPPNP)-DNA structure revealed no polymers. Instead, ParA-AMPPNP dimerization creates a multifaceted DNA-binding surface, allowing it to preferentially bind high-density DNA regions (HDRs). DNA-bound ParA-AMPPNP adopts a dimer conformation distinct from the ATP sandwich dimer, optimized for DNA association. Our ParA-AMPPNP-ParB structure reveals that ParB binds at the ParA dimer interface, stabilizing the ATPase-competent ATP sandwich dimer, ultimately driving ParA DNA dissociation. Thus, the data indicate how harnessing a conformationally adaptive dimer can drive large-scale cargo movement without the requirement for polymers and suggest a segregation mechanism by which ParA-ATP dimers equilibrate to HDRs shown to be localized near cell poles of dividing chromosomes, thus mediating equipartition of attached ParB-DNA substrates.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Chromosome Segregation , DNA, Archaeal/chemistry , DNA, Bacterial/chemistry , Models, Molecular , Adenosine Triphosphatases/metabolism , Adenylyl Imidodiphosphate/chemistry , Adenylyl Imidodiphosphate/metabolism , Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Bacterial Proteins/genetics , Crystallization , DNA, Archaeal/metabolism , DNA, Bacterial/metabolism , Enzyme Activation , Protein Binding , Protein Multimerization , Protein Structure, Quaternary , Thermus thermophilus/chemistry , Thermus thermophilus/genetics , Thermus thermophilus/metabolism
18.
J Mol Biol ; 429(9): 1406-1423, 2017 05 05.
Article in English | MEDLINE | ID: mdl-28363677

ABSTRACT

The 90-kDa heat shock protein (Hsp90) chaperones the late folding steps of many protein kinases, transcription factors, and a diverse set of other protein clients not related in sequence and structure. Hsp90's interaction with clients appears to be coupled to a series of conformational changes. How these conformational changes contribute to its chaperone activity is currently unclear. Using crosslinking, hydrogen exchange mass spectrometry, and fluorescence experiments, we demonstrate here that the N-terminal domain of Hsp90 rotates by approximately 180° as compared to the crystal structure of yeast Hsp90 in complex with Sba1 and AMPPNP. Surprisingly, Aha1 but not Sba1 suppresses this rotation in the presence of AMPPNP but not in its absence. A minimum length of the largely unstructured linker between N-terminal and middle domain is necessary for this rotation, and interfering with the rotation strongly affects the interaction with Aha1 and the intrinsic and Aha1-stimulated ATPase activity. Surprisingly, suppression of the rotation only affects the activity of some clients and does not compromise yeast viability.


Subject(s)
HSP90 Heat-Shock Proteins/chemistry , HSP90 Heat-Shock Proteins/metabolism , Protein Folding , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Adenylyl Imidodiphosphate/chemistry , Adenylyl Imidodiphosphate/metabolism , Chaperonins/chemistry , Chaperonins/metabolism , Mass Spectrometry , Microbial Viability , Models, Molecular , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Protein Conformation , Saccharomyces cerevisiae/physiology , Spectrometry, Fluorescence
19.
Nat Struct Mol Biol ; 24(3): 300-308, 2017 03.
Article in English | MEDLINE | ID: mdl-28191894

ABSTRACT

The minichromosome maintenance complex (MCM) hexameric complex (Mcm2-7) forms the core of the eukaryotic replicative helicase. During G1 phase, two Cdt1-Mcm2-7 heptamers are loaded onto each replication origin by the origin-recognition complex (ORC) and Cdc6 to form an inactive MCM double hexamer (DH), but the detailed loading mechanism remains unclear. Here we examine the structures of the yeast MCM hexamer and Cdt1-MCM heptamer from Saccharomyces cerevisiae. Both complexes form left-handed coil structures with a 10-15-Å gap between Mcm5 and Mcm2, and a central channel that is occluded by the C-terminal domain winged-helix motif of Mcm5. Cdt1 wraps around the N-terminal regions of Mcm2, Mcm6 and Mcm4 to stabilize the whole complex. The intrinsic coiled structures of the precursors provide insights into the DH formation, and suggest a spring-action model for the MCM during the initial origin melting and the subsequent DNA unwinding.


Subject(s)
Cell Cycle Proteins/chemistry , DNA-Binding Proteins/chemistry , Minichromosome Maintenance Proteins/chemistry , Protein Multimerization , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/metabolism , Adenylyl Imidodiphosphate/chemistry , Amino Acid Motifs , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/ultrastructure , Cryoelectron Microscopy , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/ultrastructure , Minichromosome Maintenance Proteins/metabolism , Minichromosome Maintenance Proteins/ultrastructure , Models, Molecular , Protein Domains , Protein Structure, Secondary , Protein Subunits/chemistry , Protein Subunits/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/ultrastructure , Zinc Fingers
20.
Proteins ; 85(1): 117-124, 2017 01.
Article in English | MEDLINE | ID: mdl-27802586

ABSTRACT

The heart-specific isoform of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (PFKFB2) is an important regulator of glycolytic flux in cardiac cells. Here, we present the crystal structures of two PFKFB2 orthologues, human and bovine, at resolutions of 2.0 and 1.8 Å, respectively. Citrate, a TCA cycle intermediate and well-known inhibitor of PFKFB2, co-crystallized in the 2-kinase domains of both orthologues, occupying the fructose-6-phosphate binding-site and extending into the γ-phosphate binding pocket of ATP. This steric and electrostatic occlusion of the γ-phosphate site by citrate proved highly consequential to the binding of co-complexed ATP analogues. The bovine structure, which co-crystallized with ADP, closely resembled the overall structure of other PFKFB isoforms, with ADP mimicking the catalytic binding mode of ATP. The human structure, on the other hand, co-complexed with AMPPNP, which, unlike ADP, contains a γ-phosphate. The presence of this γ-phosphate made adoption of the catalytic ATP binding mode impossible for AMPPNP, forcing the analogue to bind atypically with concomitant conformational changes to the ATP binding-pocket. Inhibition kinetics were used to validate the structural observations, confirming citrate's inhibition mechanism as competitive for F6P and noncompetitive for ATP. Together, these structural and kinetic data establish a molecular basis for citrate's negative feed-back loop of the glycolytic pathway via PFKFB2. Proteins 2016; 85:117-124. © 2016 Wiley Periodicals, Inc.


Subject(s)
Adenosine Diphosphate/chemistry , Adenosine Triphosphate/chemistry , Citric Acid/chemistry , Fructosephosphates/chemistry , Isoenzymes/chemistry , Myocardium/chemistry , Phosphofructokinase-2/chemistry , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Adenylyl Imidodiphosphate/chemistry , Adenylyl Imidodiphosphate/metabolism , Animals , Binding Sites , Cattle , Citric Acid/metabolism , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Fructosephosphates/metabolism , Gene Expression , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , Models, Molecular , Myocardium/enzymology , Phosphofructokinase-2/genetics , Phosphofructokinase-2/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Species Specificity , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...