Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Mol Cell Biol ; 43(5): 223-240, 2023.
Article in English | MEDLINE | ID: mdl-37154023

ABSTRACT

Glutamyl-prolyl-tRNA synthetase 1 (EPRS1) is known to associated with fibrosis through its catalytic activity to produce prolyl-tRNA. Although its catalytic inhibitor halofuginone (HF) has been known to inhibit the TGF-ß pathway as well as to reduce prolyl-tRNA production for the control of fibrosis, the underlying mechanism how EPRS1 regulates the TGF-ß pathway was not fully understood. Here, we show a noncatalytic function of EPRS1 in controlling the TGF-ß pathway and hepatic stellate cell activation via its interaction with TGF-ß receptor I (TßRI). Upon stimulation with TGF-ß, EPRS1 is phosphorylated by TGF-ß-activated kinase 1 (TAK1), leading to its dissociation from the multi-tRNA synthetase complex and subsequent binding with TßRI. This interaction increases the association of TßRI with SMAD2/3 while decreases that of TßRI with SMAD7. Accordingly, EPRS1 stabilizes TßRI by preventing the ubiquitin-mediated degradation of TßRI. HF disrupts the interaction between EPRS1 and TßRI, and reduces TßRI protein levels, leading to inhibition of the TGF-ß pathway. In conclusion, this work suggests the novel function of EPRS1 involved in the development of fibrosis by regulating the TGF-ß pathway and the antifibrotic effects of HF by controlling both of EPRS1 functions.


Subject(s)
Hepatic Stellate Cells , Receptors, Transforming Growth Factor beta , Humans , Hepatic Stellate Cells/metabolism , Receptor, Transforming Growth Factor-beta Type I/metabolism , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/metabolism , Fibrosis , Signal Transduction/physiology
2.
EMBO Mol Med ; 15(7): e16940, 2023 07 10.
Article in English | MEDLINE | ID: mdl-37212275

ABSTRACT

Prolyl-tRNA synthetase 1 (PARS1) has attracted much interest in controlling pathologic accumulation of collagen containing high amounts of proline in fibrotic diseases. However, there are concerns about its catalytic inhibition for potential adverse effects on global protein synthesis. We developed a novel compound, DWN12088, whose safety was validated by clinical phase 1 studies, and therapeutic efficacy was shown in idiopathic pulmonary fibrosis model. Structural and kinetic analyses revealed that DWN12088 binds to catalytic site of each protomer of PARS1 dimer in an asymmetric mode with different affinity, resulting in decreased responsiveness at higher doses, thereby expanding safety window. The mutations disrupting PARS1 homodimerization restored the sensitivity to DWN12088, validating negative communication between PARS1 promoters for the DWN12088 binding. Thus, this work suggests that DWN12088, an asymmetric catalytic inhibitor of PARS1 as a novel therapeutic agent against fibrosis with enhanced safety.


Subject(s)
Amino Acyl-tRNA Synthetases , Humans , Amino Acyl-tRNA Synthetases/chemistry , Amino Acyl-tRNA Synthetases/genetics , Amino Acyl-tRNA Synthetases/metabolism , Fibrosis , Proline/genetics , Proline/metabolism , Protein Biosynthesis
3.
IUCrJ ; 10(Pt 2): 220-232, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36862488

ABSTRACT

PsEst3, a psychrophilic esterase obtained from Paenibacillus sp. R4, which was isolated from the permafrost of Alaska, exhibits relatively high activity at low temperatures. Here, crystal structures of PsEst3 complexed with various ligands were generated and studied at atomic resolution, and biochemical studies were performed to analyze the structure-function relationship of PsEst3. Certain unique characteristics of PsEst3 distinct from those of other classes of lipases/esterases were identified. Firstly, PsEst3 contains a conserved GHSRA/G pentapeptide sequence in the GxSxG motif around the nucleophilic serine. Additionally, it contains a conserved HGFR/K consensus sequence in the oxyanion hole, which is distinct from that in other lipase/esterase families, as well as a specific domain composition (for example a helix-turn-helix motif) and a degenerative lid domain that exposes the active site to the solvent. Secondly, the electrostatic potential of the active site in PsEst3 is positive, which may cause unintended binding of negatively charged chemicals in the active site. Thirdly, the last residue of the oxyanion hole-forming sequence, Arg44, separates the active site from the solvent by sealing the acyl-binding pocket, suggesting that PsEst3 is an enzyme that is customized to sense an unidentified substrate that is distinct from those of classical lipases/esterases. Collectively, this evidence strongly suggests that PsEst3 belongs to a distinct family of esterases.


Subject(s)
Esterases , Paenibacillus , Esterases/genetics , Paenibacillus/genetics , Lipase/genetics , Alaska , Solvents
4.
Molecules ; 27(9)2022 May 04.
Article in English | MEDLINE | ID: mdl-35566285

ABSTRACT

The stress-responsive, SK5 subclass, dehydrin gene, CaDHN, has been identified from the Arctic mouse-ear chickweed Cerastium arcticum. CaDHN contains an unusual single cysteine residue (Cys143), which can form intermolecular disulfide bonds. Mutational analysis and a redox experiment confirmed that the dimerization of CaDHN was the result of an intermolecular disulfide bond between the cysteine residues. The biochemical and physiological functions of the mutant C143A were also investigated by in vitro and in vivo assays using yeast cells, where it enhanced the scavenging of reactive oxygen species (ROS) by neutralizing hydrogen peroxide. Our results show that the cysteine residue in CaDHN helps to enhance C. arcticum tolerance to abiotic stress by regulating the dimerization of the intrinsically disordered CaDHN protein, which acts as a defense mechanism against extreme polar environments.


Subject(s)
Caryophyllaceae , Cysteine , Cysteine/chemistry , Disulfides/chemistry , Hydrogen Peroxide , Oxidation-Reduction
5.
Cell Rep ; 35(4): 109031, 2021 04 27.
Article in English | MEDLINE | ID: mdl-33910001

ABSTRACT

Leucyl-tRNA synthetase 1 (LARS1) mediates activation of leucine-dependent mechanistic target of rapamycin complex 1 (mTORC1) as well as ligation of leucine to its cognate tRNAs, yet its mechanism of leucine sensing is poorly understood. Here we describe leucine binding-induced conformational changes of LARS1. We determine different crystal structures of LARS1 complexed with leucine, ATP, and a reaction intermediate analog, leucyl-sulfamoyl-adenylate (Leu-AMS), and find two distinct functional states of LARS1 for mTORC1 activation. Upon leucine binding to the synthetic site, H251 and R517 in the connective polypeptide and 50FPYPY54 in the catalytic domain change the hydrogen bond network, leading to conformational change in the C-terminal domain, correlating with RagD association. Leucine binding to LARS1 is increased in the presence of ATP, further augmenting leucine-dependent interaction of LARS1 and RagD. Thus, this work unveils the structural basis for leucine-dependent long-range communication between the catalytic and RagD-binding domains of LARS1 for mTORC1 activation.


Subject(s)
Leucine-tRNA Ligase/metabolism , Leucine/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Humans , Models, Molecular , Signal Transduction
6.
Genes (Basel) ; 12(2)2021 02 03.
Article in English | MEDLINE | ID: mdl-33546197

ABSTRACT

The cryoprotection of cell activity is a key determinant in frozen-dough technology. Although several factors that contribute to freezing tolerance have been reported, the mechanism underlying the manner in which yeast cells respond to freezing and thawing (FT) stress is not well established. Therefore, the present study demonstrated the relationship between DaMDHAR encoding monodehydroascorbate reductase from Antarctic hairgrass Deschampsia antarctica and stress tolerance to repeated FT cycles (FT2) in transgenic yeast Saccharomyces cerevisiae. DaMDHAR-expressing yeast (DM) cells identified by immunoblotting analysis showed high tolerance to FT stress conditions, thereby causing lower damage for yeast cells than wild-type (WT) cells with empty vector alone. To detect FT2 tolerance-associated genes, 3'-quant RNA sequencing was employed using mRNA isolated from DM and WT cells exposed to FT (FT2) conditions. Approximately 332 genes showed ≥2-fold changes in DM cells and were classified into various groups according to their gene expression. The expressions of the changed genes were further confirmed using western blot analysis and biochemical assay. The upregulated expression of 197 genes was associated with pentose phosphate pathway, NADP metabolic process, metal ion homeostasis, sulfate assimilation, ß-alanine metabolism, glycerol synthesis, and integral component of mitochondrial and plasma membrane (PM) in DM cells under FT2 stress, whereas the expression of the remaining 135 genes was partially related to protein processing, selenocompound metabolism, cell cycle arrest, oxidative phosphorylation, and α-glucoside transport under the same condition. With regard to transcription factors in DM cells, MSN4 and CIN5 were activated, but MSN2 and MGA1 were not. Regarding antioxidant systems and protein kinases in DM cells under FT stress, CTT1, GTO, GEX1, and YOL024W were upregulated, whereas AIF1, COX2, and TRX3 were not. Gene activation represented by transcription factors and enzymatic antioxidants appears to be associated with FT2-stress tolerance in transgenic yeast cells. RCK1, MET14, and SIP18, but not YPK2, have been known to be involved in the protein kinase-mediated signalling pathway and glycogen synthesis. Moreover, SPI18 and HSP12 encoding hydrophilin in the PM were detected. Therefore, it was concluded that the genetic network via the change of gene expression levels of multiple genes contributing to the stabilization and functionality of the mitochondria and PM, not of a single gene, might be the crucial determinant for FT tolerance in DaMDAHR-expressing transgenic yeast. These findings provide a foundation for elucidating the DaMDHAR-dependent molecular mechanism of the complex functional resistance in the cellular response to FT stress.


Subject(s)
Freezing/adverse effects , NADH, NADPH Oxidoreductases/genetics , Saccharomyces cerevisiae/genetics , Gene Expression Regulation, Fungal/genetics , Gene Regulatory Networks/genetics , Poaceae/enzymology , Stress, Physiological/genetics , Transcription Factors/genetics
7.
Protein Sci ; 30(4): 899-907, 2021 04.
Article in English | MEDLINE | ID: mdl-33599047

ABSTRACT

In the model organism Escherichia coli and related species, the general stress response relies on tight regulation of the intracellular levels of the promoter specificity subunit RpoS. RpoS turnover is exclusively dependent on RssB, a two-domain response regulator that functions as an adaptor that delivers RpoS to ClpXP for proteolysis. Here, we report crystal structures of the receiver domain of RssB both in its unphosphorylated form and bound to the phosphomimic BeF3- . Surprisingly, we find only modest differences between these two structures, suggesting that truncating RssB may partially activate the receiver domain to a "meta-active" state. Our structural and sequence analysis points to RssB proteins not conforming to either the Y-T coupling scheme for signaling seen in prototypical response regulators, such as CheY, or to the signaling model of the less understood FATGUY proteins.


Subject(s)
DNA-Binding Proteins/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/chemistry , Signal Transduction , Transcription Factors/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endopeptidase Clp/chemistry , Endopeptidase Clp/genetics , Endopeptidase Clp/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Sigma Factor/chemistry , Sigma Factor/genetics , Sigma Factor/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
8.
IUCrJ ; 7(Pt 1): 90-99, 2020 Jan 01.
Article in English | MEDLINE | ID: mdl-31949908

ABSTRACT

MICAL is an oxidoreductase that participates in cytoskeleton reorganization via actin disassembly in the presence of NADPH. Although three MICALs (MICAL1, MICAL2 and MICAL3) have been identified in mammals, only the structure of mouse MICAL1 has been reported. Here, the first crystal structure of human MICAL3, which contains the flavin-containing monooxygenase (FMO) and calponin-homology (CH) domains, is reported. MICAL3 has an FAD/NADP-binding Rossmann-fold domain for mono-oxygenase activity like MICAL1. The FMO and CH domains of both MICAL3 and MICAL1 are highly similar in structure, but superimposition of the two structures shows a different relative position of the CH domain in the asymmetric unit. Based on kinetic analyses, the catalytic efficiency of MICAL3 dramatically increased on adding F-actin only when the CH domain was available. However, this did not occur when two residues, Glu213 and Arg530, were mutated in the FMO and CH domains, respectively. Overall, MICAL3 is structurally highly similar to MICAL1, which suggests that they may adopt the same catalytic mechanism, but the difference in the relative position of the CH domain produces a difference in F-actin substrate specificity.

9.
IUCrJ ; 5(Pt 6): 866-879, 2018 Nov 01.
Article in English | MEDLINE | ID: mdl-30443370

ABSTRACT

Poly(ADP-ribose) polymerase 1 (PARP-1), an enzyme that modifies nuclear proteins by poly(ADP-ribosyl)ation, regulates various cellular activities and restricts the lytic replication of oncogenic gammaherpesviruses by inhibiting the function of replication and transcription activator (RTA), a key switch molecule of the viral life cycle. A viral PARP-1-interacting protein (vPIP) encoded by murine gammaherpesvirus 68 (MHV-68) orf49 facilitates lytic replication by disrupting interactions between PARP-1 and RTA. Here, the structure of MHV-68 vPIP was determined at 2.2 Šresolution. The structure consists of 12 α-helices with characteristic N-terminal ß-strands (Nß) and forms a V-shaped-twist dimer in the asymmetric unit. Structure-based mutagenesis revealed that Nß and the α1 helix (residues 2-26) are essential for the nuclear localization and function of vPIP; three residues were then identified (Phe5, Ser12 and Thr16) that were critical for the function of vPIP and its interaction with PARP-1. A recombinant MHV-68 harboring mutations of these three residues showed severely attenuated viral replication both in vitro and in vivo. Moreover, ORF49 of Kaposi's sarcoma-associated herpesvirus also directly interacted with PARP-1, indicating a conserved mechanism of action of vPIPs. The results elucidate the novel molecular mechanisms by which oncogenic gammaherpesviruses overcome repression by PARP-1 using vPIPs.

10.
Sci Rep ; 8(1): 15075, 2018 10 10.
Article in English | MEDLINE | ID: mdl-30305716

ABSTRACT

Opisthorchis viverrini, a parasitic trematode, was recategorized as a group 1 biological carcinogen because it causes opisthorchiasis, which may result in cholangiocarcinoma. A new strategy for controlling opisthorchiasis is needed because of issues such as drug resistance and reinfection. Triosephosphate isomerase (TIM), a key enzyme in energy metabolism, is regarded as a potential drug target and vaccine candidate against various pathogens. Here, we determined the crystal structures of wild-type and 3 variants of TIMs from O. viverrini (OvTIM) at high resolution. The unique tripeptide of parasite trematodes, the SAD motif, was located on the surface of OvTIM and contributed to forming a 310-helix of the following loop in a sequence-independent manner. Through thermal stability and structural analyses of OvTIM variants, we found that the SAD motif induced local structural alterations of the surface and was involved in the overall stability of OvTIM in a complementary manner with another parasite-specific residue, N115. Comparison of the surface characteristics between OvTIM and Homo sapiens TIM (HsTIM) and structure-based epitope prediction suggested that the SAD motif functions as an epitope.


Subject(s)
Epitopes/chemistry , Opisthorchis/enzymology , Triose-Phosphate Isomerase/chemistry , Amino Acid Motifs , Amino Acid Sequence , Animals , Enzyme Stability , Humans , Models, Molecular , Temperature
11.
Cell Chem Biol ; 25(4): 426-438.e4, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29429898

ABSTRACT

Kynurenine 3-monooxygenase (KMO) inhibitors have been developed for the treatment of neurodegenerative disorders. The mechanisms of flavin reduction and hydrogen peroxide production by KMO inhibitors are unknown. Herein, we report the structure of human KMO and crystal structures of Saccharomyces cerevisiae (sc) and Pseudomonas fluorescens (pf) KMO with Ro 61-8048. Proton transfer in the hydrogen bond network triggers flavin reduction in p-hydroxybenzoate hydroxylase, but the mechanism triggering flavin reduction in KMO is different. Conformational changes via π-π interactions between the loop above the flavin and substrate or non-substrate effectors lead to disorder of the C-terminal α helix in scKMO and shifts of domain III in pfKMO, stimulating flavin reduction. Interestingly, Ro 61-8048 has two different binding modes. It acts as a competitive inhibitor in scKMO and as a non-substrate effector in pfKMO. These findings provide understanding of the catalytic cycle of KMO and insight for structure-based drug design of KMO inhibitors.


Subject(s)
Enzyme Inhibitors/pharmacology , Hydrogen Peroxide/metabolism , Kynurenine 3-Monooxygenase/antagonists & inhibitors , Kynurenine 3-Monooxygenase/metabolism , Pseudomonas fluorescens/enzymology , Saccharomyces cerevisiae/enzymology , Sulfonamides/pharmacology , Thiazoles/pharmacology , Amino Acid Sequence , Animals , Flavins/metabolism , Humans , Kynurenine 3-Monooxygenase/chemistry , Molecular Docking Simulation , Oxidation-Reduction/drug effects , Protein Conformation/drug effects , Pseudomonas fluorescens/chemistry , Saccharomyces cerevisiae/chemistry , Sequence Alignment
12.
Biochem Biophys Res Commun ; 490(2): 400-405, 2017 08 19.
Article in English | MEDLINE | ID: mdl-28623126

ABSTRACT

tRNA maturation involves several steps, including processing, splicing, CCA addition, and posttranscriptional modifications. tRNAHis guanylyltransferase (Thg1) is the only enzyme known to catalyze templated nucleotide addition in the 3'-5' direction, unlike other DNA and RNA polymerases. For a better understanding of its unique catalytic mechanism at the molecular level, we determined the crystal structure of GTP-bound Thg1 from Saccharomyces cerevisiae at the maximum resolution of 3.0 Å. The structure revealed the enzyme to have a tetrameric conformation that is well conserved among different species, and the GTP molecule was clearly bound at the active site, coordinating with two magnesium ions. In addition, two flexible protomers at the potential binding site (PBS) for tRNAHis were observed. We suggest that the PBS of the tetramer could also be one of the sites for interaction with partner proteins.


Subject(s)
Nucleotidyltransferases/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/chemistry , Crystallography, X-Ray , Guanosine Triphosphate/metabolism , Molecular Docking Simulation , Nucleotidyltransferases/metabolism , Protein Conformation , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
13.
Sci Rep ; 7(1): 1764, 2017 05 11.
Article in English | MEDLINE | ID: mdl-28496122

ABSTRACT

Survival of Clonorchis sinensis, a cause of human clonorchiasis, requires tegument proteins, which are localized to the tegumental outer surface membrane. These proteins play an important role in a host response and parasite survival. Thus, these proteins are interesting molecular targets for vaccine and drug development. Here, we have determined two crystal structures of the calmodulin like domain (amino acid [aa] positions 1-81) and dynein light chain (DLC)-like domain (aa 83-177) of a 20.8-kDa tegumental-allergen-like protein from Clonorchis sinensis (CsTAL3). The calmodulin like domain has two Ca2+-binding sites (named CB1 and CB2), but Ca2+ binds to only one site, CB1. The DLC-like domain has a dimeric conformation; the interface is formed mainly by hydrogen bonds between the main chain atoms. In addition, we have determined full-length structure of CsTAL3 in solution and showed the conformational change of CsTAL3 induced by Ca2+ ion binding using small-angle X-ray scattering analysis and molecular dynamics simulations. The Ca2+-bound form has a more extended conformation than the Ca2+-free from does. These structural and biochemical analyses will advance the understanding of the biology of this liver fluke and may contribute to our understanding of the molecular mechanism of calcium-responsive and tegumental-allergen-like proteins.


Subject(s)
Allergens/chemistry , Clonorchis sinensis/chemistry , Helminth Proteins/chemistry , Amino Acid Sequence , Animals , Models, Molecular , Molecular Weight , Protein Domains , Protein Multimerization , Scattering, Small Angle , Sequence Alignment , Solutions , X-Ray Diffraction
14.
Nucleic Acids Res ; 44(19): 9483-9493, 2016 Nov 02.
Article in English | MEDLINE | ID: mdl-27596595

ABSTRACT

GTP and branched-chain amino acids (BCAAs) are metabolic sensors that are indispensable for the determination of the metabolic status of cells. However, their molecular sensing mechanism remains unclear. CodY is a unique global transcription regulator that recognizes GTP and BCAAs as specific signals and affects expression of more than 100 genes associated with metabolism. Herein, we report the first crystal structures of the full-length CodY complex with sensing molecules and describe their functional states. We observed two different oligomeric states of CodY: a dimeric complex of CodY from Staphylococcus aureus with the two metabolites GTP and isoleucine, and a tetrameric form (apo) of CodY from Bacillus cereus Notably, the tetrameric state shows in an auto-inhibitory manner by blocking the GTP-binding site, whereas the binding sites of GTP and isoleucine are clearly visible in the dimeric state. The GTP is located at a hinge site between the long helical region and the metabolite-binding site. Together, data from structural and electrophoretic mobility shift assay analyses improve understanding of how CodY senses GTP and operates as a DNA-binding protein and a pleiotropic transcription regulator.


Subject(s)
Bacterial Proteins/chemistry , Guanosine Triphosphate/chemistry , Models, Molecular , Repressor Proteins/chemistry , Transcription Factors/chemistry , Amino Acid Sequence , Bacterial Proteins/metabolism , Binding Sites , DNA/chemistry , DNA/metabolism , Guanosine Triphosphate/metabolism , Protein Conformation , Protein Interaction Domains and Motifs , Repressor Proteins/metabolism , Structure-Activity Relationship , Transcription Factors/metabolism
15.
Arch Biochem Biophys ; 606: 143-50, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27495739

ABSTRACT

RNase H fold protein PF2046 of Pyrococcus furiosus is a 3'-5' ssDNA exonuclease that cleaves after the second nucleotide from the 3' end of ssDNA and prefers poly-dT over poly-dA as a substrate. In our crystal structure of PF2046 complexed with an oligonucleotide of four thymidine nucleotides (dT4), PF2046 accommodates dT4 tightly in a groove and imposes steric hindrance on dT4 mainly by Phe220 such that dT4 assumes the A-form. As poly-dA prefer B-form due to the stereochemical restrictions, the A-form ssDNA binding by PF2046 should disfavor the processing of poly-dA. Phe220 variants display reduced activity toward poly-dA and the A-form appears to be a prerequisite for the processing by PF2046.


Subject(s)
Bacterial Proteins/chemistry , DNA, Single-Stranded/chemistry , Pyrococcus furiosus/enzymology , Ribonuclease H/chemistry , Catalytic Domain , Crystallization , Crystallography, X-Ray , DNA Repair , Exonucleases/chemistry , Hydrogen-Ion Concentration , Mutation , Nucleic Acid Conformation , Oligodeoxyribonucleotides , Substrate Specificity , X-Ray Diffraction
16.
Biochemistry ; 55(36): 5117-27, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27551953

ABSTRACT

Many bacteria, particularly pathogens, possess methionine sulfoxide reductase A (MsrA) and B (MsrB) as a fusion form (MsrAB). However, it is not clear why they possess a fusion MsrAB form rather than the separate enzymes that exist in most organisms. In this study, we performed biochemical and kinetic analyses of MsrAB from Treponema denticola (TdMsrAB), single-domain forms (TdMsrA and TdMsrB), and catalytic Cys mutants (TdMsrAB(C11S) and TdMsrAB(C285S)). We found that the catalytic efficiency of both MsrA and MsrB increased after fusion of the domains and that the linker region (iloop) that connects TdMsrA and TdMsrB is required for the higher catalytic efficiency of TdMsrAB. We also determined the crystal structure of TdMsrAB at 2.3 Å, showing that the iloop mainly interacts with TdMsrB via hydrogen bonds. Further kinetic analysis using the iloop mutants revealed that the iloop-TdMsrB interactions are critical to MsrB and MsrA activities. We also report the structure in which an oxidized form of dithiothreitol, an in vitro reductant for MsrA and MsrB, is present in the active site of TdMsrA. Collectively, the results of this study reveal an essential role of the iloop in maintaining the higher catalytic efficiency of the MsrAB fusion enzyme and provide a better understanding of why the MsrAB enzyme exists as a fused form.


Subject(s)
Bacterial Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Biocatalysis , Circular Dichroism , Recombinant Fusion Proteins/chemistry , Sequence Homology, Amino Acid
17.
Biochem Biophys Res Commun ; 465(4): 817-24, 2015 Oct 02.
Article in English | MEDLINE | ID: mdl-26315269

ABSTRACT

The SET domain of LegAS4, a type IV secretion system effector of Legionella pneumophila, is a eukaryotic protein motif involved in histone methylation and epigenetic modulation. The SET domain of LegAS4 is involved in the modification of Lys4 of histone H3 (H3K4) in the nucleolus of the host cell, thereby enhancing heterochromatic rDNA transcription. Moreover, LegAS4 contains an ankyrin repeat domain of unknown function at its C-terminal region. Here, we report the crystal structure of LegAS4 in complex with S-adenosyl-l-methionine (SAM). Our data indicate that the ankyrin repeats interact extensively with the SET domain, especially with the SAM-binding amino acids, through conserved residues. Conserved surface analysis marks Glu159, Glu203, and Glu206 on the SET domain serve as candidate residues involved in interaction with the positively charged histone tail. Conserved surface residues on the ankyrin repeat domain surround a small pocket, which is suspected to serve as a binding site for an unknown ligand.


Subject(s)
Bacterial Proteins/chemistry , Legionella pneumophila/chemistry , Type IV Secretion Systems/chemistry , Amino Acid Sequence , Ankyrin Repeat , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Crystallography, X-Ray , Histones/genetics , Histones/metabolism , Legionella pneumophila/genetics , Legionella pneumophila/metabolism , Models, Molecular , Molecular Sequence Data , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , S-Adenosylmethionine/chemistry , S-Adenosylmethionine/metabolism , Structural Homology, Protein , Type IV Secretion Systems/genetics , Type IV Secretion Systems/metabolism
18.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 10): 2136-45, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24100331

ABSTRACT

Aminoacyl-tRNA synthetases recognize cognate amino acids and tRNAs from their noncognate counterparts and catalyze the formation of aminoacyl-tRNAs. Halofuginone (HF), a coccidiostat used in veterinary medicine, exerts its effects by acting as a high-affinity inhibitor of the enzyme glutamyl-prolyl-tRNA synthetase (EPRS). In order to elucidate the precise molecular basis of this inhibition mechanism of human EPRS, the crystal structures of the prolyl-tRNA synthetase domain of human EPRS (hPRS) at 2.4 Šresolution (hPRS-apo), of hPRS complexed with ATP and the substrate proline at 2.3 Šresolution (hPRS-sub) and of hPRS complexed with HF at 2.62 Šresolution (hPRS-HF) are presented. These structures show plainly that motif 1 functions as a cap in hPRS, which is loosely opened in hPRS-apo, tightly closed in hPRS-sub and incorrectly closed in hPRS-HF. In addition, the structural analyses are consistent with more effective binding of hPRS to HF with ATP. Mutagenesis and biochemical analysis confirmed the key roles of two residues, Phe1097 and Arg1152, in the HF inhibition mechanism. These structures will lead to the development of more potent and selective hPRS inhibitors for promoting inflammatory resolution.


Subject(s)
Adenosine Triphosphate/chemistry , Amino Acyl-tRNA Synthetases/antagonists & inhibitors , Amino Acyl-tRNA Synthetases/chemistry , Piperidines/pharmacology , Proline/chemistry , Quinazolinones/pharmacology , Adenosine Triphosphate/antagonists & inhibitors , Adenosine Triphosphate/genetics , Catalytic Domain/drug effects , Catalytic Domain/genetics , Crystallography, X-Ray , Glutamate-tRNA Ligase/antagonists & inhibitors , Glutamate-tRNA Ligase/chemistry , Glutamate-tRNA Ligase/genetics , Humans , Mutagenesis , Piperidines/chemistry , Proline/antagonists & inhibitors , Proline/genetics , Protein Binding/drug effects , Protein Binding/genetics , Protein Conformation/drug effects , Protein Synthesis Inhibitors/chemistry , Protein Synthesis Inhibitors/pharmacology , Quinazolinones/chemistry , Substrate Specificity/drug effects , Substrate Specificity/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...