Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Front Mol Biosci ; 8: 704813, 2021.
Article in English | MEDLINE | ID: mdl-34869578

ABSTRACT

DNA polymerase plays a critical role in passing the genetic information of any living organism to its offspring. DNA polymerase from enterobacteria phage RB69 (RB69pol) has both polymerization and exonuclease activities and has been extensively studied as a model system for B-family DNA polymerases. Many binary and ternary complex structures of RB69pol are known, and they all contain a single polymerase-primer/template (P/T) DNA complex. Here, we report a crystal structure of the exonuclease-deficient RB69pol with the P/T duplex in a dimeric form at a resolution of 2.2 Å. The structure includes one new closed ternary complex with a single divalent metal ion bound and one new open binary complex in the pre-insertion state with a vacant dNTP-binding pocket. These complexes suggest that initial binding of the correct dNTP in the open state is much weaker than expected and that initial binding of the second divalent metal ion in the closed state is also much weaker than measured. Additional conformational changes are required to convert these complexes to high-affinity states. Thus, the measured affinities for the correct incoming dNTP and divalent metal ions are average values from many conformationally distinctive states. Our structure provides new insights into the order of the complex assembly involving two divalent metal ions. The biological relevance of specific interactions observed between one RB69pol and the P/T duplex bound to the second RB69pol observed within this dimeric complex is discussed.

2.
Biochem Biophys Res Commun ; 506(1): 102-107, 2018 11 17.
Article in English | MEDLINE | ID: mdl-30336976

ABSTRACT

Ubiquitin-conjugating enzymes (E2) form thioester bonds with ubiquitin (Ub), which are subsequently transferred to target proteins for cellular progress. Ube2K/E2-25K (a class II E2 enzyme) contains a C-terminal ubiquitin-associated (UBA) domain that has been suggested to control ubiquitin recognition, dimerization, or poly-ubiquitin chain formation. Ube2K is a special E2 because it synthesizes K48-linked poly-ubiquitin chains without E3 ubiquitin ligase. We found that a novel interaction between the acceptor di-Ub (Ub2) and the auxiliary Ube2K promotes the discharging reaction and production of tri-Ub (Ub3), probably by guiding and positioning the K48 (in the distal Ub) of the acceptor Ub2 in the active site. We also determined the crystal structure of Ube2K-Ub2 at 2.47 Šresolution. Based on our structural and biochemical data, we proposed a structural model of Ub3 synthesis by Ube2K without E3.


Subject(s)
Lysine/chemistry , Ubiquitin-Activating Enzymes/chemistry , Ubiquitin-Conjugating Enzymes/chemistry , Ubiquitins/chemistry , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Lysine/metabolism , Molecular Docking Simulation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitins/genetics , Ubiquitins/metabolism
3.
Biochem Biophys Res Commun ; 486(2): 470-475, 2017 04 29.
Article in English | MEDLINE | ID: mdl-28315686

ABSTRACT

Anti-bacterial and anti-viral neuraminidase agents inhibit neuraminidase activity catalyzing the hydrolysis of terminal N-acetylneuraminic acid (Neu5Ac) from glycoconjugates and help to prevent the host pathogenesis that lead to fatal infectious diseases including influenza, bacteremia, sepsis, and cholera. Emerging antibiotic and drug resistances to commonly used anti-neuraminidase agents such as oseltamivir (Tamiflu) and zanamivir (Relenza) have highlighted the need to develop new anti-neuraminidase drugs. We obtained a serendipitous complex crystal of the catalytic domain of Clostridium perfringens neuraminidase (CpNanICD) with 2-(cyclohexylamino)ethanesulfonic acid (CHES) as a buffer. Here, we report the crystal structure of CpNanICD in complex with CHES at 1.24 Å resolution. Amphipathic CHES binds to the catalytic site of CpNanICD similar to the substrate (Neu5Ac) binding site. The 2-aminoethanesulfonic acid moiety and cyclohexyl groups of CHES interact with the cluster of three arginine residues and with the hydrophobic pocket of the CpNanICD catalytic site. In addition, a structural comparison with other bacterial and human neuraminidases suggests that CHES could serve as a scaffold for the development of new anti-neuraminidase agents targeting CpNanI.


Subject(s)
Bacterial Proteins/chemistry , Clostridium perfringens/chemistry , Enzyme Inhibitors/chemistry , Neuraminidase/chemistry , Taurine/analogs & derivatives , Amino Acid Motifs , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Catalytic Domain , Cloning, Molecular , Clostridium perfringens/enzymology , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Models, Molecular , Neuraminidase/antagonists & inhibitors , Neuraminidase/genetics , Neuraminidase/metabolism , Protein Domains , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Structural Homology, Protein , Taurine/chemistry
4.
Sci Rep ; 6: 39095, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27974828

ABSTRACT

EFhd2/Swiprosin-1 is a cytoskeletal Ca2+-binding protein implicated in Ca2+-dependent cell spreading and migration in epithelial cells. EFhd2 domain architecture includes an N-terminal disordered region, a PxxP motif, two EF-hands, a ligand mimic helix and a C-terminal coiled-coil domain. We reported previously that EFhd2 displays F-actin bundling activity in the presence of Ca2+ and this activity depends on the coiled-coil domain and direct interaction of the EFhd2 core region. However, the molecular mechanism for the regulation of F-actin binding and bundling by EFhd2 is unknown. Here, the Ca2+-bound crystal structure of the EFhd2 core region is presented and structures of mutants defective for Ca2+-binding are also described. These structures and biochemical analyses reveal that the F-actin bundling activity of EFhd2 depends on the structural rigidity of F-actin binding sites conferred by binding of the EF-hands to Ca2+. In the absence of Ca2+, the EFhd2 core region exhibits local conformational flexibility around the EF-hand domain and C-terminal linker, which retains F-actin binding activity but loses the ability to bundle F-actin. In addition, we establish that dimerisation of EFhd2 via the C-terminal coiled-coil domain, which is necessary for F-actin bundling, occurs through the parallel coiled-coil interaction.


Subject(s)
Actins/metabolism , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/metabolism , Calcium/metabolism , Calcium-Binding Proteins/genetics , Crystallography, X-Ray , Humans , Models, Molecular , Mutation , Protein Binding , Protein Domains , Protein Structure, Tertiary
5.
FEBS Lett ; 590(23): 4402-4413, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27714797

ABSTRACT

The interaction of the rhomboid pseudoprotease Derlin-1 and p97 is crucial for the retrotranslocation of polyubiquitinated substrates in the endoplasmic reticulum-associated degradation pathway. We report a 2.25 Å resolution structure of the p97 N-terminal domain (p97N) in complex with the Derlin-1 SHP motif. Remarkably, the SHP motif adopts a short, antiparallel ß-strand that interacts with the ß-sheet of p97N-a site distinct from that to which most p97 adaptor proteins bind. Mutational and biochemical analyses contributed to defining the specific interaction, demonstrating the importance of a highly conserved binding pocket on p97N and a signature motif on SHP. Our findings may also provide insights into the interactions between other SHP-containing proteins and p97N.


Subject(s)
Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Apoproteins/chemistry , Apoproteins/metabolism , Binding Sites , Conserved Sequence , Endoplasmic Reticulum-Associated Degradation , Humans , Protein Binding , Protein Domains
6.
Biochem J ; 473(18): 2863-80, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27407164

ABSTRACT

RHBDL4 is an active rhomboid that specifically recognizes and cleaves atypical, positively charged transmembrane endoplasmic reticulum-associated degradation (ERAD) substrates. Interaction of valosin-containing protein (p97/VCP) and RHBDL4 is crucial to retrotranslocate polyubiquitinated substrates for ERAD pathway. Here, we report the first complex structure of VCP-binding motif (VBM) with p97 N-terminal domain (p97N) at 1.88 Šresolution. Consistent with p97 adaptor proteins including p47-ubiquitin regulatory X (UBX), gp78-VCP-interacting motif (VIM), OTU1-UBX-like element, and FAF1-UBX, RHBDL4 VBM also binds at the interface between the two lobes of p97N. Notably, the RF residues in VBM are involved in the interaction with p97N, showing a similar interaction pattern with that of FPR signature motif in the UBX domain, although the directionality is opposite. Comparison of VBM interaction with VIM of gp78, another α-helical motif that interacts with p97N, revealed that the helix direction is inversed. Nevertheless, the conserved arginine residues in both motifs participate in the majority of the interface via extensive hydrogen bonds and ionic interactions with p97N. We identified novel VBM-binding mode to p97N that involves a combination of two types of p97-cofactor specificities observed in the UBX and VIM interactions. This highlights the induced fit model of p97N interdomain cleft upon cofactor binding to form stable p97-cofactor complexes. Our mutational and biochemical analyses in defining the specific interaction between VBM and p97N have elucidated the importance of the highly conserved VBM, applicable to other VBM-containing proteins. We also showed that RHBDL4, ubiquitins, and p97 co-operate for efficient substrate dislocation.


Subject(s)
Membrane Proteins/chemistry , Amino Acid Sequence , Animals , Humans , Protein Conformation , Sequence Homology, Amino Acid , X-Ray Diffraction
7.
Sci Rep ; 6: 19681, 2016 Jan 25.
Article in English | MEDLINE | ID: mdl-26805589

ABSTRACT

Quinolinate phosphoribosyltransferase (QPRT) catalyses the production of nicotinic acid mononucleotide, a precursor of de novo biosynthesis of the ubiquitous coenzyme nicotinamide adenine dinucleotide. QPRT is also essential for maintaining the homeostasis of quinolinic acid in the brain, a possible neurotoxin causing various neurodegenerative diseases. Although QPRT has been extensively analysed, the molecular basis of the reaction catalysed by human QPRT remains unclear. Here, we present the crystal structures of hexameric human QPRT in the apo form and its complexes with reactant or product. We found that the interaction between dimeric subunits was dramatically altered during the reaction process by conformational changes of two flexible loops in the active site at the dimer-dimer interface. In addition, the N-terminal short helix α1 was identified as a critical hexamer stabilizer. The structural features, size distribution, heat aggregation and ITC studies of the full-length enzyme and the enzyme lacking helix α1 strongly suggest that human QPRT acts as a hexamer for cooperative reactant binding via three dimeric subunits and maintaining stability. Based on our comparison of human QPRT structures in the apo and complex forms, we propose a drug design strategy targeting malignant glioma.


Subject(s)
Glioma/drug therapy , NAD/biosynthesis , Pentosyltransferases/chemistry , Catalysis , Crystallography, X-Ray , Dimerization , Drug Design , Glioma/genetics , Humans , Pentosyltransferases/metabolism , Protein Conformation, alpha-Helical
8.
EMBO Rep ; 16(10): 1318-33, 2015 10.
Article in English | MEDLINE | ID: mdl-26341627

ABSTRACT

The mitochondrial calcium uniporter (MCU) is responsible for mitochondrial calcium uptake and homeostasis. It is also a target for the regulation of cellular anti-/pro-apoptosis and necrosis by several oncogenes and tumour suppressors. Herein, we report the crystal structure of the MCU N-terminal domain (NTD) at a resolution of 1.50 Å in a novel fold and the S92A MCU mutant at 2.75 Å resolution; the residue S92 is a predicted CaMKII phosphorylation site. The assembly of the mitochondrial calcium uniporter complex (uniplex) and the interaction with the MCU regulators such as the mitochondrial calcium uptake-1 and mitochondrial calcium uptake-2 proteins (MICU1 and MICU2) are not affected by the deletion of MCU NTD. However, the expression of the S92A mutant or a NTD deletion mutant failed to restore mitochondrial Ca(2+) uptake in a stable MCU knockdown HeLa cell line and exerted dominant-negative effects in the wild-type MCU-expressing cell line. These results suggest that the NTD of MCU is essential for the modulation of MCU function, although it does not affect the uniplex formation.


Subject(s)
Calcium Channels/chemistry , Calcium Channels/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Calcium/metabolism , Calcium Channels/genetics , Calcium-Binding Proteins/metabolism , Crystallography, X-Ray , HEK293 Cells , HeLa Cells , Humans , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/genetics , Models, Molecular , Mutation , Protein Folding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Protein Structure, Tertiary
9.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 2): 416, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25664753

ABSTRACT

A correction is made to the article by Lee et al. [(2014) Acta Cryst. D70, 1357-1365].

10.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 5): 1357-65, 2014 May.
Article in English | MEDLINE | ID: mdl-24816104

ABSTRACT

Sialidase catalyzes the removal of a terminal sialic acid from glycoconjugates and plays a pivotal role in nutrition, cellular interactions and pathogenesis mediating various infectious diseases including cholera, influenza and sepsis. An array of antiviral sialidase agents have been developed and are commercially available, such as zanamivir and oseltamivir for treating influenza. However, the development of bacterial sialidase inhibitors has been much less successful. Here, natural polyphenolic geranylated flavonoids which show significant inhibitory effects against Cp-NanI, a sialidase from Clostridium perfringens, are reported. This bacterium causes various gastrointestinal diseases. The crystal structure of the Cp-NanI catalytic domain in complex with the best inhibitor, diplacone, is also presented. This structure explains how diplacone generates a stable enzyme-inhibitor complex. These results provide a structural framework for understanding the interaction between sialidase and natural flavonoids, which are promising scaffolds on which to discover new anti-sialidase agents.


Subject(s)
Clostridium perfringens/enzymology , Enzyme Inhibitors/chemistry , Flavonoids/chemistry , Neuraminidase/antagonists & inhibitors , Neuraminidase/chemistry , Catalytic Domain , Crystallography, X-Ray , Enzyme Inhibitors/pharmacology , Flavanones/chemistry , Flavanones/pharmacology , Flavonoids/pharmacology , Inhibitory Concentration 50 , Kinetics , Models, Molecular , Protein Conformation
11.
J Synchrotron Radiat ; 20(Pt 6): 984-8, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24121353

ABSTRACT

Proper cell division at the mid-site of gram-negative bacteria reflects critical regulation by the min system (MinC, MinD and MinE) of the cytokinetic Z ring, which is a polymer composed of FtsZ subunits. MinC and MinD act together to inhibit aberrantly positioned Z-ring formation. MinC consists of two domains: an N-terminal domain (MinCNTD), which interacts with FtsZ and inhibits FtsZ polymerization, and a C-terminal domain (MinCCTD), which interacts with MinD and inhibits the bundling of FtsZ filaments. These two domains reportedly function together, and both are essential for normal cell division. The full-length dimeric structure of MinC from Thermotoga maritima has been reported, and shows that MinC dimerization occurs via MinCCTD; MinCNTD is not involved in dimerization. Here the crystal structure of Escherichia coli MinCNTD (EcoMinCNTD) is reported. EcoMinCNTD forms a dimer via domain swapping between the first ß strands in each subunit. It is therefore suggested that the dimerization of full-length EcoMinC occurs via both MinCCTD and MinCNTD, and that the dimerized EcoMinCNTD likely plays an important role in inhibiting aberrant Z-ring localization.


Subject(s)
Bacterial Proteins/chemistry , Amino Acid Sequence , Bacterial Proteins/genetics , Cloning, Molecular , Crystallography, X-Ray , Dimerization , Models, Molecular , Molecular Sequence Data , Protein Conformation , Sequence Homology, Amino Acid , Thermotoga maritima/chemistry
12.
Biochem Biophys Res Commun ; 438(4): 765-71, 2013 Sep 06.
Article in English | MEDLINE | ID: mdl-23891752

ABSTRACT

We recently reported that glutamate carboxypeptidase II (GCPII) has a new physiological function degrading amyloid-ß (Aß), distinct from its own hydrolysis activity in N-acetyl-L-aspartyl-L-glutamate (NAAG); however, its underlying mechanism remains undiscovered. Using site-directed mutagenesis and S1 pocket-specific chemical inhibitor (compound 2), which was developed for the present study based on in sillico computational modeling, we discovered that the Aß degradation occurs through S1 pocket but not through S1' pocket responsible for NAAG hydrolysis. Treatment with compound 2 prevented GCPII from Aß degradation without any impairment in NAAG hydrolysis. Likewise, 2-PMPA (specific GCPII inhibitor developed targeting S1' pocket) completely blocked the NAAG hydrolysis without any effect on Aß degradation. Pre-incubation with NAAG and Aß did not affect Aß degradation and NAAG hydrolysis, respectively. These data suggest that GCPII has two distinctive binding sites for two different substrates and that Aß degradation occurs through binding to S1 pocket of GCPII.


Subject(s)
Alzheimer Disease/enzymology , Amyloid beta-Peptides/metabolism , Glutamate Carboxypeptidase II/metabolism , Proteolysis , Alzheimer Disease/metabolism , Animals , Binding Sites/drug effects , Cell Line, Tumor , Dipeptides/metabolism , Enzyme Inhibitors/pharmacology , Glutamate Carboxypeptidase II/antagonists & inhibitors , Glutamate Carboxypeptidase II/chemistry , Glutamate Carboxypeptidase II/genetics , Glutamic Acid/metabolism , Humans , Mice , Mice, Transgenic , Molecular Docking Simulation , Mutagenesis, Site-Directed , Organophosphorus Compounds/pharmacology , Proteolysis/drug effects
13.
PLoS One ; 8(4): e62027, 2013.
Article in English | MEDLINE | ID: mdl-23626766

ABSTRACT

We have determined the crystal structure of porcine quinolinate phosphoribosyltransferase (QAPRTase) in complex with nicotinate mononucleotide (NAMN), which is the first crystal structure of a mammalian QAPRTase with its reaction product. The structure was determined from protein obtained from the porcine kidney. Because the full protein sequence of porcine QAPRTase was not available in either protein or nucleotide databases, cDNA was synthesized using reverse transcriptase-polymerase chain reaction to determine the porcine QAPRTase amino acid sequence. The crystal structure revealed that porcine QAPRTases have a hexameric structure that is similar to other eukaryotic QAPRTases, such as the human and yeast enzymes. However, the interaction between NAMN and porcine QAPRTase was different from the interaction found in prokaryotic enzymes, such as those of Helicobacter pylori and Mycobacterium tuberculosis. The crystal structure of porcine QAPRTase in complex with NAMN provides a structural framework for understanding the unique properties of the mammalian QAPRTase active site and designing new antibiotics that are selective for the QAPRTases of pathogenic bacteria, such as H. pylori and M. tuberculosis.


Subject(s)
Kidney/chemistry , Nicotinamide Mononucleotide/analogs & derivatives , Pentosyltransferases/chemistry , Animals , Catalytic Domain , Crystallography, X-Ray , DNA, Complementary/genetics , Helicobacter pylori/chemistry , Helicobacter pylori/enzymology , Humans , Kidney/enzymology , Models, Molecular , Mycobacterium tuberculosis/chemistry , Mycobacterium tuberculosis/enzymology , Nicotinamide Mononucleotide/chemistry , Pentosyltransferases/genetics , Protein Interaction Domains and Motifs , Protein Multimerization , Species Specificity , Structural Homology, Protein , Swine
14.
Article in English | MEDLINE | ID: mdl-23385756

ABSTRACT

Upc2, a zinc-cluster transcription factor, is a regulator of ergosterol biosynthesis in yeast. In response to sterol levels, the transcriptional activity of Upc2 is controlled by the C-terminal domain. In this study, the C-terminal regulatory domain of Upc2 from Saccharomyces cerevisiae was purified and crystallized by the vapour-diffusion method. To improve the diffraction quality of Upc2 crystals, a Upc2 fusion protein in which 11 residues of the variable loop (residues 715-725) were replaced by T4 lysozymes in Upc2 (Upc2-T4L) was engineered. The Upc2-T4L crystals diffracted to 2.9 Å resolution using synchrotron radiation. The crystal was trigonal, belonging to space group P3(2) with unit-cell parameters a = 67.2, b = 67.2, c = 257.5 Å. The Matthews coefficient was determined to be 3.41 Å(3) Da(-1) with two molecules in the asymmetric unit. Initial attempts to solve the structure by the single-anomalous dispersion technique using selenomethionine were successful.


Subject(s)
Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/metabolism , Sterols/metabolism , Trans-Activators/chemistry , Amino Acid Sequence , Crystallization , Crystallography, X-Ray , Molecular Sequence Data , Protein Structure, Tertiary , Sequence Alignment , Transcription Factors/chemistry
15.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 68(Pt 12): 1488-90, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23192029

ABSTRACT

Quinolinate phosphoribosyltransferase (QAPRTase) is a key enzyme in NAD biosynthesis; it catalyzes the formation of nicotinate mononucleotide (NAMN) from quinolinate and 5-phosphoribosyl-1-pyrophosphate. In order to elucidate the mechanism of NAMN biosynthesis, crystals of Sus scrofa QAPRTase (Ss-QAPRTase) purified from porcine kidney in complex with NAMN were obtained and diffraction data were collected and processed to 2.1 Šresolution. The Ss-QAPRTase-NAMN cocrystals belonged to space group P321, with unit-cell parameters a=119.1, b=119.1, c=93.7 Å, γ=120.0°. The Matthews coefficient and the solvent content were estimated as 3.10 Å3 Da(-1) and 60.3%, respectively, assuming the presence of two molecules in the asymmetric unit.


Subject(s)
Kidney/enzymology , Nicotinamide Mononucleotide/analogs & derivatives , Pentosyltransferases/chemistry , Animals , Crystallography, X-Ray , Models, Molecular , Nicotinamide Mononucleotide/chemistry , Nicotinamide Mononucleotide/metabolism , Pentosyltransferases/metabolism , Protein Conformation , Swine/metabolism
16.
Mol Pharmacol ; 82(2): 143-55, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22547262

ABSTRACT

As previously reported, the activity of the large-conductance calcium (Ca(2+))-activated potassium (K(+)) (BK(Ca)) channel is strongly potentiated from the extracellular side of the cell membrane by certain benzofuroindole derivatives. Here, the mechanism of action of one of the most potent activators, 4-chloro-7-(trifluoromethyl)-10H-benzofuro[3,2-b]indole-1-carboxylic acid (CTBIC), is characterized. This compound, Compound 22 in the previous report (Chembiochem 6:1745-1748, 2005), potentiated the activity of the channel by shifting its conductance-voltage relationship toward the more negative direction. Cotreatment with CTBIC reduced the affinity of charybdotoxin, a peptide pore-blocker, whereas that of tetraethylammonium, a small pore-blocking quaternary ammonium, was not significantly altered. Guided by these results, scanning mutagenesis of the outer vestibule of the BK(Ca) channel was launched to uncover the molecular determinants that affect CTBIC binding. Alanine substitution of several amino acid residues in the turret region and the S6 helix of the channel decreased potentiation by CTBIC. Homology modeling and molecular dynamics simulation showed that some of these residues formed a CTBIC binding pocket between two adjacent α-subunits in the outer vestibule of the channel. Thus, it can be envisioned that benzofuroindole derivatives stabilize the open conformation of the channel by binding to the residues clustered across the extracellular part of the subunit interface. The present results indicate that the interface between different α-subunits of the BK(Ca) channel may play a critical role in the modulation of channel activity. Therefore, this interface represents a potential therapeutic target site for the regulation of K(+) channels.


Subject(s)
Indoles/metabolism , Potassium Channels/genetics , Potassium Channels/metabolism , Amino Acid Sequence , Animals , Binding Sites/physiology , Dose-Response Relationship, Drug , Female , Indoles/chemistry , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits , Molecular Sequence Data , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/metabolism , Potassium Channels/chemistry , Protein Binding/physiology , Rats , Xenopus laevis
17.
Article in English | MEDLINE | ID: mdl-21505255

ABSTRACT

Methylophaga aminisulfidivorans MP(T) is a marine methylotrophic bacterium that utilizes C(1) compounds such as methanol as a carbon and energy source. The released electron from oxidation flows through a methanol-oxidizing system (MOX) consisting of a series of electron-transfer proteins encoded by the mox operon. One of the key enzymes in the pathway is methanol dehydrogenase (MDH), which contains the prosthetic group pyrroloquinoline quinone (PQQ) and converts methanol to formaldehyde in the periplasm by transferring two electrons from the oxidation of one methanol molecule to the electron acceptor cytochrome c(L). In order to obtain molecular insights into the oxidation mechanism, a native heterotetrameric α(2)ß(2) MDH complex was directly purified from M. aminisulfidivorans MP(T) grown in the presence of methanol and crystallized. The crystal diffracted to 1.7 Šresolution and belonged to the monoclinic space group P2(1) (unit-cell parameters a = 63.9, b = 109.5, c = 95.6 Å, ß = 100.5°). The asymmetric unit of the crystal contained one heterotetrameric complex, with a calculated Matthews coefficient of 2.24 Å(3) Da(-1) and a solvent content of 45.0%.


Subject(s)
Alcohol Oxidoreductases/chemistry , Piscirickettsiaceae/enzymology , Alcohol Oxidoreductases/isolation & purification , Crystallization , Crystallography, X-Ray
18.
Eur J Med Chem ; 46(4): 1153-64, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21330015

ABSTRACT

NAmPRTase (PBEF/Visfatin) plays a pivotal role in the salvage pathway of NAD(+) biosynthesis. NAmPRTase has been an attractive target for anti-cancer agents that induce apoptosis of tumor cells via a declining plasma NAD(+) level. In this report, a series of structural analogs of FK866 (1), a known NAmPRTase inhibitor, was synthesized and tested for inhibitory activities against the proliferation of cancer cells and human NAmPRTase. Among them, compound 7 showed similar anti-cancer and enzyme inhibitory activities to compound 1. Further investigation of compound 7 with X-ray analysis revealed a co-crystal structure in complex with human NAmPRTase, suggesting that Asp219 in the active site of the enzyme could contribute to an additional interaction with the pyrrole nitrogen of compound 7.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Acrylamides/chemical synthesis , Acrylamides/chemistry , Acrylamides/pharmacology , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Cell Survival/drug effects , Crystallography, X-Ray , Enzyme Assays , Enzyme Inhibitors/chemical synthesis , Humans , Hydrogen Bonding , Models, Molecular , Nicotinamide Phosphoribosyltransferase/chemistry , Nicotinamide Phosphoribosyltransferase/metabolism , Piperidines/chemical synthesis , Piperidines/chemistry , Piperidines/pharmacology , Protein Conformation , Ribose/chemistry
19.
Article in English | MEDLINE | ID: mdl-21206019

ABSTRACT

Quinolinate phosphoribosyltransferase (QPRTase) is a key NAD-biosynthetic enzyme which catalyzes the transfer of quinolinic acid to 5-phosphoribosyl-1-pyrophosphate, yielding nicotinic acid mononucleotide. Homo sapiens QPRTase (Hs-QPRTase) appeared as a hexamer during purification and the protein was crystallized. Diffraction data were collected and processed at 2.8 Šresolution. Native Hs-QPRTase crystals belonged to space group P2(1), with unit-cell parameters a=76.2, b=137.1, c=92.7 Å, ß=103.8°. Assuming the presence of six molecules in the asymmetric unit, the calculated Matthews coefficient is 2.46 Å3 Da(-1), which corresponds to a solvent content of 49.9%.


Subject(s)
Pentosyltransferases/chemistry , Protein Structure, Quaternary , Animals , Crystallization , Crystallography, X-Ray , Humans , Molecular Sequence Data , NAD/biosynthesis , Pentosyltransferases/metabolism
20.
Prog Biophys Mol Biol ; 105(3): 145-61, 2011 May.
Article in English | MEDLINE | ID: mdl-20934451

ABSTRACT

Ryanodine receptors (RyRs) are intracellular Ca(2+) release channels (CRCs) that play a pivotal role in cellular Ca(2+) signaling. In striated muscles, RyR-mediated Ca(2+) release from the sarcoplasmic reticulum (SR) induces elevation of cytosolic Ca(2+) concentration and subsequent muscle contraction. Evidence from various sources suggests that RyRs in homo-tetrameric conformation form a large conductance Ca(2+) permeable channel in the central pore and large cytoplasmic domains. RyRs form a large assembly with various cytosolic and luminal proteins. A number of papers have been published concerning the functions of RyRs and the regulation of the associated proteins, but the three dimensional (3D) structure of the assembly has not been addressed in detail. In this paper, we have attempted to establish a 3D-map for the assembly of RyRs by considering published cryo-EM data, available X-ray crystallographic information and molecular modeling methods.


Subject(s)
Protein Multimerization , Protein Structure, Quaternary , Ryanodine Receptor Calcium Release Channel/chemistry , Systems Biology/methods , Animals , Humans , Protein Interaction Mapping , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...