Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Chembiochem ; : e202400258, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38887142

ABSTRACT

S­adenosyl-l-methionine-dependent methyltransferases (MTs) are involved in the C-methylation of a variety of natural products. The MTs SgvM from Streptomyces griseoviridis and MrsA from Pseudomonas syringae pv. syringae catalyze the methylation of the ß-carbon atom of α-keto acids in the biosynthesis of the antibiotic natural products viridogrisein and 3­methylarginine, respectively. MrsA shows high substrate selectivity for 5­guanidino-2-oxovalerate, while other α-keto acids, such as the SgvM substrates 4-methyl-2-oxovalerate, 2-oxovalerate, and phenylpyruvate, are not accepted. Here we report the crystal structures of SgvM and MrsA in the apo form bound with substrate or S­adenosyl-l-methionine. By investigating key residues for substrate recognition in the active sites of both enzymes and engineering MrsA by site-directed mutagenesis, the substrate range of MrsA was extended to accept α­keto acid substrates of SgvM with uncharged and lipophilic ß­residues. Our results showcase the transfer of the substrate scope of α-keto acid MTs from different biosynthetic pathways by rational design.

2.
J Struct Biol ; 213(4): 107794, 2021 12.
Article in English | MEDLINE | ID: mdl-34506908

ABSTRACT

The S-adenosyl-L-methionine-dependent methyltransferase Rv0560c of Mycobacterium tuberculosis belongs to an orthologous group of heterocyclic toxin methyltransferases (Htm) which likely contribute to resistance of mycobacteria towards antimicrobial natural compounds as well as drugs. HtmM.t. catalyzes the methylation of the Pseudomonas aeruginosa toxin 2-heptyl-1-hydroxyquinolin-4(1H)-one (also known as 2-heptyl-4-hydroxyquinoline N-oxide), a potent inhibitor of respiratory electron transfer, its 1-hydroxyquinolin-4(1H)-one core (QNO), structurally related (iso)quinolones, and some mycobactericidal compounds. In this study, crystal structures of HtmM.t. in complex with S-adenosyl-L-homocysteine (SAH) and the methyl-accepting substrates QNO or 4-hydroxyisoquinoline-1(2H)-one, or the methylated product 1-methoxyquinolin-4(1H)-one, were determined at < 1.9 Å resolution. The monomeric protein exhibits the typical Rossmann fold topology and conserved residues of class I methyltransferases. Its SAH binding pocket is connected via a short tunnel to a large solvent-accessible cavity, which accommodates the methyl-accepting substrate. Residues W44, F168, and F208 in connection with F212 form a hydrophobic clamp around the heteroaromatic ring of the methyl-accepting substrate and likely play a major role in substrate positioning. Structural and biochemical data suggest that H139 and T136 are key active site residues, with H139 acting as general base that activates the methyl-accepting hydroxy group. Our structural data may contribute to the design of Htm inhibitors or of antimycobacterial drugs unamenable for methylation.


Subject(s)
Bacterial Proteins/metabolism , Hydroxyquinolines/metabolism , Methyltransferases/metabolism , Mycobacterium tuberculosis/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites/genetics , Biocatalysis , Catalytic Domain/genetics , Crystallography, X-Ray , Hydroxyquinolines/chemistry , Methylation , Methyltransferases/chemistry , Methyltransferases/genetics , Models, Chemical , Models, Molecular , Molecular Structure , Mutagenesis, Site-Directed , Mycobacterium tuberculosis/genetics , Protein Conformation , S-Adenosylmethionine/chemistry , S-Adenosylmethionine/metabolism , Sequence Homology, Amino Acid , Substrate Specificity
3.
Proc Natl Acad Sci U S A ; 115(13): 3350-3355, 2018 03 27.
Article in English | MEDLINE | ID: mdl-29531036

ABSTRACT

Inorganic polyphosphate is a ubiquitous, linear biopolymer built of up to thousands of phosphate residues that are linked by energy-rich phosphoanhydride bonds. Polyphosphate kinases of the family 2 (PPK2) use polyphosphate to catalyze the reversible phosphorylation of nucleotide phosphates and are highly relevant as targets for new pharmaceutical compounds and as biocatalysts for cofactor regeneration. PPK2s can be classified based on their preference for nucleoside mono- or diphosphates or both. The detailed mechanism of PPK2s and the molecular basis for their substrate preference is unclear, which is mainly due to the lack of high-resolution structures with substrates or substrate analogs. Here, we report the structural analysis and comparison of a class I PPK2 (ADP-phosphorylating) and a class III PPK2 (AMP- and ADP-phosphorylating), both complexed with polyphosphate and/or nucleotide substrates. Together with complementary biochemical analyses, these define the molecular basis of nucleotide specificity and are consistent with a Mg2+ catalyzed in-line phosphoryl transfer mechanism. This mechanistic insight will guide the development of PPK2 inhibitors as potential antibacterials or genetically modified PPK2s that phosphorylate alternative substrates.


Subject(s)
Deinococcus/enzymology , Phosphotransferases (Phosphate Group Acceptor)/chemistry , Phosphotransferases (Phosphate Group Acceptor)/metabolism , Polyphosphates/metabolism , Crystallography, X-Ray , Kinetics , Ligands , Phosphorylation , Protein Conformation , Substrate Specificity
4.
Angew Chem Int Ed Engl ; 60(52): 27277-27281, 2021 12 20.
Article in English | MEDLINE | ID: mdl-34612584

ABSTRACT

NADH:ubiquinone oxidoreductase, respiratory complex I, plays a central role in cellular energy metabolism. As a major source of reactive oxygen species (ROS) it affects ageing and mitochondrial dysfunction. The novel inhibitor NADH-OH specifically blocks NADH oxidation and ROS production by complex I in nanomolar concentrations. Attempts to elucidate its structure by NMR spectroscopy have failed. Here, by using X-ray crystallographic analysis, we report the structure of NADH-OH bound in the active site of a soluble fragment of complex I at 2.0 Šresolution. We have identified key amino acid residues that are specific and essential for binding NADH-OH. Furthermore, the structure sheds light on the specificity of NADH-OH towards the unique Rossmann-fold of complex I and indicates a regulatory role in mitochondrial ROS generation. In addition, NADH-OH acts as a lead-structure for the synthesis of a novel class of ROS suppressors.


Subject(s)
Electron Transport Complex I/antagonists & inhibitors , Enzyme Inhibitors/chemistry , NAD/analogs & derivatives , Aquifex/enzymology , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Electron Transport Complex I/chemistry , Electron Transport Complex I/metabolism , Enzyme Inhibitors/pharmacology , Humans , Hydrogen Bonding , Models, Molecular , NAD/chemistry , NAD/metabolism , NAD/pharmacology , Protein Binding
5.
J Biol Inorg Chem ; 23(7): 1049-1056, 2018 10.
Article in English | MEDLINE | ID: mdl-30141094

ABSTRACT

Nitrogenases catalyze the biological fixation of inert N2 into bioavailable ammonium. They are bipartite systems consisting of the catalytic dinitrogenase and a complementary reductase, the Fe protein that is also the site where ATP is hydrolyzed to drive the reaction forward. Three different subclasses of dinitrogenases are known, employing either molybdenum, vanadium or only iron at their active site cofactor. Although in all these classes the mode and mechanism of interaction with Fe protein is conserved, each one encodes its own orthologue of the reductase in the corresponding gene cluster. Here we present the 2.2 Å resolution structure of VnfH from Azotobacter vinelandii, the Fe protein of the alternative, vanadium-dependent nitrogenase system, in its ADP-bound state. VnfH adopts the same conformation that was observed for NifH, the Fe protein of molybdenum nitrogenase, in complex with ADP, representing a state of the functional cycle that is ready for reduction and subsequent nucleotide exchange. The overall similarity of NifH and VnfH confirms the experimentally determined cross-reactivity of both ATP-hydrolyzing reductases.


Subject(s)
Azotobacter vinelandii/enzymology , Nitrogenase/chemistry , Crystallography, X-Ray , Models, Molecular , Nitrogenase/isolation & purification , Nitrogenase/metabolism
6.
Angew Chem Int Ed Engl ; 56(41): 12476-12480, 2017 10 02.
Article in English | MEDLINE | ID: mdl-28766825

ABSTRACT

Bromodomain and extra-terminal domain (BET) inhibitors are widely used both as chemical tools to study the biological role of their targets in living organisms and as candidates for drug development against several cancer variants and human disorders. However, non-BET bromodomains such as those in p300 and CBP are less studied. XDM-CBP is a highly potent and selective inhibitor for the bromodomains of CBP and p300 derived from a pan-selective BET BRD-binding fragment. Along with X-ray crystal-structure analysis and thermodynamic profiling, XDM-CBP was used in screenings of several cancer cell lines in vitro to study its inhibitory potential on cancer cell proliferation. XDM-CBP is demonstrated to be a potent and selective CBP/p300 inhibitor that acts on specific cancer cell lines, in particular malignant melanoma, breast cancer, and leukemia.

7.
J Struct Biol ; 193(3): 157-161, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26780475

ABSTRACT

Septins are a conserved family of GTP-binding proteins that assemble into a highly ordered array of filaments at the mother bud neck in Saccharomyces cerevisiae cells. Many molecular functions and mechanisms of the septins in S. cerevisiae were already uncovered. However, structural information is only available from modeling the crystallized subunits of the human septins into the EM cryomicroscopy data of the yeast hetero-octameric septin rod. Octameric rods are the building block of septin filaments in yeast. We present here the first crystal structure of Cdc11, the terminal subunit of the octameric rod and discuss its structure in relation to its human homologues. Size exclusion chromatography analysis revealed that Cdc11 forms homodimers through its C-terminal coiled coil tail.


Subject(s)
Cell Cycle Proteins/chemistry , Cytoskeletal Proteins/chemistry , GTP-Binding Proteins/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Septins/chemistry , Cryoelectron Microscopy , Crystallography, X-Ray , Guanosine Triphosphate/chemistry , Humans , Protein Binding , Protein Conformation , Protein Multimerization , Saccharomyces cerevisiae/chemistry , Septins/metabolism
8.
J Am Chem Soc ; 138(1): 239-47, 2016 Jan 13.
Article in English | MEDLINE | ID: mdl-26654855

ABSTRACT

The two-component metalloprotein nitrogenase catalyzes the reductive fixation of atmospheric dinitrogen into bioavailable ammonium in diazotrophic prokaryotes. The process requires an efficient energy metabolism, so that although the metal clusters of nitrogenase rapidly decompose in the presence of dioxygen, many free-living diazotrophs are obligate aerobes. In order to retain the functionality of the nitrogen-fixing enzyme, some of these are able to rapidly "switch-off" nitrogenase, by shifting the enzyme into an inactive but oxygen-tolerant state. Under these conditions the two components of nitrogenase form a stable, ternary complex with a small [2Fe:2S] ferredoxin termed FeSII or the "Shethna protein II". Here we have produced and isolated Azotobacter vinelandii FeS II and have determined its three-dimensional structure to 2.1 Å resolution by X-ray diffraction. In the crystals, the dimeric protein was present in two distinct states that differ in the conformation of an extended loop in close proximity to the iron-sulfur cluster. We show that this rearrangement is redox-dependent and forms the molecular basis for oxygen-dependent conformational protection of nitrogenase. Protection assays highlight that FeSII binds to a preformed complex of MoFe and Fe protein upon activation, primarily through electrostatic interactions. The surface properties and known complexes of nitrogenase component proteins allow us to propose a model of the conformationally protected ternary complex of nitrogenase.


Subject(s)
Bacterial Proteins/chemistry , Iron-Sulfur Proteins/chemistry , Nitrogenase/chemistry , Oxygen/chemistry , Amino Acid Sequence , Models, Molecular , Molecular Sequence Data , Protein Conformation , Sequence Homology, Amino Acid , X-Ray Diffraction
9.
Opt Express ; 24(8): 8539-46, 2016 Apr 18.
Article in English | MEDLINE | ID: mdl-27137291

ABSTRACT

The implementation and engineering of bright and coherent solid state quantum light sources is key for the realization of both on chip and remote quantum networks. Despite tremendous efforts for more than 15 years, the combination of these two key prerequisites in a single, potentially scalable device is a major challenge. Here, we report on the observation of bright single photon emission generated via pulsed, resonance fluorescence conditions from a single quantum dot (QD) deterministically centered in a micropillar cavity device via cryogenic optical lithography. The brightness of the QD fluorescence is greatly enhanced on resonance with the fundamental mode of the pillar, leading to an overall device efficiency of η = (74 ± 4) % for a single photon emission as pure as g(2)(0) = 0.0092 ± 0.0004. The combination of large Purcell enhancement and resonant pumping conditions allows us to observe a two-photon wave packet overlap up to ν = (88 ± 3) %.

10.
Angew Chem Int Ed Engl ; 55(50): 15531-15534, 2016 12 12.
Article in English | MEDLINE | ID: mdl-27874239

ABSTRACT

Phloroglucinol reductases (PGRs) are involved in anaerobic degradation in bacteria, in which they catalyze the dearomatization of phloroglucinol into dihydrophloroglucinol. We identified three PGRs, from different bacterial species, that are members of the family of NAD(P)H-dependent short-chain dehydrogenases/reductases (SDRs). In addition to catalyzing the reduction of the physiological substrate, the three enzymes exhibit activity towards 2,4,6-trihydroxybenzaldehyde, 2,4,6-trihydroxyacetophenone, and methyl 2,4,6-trihydroxybenzoate. Structural elucidation of PGRcl and comparison to known SDRs revealed a high degree of conservation. Several amino acid positions were identified as being conserved within the PGR subfamily and might be involved in substrate differentiation. The results enable the enzymatic dearomatization of monoaromatic phenol derivatives and provide insight into the functional diversity that may be found in families of enzymes displaying a high degree of structural homology.


Subject(s)
Bacteria/enzymology , Oxidoreductases Acting on CH-CH Group Donors/chemistry , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Acetophenones/metabolism , Bacteria/chemistry , Bacteria/metabolism , Benzaldehydes/metabolism , Biocatalysis , Gallic Acid/analogs & derivatives , Gallic Acid/metabolism , Protein Conformation , Substrate Specificity
11.
Angew Chem Int Ed Engl ; 55(6): 2252-6, 2016 Feb 05.
Article in English | MEDLINE | ID: mdl-26748890

ABSTRACT

Sirtuins are NAD(+)-dependent protein deacylases that cleave off acetyl groups, as well as other acyl groups, from the ɛ-amino group of lysines in histones and other substrate proteins. Dysregulation of human Sirt2 activity has been associated with the pathogenesis of cancer, inflammation, and neurodegeneration, thus making Sirt2 a promising target for pharmaceutical intervention. Here, based on a crystal structure of Sirt2 in complex with an optimized sirtuin rearranging ligand (SirReal) that shows improved potency, water solubility, and cellular efficacy, we present the development of the first Sirt2-selective affinity probe. A slow dissociation of the probe/enzyme complex offers new applications for SirReals, such as biophysical characterization, fragment-based screening, and affinity pull-down assays. This possibility makes the SirReal probe an important tool for studying sirtuin biology.


Subject(s)
Molecular Probes/analysis , Molecular Probes/chemistry , Sirtuin 2/analysis , Sirtuin 2/chemistry , Crystallography, X-Ray , Humans , Ligands , Models, Molecular , Molecular Probes/chemical synthesis , Molecular Structure , Sirtuin 2/metabolism , Solubility , Structure-Activity Relationship
12.
Opt Express ; 23(26): 32977-85, 2015 Dec 28.
Article in English | MEDLINE | ID: mdl-26831965

ABSTRACT

We report on the observation of bright emission of single photons under pulsed resonance fluorescence conditions from a single quantum dot (QD) in a micropillar cavity. The brightness of the QD fluorescence is greatly enhanced via the coupling to the fundamental mode of a micropillar, allowing us to determine a single photon extraction efficiency of (20.7 ± 0.8) % per linear polarization basis. This yields an overall extraction efficiency of (41.4 ± 1.5) % in our device. We observe the first Rabi-oscillation in a weakly coupled quantum dot-micropillar system under coherent pulsed optical excitation, which enables us to deterministically populate the excited QD state. In this configuration, we probe the single photon statistics of the device yielding g(2)(0) = 0.072 ± 0.011 at a QD-cavity detuning of 75 µeV.

13.
Angew Chem Int Ed Engl ; 54(9): 2811-5, 2015 Feb 23.
Article in English | MEDLINE | ID: mdl-25581707

ABSTRACT

The structures of the O-glycosyltransferase LanGT2 and the engineered, C-C bond-forming variant LanGT2S8Ac show how the replacement of a single loop can change the functionality of the enzyme. Crystal structures of the enzymes in complex with a nonhydrolyzable nucleotide-sugar analogue revealed that there is a conformational transition to create the binding sites for the aglycon substrate. This induced-fit transition was explored by molecular docking experiments with various aglycon substrates.


Subject(s)
Glycosyltransferases/metabolism , Crystallography, X-Ray , Glycosylation , Glycosyltransferases/chemistry , Molecular Docking Simulation , Protein Conformation , Protein Engineering
14.
J Struct Biol ; 186(3): 335-48, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24631970

ABSTRACT

The design and selection of peptides targeting cellular proteins is challenging and often yields candidates with undesired properties. Therefore we deployed a new selection system based on the twin-arginine translocase (TAT) pathway of Escherichia coli, named hitchhiker translocation (HiT) selection. A pool of α-helix encoding sequences was designed and selected for interference with the coiled coil domain (CC) of a melanoma-associated basic-helix-loop-helix-leucine-zipper (bHLHLZ) protein, the microphthalmia associated transcription factor (MITF). One predominant sequence (iM10) was enriched during selection and showed remarkable protease resistance, high solubility and thermal stability while maintaining its specificity. Furthermore, it exhibited nanomolar range affinity towards the target peptide. A mutation screen indicated that target-binding helices of increased homodimer stability and improved expression rates were preferred in the selection process. The crystal structure of the iM10/MITF-CC heterodimer (2.1Å) provided important structural insights and validated our design predictions. Importantly, iM10 did not only bind to the MITF coiled coil, but also to the markedly more stable HLHLZ domain of MITF. Characterizing the selected variants of the semi-rational library demonstrated the potential of the innovative bacterial selection approach.


Subject(s)
Escherichia coli Proteins/chemistry , Membrane Transport Proteins/chemistry , Microphthalmia-Associated Transcription Factor/chemistry , Protein Engineering/methods , Recombinant Proteins/chemistry , Amino Acid Sequence , Base Sequence , Crystallography, X-Ray , Endopeptidase K/metabolism , Escherichia coli Proteins/genetics , Leucine Zippers , Membrane Transport Proteins/genetics , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Models, Molecular , Molecular Sequence Data , Mutation , Peptide Library , Protein Multimerization , Protein Stability , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Solubility
15.
Extremophiles ; 18(4): 641-51, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24794033

ABSTRACT

4-Hydroxyphenylpyruvate dioxygenase (Hpd, EC 1.13.11.27) catalyzes the conversion of 4-hydroxyphenylpyruvate into homogentisate in the second step of oxidative tyrosine catabolism. This pathway is known from bacteria and eukaryotes, but so far no archaeal Hpd has been described. Here, we report the biochemical characterization of an Hpd from the extremophilic archaeon Picrophilus torridus (Pt_Hpd), together with its three-dimensional structure at a resolution of 2.6 Å. Two pH optima were observed at 50 °C: pH 4.0 (close to native conditions) and pH 7.0. The enzyme showed only moderate thermostability and was inactivated with a half-life of ~1.5 h even under optimal reaction conditions. At the ideal physiological growth conditions of P. torridus, Pt_Hpd was inactive after 1 h, showing that the enzyme is protected in vivo from denaturation and/or is only partially adapted to the harsh environmental conditions in the cytosol of P. torridus. The influence of different additives on the activity was investigated. Pt_Hpd exhibited a turnover number k(cat) of 9.9 ± 0.6 s(-1) and a substrate binding affinity K(m) of 142 ± 23 µM. In addition, substrate inhibition with a binding affinity K(i) of 1.9 ± 0.3 mM was observed. Pt_Hpd is compared with isoenzymes from other species and the putative bacterial origin of the gene is discussed.


Subject(s)
4-Hydroxyphenylpyruvate Dioxygenase/chemistry , Archaeal Proteins/chemistry , Thermoplasmales/enzymology , 4-Hydroxyphenylpyruvate Dioxygenase/genetics , 4-Hydroxyphenylpyruvate Dioxygenase/metabolism , Amino Acid Sequence , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Cloning, Molecular , Kinetics , Molecular Sequence Data , Phylogeny , Protein Conformation
16.
Commun Biol ; 7(1): 48, 2024 01 06.
Article in English | MEDLINE | ID: mdl-38184752

ABSTRACT

The septins of the yeast Saccharomyces cerevisiae assemble into hetero-octameric rods by alternating interactions between neighboring G-domains or N- and C-termini, respectively. These rods polymerize end to end into apolar filaments, forming a ring beneath the prospective new bud that expands during the cell cycle into an hourglass structure. The hourglass finally splits during cytokinesis into a double ring. Understanding these transitions as well as the plasticity of the higher order assemblies requires a detailed knowledge of the underlying structures. Here we present the first X-ray crystal structure of a tetrameric Shs1-Cdc12-Cdc3-Cdc10 complex at a resolution of 3.2 Å. Close inspection of the NC-interfaces of this and other septin structures reveals a conserved contact motif that is essential for NC-interface integrity of yeast and human septins in vivo and in vitro. Using the tetrameric structure in combination with AlphaFold-Multimer allowed us to propose a model of the octameric septin rod.


Subject(s)
Saccharomyces cerevisiae , Septins , Humans , Cell Division , Cell Cycle , Cytokinesis
17.
BMC Struct Biol ; 13: 22, 2013 Oct 18.
Article in English | MEDLINE | ID: mdl-24134203

ABSTRACT

BACKGROUND: Methionine adenosyltransferases catalyse the synthesis of S-adenosylmethionine, a cofactor abundant in all domains of life. In contrast to the enzymes from bacteria and eukarya that show high sequence similarity, methionine adenosyltransferases from archaea diverge on the amino acid sequence level and only few conserved residues are retained. RESULTS: We describe the initial characterisation and the crystal structure of the methionine adenosyltransferase from the hyperthermophilic archaeon Thermococcus kodakarensis. As described for other archaeal methionine adenosyltransferases the enzyme is a dimer in solution and shows high temperature stability. The overall structure is very similar to that of the bacterial and eukaryotic enzymes described, with some additional features that might add to the stability of the enzyme. Compared to bacterial and eukaryotic structures, the active site architecture is largely conserved, with some variation in the substrate/product-binding residues. A flexible loop that was not fully ordered in previous structures without ligands in the active side is clearly visible and forms a helix that leaves an entrance to the active site open. CONCLUSIONS: The similar three-dimensional structures of archaeal and bacterial or eukaryotic methionine adenosyltransferases support that these enzymes share an early common ancestor from which they evolved independently, explaining the low similarity in their amino acid sequences. Furthermore, methionine adenosyltransferase from T. kodakarensis is the first structure without any ligands bound in the active site where the flexible loop covering the entrance to the active site is fully ordered, supporting a mechanism postulated earlier for the methionine adenosyltransferase from E. coli. The structure will serve as a starting point for further mechanistic studies and permit the generation of enzyme variants with different characteristics by rational design.


Subject(s)
Methionine Adenosyltransferase/chemistry , Methionine Adenosyltransferase/metabolism , S-Adenosylmethionine/metabolism , Thermococcus/enzymology , Amino Acid Sequence , Catalytic Domain , Circular Dichroism , Cloning, Molecular , Enzyme Stability , Escherichia coli/enzymology , Escherichia coli/genetics , Methionine Adenosyltransferase/genetics , Models, Molecular , Protein Conformation , Protein Multimerization , Protein Structure, Tertiary , Sequence Alignment , Thermococcus/genetics
18.
Biochemistry ; 51(13): 2747-56, 2012 Apr 03.
Article in English | MEDLINE | ID: mdl-22417533

ABSTRACT

The metal-reducing δ-proteobacterium Geobacter sulfurreducens produces a large number of c-type cytochromes, many of which have been implicated in the transfer of electrons to insoluble metal oxides. Among these, the dihemic MacA was assigned a central role. Here we have produced G. sulfurreducens MacA by recombinant expression in Escherichia coli and have solved its three-dimensional structure in three different oxidation states. Sequence comparisons group MacA into the family of diheme cytochrome c peroxidases, and the protein indeed showed hydrogen peroxide reductase activity with ABTS(-2) as an electron donor. The observed K(M) was 38.5 ± 3.7 µM H(2)O(2) and v(max) was 0.78 ± 0.03 µmol of H(2)O(2)·min(-1)·mg(-1), resulting in a turnover number k(cat) = 0.46 · s(-1). In contrast, no Fe(III) reductase activity was observed. MacA was found to display electrochemical properties similar to other bacterial diheme peroxidases, in addition to the ability to electrochemically mediate electron transfer to the soluble cytochrome PpcA. Differences in activity between CcpA and MacA can be rationalized with structural variations in one of the three loop regions, loop 2, that undergoes conformational changes during reductive activation of the enzyme. This loop is adjacent to the active site heme and forms an open loop structure rather than a more rigid helix as in CcpA. For the activation of the protein, the loop has to displace the distal ligand to the active site heme, H93, in loop 1. A H93G variant showed an unexpected formation of a helix in loop 2 and disorder in loop 1, while a M297H variant that altered the properties of the electron transfer heme abolished reductive activation.


Subject(s)
Cytochrome-c Peroxidase/metabolism , Geobacter/enzymology , Base Sequence , Biocatalysis , Cytochrome-c Peroxidase/chemistry , Cytochrome-c Peroxidase/genetics , DNA Primers , Electrochemistry , Models, Molecular , Mutagenesis, Site-Directed , Oxidation-Reduction
19.
Bioorg Med Chem Lett ; 22(1): 271-7, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22153941

ABSTRACT

Directed screening has identified a novel series of non-zinc binding MMP13 inhibitors that possess good levels of activity whilst demonstrating excellent selectivity over related MMPs. A lead optimisation campaign has delivered compounds with enhanced MMP13 potency, good selectivity and acceptable bioavailability profiles leading to a predicted twice-a-day dosing regimen in man.


Subject(s)
Chemistry, Pharmaceutical/methods , Enzyme Inhibitors/pharmacology , Matrix Metalloproteinase 13/chemistry , Matrix Metalloproteinase Inhibitors , Zinc/chemistry , Animals , Dogs , Drug Design , Humans , Inhibitory Concentration 50 , Models, Chemical , Models, Molecular , Osteoarthritis/drug therapy , Rats , Solubility , Structure-Activity Relationship
20.
Bioorg Med Chem Lett ; 22(12): 3879-83, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22608965

ABSTRACT

A novel, potent and selective quinazolinone series of inhibitors of p38α MAP kinase has been identified. Modifications designed to address the issues of poor aqueous solubility and high plasma protein binding as well as embedded aniline functionalities resulted in the identification of a clinical candidate N-cyclopropyl-4-methyl-3-[6-(4-methylpiperazin-1-yl)-4-oxoquinazolin-3(4H)-yl]benzamide (AZD6703). Optimisation was guided by understanding of the binding modes from X-ray crystallographic studies which showed a switch from DFG 'out' to DFG 'in' as the inhibitor size was reduced to improve overall properties.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Piperazines/chemical synthesis , Protein Kinase Inhibitors/chemical synthesis , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Blood Proteins/chemistry , Crystallography, X-Ray , Dogs , Drug Discovery , Humans , Inflammation/drug therapy , Mitogen-Activated Protein Kinase 14/metabolism , Models, Molecular , Molecular Weight , Piperazines/chemistry , Piperazines/pharmacology , Protein Binding , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Rats , Solubility , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL