Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
iScience ; 27(4): 109458, 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38571760

ABSTRACT

Histone chaperones-structurally diverse, non-catalytic proteins enriched with acidic intrinsically disordered regions (IDRs)-protect histones from spurious nucleic acid interactions and guide their deposition into and out of nucleosomes. Despite their conservation and ubiquity, the function of the chaperone acidic IDRs remains unclear. Here, we show that the Xenopus laevis Npm2 and Nap1 acidic IDRs are substrates for TTLL4 (Tubulin Tyrosine Ligase Like 4)-catalyzed post-translational glutamate-glutamylation. We demonstrate that to bind, stabilize, and deposit histones into nucleosomes, chaperone acidic IDRs function as DNA mimetics. Our biochemical, computational, and biophysical studies reveal that glutamylation of these chaperone polyelectrolyte acidic stretches functions to enhance DNA electrostatic mimicry, promoting the binding and stabilization of H2A/H2B heterodimers and facilitating nucleosome assembly. This discovery provides insights into both the previously unclear function of the acidic IDRs and the regulatory role of post-translational modifications in chromatin dynamics.

2.
bioRxiv ; 2023 Sep 19.
Article in English | MEDLINE | ID: mdl-37790377

ABSTRACT

Histone chaperones-structurally diverse, non-catalytic proteins enriched with acidic intrinsically disordered regions (IDRs)-protect histones from spurious nucleic acid interactions and guide their deposition into and out of nucleosomes. Despite their conservation and ubiquity, the function of the chaperone acidic IDRs remains unclear. Here, we show that the Xenopus laevis Npm2 and Nap1 acidic IDRs are substrates for TTLL4 (Tubulin Tyrosine Ligase Like 4)-catalyzed post-translational glutamate-glutamylation. We demonstrate that, to bind, stabilize, and deposit histones into nucleosomes, chaperone acidic IDRs function as DNA mimetics. Our biochemical, computational, and biophysical studies reveal that glutamylation of these chaperone polyelectrolyte acidic stretches functions to enhance DNA electrostatic mimicry, promoting the binding and stabilization of H2A/H2B heterodimers and facilitating nucleosome assembly. This discovery provides insights into both the previously unclear function of the acidic IDRs and the regulatory role of post-translational modifications in chromatin dynamics.

3.
iScience ; 24(9): 102971, 2021 Sep 24.
Article in English | MEDLINE | ID: mdl-34505004

ABSTRACT

Protein arginine methyltransferases (PRMTs) catalyze the post-translational monomethylation (Rme1), asymmetric (Rme2a), or symmetric (Rme2s) dimethylation of arginine. To determine the cellular consequences of type I (Rme2a) and II (Rme2s) PRMTs, we developed and integrated multiple approaches. First, we determined total cellular dimethylarginine levels, revealing that Rme2s was ∼3% of total Rme2 and that this percentage was dependent upon cell type and PRMT inhibition status. Second, we quantitatively characterized in vitro substrates of the major enzymes and expanded upon PRMT substrate recognition motifs. We also compiled our data with publicly available methylarginine-modified residues into a comprehensive database. Third, we inhibited type I and II PRMTs and performed proteomic and transcriptomic analyses to reveal their phenotypic consequences. These experiments revealed both overlapping and independent PRMT substrates and cellular functions. Overall, this study expands upon PRMT substrate diversity, the arginine methylome, and the complex interplay of type I and II PRMTs.

4.
J Biol Chem ; 294(35): 13158-13170, 2019 08 30.
Article in English | MEDLINE | ID: mdl-31315931

ABSTRACT

Iron-sulfur clusters are protein cofactors with an ancient evolutionary origin. These clusters are best known for their roles in redox proteins such as ferredoxins, but some iron-sulfur clusters have nonredox roles in the active sites of enzymes. Such clusters are often prone to oxidative degradation, making the enzymes difficult to characterize. Here we report a structural and functional characterization of dihydroxyacid dehydratase (DHAD) from Mycobacterium tuberculosis (Mtb), an essential enzyme in the biosynthesis of branched-chain amino acids. Conducting this analysis under fully anaerobic conditions, we solved the DHAD crystal structure, at 1.88 Å resolution, revealing a 2Fe-2S cluster in which one iron ligand is a potentially exchangeable water molecule or hydroxide. UV and EPR spectroscopy both suggested that the substrate binds directly to the cluster or very close to it. Kinetic analysis implicated two ionizable groups in the catalytic mechanism, which we postulate to be Ser-491 and the iron-bound water/hydroxide. Site-directed mutagenesis showed that Ser-491 is essential for activity, and substrate docking indicated that this residue is perfectly placed for proton abstraction. We found that a bound Mg2+ ion 6.5 Å from the 2Fe-2S cluster plays a key role in substrate binding. We also identified a putative entry channel that enables access to the cluster and show that Mtb-DHAD is inhibited by a recently discovered herbicide, aspterric acid, that, given the essentiality of DHAD for Mtb survival, is a potential lead compound for the design of novel anti-TB drugs.


Subject(s)
Amino Acids, Branched-Chain/biosynthesis , Hydro-Lyases/metabolism , Iron-Sulfur Proteins/metabolism , Mycobacterium tuberculosis/chemistry , Amino Acids, Branched-Chain/chemistry , Binding Sites , Hydro-Lyases/chemistry , Iron-Sulfur Proteins/chemistry , Models, Molecular , Molecular Conformation , Mycobacterium tuberculosis/metabolism
5.
Biochemistry ; 56(31): 4044-4052, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28700220

ABSTRACT

l-Aspartate oxidase, encoded by the nadB gene, is the first enzyme in the de novo synthesis of NAD+ in bacteria. This FAD-dependent enzyme catalyzes the oxidation of l-aspartate to generate iminoaspartate and reduced flavin. Distinct from most amino acid oxidases, it can use either molecular oxygen or fumarate to reoxidize the reduced enzyme. Sequence alignments and the three-dimensional crystal structure have revealed that the overall fold and catalytic residues of NadB closely resemble those of the succinate dehydrogenase/fumarate reductase family rather than those of the prototypical d-amino acid oxidases. This suggests that the enzyme can catalyze amino acid oxidation via typical amino acid oxidase chemistry, involving the removal of protons from the α-amino group and the transfer of the hydride from C2, or potentially deprotonation at C3 followed by transfer of the hydride from C2, similar to chemistry occurring during succinate oxidation. We have investigated this potential mechanistic ambiguity using a combination of primary, solvent, and multiple deuterium kinetic isotope effects in steady state experiments. Our results indicate that the chemistry is similar to that of typical amino acid oxidases in which the transfer of the hydride from C2 of l-aspartate to FAD is rate-limiting and occurs in a concerted manner with respect to deprotonation of the α-amine. Together with previous kinetic and structural data, we propose that NadB has structurally evolved from succinate dehydrogenase/fumarate reductase-type enzymes to gain the new functionality of oxidizing amino acids while retaining the ability to reduce fumarate.


Subject(s)
Amino Acid Oxidoreductases/metabolism , Aspartic Acid/metabolism , Coenzymes/metabolism , Escherichia coli K12/enzymology , Escherichia coli Proteins/metabolism , Flavin-Adenine Dinucleotide/metabolism , Models, Molecular , Algorithms , Amino Acid Oxidoreductases/chemistry , Amino Acid Oxidoreductases/genetics , Animals , Aspartic Acid/chemistry , Binding Sites , Biocatalysis , Catalytic Domain , Coenzymes/chemistry , Deuterium Exchange Measurement , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Flavin-Adenine Dinucleotide/chemistry , Hydrogen-Ion Concentration , Kinetics , Malate Dehydrogenase/metabolism , Oxidation-Reduction , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sus scrofa
6.
Infect Immun ; 85(4)2017 04.
Article in English | MEDLINE | ID: mdl-28115505

ABSTRACT

Tuberculosis (TB) due to Mycobacterium tuberculosis remains a major global infectious disease problem, and a more efficacious vaccine is urgently needed for the control and prevention of disease caused by this organism. We previously reported that a genetically modified strain of Mycobacterium smegmatis called IKEPLUS is a promising TB vaccine candidate. Since protective immunity induced by IKEPLUS is dependent on antigen-specific CD4+ T cell memory, we hypothesized that the specificity of the CD4+ T cell response was a critical feature of this protection. Using in vitro assays of interferon gamma production (enzyme-linked immunosorbent spot [ELISPOT] assays) by splenocytes from IKEPLUS-immunized C57BL/6J mice, we identified an immunogenic peptide within the mycobacterial ribosomal large subunit protein RplJ, encoded by the Rv0651 gene. In a complementary approach, we generated major histocompatibility complex (MHC) class II-restricted T cell hybridomas from IKEPLUS-immunized mice. Screening of these T cell hybridomas against IKEPLUS and ribosomes enriched from IKEPLUS suggested that the CD4+ T cell response in IKEPLUS-immunized mice was dominated by the recognition of multiple components of the mycobacterial ribosome. Importantly, CD4+ T cells specific for mycobacterial ribosomes accumulate to significant levels in the lungs of IKEPLUS-immunized mice following aerosol challenge with virulent M. tuberculosis, consistent with a role for these T cells in protective host immunity in TB. The identification of CD4+ T cell responses to defined ribosomal protein epitopes expands the range of antigenic targets for adaptive immune responses to M. tuberculosis and may help to inform the design of more effective vaccines against tuberculosis.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Proteins/immunology , CD4-Positive T-Lymphocytes/immunology , Mycobacterium/immunology , Tuberculosis/immunology , Tuberculosis/microbiology , Amino Acid Sequence , Animals , Antigens, Bacterial/chemistry , Bacterial Proteins/chemistry , CD4-Positive T-Lymphocytes/metabolism , Cytokines/metabolism , Disease Models, Animal , Epitope Mapping , Epitopes, T-Lymphocyte/immunology , Female , Histocompatibility Antigens Class II/chemistry , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Immunization , Mice , Mycobacterium/pathogenicity , Peptides/chemistry , Peptides/immunology , Ribosomal Proteins/immunology , T-Cell Antigen Receptor Specificity/immunology , Tuberculosis/mortality , Virulence
7.
Biochemistry ; 55(45): 6295-6303, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27780341

ABSTRACT

The biosynthetic pathway of the branched-chain amino acids is essential for Mycobacterium tuberculosis growth and survival. We report here the kinetic and chemical mechanism of the pyridoxal 5'-phosphate (PLP)-dependent branched-chain aminotransferase, IlvE, from M. tuberculosis (MtIlvE). This enzyme is responsible for the final step of the synthesis of the branched-chain amino acids isoleucine, leucine, and valine. As seen in other aminotransferases, MtIlvE displays a ping-pong kinetic mechanism. pK values were identified from the pH dependence on V as well as V/K, indicating that the phosphate ester of the PLP cofactor, and the α-amino group from l-glutamate and the active site Lys204, play roles in acid-base catalysis and binding, respectively. An intrinsic primary kinetic isotope effect was identified for the α-C-H bond cleavage of l-glutamate. Large solvent kinetic isotope effect values for the ping and pong half-reactions were also identified. The absence of a quininoid intermediate in combination with the Dkobs in our multiple kinetic isotope effects under single-turnover conditions suggests a concerted type of mechanism. The deprotonation of C2 of l-glutamate and the protonation of C4' of the PLP cofactor happen synchronously in the ping half-reaction. A chemical mechanism is proposed on the basis of the results obtained here.


Subject(s)
Bacterial Proteins/chemistry , Mycobacterium tuberculosis/enzymology , Protein Conformation , Transaminases/chemistry , Bacterial Proteins/metabolism , Biocatalysis , Biosynthetic Pathways , Catalytic Domain , Glutamic Acid/chemistry , Glutamic Acid/metabolism , Hydrogen-Ion Concentration , Isoleucine/chemistry , Isoleucine/metabolism , Kinetics , Leucine/chemistry , Leucine/metabolism , Lysine/chemistry , Lysine/metabolism , Models, Chemical , Models, Molecular , Molecular Structure , Pyridoxal Phosphate/chemistry , Pyridoxal Phosphate/metabolism , Transaminases/metabolism , Valine/chemistry , Valine/metabolism
8.
Transgenic Res ; 24(5): 783-90, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26264890

ABSTRACT

The low-level presence (LLP) of genetically engineered (GE) seeds that have been approved in the country of origin but not the country of import presents challenges for regulators in both seed importing and exporting countries, as well as for the international seed trade and the farmers who rely on it. In addition to legal, financial and regulatory challenges, such LLP situations in seed may also require an environmental risk assessment by the country of import. Such assessments have typically been informed by the national framework established to support decisions related to wide scale cultivation, and frequently do not take into account the low environmental exposure and prior regulatory history of the GE plant. In addition, such assessment processes may not be well suited to the decision-making timeframe that is necessary when dealing with an LLP situation in imported seed. In order to facilitate regulatory decision making, this paper proposes a set of scientific criteria for identifying GE crop plants that are expected to pose a low or negligible risk to the environment under LLP conditions in seed. Regulatory decision makers in some importing countries may decide to use these criteria to assist in risk analysis associated with LLP situations they are experiencing or could experience in the future, and might choose to proactively apply the criteria to identify existing GE plants with regulatory approvals in other countries that would be expected to pose low risk under conditions of LLP in seed.


Subject(s)
Environmental Health , Plants, Genetically Modified , Seeds/genetics , Risk Assessment
9.
Biochemistry ; 50(26): 5883-92, 2011 Jul 05.
Article in English | MEDLINE | ID: mdl-21627103

ABSTRACT

Recent proteomics studies have revealed that protein acetylation is an abundant and evolutionarily conserved post-translational modification from prokaryotes to eukaryotes. Although an astonishing number of acetylated proteins have been identified in those studies, the acetyltransferases that target these proteins remain largely unknown. Here we characterized MSMEG_5458, one of the GCN5-related N-acetyltransferases (GNAT's) in Mycobacterium smegmatis, and show that it is a protein acetyltransferase (MsPat) that specifically acetylates the ε-amino group of a highly conserved lysine residue in acetyl-CoA synthetase (ACS) with a k(cat)/K(m) of nearly 10(4) M(-1) s(-1). This acetylation results in the inactivation of ACS activity. Lysine acetylation by MsPat is dependent on 3',5'-cyclic adenosine monophosphate (cAMP), an important second messenger, indicating that MsPat is a downstream target of the intracellular cAMP signaling pathway. To the best of our knowledge, this is the first protein acetyltransferase in mycobacteria that both is dependent on cAMP and targets a central metabolic enzyme by a specific post-translational modification. Since cAMP is synthesized by adenylate cyclases (AC's) that sense various environmental signals, we hypothesize that the acetylation and inactivation of ACS is important for mycobacteria to adjust to environmental changes. In addition, we show that Rv1151c, a sirtuin-like deacetylase in Mycobacterium tuberculosis, reactivates acetylated ACS through an NAD(+)-dependent deacetylation. Therefore, Pat and the sirtuin-like deacetylase in mycobacteria constitute a reversible acetylation system that regulates the activity of ACS.


Subject(s)
Acetate-CoA Ligase/metabolism , Cyclic AMP/metabolism , Mycobacterium tuberculosis/enzymology , Acetate-CoA Ligase/chemistry , Acetylation , Amino Acid Sequence , Amino Acids/metabolism , Carbohydrate Metabolism , Enzyme Activation , Histone Acetyltransferases/chemistry , Histone Acetyltransferases/metabolism , Lysine , Molecular Sequence Data , Mycobacterium smegmatis/enzymology , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Sirtuins/chemistry , Sirtuins/metabolism
10.
J Biol Chem ; 286(28): 25265-73, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21597116

ABSTRACT

QnrB1 is a plasmid-encoded pentapeptide repeat protein (PRP) that confers a moderate degree of resistance to fluoroquinolones. Its gene was cloned into an expression vector with an N-terminal polyhistidine tag, and the protein was purified by nickel affinity chromatography. The structure of QnrB1 was determined by a combination of trypsinolysis, surface mutagenesis, and single anomalous dispersion phasing. QnrB1 folds as a right-handed quadrilateral ß-helix with a highly asymmetric dimeric structure typical of PRP-topoisomerase poison resistance factors. The threading of pentapeptides into the ß-helical fold is interrupted by two noncanonical PRP sequences that produce outward projecting loops that interrupt the regularity of the PRP surface. Deletion of the larger upper loop eliminated the protective effect of QnrB1 on DNA gyrase toward inhibition by quinolones, whereas deletion of the smaller lower loop drastically reduced the protective effect. These loops are conserved among all plasmid-based Qnr variants (QnrA, QnrC, QnrD, and QnrS) and some chromosomally encoded Qnr varieties. A mechanism in which PRP-topoisomerase poison resistance factors bind to and disrupt the quinolone-DNA-gyrase interaction is proposed.


Subject(s)
Bacterial Proteins/chemistry , Drug Resistance, Bacterial/physiology , Enterococcus faecalis/chemistry , Fluoroquinolones/chemistry , Plasmids , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , DNA Gyrase/chemistry , DNA Gyrase/genetics , DNA Gyrase/metabolism , Enterococcus faecalis/genetics , Enterococcus faecalis/metabolism , Fluoroquinolones/pharmacology , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Deletion , Structure-Activity Relationship
11.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 67(Pt 3): 296-302, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21393830

ABSTRACT

The protein AlbG is a self-resistance factor against albicidin, a nonribosomally encoded hybrid polyketide-peptide with antibiotic and phytotoxic properties produced by Xanthomonas albilineans. Primary-sequence analysis indicates that AlbG is a member of the pentapeptide-repeat family of proteins (PRP). The structure of AlbG from X. albilineans was determined at 2.0 Šresolution by SAD phasing using data collected from a single trimethyllead acetate derivative on a home source. AlbG folds into a right-handed quadrilateral ß-helix composed of approximately eight semi-regular coils. The regularity of the ß-helix is blemished by a large loop/deviation in the ß-helix between coils 4 and 5. The C-terminus of the ß-helix is capped by a dimerization module, yielding a dimer with a 110 Šsemi-collinear ß-helical axis. This method of dimer formation appears to be common to all PRP proteins that confer resistance to topoisomerase poisons and contrasts with most PRP proteins, which are typically monomeric.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Oligopeptides/chemistry , Oligopeptides/genetics , Protein Structure, Secondary , R Factors/chemistry , R Factors/genetics , Amino Acid Sequence , Bacterial Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Oligopeptides/metabolism , Organic Chemicals/chemistry , Organic Chemicals/metabolism , Protein Multimerization , R Factors/metabolism , Xanthomonas/chemistry , Xanthomonas/genetics , Xanthomonas/metabolism
12.
Antimicrob Agents Chemother ; 55(1): 110-7, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20937785

ABSTRACT

The chromosomally encoded Qnr homolog protein from Enterococcus faecalis (EfsQnr), when expressed, confers to its host a decreased susceptibility to quinolones and consists mainly of tandem repeats, which is consistent with belonging to the pentapeptide repeat family of proteins (PRPs). EfsQnr was cloned with an N-terminal 6× His tag and purified to homogeneity. EfsQnr partially protected DNA gyrase from fluoroquinolone inhibition at concentrations as low as 20 nM. EfsQnr inhibited the ATP-dependent supercoiling activity of DNA gyrase with a 50% inhibitory concentration (IC(50)) of 1.2 µM, while no significant inhibition of ATP-independent relaxation activity was observed. EfsQnr was cytotoxic when overexpressed in Escherichia coli, resulting in the clumping of cells and a loss of viability. The X-ray crystal structure of EfsQnr was determined to 1.6-Å resolution. EfsQnr exhibits the right-handed quadrilateral beta-helical fold typical of PRPs, with features more analogous to MfpA (mycobacterium fluoroquinolone resistance pentapeptide) than to the PRPs commonly found in cyanobacteria.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Proteins/pharmacology , Enterococcus faecalis/metabolism , Topoisomerase II Inhibitors , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Enterococcus faecalis/genetics , Escherichia coli/cytology , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli/ultrastructure , Microbial Viability , Microscopy, Electron, Scanning , Molecular Sequence Data , Protein Multimerization , Protein Structure, Secondary , Sequence Homology, Amino Acid
13.
Biochemistry ; 49(44): 9613-9, 2010 Nov 09.
Article in English | MEDLINE | ID: mdl-20879713

ABSTRACT

Nicotinamidase/pyrazinamidase (PncA) is involved in the NAD+ salvage pathway of Mycobacterium tuberculosis and other bacteria. In addition to hydrolyzing nicotinamide into nicotinic acid, PncA also hydrolyzes the prodrug pyrazinamide to generate the active form of the drug, pyrazinoic acid, which is an essential component of the multidrug treatment of TB. A coupled enzymatic activity assay has been developed for PncA that allows for the spectroscopic observation of enzyme activity. The enzyme activity was essentially pH-independent under the conditions tested; however, the measurement of the pH dependence of iodoacetamide alkylation revealed a pK value of 6.6 for the active site cysteine. Solvent deuterium kinetic isotope effects revealed an inverse value for kcat of 0.64, reconfirming the involvement of a thiol group in the mechanism. A mechanism is proposed for PncA catalysis that is similar to the mechanisms proposed for members of the nitrilase superfamily, in which nucleophilic attack by the active site cysteine generates a tetrahedral intermediate that collapses with the loss of ammonia and subsequent hydrolysis of the thioester bond by water completes the cycle. An inhibitor screen identified the competitive inhibitor 3-pyridine carboxaldehyde with a Ki of 290 nM. Additionally, pyrazinecarbonitrile was found to be an irreversible inactivator of PncA, with a kinact/KI of 975 M(−1) s(−1).


Subject(s)
Amidohydrolases/antagonists & inhibitors , Amidohydrolases/metabolism , Antitubercular Agents/chemistry , Antitubercular Agents/pharmacology , Mycobacterium tuberculosis/enzymology , Pyrazinamide/analogs & derivatives , Pyrazinamide/pharmacology , Amidohydrolases/genetics , Amidohydrolases/isolation & purification , Cloning, Molecular , Humans , Mycobacterium bovis/enzymology , Mycobacterium bovis/genetics , Mycobacterium tuberculosis/genetics , Tuberculosis/drug therapy
14.
Nat Chem Biol ; 6(11): 797-9, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20852636

ABSTRACT

The Mycobacterium tuberculosis enzyme Rv2275 catalyzes the formation of cyclo(L-Tyr-L-Tyr) using two molecules of Tyr-tRNA(Tyr) as substrates. The three-dimensional (3D) structure of Rv2275 was determined to 2.0-Å resolution, revealing that Rv2275 is structurally related to the class Ic aminoacyl-tRNA synthetase family of enzymes. Mutagenesis and radioactive labeling suggests a covalent intermediate in which L-tyrosine is transferred from Tyr-tRNA(Tyr) to an active site serine (Ser88) by transesterification with Glu233 serving as a critical base, catalyzing dipeptide bond formation.


Subject(s)
Dipeptides/biosynthesis , Mycobacterium tuberculosis/enzymology , Peptide Synthases/chemistry , Peptide Synthases/metabolism , Peptides, Cyclic/biosynthesis , Tyrosine-tRNA Ligase/chemistry , Biocatalysis , Catalytic Domain , Crystallography, X-Ray , Cyclization , Esterification , Glutamic Acid/chemistry , Glutamic Acid/metabolism , Models, Molecular , Protein Conformation , RNA, Transfer, Tyr/chemistry , RNA, Transfer, Tyr/metabolism , Serine/genetics , Serine/metabolism , Tyrosine/chemistry , Tyrosine/metabolism
15.
Acta Crystallogr D Biol Crystallogr ; 65(Pt 5): 462-9, 2009 May.
Article in English | MEDLINE | ID: mdl-19390151

ABSTRACT

The pentapeptide-repeat protein EfsQnr from Enterococcus faecalis protects DNA gyrase from inhibition by fluoroquinolones. EfsQnr was cloned and purified to homogeneity, but failed to produce diffraction-quality crystals in initial crystallization screens. Treatment of EfsQnr with glutaraldehyde and the strong reducing agent borane-dimethylamine resulted in a derivatized protein which produced crystals that diffracted to 1.6 A resolution; their structure was subsequently determined by single-wavelength anomalous dispersion. Analysis of the derivatized protein using Fourier transform ion cyclotron resonance mass spectrometry indicated a mass increase of 68 Da per free amino group. Electron-density maps about a limited number of structurally ordered lysines indicated that the modification was a cyclic pentylation of free amines, producing piperidine groups.


Subject(s)
Bacterial Proteins/chemistry , Cross-Linking Reagents/pharmacology , Enterococcus faecalis/chemistry , Glutaral/pharmacology , Amines/chemistry , Bacterial Proteins/drug effects , Bacterial Proteins/genetics , Boranes/pharmacology , Crystallization/methods , Crystallography, X-Ray , Dimethylamines/pharmacology , Drug Resistance, Bacterial , Humans , Lysine/chemistry , Lysine/drug effects , Mass Spectrometry/methods , Models, Molecular , Oligopeptides/chemistry , Protein Conformation , Recombinant Fusion Proteins/chemistry , Repetitive Sequences, Amino Acid
16.
J Biol Chem ; 283(52): 36290-9, 2008 Dec 26.
Article in English | MEDLINE | ID: mdl-18977756

ABSTRACT

The pentapeptide repeat is a recently discovered protein fold. Mycobacterium tuberculosis MfpA is a founding member of the pentapeptide repeat protein (PRP) family that confers resistance to the antibiotic fluoroquinolone by binding to DNA gyrase and inhibiting its activity. The size, shape, and surface potential of MfpA mimics duplex DNA. As an initial step in a comprehensive biophysical analysis of the role of PRPs in the regulation of cellular topoisomerase activity and conferring antibiotic resistance, we have explored the solution structure and refolding of MfpA by fluorescence spectroscopy, CD, and analytical centrifugation. A unique CD spectrum for the pentapeptide repeat fold is described. This spectrum reveals a native structure whose beta-strands and turns within the right-handed quadrilateral beta-helix that define the PRP fold differ from canonical secondary structure types. MfpA refolded from urea or guanidium by dialysis or dilution forms stable aggregates of monomers whose secondary and tertiary structure are not native. In contrast, MfpA refolded using a novel "time-dependent renaturation" protocol yields protein with native secondary, tertiary, and quaternary structure. The generality of "time-dependent renaturation" to other proteins and denaturation methods is discussed.


Subject(s)
Bacterial Proteins/metabolism , Mycobacterium tuberculosis/metabolism , Amino Acid Sequence , Circular Dichroism , DNA Topoisomerases, Type I/metabolism , Drug Resistance, Microbial , Guanidine/chemistry , Molecular Sequence Data , Monomeric GTP-Binding Proteins , Protein Conformation , Protein Denaturation , Protein Folding , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Spectrometry, Fluorescence/methods , Urea/chemistry
17.
Biochemistry ; 47(37): 9825-35, 2008 Sep 16.
Article in English | MEDLINE | ID: mdl-18710261

ABSTRACT

Enzymatic modification of aminoglycoside antibiotics mediated by regioselective aminoglycoside N-acetyltransferases is the predominant cause of bacterial resistance to aminoglycosides. A recently discovered bifunctional aminoglycoside acetyltransferase (AAC(6')-Ib variant, AAC(6')-Ib-cr) has been shown to catalyze the acetylation of fluoroquinolones as well as aminoglycosides. We have expressed and purified AAC(6')-Ib-wt and its bifunctional variant AAC(6')-Ib-cr in Escherichia coli and characterized their kinetic and chemical mechanism. Initial velocity and dead-end inhibition studies support an ordered sequential mechanism for the enzyme(s). The three-dimensional structure of AAC(6')-Ib-wt was determined in various complexes with donor and acceptor ligands to resolutions greater than 2.2 A. Observation of the direct, and optimally positioned, interaction between the 6'-NH 2 and Asp115 suggests that Asp115 acts as a general base to accept a proton in the reaction. The structure of AAC(6')-Ib-wt permits the construction of a molecular model of the interactions of fluoroquinolones with the AAC(6')-Ib-cr variant. The model suggests that a major contribution to the fluoroquinolone acetylation activity comes from the Asp179Tyr mutation, where Tyr179 makes pi-stacking interactions with the quinolone ring facilitating quinolone binding. The model also suggests that fluoroquinolones and aminoglycosides have different binding modes. On the basis of kinetic properties, the pH dependence of the kinetic parameters, and structural information, we propose an acid/base-assisted reaction catalyzed by AAC(6')-Ib-wt and the AAC(6')-Ib-cr variant involving a ternary complex.


Subject(s)
Acetyltransferases/chemistry , Acetyltransferases/metabolism , Acetylation , Acetyltransferases/genetics , Aminoglycosides/chemistry , Aminoglycosides/metabolism , Binding Sites , Escherichia coli/genetics , Escherichia coli/metabolism , Fluoroquinolones/chemistry , Fluoroquinolones/metabolism , Hydrogen-Ion Concentration , Kinetics , Models, Molecular , Mutation , Protein Conformation , Structure-Activity Relationship , Substrate Specificity
18.
Biochemistry ; 46(24): 7187-95, 2007 Jun 19.
Article in English | MEDLINE | ID: mdl-17516632

ABSTRACT

The N1-acetylation of spermidine and spermine by spermidine/spermine acetyltransferase (SSAT) is a crucial step in the regulation of the cellular polyamine levels in eukaryotic cells. Altered polyamine levels are associated with a variety of cancers as well as other diseases, and key enzymes in the polyamine pathway, including SSAT, are being explored as potential therapeutic drug targets. We have expressed and purified human SSAT in Escherichia coli and characterized its kinetic and chemical mechanism. Initial velocity and inhibition studies support a random sequential mechanism for the enzyme. The bisubstrate analogue, N1-spermine-acetyl-coenzyme A, exhibited linear, competitive inhibition against both substrates with a true Ki of 6 nM. The pH-activity profile was bell-shaped, depending on the ionization state of two groups exhibiting apparent pKa values of 7.27 and 8.87. The three-dimensional crystal structure of SSAT with bound bisubstrate inhibitor was determined at 2.3 A resolution. The structure of the SSAT-spermine-acetyl-coenzyme A complex suggested that Tyr140 acts as general acid and Glu92, through one or more water molecules, acts as the general base during catalysis. On the basis of kinetic properties, pH dependence, and structural information, we propose an acid/base-assisted reaction catalyzed by SSAT, involving a ternary complex.


Subject(s)
Acetyltransferases/chemistry , Acetyltransferases/metabolism , Acetyl Coenzyme A/analogs & derivatives , Acetyl Coenzyme A/metabolism , Acetyltransferases/genetics , Base Sequence , Crystallography, X-Ray , DNA, Complementary/genetics , Humans , Hydrogen-Ion Concentration , In Vitro Techniques , Kinetics , Models, Molecular , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spermine/analogs & derivatives , Spermine/metabolism , Substrate Specificity
19.
J Hered ; 98(2): 183-7, 2007.
Article in English | MEDLINE | ID: mdl-17400586

ABSTRACT

The closest wild relatives of maize, Zea mays ssp. mays are various Zea taxa known as "teosinte." Hybrids between maize and the teosinte taxon, Zea mays ssp. mexicana, often occur when the 2 are sympatric in Mexico. Measuring the spontaneous hybridization rate of the 2 taxa would shed light on the mechanisms contributing to the evolution and persistence of these hybrid swarms. We conducted a series of field experiments in Riverside, CA, to measure the natural hybridization rates between maize and 2 teosinte taxa, Z. m. ssp. mexicana and Zea mays ssp. parviglumis. We planted teosinte within and near maize plantations. Hybrids were identified by progeny testing for a maize-specific herbicide resistance allele and a teosinte-specific allozyme allele. Hybridity was confirmed by growing putative hybrid progeny to maturity to evaluate whether they had the characteristic morphology of maize x teosinte hybrids. We found that maize and Z. m. ssp. mexicana naturally hybridize at a low rate (<1%), whereas Z. m. ssp. parviglumis hybridizes with the crop at a high rate (>>50%).


Subject(s)
Hybridization, Genetic , Zea mays/genetics , Phenotype
20.
Protein Sci ; 16(4): 755-60, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17384236

ABSTRACT

The Nostoc punctiforme genes Np275 and Np276 are two adjacently encoded proteins of 98 and 75 amino acids in length and exhibit sequences composed of tandem pentapeptide repeats. The structures of Np275 and a fusion of Np275 and Np276 were determined to 2.1 and 1.5 A, respectively. The two Nostoc proteins fold as highly symmetric right-handed quadrilateral beta-helices similar to the mycobacterial protein MfpA implicated in fluoroquinolone resistance and DNA gyrase inhibition. The sequence composition of the intervening coding region and the ability to express a fused protein by removing the stop codon for Np275 suggests Np275 and Np276 were recently part of a larger ancestral pentapeptide repeat protein.


Subject(s)
Bacterial Proteins/chemistry , Nostoc/chemistry , Oligopeptides/chemistry , Amino Acid Sequence , Base Sequence , DNA Primers , Models, Molecular , Molecular Sequence Data , Protein Conformation , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...