Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
mBio ; 12(6): e0267921, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34749534

ABSTRACT

During nutrient limitation, bacteria produce the alarmones (p)ppGpp as effectors of a stress signaling network termed the stringent response. RsgA, RbgA, Era, and HflX are four ribosome-associated GTPases (RA-GTPases) that bind to (p)ppGpp in Staphylococcus aureus. These enzymes are cofactors in ribosome assembly, where they cycle between the ON (GTP-bound) and OFF (GDP-bound) ribosome-associated states. Entry into the OFF state occurs upon hydrolysis of GTP, with GTPase activity increasing substantially upon ribosome association. When bound to (p)ppGpp, GTPase activity is inhibited, reducing 70S ribosome assembly and growth. Here, we determine how (p)ppGpp impacts RA-GTPase-ribosome interactions. We show that RA-GTPases preferentially bind to 5'-diphosphate-containing nucleotides GDP and ppGpp over GTP, which is likely exploited as a regulatory mechanism within the cell to shut down ribosome biogenesis during stress. Stopped-flow fluorescence and association assays reveal that when bound to (p)ppGpp, the association of RA-GTPases to ribosomal subunits is destabilized, both in vitro and within bacterial cells. Consistently, structural analysis of the ppGpp-bound RA-GTPase RsgA reveals an OFF-state conformation similar to the GDP-bound state, with the G2/switch I loop adopting a conformation incompatible with ribosome association. Altogether, we highlight (p)ppGpp-mediated inhibition of RA-GTPases as a major mechanism of stringent response-mediated ribosome assembly and growth control. IMPORTANCE The stringent response is a bacterial signaling network that utilizes the nucleotides pppGpp and ppGpp to reprogram cells in order to survive nutritional stresses. However, much about how these important nucleotides control cellular reprogramming is unknown. Our previous work revealed that (p)ppGpp can bind to and inhibit the enzymatic activity of four ribosome-associated GTPases (RA-GTPases), enzymes that facilitate maturation of the 50S and 30S ribosomal subunits. Here, we examine how this occurs mechanistically and demonstrate that this interaction prevents the accommodation of RA-GTPases on ribosomal subunits both in vitro and within bacterial cells, with the ppGpp-bound state structurally mimicking the inactive GDP-bound conformation of the enzyme. We additionally reveal that these GTPase enzymes have a greater affinity for OFF-state-inducing nucleotides, which is a mechanism likely to control ribosome assembly during growth. With this, we further our understanding of how ribosome function is controlled by (p)ppGpp, enabling bacterial survival during stress.


Subject(s)
Bacterial Proteins/metabolism , GTP Phosphohydrolases/metabolism , Ribosome Subunits/metabolism , Staphylococcus aureus/enzymology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , GTP Phosphohydrolases/chemistry , GTP Phosphohydrolases/genetics , Guanosine Pentaphosphate/chemistry , Guanosine Pentaphosphate/metabolism , Guanosine Tetraphosphate/chemistry , Guanosine Tetraphosphate/metabolism , Models, Molecular , Protein Binding , Ribosome Subunits/chemistry , Ribosome Subunits/genetics , Staphylococcus aureus/chemistry , Staphylococcus aureus/genetics
2.
Mol Cell ; 81(15): 3160-3170.e9, 2021 08 05.
Article in English | MEDLINE | ID: mdl-34174184

ABSTRACT

RelA-SpoT Homolog (RSH) enzymes control bacterial physiology through synthesis and degradation of the nucleotide alarmone (p)ppGpp. We recently discovered multiple families of small alarmone synthetase (SAS) RSH acting as toxins of toxin-antitoxin (TA) modules, with the FaRel subfamily of toxSAS abrogating bacterial growth by producing an analog of (p)ppGpp, (pp)pApp. Here we probe the mechanism of growth arrest used by four experimentally unexplored subfamilies of toxSAS: FaRel2, PhRel, PhRel2, and CapRel. Surprisingly, all these toxins specifically inhibit protein synthesis. To do so, they transfer a pyrophosphate moiety from ATP to the tRNA 3' CCA. The modification inhibits both tRNA aminoacylation and the sensing of cellular amino acid starvation by the ribosome-associated RSH RelA. Conversely, we show that some small alarmone hydrolase (SAH) RSH enzymes can reverse the pyrophosphorylation of tRNA to counter the growth inhibition by toxSAS. Collectively, we establish RSHs as RNA-modifying enzymes.


Subject(s)
Bacterial Toxins/metabolism , Guanosine Pentaphosphate/metabolism , Ligases/metabolism , RNA, Transfer/metabolism , Bacterial Toxins/genetics , Bacterial Toxins/pharmacology , Gram-Positive Asporogenous Rods/chemistry , Gram-Positive Asporogenous Rods/metabolism , Guanosine Pentaphosphate/chemistry , Ligases/chemistry , Ligases/genetics , Phosphorylation/drug effects , Protein Biosynthesis/drug effects , Protein Biosynthesis/physiology , Protein Synthesis Inhibitors/pharmacology , Pyrophosphatases , Ribosomes/metabolism
3.
Protein Expr Purif ; 177: 105760, 2021 01.
Article in English | MEDLINE | ID: mdl-33002609

ABSTRACT

Resistance to antibiotics is a serious concern to treat infectious diseases and also, for food preservation. Existing antibiotics generally inhibit enzymes participating in key bacterial processes, such as formation of cell wall, replication, transcription and translation. However, bacteria have rapidly evolved new mechanisms to combat these antibiotics and it hence becomes indispensable to identify newer targets and identify/design inhibitors against them. Another concern is that most antibiotics are broad spectrum; they largely bind and inhibit the active site of the target enzyme. Rel proteins, which synthesize (and hydrolyze) (p)ppGpp in response to a variety of stress encountered by bacteria, is a profitable target owing to its distinct absence in humans and an intricate regulation of the catalytic activities. Inactivation of (p)ppGpp synthesis by Rel, disables bacterial survival in Mycobacterium tuberculosis and Staphylococcus aureus, while inactivating the hydrolysis activity was lethal. The poor MIC values of the currently known Rel inhibitors present a distinct opportunity to develop better inhibitors and warrants a detailed structural characterization and understanding of the complex regulation in Rel proteins. It will open new avenues for the design of effective, species-specific inhibitors. In an attempt to identify unique sites for inhibitor design using structure-based approaches, we initiate a study of Rel homologues from four different pathogenic bacteria, in order to compare their attributes with well characterized Rel homologues. Here, we present cloning, over-expression, purification and preliminary characterization of these four homologues; and suggest similarities and differences that can be exploited for inhibitor design.


Subject(s)
Guanosine Pentaphosphate/chemistry , Ligases/chemistry , Pyrophosphatases/chemistry , Amino Acid Sequence , Binding Sites , Cloning, Molecular , Computational Biology/methods , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Guanosine Pentaphosphate/metabolism , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/metabolism , Klebsiella pneumoniae/pathogenicity , Ligases/genetics , Ligases/metabolism , Listeria monocytogenes/genetics , Listeria monocytogenes/metabolism , Listeria monocytogenes/pathogenicity , Models, Molecular , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Mycobacterium tuberculosis/pathogenicity , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/metabolism , Pseudomonas aeruginosa/pathogenicity , Pyrophosphatases/genetics , Pyrophosphatases/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Shigella flexneri/genetics , Shigella flexneri/metabolism , Shigella flexneri/pathogenicity , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Staphylococcus aureus/pathogenicity , Structural Homology, Protein , Substrate Specificity , Thermodynamics
4.
FEBS J ; 287(9): 1865-1885, 2020 05.
Article in English | MEDLINE | ID: mdl-31679177

ABSTRACT

Rapid adaptation to environmental changes is crucial for bacterial survival. Almost all bacteria possess a conserved stringent response system to prompt transcriptional and metabolic responses toward stress. The adaptive process relies on alarmones, guanosine pentaphosphate (pppGpp), and tetraphosphate (ppGpp), to regulate global gene expression. The ppGpp is more potent than pppGpp in the regulatory activity, and pppGpp phosphohydrolase (GppA) plays a key role in (p)ppGpp homeostasis. Sharing a similar domain structure, GppA is indistinguishable from exopolyphosphatase (PPX), which mediates the metabolism of cellular inorganic polyphosphate. Here, our phylogenetic analysis of PPX/GppA homologs in bacteria shows a wide distribution with several distinct subfamilies, and our structural and functional analysis of Escherichia coli GppA and Helicobacter pylori PPX/GppA reveals unique properties of each homolog. These results explain how each homolog possesses its distinct functionality.


Subject(s)
Acid Anhydride Hydrolases/chemistry , Acid Anhydride Hydrolases/metabolism , Escherichia coli/enzymology , Escherichia coli/metabolism , Guanosine Pentaphosphate/metabolism , Helicobacter pylori/enzymology , Helicobacter pylori/metabolism , Amino Acid Sequence , Guanosine Pentaphosphate/chemistry , Models, Molecular , Molecular Structure , Phosphoric Monoester Hydrolases/chemistry , Phosphoric Monoester Hydrolases/metabolism , Sequence Alignment
5.
J Vis Exp ; (148)2019 06 04.
Article in English | MEDLINE | ID: mdl-31233015

ABSTRACT

The (p)ppGpp nucleotide functions as a global regulator in bacteria in response to a variety of physical and nutritional stress. It has a rapid onset, in seconds, which leads to accumulation of levels that approach or exceed those of GTP pools. Stress reversal occasions a rapid disappearance of (p)ppGpp, often with a half-life of less than a minute. The presence of (p)ppGpp results in alterations of cellular gene expression and metabolism that counter the damaging effects of stress. Gram-negative and Gram-positive bacteria have different response mechanisms, but both depend on (p)ppGpp concentration. In any event, there is a need to simultaneously monitor many radiolabeled bacterial cultures at time intervals that may vary from 10 seconds to hours during critical stress transition periods. This protocol addresses this technical challenge. The method takes advantage of temperature- and shaker-controlled microtiter dish incubators that allow parallel monitoring of growth (absorbance) and rapid sampling of uniformly phosphate-radiolabeled cultures to resolve and quantitate nucleotide pools by thin-layer chromatography on PEI-cellulose. Small amounts of sample are needed for multiple technical and biological replicates of analyses. Complex growth transitions, such as diauxic growth and rapid (p)ppGpp turnover rates can be quantitatively assessed by this method.


Subject(s)
Chromatography, Thin Layer , Escherichia coli/metabolism , Guanosine Pentaphosphate/metabolism , Phosphorus Radioisotopes , Escherichia coli/genetics , Gene Expression Regulation, Bacterial , Guanosine Pentaphosphate/chemistry , Stress, Physiological/genetics
6.
Mol Cell ; 74(6): 1239-1249.e4, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31023582

ABSTRACT

The stringent response alarmones pppGpp and ppGpp are essential for rapid adaption of bacterial physiology to changes in the environment. In Escherichia coli, the nucleosidase PpnN (YgdH) regulates purine homeostasis by cleaving nucleoside monophosphates and specifically binds (p)ppGpp. Here, we show that (p)ppGpp stimulates the catalytic activity of PpnN both in vitro and in vivo causing accumulation of several types of nucleobases during stress. The structure of PpnN reveals a tetramer with allosteric (p)ppGpp binding sites located between subunits. pppGpp binding triggers a large conformational change that shifts the two terminal domains to expose the active site, providing a structural rationale for the stimulatory effect. We find that PpnN increases fitness and adjusts cellular tolerance to antibiotics and propose a model in which nucleotide levels can rapidly be adjusted during stress by simultaneous inhibition of biosynthesis and stimulation of degradation, thus achieving a balanced physiological response to constantly changing environments.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli/genetics , Gene Expression Regulation, Bacterial , Guanosine Pentaphosphate/chemistry , Guanosine Tetraphosphate/chemistry , N-Glycosyl Hydrolases/chemistry , Allosteric Regulation , Amino Acid Sequence , Anti-Bacterial Agents/pharmacology , Binding Sites , Crystallography, X-Ray , Escherichia coli/drug effects , Escherichia coli/enzymology , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Guanosine Pentaphosphate/metabolism , Guanosine Tetraphosphate/metabolism , Kinetics , Models, Molecular , N-Glycosyl Hydrolases/genetics , N-Glycosyl Hydrolases/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization , Sequence Alignment , Sequence Homology, Amino Acid , Stress, Physiological , Substrate Specificity
7.
Chembiochem ; 20(13): 1717-1721, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30843657

ABSTRACT

Guanosine penta- or tetraphosphate (pppGpp or ppGpp, respectively) is a nucleotide signalling molecule with a marked effect on bacterial physiology during stress. Its accumulation slows down cell metabolism and replication, supposedly leading to the formation of the antibiotic-tolerant persister phenotype. A specifically tailored fluorescent chemosensor, PyDPA, allows the detection of (p)ppGpp in solution with high selectivity, relative to that of other nucleotides. Herein, an optimised synthetic approach is presented that improves the overall yield from 9 to 67 % over 7 steps. The simplicity and robustness of this approach will allow groups investigating the many facets of (p)ppGpp easy access to this probe.


Subject(s)
Coordination Complexes/chemical synthesis , Fluorescent Dyes/chemical synthesis , Guanosine Pentaphosphate/analysis , Guanosine Tetraphosphate/analysis , Pyrenes/chemical synthesis , Fluorescence , Guanosine Pentaphosphate/chemistry , Guanosine Tetraphosphate/chemistry , Zinc/chemistry
8.
Biochim Biophys Acta Gene Regul Mech ; 1861(8): 731-742, 2018 08.
Article in English | MEDLINE | ID: mdl-30012465

ABSTRACT

Precise regulation of gene expression is crucial for bacteria to respond to changing environmental conditions. In addition to protein factors affecting RNA polymerase (RNAP) activity, second messengers play an important role in transcription regulation, such as well-known effectors of the stringent response: guanosine 5'triphosphate-3'diphosphate and guanosine 3', 5'-bis(diphosphate) [(p)ppGpp]. Although much is known about importance of the 5' and 3' moieties of (p)ppGpp, the role of the guanine base remains somewhat cryptic. Here, we use (p)ppGpp's adenine analogs [(p)ppApp] to investigate how the nucleobase contributes to determine its binding site and transcriptional regulation. We determined X-ray crystal structure of Escherichia coli RNAP-(p)ppApp complex, which shows the analogs bind near the active site and switch regions of RNAP. We have also explored the regulatory effects of (p)ppApp on transcription initiating from the well-studied E. coli rrnB P1 promoter to assess and compare properties of (p)ppApp with (p)ppGpp. We demonstrate that contrary to (p)ppGpp, (p)ppApp activates transcription at this promoter and DksA hinders this effect. Moreover, pppApp exerts a stronger effect than ppApp. We also show that when ppGpp and pppApp are present together, the outcome depends on which one of them was pre-incubated with RNAP first. This behavior suggests a surprising Yin-Yang like reciprocal plasticity of RNAP responses at a single promoter, occasioned simply by pre-exposure to one or the other nucleotide. Our observations underscore the importance of the (p)ppNpp's purine nucleobase for interactions with RNAP, which may lead to a better fundamental understanding of (p)ppGpp regulation of RNAP activity.


Subject(s)
Adenine Nucleotides/chemistry , DNA-Directed RNA Polymerases/chemistry , Escherichia coli/genetics , Gene Expression Regulation, Bacterial , Promoter Regions, Genetic , Transcriptional Activation , Adenine Nucleotides/metabolism , Binding Sites , DNA-Directed RNA Polymerases/metabolism , Escherichia coli/enzymology , Escherichia coli Proteins/metabolism , Guanosine Pentaphosphate/chemistry , Guanosine Pentaphosphate/metabolism , Models, Molecular , Structure-Activity Relationship
9.
Mol Microbiol ; 101(4): 531-44, 2016 08.
Article in English | MEDLINE | ID: mdl-27149325

ABSTRACT

The alarmones (p)ppGpp are important second messengers that orchestrate pleiotropic adaptations of bacteria and plant chloroplasts in response to starvation and stress. Here, we review our structural and mechanistic knowledge on (p)ppGpp metabolism including their synthesis, degradation and interconversion by a highly diverse set of enzymes. Increasing structural information shows how (p)ppGpp interacts with an incredibly diverse set of different targets that are essential for replication, transcription, translation, ribosome assembly and metabolism. This raises the question how the chemically rather simple (p)ppGpp is able to interact with these different targets? Structural analysis shows that the diversity of (p)ppGpp interaction with cellular targets critically relies on the conformational flexibility of the 3' and 5' phosphate moieties allowing alarmones to efficiently modulate the activity of target structures in a broad concentration range. Current approaches in the design of (p)ppGpp-analogs as future antibiotics might be aided by the comprehension of conformational flexibility exhibited by the magic dancers (p)ppGpp.


Subject(s)
Guanosine Pentaphosphate/metabolism , Animals , Bacteria/metabolism , Chloroplasts/metabolism , Guanosine Pentaphosphate/chemistry , Guanosine Tetraphosphate/chemistry , Guanosine Tetraphosphate/metabolism , Humans
10.
Genes Cells ; 20(12): 1006-16, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26606426

ABSTRACT

(p)ppGpp, a secondary messenger, is induced under stress and shows pleiotropic response. It binds to RNA polymerase and regulates transcription in Escherichia coli. More than 25 years have passed since the first discovery was made on the direct interaction of ppGpp with E. coli RNA polymerase. Several lines of evidence suggest different modes of ppGpp binding to the enzyme. Earlier cross-linking experiments suggested that the ß-subunit of RNA polymerase is the preferred site for ppGpp, whereas recent crystallographic studies pinpoint the interface of ß'/ω-subunits as the site of action. With an aim to validate the binding domain and to follow whether tetra- and pentaphosphate guanosines have different location on RNA polymerase, this work was initiated. RNA polymerase was photo-labeled with 8-azido-ppGpp/8-azido-pppGpp, and the product was digested with trypsin and subjected to mass spectrometry analysis. We observed three new peptides in the trypsin digest of the RNA polymerase labeled with 8-azido-ppGpp, of which two peptides correspond to the same pocket on ß'-subunit as predicted by X-ray structural analysis, whereas the third peptide was mapped on the ß-subunit. In the case of 8-azido-pppGpp-labeled RNA polymerase, we have found only one cross-linked peptide from the ß'-subunit. However, we were unable to identify any binding site of pppGpp on the ß-subunit. Interestingly, we observed that pppGpp at high concentration competes out ppGpp bound to RNA polymerase more efficiently, whereas ppGpp cannot titrate out pppGpp. The competition between tetraphosphate guanosine and pentaphosphate guanosine for E. coli RNA polymerase was followed by gel-based assay as well as by a new method known as DRaCALA assay.


Subject(s)
DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/metabolism , Escherichia coli/enzymology , Guanosine Pentaphosphate/metabolism , Guanosine Tetraphosphate/metabolism , Binding Sites , Escherichia coli/chemistry , Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Guanosine Pentaphosphate/chemistry , Guanosine Tetraphosphate/chemistry , Mass Spectrometry , Models, Molecular , Photoaffinity Labels/pharmacology , Protein Binding , Protein Structure, Secondary
11.
Mol Cell ; 57(4): 735-749, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25661490

ABSTRACT

The nucleotide (p)ppGpp mediates bacterial stress responses, but its targets and underlying mechanisms of action vary among bacterial species and remain incompletely understood. Here, we characterize the molecular interaction between (p)ppGpp and guanylate kinase (GMK), revealing the importance of this interaction in adaptation to starvation. Combining structural and kinetic analyses, we show that (p)ppGpp binds the GMK active site and competitively inhibits the enzyme. The (p)ppGpp-GMK interaction prevents the conversion of GMP to GDP, resulting in GMP accumulation upon amino acid downshift. Abolishing this interaction leads to excess (p)ppGpp and defective adaptation to amino acid starvation. A survey of GMKs from phylogenetically diverse bacteria shows that the (p)ppGpp-GMK interaction is conserved in members of Firmicutes, Actinobacteria, and Deinococcus-Thermus, but not in Proteobacteria, where (p)ppGpp regulates RNA polymerase (RNAP). We propose that GMK is an ancestral (p)ppGpp target and RNAP evolved more recently as a direct target in Proteobacteria.


Subject(s)
Bacteria/enzymology , Bacterial Proteins/metabolism , Evolution, Molecular , Guanosine Pentaphosphate/metabolism , Guanosine Tetraphosphate/metabolism , Guanylate Kinases/metabolism , Bacteria/genetics , Bacteria/metabolism , Binding, Competitive , Catalytic Domain , Crystallography, X-Ray , DNA-Directed RNA Polymerases/metabolism , Guanosine Pentaphosphate/chemistry , Guanosine Tetraphosphate/chemistry , Guanosine Triphosphate/metabolism , Guanylate Kinases/chemistry , Models, Biological , Species Specificity , Stress, Physiological
12.
Nucleic Acids Res ; 41(12): 6175-89, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23620295

ABSTRACT

Both ppGpp and pppGpp are thought to function collectively as second messengers for many complex cellular responses to nutritional stress throughout biology. There are few indications that their regulatory effects might be different; however, this question has been largely unexplored for lack of an ability to experimentally manipulate the relative abundance of ppGpp and pppGpp. Here, we achieve preferential accumulation of either ppGpp or pppGpp with Escherichia coli strains through induction of different Streptococcal (p)ppGpp synthetase fragments. In addition, expression of E. coli GppA, a pppGpp 5'-gamma phosphate hydrolase that converts pppGpp to ppGpp, is manipulated to fine tune differential accumulation of ppGpp and pppGpp. In vivo and in vitro experiments show that pppGpp is less potent than ppGpp with respect to regulation of growth rate, RNA/DNA ratios, ribosomal RNA P1 promoter transcription inhibition, threonine operon promoter activation and RpoS induction. To provide further insights into regulation by (p)ppGpp, we have also determined crystal structures of E. coli RNA polymerase-σ(70) holoenzyme with ppGpp and pppGpp. We find that both nucleotides bind to a site at the interface between ß' and ω subunits.


Subject(s)
Escherichia coli/metabolism , Guanosine Pentaphosphate/metabolism , Guanosine Tetraphosphate/metabolism , Arabinose/pharmacology , Bacterial Proteins/metabolism , Binding Sites , DNA-Directed RNA Polymerases/chemistry , Escherichia coli/genetics , Escherichia coli/growth & development , Guanosine Pentaphosphate/biosynthesis , Guanosine Pentaphosphate/chemistry , Guanosine Tetraphosphate/biosynthesis , Guanosine Tetraphosphate/chemistry , Hydrolases/metabolism , Ligases/metabolism , Operon , Promoter Regions, Genetic , RNA, Bacterial/biosynthesis , RNA, Ribosomal/genetics , Sigma Factor/chemistry , Sigma Factor/metabolism , Substrate Specificity
13.
Structure ; 20(9): 1447-8, 2012 Sep 05.
Article in English | MEDLINE | ID: mdl-22958638

ABSTRACT

In this issue of Structure, Rymer and colleagues present the first crystal structures of a bacterial DnaG primase with bound substrate NTPs and alarmone inhibitors. A thoughtful comparative structural analysis provides important insights into the chemical mechanism of primase.


Subject(s)
Bacterial Proteins/chemistry , DNA Primase/chemistry , Deoxyribonucleotides/chemistry , Guanosine Pentaphosphate/chemistry , Guanosine Tetraphosphate/chemistry , Staphylococcus aureus/enzymology
14.
Structure ; 20(9): 1478-89, 2012 Sep 05.
Article in English | MEDLINE | ID: mdl-22795082

ABSTRACT

Primases are DNA-dependent RNA polymerases found in all cellular organisms. In bacteria, primer synthesis is carried out by DnaG, an essential enzyme that serves as a key component of DNA replication initiation, progression, and restart. How DnaG associates with nucleotide substrates and how certain naturally prevalent nucleotide analogs impair DnaG function are unknown. We have examined one of the earliest stages in primer synthesis and its control by solving crystal structures of the S. aureus DnaG catalytic core bound to metal ion cofactors and either individual nucleoside triphosphates or the nucleotidyl alarmones, pppGpp and ppGpp. These structures, together with both biochemical analyses and comparative studies of enzymes that use the same catalytic fold as DnaG, pinpoint the predominant nucleotide-binding site of DnaG and explain how the induction of the stringent response in bacteria interferes with primer synthesis.


Subject(s)
Bacterial Proteins/chemistry , DNA Primase/chemistry , Deoxyribonucleotides/chemistry , Guanosine Pentaphosphate/chemistry , Guanosine Tetraphosphate/chemistry , Staphylococcus aureus/enzymology , Catalytic Domain , Coenzymes/chemistry , Conserved Sequence , Coordination Complexes/chemistry , Crystallography, X-Ray , Manganese/chemistry , Models, Molecular , Protein Binding , Protein Structure, Secondary , Stress, Physiological
15.
J Cell Physiol ; 220(2): 297-302, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19391118

ABSTRACT

Guanosine 5'-(tri)diphosphate, 3'-diphosphate [(p) ppGpp] is a small nucleic acid that helps bacteria survive in limited environments. Gene chip shows that (p) ppGpp is a global transcription-regulator of genes related to important bacterial metabolic processes. Therefore, more attention should be focused on the molecular mechanisms of (p) ppGpp, as it is the foundation to understanding how bacteria adapt to extreme circumstances through the stringent response.


Subject(s)
Bacteria , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Guanosine Pentaphosphate/metabolism , Adaptation, Physiological , Bacteria/genetics , Bacteria/metabolism , Bacterial Proteins/genetics , DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Guanosine Pentaphosphate/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Ligases/genetics , Ligases/metabolism , Models, Molecular , Molecular Structure , Plant Proteins/genetics , Plant Proteins/metabolism , Protein Conformation , Pyrophosphatases/genetics , Pyrophosphatases/metabolism
16.
J Mol Biol ; 375(5): 1469-76, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18155044

ABSTRACT

The crystal structure of the prototype exopolyphosphatase/guanosine pentaphosphate phosphohydrolase protein family member from Aquifex aeolicus in complex with the intracellular second messenger guanosine tetraphosphate was determined at 2.7-A resolution. The hydrolytic base is identified as E119. The dual specificity established for the Escherichia coli homolog is shown to be compatible with a common active site for guanosine pentaphosphate and polyphosphate hydrolysis. Distinct and different degrees of closure between the two domains of the enzyme are associated with substrate binding. The arginines R22 and R267, residing in different domains, are crucial for guanosine pentaphosphate specificity as they interact with the unique 3'-ribose phosphorylation.


Subject(s)
Acid Anhydride Hydrolases/chemistry , Bacteria/enzymology , Guanosine Pentaphosphate/chemistry , Guanosine Tetraphosphate/metabolism , Phosphoric Monoester Hydrolases/chemistry , Acid Anhydride Hydrolases/genetics , Acid Anhydride Hydrolases/isolation & purification , Acid Anhydride Hydrolases/metabolism , Amino Acid Sequence , Apoenzymes/chemistry , Arginine/chemistry , Binding Sites , Conserved Sequence , Crystallization , Crystallography, X-Ray , Guanosine Pentaphosphate/genetics , Guanosine Pentaphosphate/isolation & purification , Guanosine Pentaphosphate/metabolism , Hydrogen Bonding , Hydrolysis , Ligands , Models, Chemical , Models, Molecular , Molecular Sequence Data , Mutation , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/isolation & purification , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Software , Substrate Specificity
17.
Nucleosides Nucleotides Nucleic Acids ; 26(10-12): 1339-48, 2007.
Article in English | MEDLINE | ID: mdl-18066780

ABSTRACT

Synthesis of Leishmania mRNA 5'-cap analogs, m(7)Gpppm(2)(6)AmpAm (cap-2), and m(7)Gpppm(2)(6)AmpAmpCm (cap-3) is reported. Binding affinities of those cap analogs for LeishIF4E proteins were determined using fluorescence spectroscopy. Cap-3 showed similar affinity to LeishIF4Es compared to the mature trypanosomatids cap structure (cap-4).


Subject(s)
Guanosine Pentaphosphate/analogs & derivatives , Guanosine Pentaphosphate/chemical synthesis , Leishmania/metabolism , RNA Cap Analogs/chemical synthesis , RNA Caps/chemical synthesis , RNA, Protozoan/chemical synthesis , Animals , Eukaryotic Initiation Factor-4E/chemistry , Guanosine Pentaphosphate/chemistry , RNA Cap Analogs/chemistry , RNA Caps/chemistry , RNA, Protozoan/chemistry
18.
Biophys Chem ; 127(1-2): 41-50, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17188418

ABSTRACT

Bacteria respond to starvation by synthesizing a polyphosphate derivative of guanosine, (p)ppGpp, that helps the bacteria in surviving during stress. The protein in Gram-positive organisms required for (p)ppGpp synthesis is Rel, a bifunctional enzyme that carries out both synthesis and hydrolysis of this molecule. Rel shows increased pppGpp synthesis in the presence of uncharged tRNA, the effect of which is regulated by the C-terminal of Rel. We show by fluorescence resonance energy transfer that the distance between the N-terminus cysteine residue at the catalytic domain and C692 at the C-terminus increases upon the addition of uncharged tRNA. In apparent anomaly, the steady state anisotropy of the Rel protein decreases upon tRNA binding suggesting "compact conformation" vis-à-vis "open conformation" of the free Rel. We propose that the interaction between C692 and the residues present in the pppGpp synthesis site results in the regulated activity and this interaction is abrogated upon addition of uncharged tRNA. We also report here the binding of pppGpp to the C-terminal part of the protein that leads to more unfolding in this region.


Subject(s)
Guanosine Pentaphosphate/metabolism , Ligases/chemistry , Mycobacterium smegmatis/enzymology , RNA, Transfer/chemistry , Amino Acid Sequence , Catalytic Domain , Cloning, Molecular , Cysteine/chemistry , Cysteine/genetics , Escherichia coli/genetics , Feedback, Physiological , Guanosine Pentaphosphate/biosynthesis , Guanosine Pentaphosphate/chemistry , Hydrolysis , Ligands , Ligases/genetics , Ligases/metabolism , Molecular Sequence Data , Mutation , Mycobacterium smegmatis/genetics , Protein Conformation , RNA, Transfer/metabolism
19.
Biochemistry ; 39(38): 11640-8, 2000 Sep 26.
Article in English | MEDLINE | ID: mdl-10995231

ABSTRACT

Rel(Mtb) of Mycobacterium tuberculosis is responsible for the intracellular regulation of (p)ppGpp and the consequent ability of the organism to survive long-term starvation, indicating a possible role in the pathogenesis of tuberculosis. Purified Rel(Mtb) is a dual-function enzyme carrying out ATP: GTP/GDP/ITP 3'-pyrophosphoryltransferase and (p)ppGpp 3'-pyrophosphohydrolase reactions. Here we show that in the absence of biological regulators, Rel(Mtb) simultaneously catalyzes both transferase and hydrolysis at the maximal rate for each reaction, indicating the existence of two distinct active sites. The differential regulation of the opposing activities of Rel(Mtb) is dependent on the ratio of uncharged to charged tRNA and the association of Rel(Mtb) with a complex containing tRNA, ribosomes, and mRNA. A 20-fold increase in the k(cat) and a 4-fold decrease in K(ATP) and K(GTP) from basal levels for transferase activity occur when Rel(Mtb) binds to a complex containing uncharged tRNA, ribosomes, and mRNA (Rel(Mtb) activating complex or RAC). The k(cat) for hydrolysis, however, is reduced 2-fold and K(m) for pppGpp increased 2-fold from basal levels in the presence of the Rel(Mtb) activating complex. The addition of charged tRNA to this complex has the opposite effect by inhibiting transferase activity and activating hydrolysis activity. Differential control of Rel(Mtb) gives the Mtb ribosomal complex a new regulatory role in controlling cellular metabolism in response to stringent growth conditions that may be present in the dormant Mtb lesion.


Subject(s)
Ligases/metabolism , Mycobacterium tuberculosis/enzymology , RNA, Messenger/metabolism , RNA, Ribosomal/metabolism , RNA, Transfer, Amino Acyl/metabolism , Ribosomes/enzymology , Acylation , Acyltransferases/metabolism , Catalysis , Cations, Divalent/chemistry , Enzyme Activation , Guanosine Pentaphosphate/chemistry , Guanosine Pentaphosphate/metabolism , Hydrolysis , Ligases/chemistry , Macromolecular Substances , Pyrophosphatases/chemistry , Pyrophosphatases/metabolism , RNA, Transfer, Amino Acyl/chemistry , Ribosomes/metabolism , rac GTP-Binding Proteins/chemistry , rac GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL