Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Science ; 362(6418)2018 11 30.
Article in English | MEDLINE | ID: mdl-30409808

ABSTRACT

In bacteria, nascent proteins bear the pretranslationally generated N-terminal (Nt) formyl-methionine (fMet) residue. Nt-fMet of bacterial proteins is a degradation signal, termed fMet/N-degron. By contrast, proteins synthesized by cytosolic ribosomes of eukaryotes were presumed to bear unformylated Nt-Met. Here we found that the yeast formyltransferase Fmt1, although imported into mitochondria, could also produce Nt-formylated proteins in the cytosol. Nt-formylated proteins were strongly up-regulated in stationary phase or upon starvation for specific amino acids. This up-regulation strictly required the Gcn2 kinase, which phosphorylates Fmt1 and mediates its retention in the cytosol. We also found that the Nt-fMet residues of Nt-formylated proteins act as fMet/N-degrons and identified the Psh1 ubiquitin ligase as the recognition component of the eukaryotic fMet/N-end rule pathway, which destroys Nt-formylated proteins.


Subject(s)
Amino Acids/deficiency , Hydroxymethyl and Formyl Transferases/metabolism , N-Formylmethionine/metabolism , Proteolysis , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Azides/pharmacology , Cold Temperature , Cytosol/metabolism , Metabolic Networks and Pathways , Mitochondria/enzymology , N-Formylmethionine/chemistry , Peptide Elongation Factors/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/enzymology , Ubiquitin-Protein Ligases/metabolism , Up-Regulation
2.
J Leukoc Biol ; 97(4): 689-97, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25724390

ABSTRACT

Most staphylococci produce short α-type PSMs and about twice as long ß-type PSMs that are potent leukocyte attractants and toxins. PSMs are usually secreted with the N-terminal formyl group but are only weak agonists for the leukocyte FPR1. Instead, the FPR1-related FPR2 senses PSMs efficiently and is crucial for leukocyte recruitment in infection. Which structural features distinguish FPR1 from FPR2 ligands has remained elusive. To analyze which peptide properties may govern the capacities of ß-type PSMs to activate FPRs, full-length and truncated variants of such peptides from Staphylococcus aureus, Staphylococcus epidermidis, and Staphylococcus lugdunensis were synthesized. FPR2 activation was observed even for short N- or C-terminal ß-type PSM variants once they were longer than 18 aa, and this activity increased with length. In contrast, the shortest tested peptides were potent FPR1 agonists, and this property declined with increasing peptide length. Whereas full-length ß-type PSMs formed α-helices and exhibited no FPR1-specific activity, the truncated peptides had less-stable secondary structures, were weak agonists for FPR1, and required N-terminal formyl-methionine residues to be FPR2 agonists. Together, these data suggest that FPR1 and FPR2 have opposed ligand preferences. Short, flexible PSM structures may favor FPR1 but not FPR2 activation, whereas longer peptides with α-helical, amphipathic properties are strong FPR2 but only weak FPR1 agonists. These findings should help to unravel the ligand specificities of 2 critical human PRRs, and they may be important for new, anti-infective and anti-inflammatory strategies.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Toxins/chemistry , Hemolysin Proteins/chemistry , Neutrophils/drug effects , Receptors, Formyl Peptide/drug effects , Receptors, Lipoxin/drug effects , Amino Acid Sequence , Bacterial Proteins/chemical synthesis , Bacterial Proteins/pharmacology , Bacterial Toxins/pharmacology , CD11b Antigen/biosynthesis , CD11b Antigen/genetics , Calcium Signaling/drug effects , Chemotaxis, Leukocyte/drug effects , Cytotoxins/chemical synthesis , Cytotoxins/chemistry , Cytotoxins/pharmacology , HL-60 Cells , Hemolysin Proteins/chemical synthesis , Hemolysin Proteins/pharmacology , Humans , Interleukin-8/biosynthesis , Interleukin-8/genetics , Molecular Sequence Data , N-Formylmethionine/chemistry , Neutrophils/metabolism , Peptide Fragments/chemical synthesis , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Protein Folding , Protein Stability , Protein Structure, Secondary , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/metabolism , Staphylococcus aureus/chemistry , Staphylococcus epidermidis/chemistry , Staphylococcus lugdunensis/chemistry , Structure-Activity Relationship
3.
J Biol Chem ; 280(46): 38403-9, 2005 Nov 18.
Article in English | MEDLINE | ID: mdl-16166083

ABSTRACT

One molecule of glucose 6-phosphate inhibits brain hexokinase (HKI) with high affinity by binding to either one of two sites located in distinct halves of the enzyme. In addition to potent inhibition, glucose 6-phosphate releases HKI from the outer leaflet of mitochondria; however, the site of glucose 6-phosphate association responsible for the release of HKI is unclear. The incorporation of a C-terminal polyhistidine tag on HKI facilitates the rapid purification of recombinant enzyme from Escherichia coli. The tagged construct has N-formyl methionine as its first residue and has mitochondrial association properties comparable with native brain hexokinases. Release of wild-type and mutant hexokinases from mitochondria by glucose 6-phosphate follow equilibrium models, which explain the release phenomenon as the repartitioning of ligand-bound HKI between solution and the membrane. Mutations that block the binding of glucose 6-phosphate to the C-terminal half of HKI have little or no effect on the glucose 6-phosphate release. In contrast, mutations that block glucose 6-phosphate binding to the N-terminal half require approximately 7-fold higher concentrations of glucose 6-phosphate for the release of HKI. Results here implicate a primary role for the glucose 6-phosphate binding site at the N-terminal half of HKI in the release mechanism.


Subject(s)
Brain/enzymology , Glucose-6-Phosphate/metabolism , Hexokinase/chemistry , Hexokinase/genetics , Hexokinase/physiology , Mitochondria/metabolism , Adenosine Triphosphate/chemistry , Alanine/chemistry , Animals , Binding Sites , Brain/metabolism , Circular Dichroism , Dose-Response Relationship, Drug , Escherichia coli/metabolism , Histidine/chemistry , Humans , Kinetics , Ligands , Liver/enzymology , Liver/metabolism , Magnesium/chemistry , Models, Chemical , Mutation , N-Formylmethionine/chemistry , Plasmids/metabolism , Protein Binding , Protein Structure, Tertiary , Swine , Threonine/chemistry
4.
Proteins ; 58(3): 571-88, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15616985

ABSTRACT

The conformational space of the most biologically significant backbone folds of a suitable methionine peptide model was explored by density functional computational method. Using a medium [6-31G(d)] and a larger basis set [6-311++G(2d,2p)], the systematic exploration of low-energy backbone structures restricted for the "L-region" in the Ramachandran map of N-formyl-L-methioninamide results in conformers corresponding to the building units of an extended backbone structure (betaL), an inverse gamma-turn (gammaL), or a right-handed helical structure (alphaL). However, no poly-proline II type (epsilonL) fold was found, indicating that this conformer has no intrinsic stability, and highlighting the effect of molecular environment in stabilizing this backbone structure. This is in agreement with the abundance of the epsilonL-type backbone conformation of methionine found in proteins. Stability properties (DeltaE) and distinct backbone-side-chain interactions support the idea that specific intramolecular contacts are operative in the selection of the lowest energy conformers. Apart from the number of different folds, all stable conformers are within a 10 kcal x mol(-1) energy range, indicating the highly flexible behavior of methionine. This conformational feature can be important in supporting catalytic processes, facilitating protein folding and dimerization via metal ion binding. In both of the biological examples discussed (HIV-1 reverse transcriptase and PcoC copper-resistant protein), the conformational properties of Met residues were found to be of key importance. Spatial proximity to other types of residues or the same type of residue seems to be crucial for the structural integrity of a protein, whether Met is buried or exposed.


Subject(s)
Amino Acids/chemistry , Methionine/chemistry , N-Formylmethionine/chemistry , Peptides/chemistry , Proteins/chemistry , Biophysics/methods , Databases, Protein , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , HIV Reverse Transcriptase/chemistry , Hydrogen Bonding , Methionine/analogs & derivatives , Models, Biological , Models, Chemical , Models, Molecular , Models, Statistical , Molecular Conformation , Proline/chemistry , Protein Conformation , Protein Folding , Protein Structure, Secondary , Proteomics/methods , Static Electricity , Thermodynamics
5.
Cell Biol Int ; 27(8): 695-700, 2003.
Article in English | MEDLINE | ID: mdl-12867161

ABSTRACT

The chemotactic effects of six formylated, putatively bacterial peptides (fMLP, fMLPP, fMMM, fMP, fMV, and fMS) were studied. From the set of six peptides, only fMLP (one of the most effective chemoattractant peptides in mammals) elicited a significant positive chemotactic response in the eukaryotic ciliate Tetrahymena pyriformis, while the other formylated ligands, e.g. fMMM (which is also effective in mammals), had neutral or antagonistic effects in Tetrahymena. A study of their amino acid sequences points to an, as yet obscure, interaction between C-terminal f-Met and N-terminal aromatic Phe. Some optimal physicochemical characteristics (e.g. solvent exposed area, solubility) of the molecule may be responsible for this special feature of f-MLP at such a low level of phylogeny. This means that the unicellular Tetrahymena is able to select between related molecules, giving high priority to the molecule that is the most chemoattractive in mammals. The results call attention to the possible presence of f-Met receptors at a unicellular level and to the evolutionary conservation of chemotaxis-activating processes.


Subject(s)
Chemotaxis/drug effects , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , N-Formylmethionine/chemistry , Peptides/pharmacology , Tetrahymena pyriformis/drug effects , Animals , Chemotaxis/physiology , Dose-Response Relationship, Drug , Ligands , Oligopeptides/chemistry , Oligopeptides/pharmacology , Peptides/chemical synthesis , Peptides/chemistry , Tetrahymena pyriformis/physiology
6.
Protein Expr Purif ; 32(2): 317-22, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14965779

ABSTRACT

In bacteria, protein expression initiates with a formyl-methionine group. Addition of the antibiotic actinonin, a known peptide deformylase inhibitor, at the time of induction of protein expression results in the retention of the formyl group by the overexpressed protein. In addition, because deformylation is a prerequisite for removal of the initiating methionine, this post-translational processing step is also prevented by actinonin, and the N-formyl methionine residue is retained by proteins from which it is normally removed. We have demonstrated the applicability of this system for obtaining N-modified forms of several different proteins and use one of these modified molecules to show that the N-terminal amino group is not required for ClpXP degradation of proteins bearing an N-terminal recognition signal.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , N-Formylmethionine/chemistry , Amidohydrolases/antagonists & inhibitors , Amidohydrolases/metabolism , Chromatography, High Pressure Liquid , Electrophoresis, Polyacrylamide Gel , Escherichia coli Proteins/biosynthesis , Escherichia coli Proteins/genetics , Escherichia coli Proteins/isolation & purification , Gene Expression , Hydroxamic Acids/pharmacology , N-Formylmethionine/metabolism , Protein Processing, Post-Translational/drug effects , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Serine Endopeptidases/metabolism , Spectrometry, Mass, Electrospray Ionization , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods
7.
EMBO J ; 19(19): 5233-40, 2000 Oct 02.
Article in English | MEDLINE | ID: mdl-11013225

ABSTRACT

The interaction between fMet-tRNA(f)(Met) and Bacillus stearothermophilus translation initiation factor IF2 has been characterized. We demonstrate that essentially all thermodynamic determinants governing the stability and the specificity of this interaction are localized within the acceptor hexanucleotide fMet-3'ACCAAC of the initiator tRNA and a fairly small area at the surface of the beta-barrel structure of the 90-amino acid C-terminal domain of IF2 (IF2 C-2). A weak but specific interaction between IF2 C-2 and formyl-methionyl was also demonstrated. The surface of IF2 C-2 interacting with fMet-tRNA(f)(Met) has been mapped using two independent approaches, site- directed mutagenesis and NMR spectroscopy, which yielded consistent results. The binding site comprises C668 and G715 located in a groove accommodating the methionyl side-chain, R700, in the vicinity of the formyl group, Y701 and K702 close to the acyl bond between fMet and tRNA(f)(Met), and the surface lined with residues K702-S660, along which the acceptor arm of the initiator tRNA spans in the direction 3' to 5'.


Subject(s)
Geobacillus stearothermophilus/chemistry , Peptide Initiation Factors/chemistry , Protein Biosynthesis , RNA, Transfer, Met/chemistry , Binding Sites , Geobacillus stearothermophilus/metabolism , Magnetic Resonance Spectroscopy , Models, Molecular , Mutagenesis, Site-Directed , N-Formylmethionine/chemistry , N-Formylmethionine/metabolism , Peptide Initiation Factors/metabolism , Prokaryotic Initiation Factor-2 , Protein Conformation , RNA, Transfer, Met/metabolism , Thermodynamics
8.
J Mol Biol ; 286(1): 71-81, 1999 Feb 12.
Article in English | MEDLINE | ID: mdl-9931250

ABSTRACT

The effect on translational pausing of a hydrophobic probe, coumarin, at the N terminus of nascent peptides was investigated. Two different proteins, bacterial chloramphenicol acetyltransferase and bovine rhodanese, were synthesized by coupled transcription/translation in a cell-free system derived from Escherichia coli. Protein synthesis was initiated with N-formyl-Met-tRNAf or N-acetyl-S-coumarin-Met-tRNAf. Cotranslational incorporation of the coumarin derivative generated nascent polypeptides with a hydrophobic residue at their N termini. The effect of the two N-terminal groups on the size distribution and quantity of the peptides formed by translational pausing was investigated. The N-terminal coumarin caused an accumulation of nascent chloramphenicol acetyltransferase peptides in the mass range of 3.5-4.0 kDa that reflects a delay in translation at this point. No similar effect on rhodanese pause-site peptides was observed. This effect on translational pausing cannot be explained by either mRNA secondary structure or rare codons and tRNA abundance. It is suggested that the effect of N-terminal coumarin on translational pausing is the result of the interaction of the nascent peptide with components of the large ribosomal subunit along the path it follows between the peptidyl transferase center and the exit site on the distal surface.


Subject(s)
Chloramphenicol O-Acetyltransferase/genetics , Protein Biosynthesis , Thiosulfate Sulfurtransferase/genetics , Animals , Cattle , Chloramphenicol O-Acetyltransferase/biosynthesis , Coumarins/chemistry , Coumarins/pharmacology , Escherichia coli , N-Formylmethionine/chemistry , N-Formylmethionine/metabolism , Peptide Biosynthesis , Peptides/metabolism , Protein Biosynthesis/drug effects , RNA, Messenger/metabolism , Ribosomes/metabolism , Thiosulfate Sulfurtransferase/biosynthesis
9.
J Mol Biol ; 262(3): 375-86, 1996 Sep 27.
Article in English | MEDLINE | ID: mdl-8845003

ABSTRACT

Escherichia coli peptide deformylase, a member of the zinc metalloproteases family, is made up of an active core domain composed of 147 residues and of an additional and dispensable C-terminal tail of 21 residues. The three-dimensional structure of the catalytic core could be studied by NMR. 1H and 15N NMR resonances assignments were obtained by two-dimensional and three-dimensional heteronuclear spectroscopy. The structure could be calculated using a set of 1015 restraints for the 147 residues of the enzyme. The overall structure is composed of a series of antiparallel beta-strands which surround two perpendicular alpha-helices. The C-terminal helix contains the HEXXH motif, which is crucial for activity. This helical arrangement and the way the histidines bind the zinc ion clearly are structurally reminiscent of the other members of the metalloprotease family, such as thermolysin or metzincins. Nevertheless, the overall arrangement of secondary and tertiary structures of peptide deformylase and the positioning of its third zinc ligand (a cysteine) are quite different from those of the other members of the family. These discrepancies, together with several biochemical differences, lead us to propose that peptide deformylase is the first example of a new class of the zinc-metalloproteases family. Studies of the interaction of peptide deformylase with either an inhibitor of the reaction or a product of the catalysed reaction, Met-Ala-Ser, as well as comparisons with the structures of other enzymes of the family, have enabled us to delineate the area corresponding to their binding site. The structural basis of the specificity of recognition of the formyl group is discussed in the context of the protease superfamily.


Subject(s)
Amidohydrolases , Aminopeptidases/chemistry , Metalloendopeptidases/classification , Zinc/metabolism , Amino Acid Sequence , Aminopeptidases/antagonists & inhibitors , Aminopeptidases/metabolism , Binding Sites , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Magnetic Resonance Spectroscopy , Metalloendopeptidases/chemistry , Models, Molecular , Molecular Sequence Data , N-Formylmethionine/chemistry , N-Formylmethionine/metabolism , Peptides/metabolism , Protein Conformation , Substrate Specificity
10.
Peptides ; 17(1): 13-6, 1996.
Article in English | MEDLINE | ID: mdl-8822504

ABSTRACT

Endopeptidase 24.11 (EP 24.11), a membrane-bound cell surface enzyme, modulates chemotactic responsiveness of neutrophils to f-Met-Leu-Phe. It is unknown if the enzyme degrades potent formylmethionyl tetrapeptides or if an enzyme with similar activities, endopeptidase 24.15 (EP 24.15), degrades formylated chemotactic peptides. In a study of five formylmethionyl tetrapeptides and f-Met-Leu-Phe, we found that EP 24.11 had high affinity for all peptides evaluated, although it did not effectively degrade f-Met-Ile-Leu-Phe. EP 24.15 had high affinity for three of the tetrapeptides, and for f-Met-Leu-Phe, although, for unclear reasons, it did not degrade f-Met-Ile-Leu-Phe or f-Met-Leu-Phe, the apparent natural products of Staphylococcus aureus and Escherichia coli, respectively.


Subject(s)
Chemotactic Factors/metabolism , Metalloendopeptidases/metabolism , N-Formylmethionine/metabolism , Neprilysin/metabolism , Oligopeptides/metabolism , Amino Acid Sequence , Animals , Binding Sites , Chemotactic Factors/chemistry , Chemotactic Factors/genetics , Hydrolysis , In Vitro Techniques , Kinetics , Male , Metalloendopeptidases/antagonists & inhibitors , Molecular Sequence Data , N-Formylmethionine/chemistry , Neprilysin/antagonists & inhibitors , Oligopeptides/chemistry , Oligopeptides/genetics , Rabbits , Rats
11.
Biotechnol Genet Eng Rev ; 13: 197-216, 1996.
Article in English | MEDLINE | ID: mdl-8948113

ABSTRACT

The use of site-directed mutagenesis (Smith, 1985) to replace amino acids at any chosen position in a protein, coupled with advances in analytical procedures, has greatly advanced our understanding of biological structure-function relationships in recent years. The only limitation of conventional site-directed mutagenesis is that substitutions are restricted to the 20 naturally occurring amino acids. However, the discovery of a 21st amino acid, selenocysteine, and the development of novel in vitro translation techniques have demonstrated that considerably more site-specific replacements are possible during protein engineering. These techniques have already found a wide range of applications and have shown that the translational machinery is able to accommodate an enormously divergent range of aminoacylated tRNAs. Although these techniques are mainly restricted to in vitro systems, recent progress in our understanding of aminoacyl-tRNA synthetase-catalyzed tRNA charging suggests that it may ultimately be possible to extend this technique to growing cells.


Subject(s)
Amino Acids/chemistry , Amino Acids/genetics , Protein Biosynthesis , Protein Engineering , Amino Acyl-tRNA Synthetases/metabolism , Base Sequence , Codon/genetics , Mutagenesis, Site-Directed , N-Formylmethionine/chemistry , Proteins/chemistry , Proteins/genetics , RNA, Transfer, Amino Acyl/metabolism , Selenocysteine/genetics , Selenoproteins
13.
Biotechnology (N Y) ; 13(5): 504-6, 1995 May.
Article in English | MEDLINE | ID: mdl-9634792

ABSTRACT

Preparations of rHMfA (recombinant histone A from Methanothermus fervidus) synthesized in E. coli by the heterologous expression of the hmfA gene were found to contain a mixture of rHMfA molecules, approximately 40% that retained the N-terminal formyl-methionyl residue (f-met-rHMfA), approximately 50% that lacked the formyl moiety but retained the methionyl residue (met-rHMfA), and only approximately 10% that had lost both components of the protein synthesis initiating amino acid residue and therefore had the same N-terminal sequence as native HMfA molecules synthesized in Mt. fervidus. Expression of the hmfA gene in E. coli cells grown in the presence of trimethoprim and thymidine, coupled with the concurrent over-expression of a methionine aminopeptidase-encoding map gene, has been shown to overcome this N-terminal heterogeneity problem and to result in rHMfA preparations in which > 85% of the molecules have the fully processed, native N-terminal sequence. This procedure should be generally useful for ensuring N-terminal processing of recombinant proteins synthesized in E. coli.


Subject(s)
Histones/genetics , N-Formylmethionine/chemistry , Protein Processing, Post-Translational , Archaea , Escherichia coli , Histones/biosynthesis , Histones/chemistry , Recombinant Proteins/biosynthesis
14.
Pept Res ; 6(6): 298-307, 1993.
Article in English | MEDLINE | ID: mdl-8292847

ABSTRACT

In order to probe the role of peptide backbone conformation on the biological activity of chemotactic peptides through conformationally constrained peptides, we synthesized the following three analogs of N-formyl-Met-Leu-Phe-OH (fMLF) containing dehydrophenylalanine (delta ZPhe) and dehydroleucine (delta ZLeu): formyl-Met-delta ZPhe-Phe-OCH3 (1), formyl-Met-delta ZLeu-Phe-OCH3 (2) and formyl-Met-delta ZPhe-delta ZPhe-OCH3 (3) and studied their conformational behavior in solution by 1H NMR and IR spectroscopy. The conformation of (1) was also examined by x-ray diffraction methods. Biological activity of these analogs was assessed for their ability to induce the release of beta-glucosaminidase from rabbit neutrophils. In addition, the chemotactic activity of analog (2) was also determined. We found that, in the solid state, (1) favors a type II beta-turn structure, stabilized by a 4-->1 intramolecular hydrogen bond. A similar structure was reported recently for (2) also. 1H NMR studies in solution suggest that the Phe NH is solvent shielded in both (1) and (2) and that a major population of peptide molecule exists in an intramolecular hydrogen bond stabilized type II beta-turn conformation. None of the NH groups in (3) and another analog, formyl-Met-Phe-Phe-OCH3 (4), appear solvent shielded, favoring an extended structure for these analogs. Analogs (2) and (4) are highly active indicating that both extended and beta-turn backbone conformations may be compatible with high activity and that the phenylalanine ring in the middle position is well accepted. Highly reduced activities of (1) and (3) suggest that delta ZPhe residue in position 2, irrespective of the preferred peptide backbone conformation, is not acceptable for high bioactivity. These results suggest that an induced fit mechanism may possibly be the most relevant one, but the nature and the topography of the side chains, particularly the middle residue, may be crucial for appropriate receptor ligand interactions.


Subject(s)
Leucine/analogs & derivatives , N-Formylmethionine/chemistry , Oligopeptides/chemistry , Phenylalanine/analogs & derivatives , Animals , Hexosaminidases/metabolism , Leucine/chemistry , Magnetic Resonance Spectroscopy , Molecular Structure , N-Formylmethionine Leucyl-Phenylalanine/chemistry , Neutrophils/drug effects , Neutrophils/enzymology , Oligopeptides/pharmacology , Phenylalanine/chemistry , Protein Conformation , Rabbits , Solutions , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...