Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
Angew Chem Int Ed Engl ; 60(9): 4666-4672, 2021 02 23.
Article in English | MEDLINE | ID: mdl-33935588

ABSTRACT

Radical S-adenosyl-l-methionine (SAM) enzymes initiate biological radical reactions with the 5'-deoxyadenosyl radical (5'-dAdo•). A [4Fe-4S]+ cluster reductively cleaves SAM to form the Ω organometallic intermediate in which the 5'-deoxyadenosyl moiety is directly bound to the unique iron of the [4Fe-4S] cluster, with subsequent liberation of 5'-dAdo•. Here we present synthesis of the SAM analog S-adenosyl-l-ethionine (SAE) and show SAE is a mechanistically-equivalent SAM-alternative for HydG, both supporting enzymatic turnover of substrate tyrosine and forming the organometallic intermediate Ω. Photolysis of SAE bound to HydG forms an ethyl radical trapped in the active site. The ethyl radical withstands prolonged storage at 77 K and its EPR signal is only partially lost upon annealing at 100 K, making it significantly less reactive than the methyl radical formed by SAM photolysis. Upon annealing above 77K, the ethyl radical adds to the [4Fe-4S]2+ cluster, generating an ethyl-[4Fe-4S]3+ organometallic species termed ΩE.


Subject(s)
Escherichia coli Proteins/metabolism , Ethionine/metabolism , Trans-Activators/metabolism , Biocatalysis , Electron Spin Resonance Spectroscopy , Escherichia coli Proteins/chemistry , Ethionine/analogs & derivatives , Ethionine/chemistry , Free Radicals/chemistry , Free Radicals/metabolism , Models, Molecular , Molecular Structure , Trans-Activators/chemistry
2.
Spectrochim Acta A Mol Biomol Spectrosc ; 223: 117328, 2019 Dec 05.
Article in English | MEDLINE | ID: mdl-31295632

ABSTRACT

Ethionine is an S-ethyl analog of methionine (Met) having a small change in structure. But it is a chemical carcinogen and an antagonist of Met, thus displaying a disparate biological profile. The oxidations of ethionine by biologically important oxidants have not been exploited. Oxidations of dl-ethionine by Pt(IV) anticancer model complexes trans-[PtX2(CN4)]2- (X = Cl or Br) were thus analyzed by time-resolved and stopped-flow spectral techniques. Overall second-order kinetics was established, being first-order in [Pt(IV)] and [Ethionine]tot (the total concentration of ethionine); the observed second-order rate constant k' versus pH profiles were obtained. A stoichiometry of Δ[Pt(IV)]:Δ[Ethionine]tot = 1:1 was unraveled, indicating that ethionine was oxidized to ethionine-sulfoxide which was confirmed by NMR spectroscopic and high-resolution mass spectral analyses. In the proposed reaction mechanism which is similar to that for the oxidation of Met by the same Pt(IV) compounds, the rate-determining steps are rationalized in terms of a bridge formation between one of the coordinated halides in [PtX2(CN4)]2- and the sulfur atom in ethionine, followed by an X+ transfer. Moreover, a large rate enhancement for the reaction of ethionine with [PtBr2(CN4)]2- compared with [PtCl2(CN4)]2- strongly supports an X+ transfer mechanism. Furthermore, a combined quantum-mechanical/molecular-mechanical (QM/MM) method was utilized to simulate a Cl+ transfer mechanism from trans-[PtCl2(CN)4]2- to ethionine. The simulations unraveled the energetically stable structures of reactants and products, which favor the Cl+ transfer process. Rate constants of the rate-determining steps have been derived. Ratios of k (ethionine)/k (Met) are between 2.2 and 2.6 obtained for the three protolytic species of ethionine and Met; the enhanced reactivity might be partially responsible for the disparate biological profiles.


Subject(s)
Antineoplastic Agents/pharmacology , Ethionine/chemistry , Models, Theoretical , Platinum/pharmacology , Spectrum Analysis , Hydrogen-Ion Concentration , Kinetics , Molecular Conformation , Oxidation-Reduction , Proton Magnetic Resonance Spectroscopy , Time Factors
3.
Biochem Biophys Res Commun ; 482(4): 1346-1352, 2017 Jan 22.
Article in English | MEDLINE | ID: mdl-27939882

ABSTRACT

Acute pancreatitis has several underlying etiologies, and results in consequences ranging from mild to complex multi-organ failure. The wide range of pathology suggests a genetic predisposition for progression. We compared the susceptibility to acute pancreatitis in BALB/c and FVB/N mice, coupled with proteomic analysis, in order to identify potential protein associations with pancreatitis progression. METHODS: Pancreatitis was induced in BALB/c and FVB/N mice by administration of cerulein or feeding a choline-deficient, ethionine-supplemented (CDE) diet. Histology and changes in serum amylase were examined. Proteome profiling in cerulein-treated mice was performed using 2-dimensional differential in gel electrophoresis (2D-DIGE) followed by mass spectrometry analysis and biochemical validation. RESULTS: Male and female FVB/N mice manifested more severe cerulein-induced pancreatitis as compared with BALB/c mice, but both strains were similarly susceptible to CDE-induced pancreatitis. Few of the 2D-DIGE alterations were validated by immunoblotting. Clusterin was markedly up-regulated after cerulein-induced pancreatitis in FVB/N but less-so in BALB/c mice. Pyrroline-5-carboxylate reductase (Pycr1), an enzyme involved in proline biosynthesis, had higher basal levels in FVB/N male and female mouse pancreata compared with BALB/c pancreata, and was relatively more resistant to degradation in FVB/N pancreata. However, serum and pancreas tissue proline levels were similar in the two strains. CONCLUSION: FVB/N is more susceptible than BALB/c mice to cerulein-induced but not CDE-induced pancreatitis. Most of the 2D-DIGE alterations in the two strains likely relate to posttranslational modifications rather than protein level differences. Clusterin levels increase dramatically in association with pancreatitis severity, while Pycr1 is higher in FVB/N versus BALB/c pancreata basally and after induction of pancreatitis. Changes in proline metabolism may represent a novel potential genetic modifier in the context of pancreatitis.


Subject(s)
Clusterin/genetics , Gene Expression Regulation , Genetic Predisposition to Disease , Pancreatitis/genetics , Pyrroline Carboxylate Reductases/genetics , Amylases/blood , Animals , Ceruletide/chemistry , Choline/chemistry , Clusterin/metabolism , Disease Models, Animal , Ethionine/chemistry , Female , Male , Mice , Mice, Inbred BALB C , Pancreatitis/metabolism , Proline/chemistry , Protein Processing, Post-Translational , Proteome , Pyrroline Carboxylate Reductases/metabolism , Species Specificity , delta-1-Pyrroline-5-Carboxylate Reductase
4.
J Am Chem Soc ; 138(9): 2877-80, 2016 Mar 09.
Article in English | MEDLINE | ID: mdl-26901520

ABSTRACT

Identifying an enzyme's substrates is essential to understand its function, yet it remains challenging. A fundamental impediment is the transient interactions between an enzyme and its substrates. In contrast, tight binding is often observed for multisubstrate-adduct inhibitors due to synergistic interactions. Extending this venerable concept to enzyme-catalyzed in situ adduct formation, unknown substrates were affinity-captured by an S-adenosyl-methionine (AdoMet, SAM)-dependent methyltransferase (MTase). Specifically, the electrophilic methyl sulfonium (alkyl donor) in AdoMet is replaced with a vinyl sulfonium (Michael acceptor) in S-adenosyl-vinthionine (AdoVin). Via an addition reaction, AdoVin and the nucleophilic substrate form a covalent bisubstrate-adduct tightly complexed with thiopurine MTase (2.1.1.67). As such, an unknown substrate was readily identified from crude cell lysates. Moreover, this approach is applicable to other systems, even if the enzyme is unknown.


Subject(s)
Ethionine/analogs & derivatives , Methyltransferases/chemistry , Methyltransferases/metabolism , S-Adenosylmethionine/chemistry , S-Adenosylmethionine/metabolism , Chromatography, High Pressure Liquid , Click Chemistry , Ethionine/chemistry , Ethionine/metabolism , Humans , Spectrophotometry, Ultraviolet , Substrate Specificity
5.
J Proteomics ; 130: 190-9, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26370163

ABSTRACT

Staphylococcus epidermidis is an opportunistic pathogen that is an emerging risk factor in hospitals worldwide and is often difficult to eradicate as virulent strains produce a protective biofilm matrix. We utilized cell shaving proteomics to profile surface-exposed proteins from two fully genome sequenced S. epidermidis strains: the avirulent, non-biofilm forming ATCC12228 and the virulent, strongly adherent biofilm forming ATCC35984 (RP62A). A false positive control strategy was employed to calculate the probabilities of proteins being truly surface-exposed. A total of 78 surface-exposed proteins were identified, of which only 19 proteins were common to ATCC12228 and RP62A, and which thus represents the core surfaceome. S. epidermidis RP62A displayed additional proteins involved in biofilm formation (cell wall-associated Bhp and intercellular adhesion protein IcaB), surface antigenicity, peptidoglycan biosynthesis and antibiotic resistance. We concurrently profiled whole cell proteomes of the two strains using iTRAQ quantitation and LC-MS/MS. A total of 1610 proteins were confidently identified (representing 64% of the theoretical S. epidermidis proteome). One hundred and ninety one proteins were differentially abundant between strains. Proteins associated with RP62A were clustered into functions including Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-mediated defense, sulfate assimilation, antibiotic resistance and biofilm formation. Validation of the sulfate assimilation and cysteine/methionine biosynthesis pathways showed RP62A contained elevated levels (~25% increase) of methionine that are likely linked to biofilm formation. BIOLOGICAL SIGNIFICANCE: Cell shaving and quantitative proteomics identified proteins associated with a biofilm-forming, virulent strain of S. epidermidis (RP62A). These proteins show RP62A maintains an active CRISPR-mediated defense, as well as heightened antibiotic resistance in comparison to a non-virulent, non-biofilm forming strain. Increased abundances of sulfate assimilation proteins lead to elevated intracellular methionine. Proteins and their exposed peptides identified on the surface of S. epidermidis RP62A may be useful vaccine antigens in clinical settings if administered in at-risk patients prior to surgical implantations.


Subject(s)
Cell Membrane/metabolism , Proteomics/methods , Staphylococcus epidermidis/metabolism , Anti-Bacterial Agents/chemistry , Bacterial Proteins/chemistry , Biofilms , CRISPR-Cas Systems , Drug Resistance, Bacterial , Ethionine/chemistry , Mass Spectrometry , Methionine/chemistry , Phenotype , Polysaccharides/chemistry , Proteome/metabolism , Species Specificity , Sulfates/chemistry , Tandem Mass Spectrometry , Trypsin/chemistry , Virulence
6.
Biochem Biophys Res Commun ; 456(1): 135-8, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25446111

ABSTRACT

Free fatty acids (FFAs) are dietary nutrients which act as ligands for FFAs receptors. G-protein-coupled receptor 120 (GPR120) and GPR40 are activated by long and medium chain FFAs. In the present study, we investigated the role of the GPR120 and GPR40 in cell motile activity stimulated by ethionine in rat liver epithelial WB-F344 cells. Cells were treated with ethionine at a concentration of 10µM every 24h for 2days. The expression levels of the Gpr120 and Gpr40 genes in WB-F344 cells treated ethionine were significantly higher than those in untreated cells. In cell motility assay, the cell motile activity of WB-F344 cells was markedly elevated by ethionine, compared with untreated cells. To evaluate the effects of GPR120 on the cell motile activity by ethionine, we established GPR120 knockdown cells from WB-F344 cells. The cell motile activity stimulated by ethionine was significantly suppressed by GPR120 knockdown. In addition, a potent GPR40 antagonist GW1100 enhanced the cell motile activity by ethionine. These results suggest that opposite effects of GPR120 and GPR40 may be involved in the cell motile activity stimulated by ethionine in WB-F344 cells.


Subject(s)
Epithelial Cells/metabolism , Ethionine/chemistry , Liver/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Movement , Cell Proliferation , Male , Polymerase Chain Reaction , Rats , Rats, Inbred F344
7.
Biochim Biophys Acta ; 1844(9): 1453-62, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24780582

ABSTRACT

Human cystathionine ß-synthase (hCBS) is a key enzyme of sulfur amino acid metabolism, controlling the commitment of homocysteine to the transsulfuration pathway and antioxidant defense. Mutations in hCBS cause inherited homocystinuria (HCU), a rare inborn error of metabolism characterized by accumulation of toxic homocysteine in blood and urine. hCBS is a complex multidomain and oligomeric protein whose activity and stability are independently regulated by the binding of S-adenosyl-methionine (SAM) to two different types of sites at its C-terminal regulatory domain. Here we study the role of surface electrostatics on the complex regulation and stability of hCBS using biophysical and biochemical procedures. We show that the kinetic stability of the catalytic and regulatory domains is significantly affected by the modulation of surface electrostatics through noticeable structural and energetic changes along their denaturation pathways. We also show that surface electrostatics strongly affect SAM binding properties to those sites responsible for either enzyme activation or kinetic stabilization. Our results provide new insight into the regulation of hCBS activity and stability in vivo with implications for understanding HCU as a conformational disease. We also lend experimental support to the role of electrostatic interactions in the recently proposed binding modes of SAM leading to hCBS activation and kinetic stabilization.


Subject(s)
Adenosine/analogs & derivatives , Cystathionine beta-Synthase/chemistry , Ethionine/analogs & derivatives , Protein Subunits/chemistry , Adenosine/chemistry , Adenosine/metabolism , Catalytic Domain , Cystathionine beta-Synthase/metabolism , Enzyme Activation , Ethionine/chemistry , Ethionine/metabolism , Humans , Kinetics , Ligands , Protein Binding , Protein Multimerization , Protein Stability , Protein Structure, Secondary , Protein Subunits/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sodium Chloride/chemistry , Static Electricity , Surface Properties , Thermodynamics , Urea/chemistry
8.
J Biol Chem ; 283(27): 18861-72, 2008 Jul 04.
Article in English | MEDLINE | ID: mdl-18400755

ABSTRACT

Maturation of the [FeFe]-hydrogenase active site depends on at least the expression of three gene products called HydE, HydF, and HydG. We have solved the high resolution structure of recombinant, reconstituted S-adenosine-L-methionine-dependent HydE from Thermotoga maritima. Besides the conserved [Fe(4)S(4)] cluster involved in the radical-based reaction, this HydE was reported to have a second [Fe(4)S(4)] cluster coordinated by three Cys residues. However, in our crystals, depending on the reconstitution and soaking conditions, this second cluster is either a [Fe(2)S(2)] center, with water occupying the fourth ligand site or is absent. We have carried out site-directed mutagenesis studies on the related HydE from Clostridium acetobutylicum, along with in silico docking and crystal soaking experiments, to define the active site region and three anion-binding sites inside a large, positive cavity, one of which binds SCN(-) with high affinity. Although the overall triose-phosphate isomerase-barrel structure of HydE is very similar to that of biotin synthase, the residues that line the internal cavity are significantly different in the two enzymes.


Subject(s)
Bacterial Proteins/chemistry , Iron-Sulfur Proteins/chemistry , Iron/chemistry , Thermotoga maritima/enzymology , Adenosine/analogs & derivatives , Adenosine/chemistry , Adenosine/genetics , Adenosine/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites/physiology , Clostridium acetobutylicum/enzymology , Clostridium acetobutylicum/genetics , Crystallography, X-Ray , Ethionine/analogs & derivatives , Ethionine/chemistry , Ethionine/genetics , Ethionine/metabolism , Free Radicals/chemistry , Free Radicals/metabolism , Iron/metabolism , Iron-Sulfur Proteins/genetics , Iron-Sulfur Proteins/metabolism , Protein Structure, Tertiary/physiology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Structural Homology, Protein , Thermotoga maritima/genetics , Water/chemistry , Water/metabolism
9.
Biochim Biophys Acta ; 1774(8): 1014-9, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17604705

ABSTRACT

A new type II 6His-Eco29kI DNA methyltransferase was tested for methylation site (CC(Me)GCGG) and catalytic reaction optimal conditions. With high substrate concentrations, an inhibitory effect of DNA, but not AdoMet, on its activity was observed. Isotope partitioning and substrate preincubation assays showed that the enzyme-AdoMet complex is catalytically active. Considering effect of different concentrations of DNA and AdoMet on initial velocity, ping-pong mechanisms were ruled out. According to data obtained, the enzyme appears to work by preferred ordered bi-bi mechanism with AdoMet as leading substrate.


Subject(s)
DNA Methylation , DNA-Cytosine Methylases/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/enzymology , Adenosine/analogs & derivatives , Adenosine/chemistry , Binding Sites , Catalysis , Ethionine/analogs & derivatives , Ethionine/chemistry , Kinetics , Substrate Specificity
10.
Analyst ; 125(2): 231-4, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10820888

ABSTRACT

The enantiomeric separation of three selenoamino acids, D,L-selenomethionine, D,L-selenoethionine and D,L-selenocystine is described. Both sulfated beta-cyclodextrin and vancomycin have been successfully used to separate all enantiomers of the compounds with UV detection. Reproducible separations, in terms of peak area and migration time were obtained using sulfated beta-cyclodextrin with reversed polarity and UV detection. With vancomycin as a chiral selector, reversed polarity was found to be more reproducible than positive polarity in terms of peak migration times.


Subject(s)
Electrophoresis, Capillary , Selenium Compounds/chemistry , Vancomycin/chemistry , Cystine/analogs & derivatives , Cystine/chemistry , Ethionine/chemistry , Organoselenium Compounds/chemistry , Selenomethionine/chemistry , Stereoisomerism , Sulfur/metabolism , Ultraviolet Rays
11.
Protein Eng ; 13(1): 59-66, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10679531

ABSTRACT

The interactions between the abundant methionine residues of the calcium regulatory protein calmodulin (CaM) and several of its binding targets were probed using fluorescence spectroscopy. Tryptophan steady-state fluorescence from peptides encompassing the CaM-binding domains of the target proteins myosin light chain kinase (MLCK), cyclic nucleotide phosphodiesterase (PDE) and caldesmon site A and B (CaD A, CaD B), and the model peptide melittin showed Ca(2+)-dependent blue-shifts in their maximum emission wavelength when complexed with wild-type CaM. Blue-shifts were also observed for complexes in which the CaM methionine residues were replaced by selenomethionine, norleucine and ethionine, and when a quadruple methionine to leucine C-terminal mutant of CaM was studied. Quenching of the tryptophan fluorescence intensity was observed with selenomethionine, but not with norleucine or ethionine substituted protein. Fluorescence quenching studies with added potassium iodide (KI) demonstrate that the non-native proteins limit the solvent accessibility of the Trp in the MLCK peptide to levels close to that of the wild-type CaM-MLCK interaction. Our results show that the methionine residues from CaM are highly sensitive to the target peptide in question, confirming the importance of their role in binding interactions. In addition, we provide evidence that the nature of binding in the CaM-CaD B complex is unique compared with the other complexes studied, as the Trp residue of this peptide remains partially solvent exposed upon binding to CaM.


Subject(s)
Calmodulin-Binding Proteins/chemistry , Calmodulin-Binding Proteins/metabolism , Calmodulin/metabolism , Methionine/analogs & derivatives , Tryptophan/chemistry , 3',5'-Cyclic-AMP Phosphodiesterases/chemistry , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Amino Acid Sequence , Binding Sites , Calmodulin/chemistry , Calmodulin-Binding Proteins/isolation & purification , Ethionine/chemistry , Fluorescence , Melitten/chemistry , Melitten/metabolism , Molecular Sequence Data , Myosin-Light-Chain Kinase/chemistry , Myosin-Light-Chain Kinase/metabolism , Norleucine/chemistry , Peptide Fragments/chemistry , Peptide Fragments/isolation & purification , Peptide Fragments/metabolism , Spectrometry, Fluorescence
12.
Protein Sci ; 8(1): 113-21, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10210190

ABSTRACT

Calmodulin (CaM) is a 148-residue regulatory calcium-binding protein that activates a wide range of target proteins and enzymes. Calcium-saturated CaM has a bilobal structure, and each domain has an exposed hydrophobic surface region where target proteins are bound. These two "active sites" of calmodulin are remarkably rich in Met residues. Here we have biosynthetically substituted (up to 90% incorporation) the unnatural amino acids ethionine (Eth) and norleucine (Nle) for the nine Met residues of CaM. The substituted proteins bind in a calcium-dependent manner to hydrophobic matrices and a synthetic peptide, encompassing the CaM-binding domain of myosin light-chain kinase (MLCK). Infrared and circular dichroism spectroscopy show that there are essentially no changes in the secondary structure of these proteins compared to wild-type CaM (WT-CaM). One- and two-dimensional NMR studies of the Eth-CaM and Nle-CaM proteins reveal that, while the core of the proteins is relatively unaffected by the substitutions, the two hydrophobic interaction surfaces adjust to accommodate the Eth and Nle residues. Enzyme activation studies with MLCK show that Eth-CaM and Nle-CaM activate the enzyme to 90% of its maximal activity, with little changes in dissociation constant. For calcineurin only 50% activation was obtained, and the K(D) for Nle-CaM also increased 3.5-fold compared with WT-CaM. These data show that the "active site" Met residues of CaM play a distinct role in the activation of different target enzymes, in agreement with site-directed mutagenesis studies of the Met residues of CaM.


Subject(s)
Calmodulin/chemistry , Ethionine/chemistry , Methionine/chemistry , Norleucine/chemistry , Amino Acid Substitution , Amino Acids/analysis , Animals , Calcineurin/metabolism , Calmodulin/metabolism , Circular Dichroism , Electrophoresis, Polyacrylamide Gel , Magnetic Resonance Spectroscopy , Myosin-Light-Chain Kinase/metabolism , Spectroscopy, Fourier Transform Infrared
13.
Eur J Biochem ; 230(2): 788-96, 1995 Jun 01.
Article in English | MEDLINE | ID: mdl-7607253

ABSTRACT

We have utilized a T7 polymerase/promoter system for the high-level incorporation of methionine analogs with suitable labels for structural research (X-ray and NMR studies) on recombinant annexin V produced in Escherichia coli. Here, we describe, to our knowledge, the first biosynthetic high-level substitution of methionine by 2-aminohexanoic acid (norleucine), ethionine and telluromethionine in a protein. The replacement has been confirmed by electrospray mass spectroscopy, amino acid analysis and X-ray structural analysis. Conditions for expression were optimized concerning the frequency of appearance of revertants, high-level replacement and maximal protein yield. For the incorporation of norleucine and ethionine, E. coli B834 (DE3)(hsd metB), which is auxotrophic for methionine, was grown under methionine-limited conditions with an excess of the analog in the culture medium, and the expression of protein under the control of the T7 promoter was induced after the methionine supply had been exhausted. The factor limiting the high-level incorporation of telluromethionine into protein is its sensitivity towards oxidation. To overcome this problem, bacteria were grown with a limited amount of methionine, harvested after its exhaustion and resuspended in fresh media without methionine; telluromethionine was added and protein synthesis induced. Under these conditions, significant amounts of protein can be expressed before telluromethionine has been completely degraded (within hours). Biosynthetic incorporation of heavy atoms such as tellurium into recombinant proteins can accelerate the process of obtaining heavy-atom derivatives suitable for X-ray structural analysis, supplementing the traditional trial-and-error preparation of heavy-atom derivatives for the method of multiple isomorphous replacement. Furthermore, the successful high-level incorporation of amino acid analogs can provide single-atom mutations for the detailed study of the structure and function of proteins.


Subject(s)
Annexin A5/chemistry , Methionine/analogs & derivatives , Amino Acids/analysis , Annexin A5/genetics , Chromatography, High Pressure Liquid , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli , Ethionine/chemistry , Fourier Analysis , Humans , Mass Spectrometry/methods , Methionine/chemistry , Methionine/genetics , Norleucine/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Selenomethionine/chemistry , Tellurium/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL