Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters











Publication year range
1.
J Biol Chem ; 299(10): 105161, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37586588

ABSTRACT

Chorismate mutase (CM) and cyclohexadienyl dehydratase (CDT) catalyze two subsequent reactions in the intracellular biosynthesis of l-phenylalanine (Phe). Here, we report the discovery of novel and extremely rare bifunctional fusion enzymes, consisting of fused CM and CDT domains, which are exported from the cytoplasm. Such enzymes were found in only nine bacterial species belonging to non-pathogenic γ- or ß-Proteobacteria. In γ-proteobacterial fusion enzymes, the CM domain is N-terminal to the CDT domain, whereas the order is inverted in ß-Proteobacteria. The CM domains share 15% to 20% sequence identity with the AroQγ class CM holotype of Mycobacterium tuberculosis (∗MtCM), and the CDT domains 40% to 60% identity with the exported monofunctional enzyme of Pseudomonas aeruginosa (PheC). In vitro kinetics revealed a Km <7 µM, much lower than for ∗MtCM, whereas kinetic parameters are similar for CDT domains and PheC. There is no feedback inhibition of CM or CDT by the pathway's end product Phe, and no catalytic benefit of the domain fusion compared with engineered single-domain constructs. The fusion enzymes of Aequoribacter fuscus, Janthinobacterium sp. HH01, and Duganella sacchari were crystallized and their structures refined to 1.6, 1.7, and 2.4 Å resolution, respectively. Neither the crystal structures nor the size-exclusion chromatography show evidence for substrate channeling or higher oligomeric structure that could account for the cooperation of CM and CDT active sites. The genetic neighborhood with genes encoding transporter and substrate binding proteins suggests that these exported bifunctional fusion enzymes may participate in signaling systems rather than in the biosynthesis of Phe.

2.
Biochemistry ; 62(3): 782-796, 2023 02 07.
Article in English | MEDLINE | ID: mdl-36705397

ABSTRACT

Unlike typical chorismate mutases, the enzyme from Mycobacterium tuberculosis (MtCM) has only low activity on its own. Remarkably, its catalytic efficiency kcat/Km can be boosted more than 100-fold by complex formation with a partner enzyme. Recently, an autonomously fully active MtCM variant was generated using directed evolution, and its structure was solved by X-ray crystallography. However, key residues were involved in crystal contacts, challenging the functional interpretation of the structural changes. Here, we address these challenges by microsecond molecular dynamics simulations, followed up by additional kinetic and structural analyses of selected sets of specifically engineered enzyme variants. A comparison of wild-type MtCM with naturally and artificially activated MtCMs revealed the overall dynamic profiles of these enzymes as well as key interactions between the C-terminus and the active site loop. In the artificially evolved variant of this model enzyme, this loop is preorganized and stabilized by Pro52 and Asp55, two highly conserved residues in typical, highly active chorismate mutases. Asp55 stretches across the active site and helps to appropriately position active site residues Arg18 and Arg46 for catalysis. The role of Asp55 can be taken over by another acidic residue, if introduced at position 88 close to the C-terminus of MtCM, as suggested by molecular dynamics simulations and confirmed by kinetic investigations of engineered variants.


Subject(s)
Chorismate Mutase , Mycobacterium tuberculosis , Chorismate Mutase/chemistry , Molecular Dynamics Simulation , Research Design , Crystallography, X-Ray
3.
Science ; 369(6502): 440-445, 2020 07 24.
Article in English | MEDLINE | ID: mdl-32703877

ABSTRACT

The rational design of enzymes is an important goal for both fundamental and practical reasons. Here, we describe a process to learn the constraints for specifying proteins purely from evolutionary sequence data, design and build libraries of synthetic genes, and test them for activity in vivo using a quantitative complementation assay. For chorismate mutase, a key enzyme in the biosynthesis of aromatic amino acids, we demonstrate the design of natural-like catalytic function with substantial sequence diversity. Further optimization focuses the generative model toward function in a specific genomic context. The data show that sequence-based statistical models suffice to specify proteins and provide access to an enormous space of functional sequences. This result provides a foundation for a general process for evolution-based design of artificial proteins.


Subject(s)
Chorismate Mutase , Evolution, Molecular , Models, Genetic , Models, Statistical , Amino Acid Sequence , Chorismate Mutase/chemistry , Chorismate Mutase/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics
4.
J Biol Chem ; 295(51): 17514-17534, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33453995

ABSTRACT

Chorismate mutase (CM), an essential enzyme at the branch-point of the shikimate pathway, is required for the biosynthesis of phenylalanine and tyrosine in bacteria, archaea, plants, and fungi. MtCM, the CM from Mycobacterium tuberculosis, has less than 1% of the catalytic efficiency of a typical natural CM and requires complex formation with 3-deoxy-d-arabino-heptulosonate 7-phosphate synthase for high activity. To explore the full potential of MtCM for catalyzing its native reaction, we applied diverse iterative cycles of mutagenesis and selection, thereby raising kcat/Km 270-fold to 5 × 105m-1s-1, which is even higher than for the complex. Moreover, the evolutionarily optimized autonomous MtCM, which had 11 of its 90 amino acids exchanged, was stabilized compared with its progenitor, as indicated by a 9 °C increase in melting temperature. The 1.5 Å crystal structure of the top-evolved MtCM variant reveals the molecular underpinnings of this activity boost. Some acquired residues (e.g. Pro52 and Asp55) are conserved in naturally efficient CMs, but most of them lie beyond the active site. Our evolutionary trajectories reached a plateau at the level of the best natural enzymes, suggesting that we have exhausted the potential of MtCM. Taken together, these findings show that the scaffold of MtCM, which naturally evolved for mediocrity to enable inter-enzyme allosteric regulation of the shikimate pathway, is inherently capable of high activity.


Subject(s)
Bacterial Proteins/metabolism , Chorismate Mutase/metabolism , Mycobacterium tuberculosis/enzymology , 3-Deoxy-7-Phosphoheptulonate Synthase/chemistry , 3-Deoxy-7-Phosphoheptulonate Synthase/metabolism , Allosteric Regulation , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Biocatalysis , Catalytic Domain , Chorismate Mutase/chemistry , Chorismate Mutase/genetics , Crystallography, X-Ray , Directed Molecular Evolution , Kinetics , Molecular Dynamics Simulation , Mutagenesis , Protein Structure, Quaternary , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Sequence Alignment , Shikimic Acid/metabolism , Transition Temperature
5.
Biochemistry ; 57(5): 557-573, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29178787

ABSTRACT

Corynebacterium glutamicum is widely used for the industrial production of amino acids, nucleotides, and vitamins. The shikimate pathway enzymes DAHP synthase (CgDS, Cg2391) and chorismate mutase (CgCM, Cgl0853) play a key role in the biosynthesis of aromatic compounds. Here we show that CgCM requires the formation of a complex with CgDS to achieve full activity, and that both CgCM and CgDS are feedback regulated by aromatic amino acids binding to CgDS. Kinetic analysis showed that Phe and Tyr inhibit CgCM activity by inter-enzyme allostery, whereas binding of Trp to CgDS strongly activates CgCM. Mechanistic insights were gained from crystal structures of the CgCM homodimer, tetrameric CgDS, and the heterooctameric CgCM-CgDS complex, refined to 1.1, 2.5, and 2.2 Å resolution, respectively. Structural details from the allosteric binding sites reveal that DAHP synthase is recruited as the dominant regulatory platform to control the shikimate pathway, similar to the corresponding enzyme complex from Mycobacterium tuberculosis.


Subject(s)
3-Deoxy-7-Phosphoheptulonate Synthase/metabolism , Chorismate Mutase/metabolism , Corynebacterium glutamicum/enzymology , Tryptophan/metabolism , Allosteric Regulation , Amino Acids, Aromatic/metabolism , Chorismate Mutase/chemistry , Corynebacterium glutamicum/chemistry , Corynebacterium glutamicum/metabolism , Crystallography, X-Ray , Enzyme Activation , Models, Molecular , Phenylalanine/metabolism , Protein Conformation , Protein Multimerization , Shikimic Acid/metabolism , Tyrosine/metabolism
6.
J Mol Biol ; 428(6): 1237-1255, 2016 Mar 27.
Article in English | MEDLINE | ID: mdl-26776476

ABSTRACT

DAHP synthase and chorismate mutase catalyze key steps in the shikimate biosynthetic pathway en route to aromatic amino acids. In Mycobacterium tuberculosis, chorismate mutase (MtCM; Rv0948c), located at the branch point toward phenylalanine and tyrosine, has poor activity on its own. However, it is efficiently activated by the first enzyme of the pathway, DAHP synthase (MtDS; Rv2178c), through formation of a non-covalent MtCM-MtDS complex. Here, we show how MtDS serves as an allosteric platform for feedback regulation of both enzymes, using X-ray crystallography, small-angle X-ray scattering, size-exclusion chromatography, and multi-angle light scattering. Crystal structures of the fully inhibited MtDS and the allosterically down-regulated MtCM-MtDS complex, solved at 2.8 and 2.7Å, respectively, reveal how effector binding at the internal MtDS subunit interfaces regulates the activity of MtDS and MtCM. While binding of all three metabolic end products to MtDS shuts down the entire pathway, the binding of phenylalanine jointly with tyrosine releases MtCM from the MtCM-MtDS complex, hence suppressing MtCM activation by 'inter-enzyme allostery'. This elegant regulatory principle, invoking a transient allosteric enzyme interaction, seems to be driven by dynamics and is likely a general strategy used by nature.


Subject(s)
3-Deoxy-7-Phosphoheptulonate Synthase/chemistry , 3-Deoxy-7-Phosphoheptulonate Synthase/metabolism , Allosteric Regulation , Chorismate Mutase/chemistry , Chorismate Mutase/metabolism , Mycobacterium tuberculosis/enzymology , Shikimic Acid/metabolism , Chromatography, Gel , Crystallography, X-Ray , Dynamic Light Scattering , Metabolic Networks and Pathways , Protein Binding , Scattering, Small Angle
7.
Proc Natl Acad Sci U S A ; 111(49): 17516-21, 2014 Dec 09.
Article in English | MEDLINE | ID: mdl-25422475

ABSTRACT

For more than half a century, transition state theory has provided a useful framework for understanding the origins of enzyme catalysis. As proposed by Pauling, enzymes accelerate chemical reactions by binding transition states tighter than substrates, thereby lowering the activation energy compared with that of the corresponding uncatalyzed process. This paradigm has been challenged for chorismate mutase (CM), a well-characterized metabolic enzyme that catalyzes the rearrangement of chorismate to prephenate. Calculations have predicted the decisive factor in CM catalysis to be ground state destabilization rather than transition state stabilization. Using X-ray crystallography, we show, in contrast, that a sluggish variant of Bacillus subtilis CM, in which a cationic active-site arginine was replaced by a neutral citrulline, is a poor catalyst even though it effectively preorganizes chorismate for the reaction. A series of high-resolution molecular snapshots of the reaction coordinate, including the apo enzyme, and complexes with substrate, transition state analog and product, demonstrate that an active site, which is only complementary in shape to a reactive substrate conformer, is insufficient for effective catalysis. Instead, as with other enzymes, electrostatic stabilization of the CM transition state appears to be crucial for achieving high reaction rates.


Subject(s)
Bacillus subtilis/enzymology , Chorismate Mutase/chemistry , Catalysis , Catalytic Domain , Chorismic Acid/chemistry , Citrulline/chemistry , Crystallization , Crystallography, X-Ray , Cyclohexanecarboxylic Acids/chemistry , Cyclohexenes/chemistry , Electrons , Escherichia coli/metabolism , Kinetics , Ligands , Models, Molecular , Oxygen/chemistry , Protein Conformation , Protein Folding , Static Electricity
8.
J Struct Biol ; 185(2): 168-77, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23537847

ABSTRACT

Computational methods have been recently applied to the design of protein-protein interfaces. Using this approach, a 61 amino acid long protein called Spider Roll was engineered to recognize the kinase domain of the human p21-activated kinase 1 (PAK1) with good specificity but modest affinity (KD=100µM). Here we show that this artificial protein can be optimized by yeast surface display and fluorescence-activated cell sorting. After three rounds of mutagenesis and screening, a diverse set of tighter binding variants was obtained. A representative binder, MSR7, has a >10(2)-fold higher affinity for PAK1 when displayed on yeast and a 6 to 11-fold advantage when produced free in solution. In contrast to the starting Spider Roll protein, however, MSR7 unexpectedly exhibits characteristics typical of partially disordered proteins, including lower α-helical content, non-cooperative thermal denaturation, and NMR data showing peak broadening and poor signal dispersion. Although conformational disorder is increasingly recognized as an important property of proteins involved in cellular signaling and regulation, it is poorly modeled by current computational methods. Explicit consideration of structural flexibility may improve future protein designs and provide deeper insight into molecular events at protein-protein interfaces.


Subject(s)
Recombinant Fusion Proteins/chemistry , p21-Activated Kinases/chemistry , Amino Acid Sequence , Computer Simulation , Directed Molecular Evolution , Flow Cytometry , Fluorescence Polarization , Humans , Intrinsically Disordered Proteins/chemistry , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Engineering , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Saccharomyces cerevisiae
9.
PLoS One ; 9(12): e116234, 2014.
Article in English | MEDLINE | ID: mdl-25551646

ABSTRACT

The shikimate pathway enzyme chorismate mutase converts chorismate into prephenate, a precursor of Tyr and Phe. The intracellular chorismate mutase (MtCM) of Mycobacterium tuberculosis is poorly active on its own, but becomes >100-fold more efficient upon formation of a complex with the first enzyme of the shikimate pathway, 3-deoxy-d-arabino-heptulosonate-7-phosphate synthase (MtDS). The crystal structure of the enzyme complex revealed involvement of C-terminal MtCM residues with the MtDS interface. Here we employed evolutionary strategies to probe the tolerance to substitution of the C-terminal MtCM residues from positions 84-90. Variants with randomized positions were subjected to stringent selection in vivo requiring productive interactions with MtDS for survival. Sequence patterns identified in active library members coincide with residue conservation in natural chorismate mutases of the AroQδ subclass to which MtCM belongs. An Arg-Gly dyad at positions 85 and 86, invariant in AroQδ sequences, was intolerant to mutation, whereas Leu88 and Gly89 exhibited a preference for small and hydrophobic residues in functional MtCM-MtDS complexes. In the absence of MtDS, selection under relaxed conditions identifies positions 84-86 as MtCM integrity determinants, suggesting that the more C-terminal residues function in the activation by MtDS. Several MtCM variants, purified using a novel plasmid-based T7 RNA polymerase gene expression system, showed that a diminished ability to physically interact with MtDS correlates with reduced activatability and feedback regulatory control by Tyr and Phe. Mapping critical protein-protein interaction sites by evolutionary strategies may pinpoint promising targets for drugs that interfere with the activity of protein complexes.


Subject(s)
Chorismate Mutase/metabolism , Directed Molecular Evolution/methods , Mycobacterium tuberculosis/metabolism , Protein Interaction Mapping/methods , 3-Deoxy-7-Phosphoheptulonate Synthase/metabolism , Amino Acid Substitution , Base Sequence , Calibration , Chorismate Mutase/genetics , Gene Library , Hydrophobic and Hydrophilic Interactions , Molecular Sequence Data , Multienzyme Complexes/metabolism , Random Allocation
10.
Nat Chem Biol ; 9(8): 494-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23748672

ABSTRACT

Evolutionary advances are often fueled by unanticipated innovation. Directed evolution of a computationally designed enzyme suggests that pronounced molecular changes can also drive the optimization of primitive protein active sites. The specific activity of an artificial retro-aldolase was boosted >4,400-fold by random mutagenesis and screening, affording catalytic efficiencies approaching those of natural enzymes. However, structural and mechanistic studies reveal that the engineered catalytic apparatus, consisting of a reactive lysine and an ordered water molecule, was unexpectedly abandoned in favor of a new lysine residue in a substrate-binding pocket created during the optimization process. Structures of the initial in silico design, a mechanistically promiscuous intermediate and one of the most evolved variants highlight the importance of loop mobility and supporting functional groups in the emergence of the new catalytic center. Such internal competition between alternative reactive sites may have characterized the early evolution of many natural enzymes.


Subject(s)
Aldehyde-Lyases/chemistry , Aldehyde-Lyases/metabolism , Computational Biology , Directed Molecular Evolution , Biocatalysis , Catalytic Domain , Computer Simulation , Models, Molecular , Molecular Structure
11.
PLoS Genet ; 9(1): e1003187, 2013.
Article in English | MEDLINE | ID: mdl-23300488

ABSTRACT

The contemporary proteinogenic repertoire contains 20 amino acids with diverse functional groups and side chain geometries. Primordial proteins, in contrast, were presumably constructed from a subset of these building blocks. Subsequent expansion of the proteinogenic alphabet would have enhanced their capabilities, fostering the metabolic prowess and organismal fitness of early living systems. While the addition of amino acids bearing innovative functional groups directly enhances the chemical repertoire of proteomes, the inclusion of chemically redundant monomers is difficult to rationalize. Here, we studied how a simplified chorismate mutase evolves upon expanding its amino acid alphabet from nine to potentially 20 letters. Continuous evolution provided an enhanced enzyme variant that has only two point mutations, both of which extend the alphabet and jointly improve protein stability by >4 kcal/mol and catalytic activity tenfold. The same, seemingly innocuous substitutions (Ile→Thr, Leu→Val) occurred in several independent evolutionary trajectories. The increase in fitness they confer indicates that building blocks with very similar side chain structures are highly beneficial for fine-tuning protein structure and function.


Subject(s)
Amino Acids , Directed Molecular Evolution , Genetic Code , Proteins/genetics , Amino Acid Sequence , Amino Acid Substitution , Amino Acids/chemistry , Amino Acids/genetics , Chorismate Mutase/chemistry , Chorismate Mutase/genetics , Methanococcales/genetics , Molecular Dynamics Simulation , Molecular Sequence Data , Point Mutation , Protein Conformation , Protein Stability , Structure-Activity Relationship
12.
Chem Biodivers ; 9(11): 2507-27, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23161632

ABSTRACT

Tuberculosis (TB) is a devastating disease that claims millions of lives every year. Hindered access or non-compliance to medication, especially in developing countries, led to drug resistance, further aggravating the situation. With current standard therapies in use for over 50 years and only few new candidates in clinical trials, there is an urgent call for new TB drugs. A powerful tool for the development of new medication is structure-guided design, combined with virtual screening or docking studies. Here, we report the results of a drug-design project, which we based on a publication that claimed the structure-guided discovery of several promising and highly active inhibitors targeting the secreted chorismate mutase (*MtCM) from Mycobacterium tuberculosis. We set out to further improve on these compounds and synthesized a series of new derivatives. Thorough evaluation of these molecules in enzymatic assays revealed, to our dismay, that neither the claimed lead compounds, nor any of the synthesized derivatives, show any inhibitory effects against *MtCM.


Subject(s)
Antitubercular Agents/chemistry , Antitubercular Agents/pharmacology , Chorismate Mutase/antagonists & inhibitors , Drug Design , Mycobacterium tuberculosis/enzymology , Chorismate Mutase/metabolism , Humans , Molecular Docking Simulation , Mycobacterium tuberculosis/drug effects , Structure-Activity Relationship , Tuberculosis/drug therapy , Tuberculosis/microbiology
13.
Protein Sci ; 21(5): 717-26, 2012 May.
Article in English | MEDLINE | ID: mdl-22407837

ABSTRACT

Enzyme catalysts of a retroaldol reaction have been generated by computational design using a motif that combines a lysine in a nonpolar environment with water-mediated stabilization of the carbinolamine hydroxyl and ß-hydroxyl groups. Here, we show that the design process is robust and repeatable, with 33 new active designs constructed on 13 different protein scaffold backbones. The initial activities are not high but are increased through site-directed mutagenesis and laboratory evolution. Mutational data highlight areas for improvement in design. Different designed catalysts give different borohydride-reduced reaction intermediates, suggesting a distribution of properties of the designed enzymes that may be further explored and exploited.


Subject(s)
Aldehyde-Lyases/chemistry , Protein Engineering/methods , Recombinant Proteins/chemistry , Aldehyde-Lyases/genetics , Aldehyde-Lyases/metabolism , Catalytic Domain , Directed Molecular Evolution/methods , Models, Molecular , Mutagenesis, Site-Directed , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
15.
Biochemistry ; 50(40): 8594-602, 2011 Oct 11.
Article in English | MEDLINE | ID: mdl-21916414

ABSTRACT

Degradation tags are short peptide sequences that target proteins for destruction by housekeeping proteases. We previously utilized the C-terminal SsrA tag in directed evolution experiments to decrease the intracellular lifetime of a growth-limiting enzyme and thereby facilitate selection of highly active variants. In this study, we examine the N-terminal RepA tag as an alternative degradation signal for laboratory evolution. Although RepA proved to be less effective than SsrA at lowering protein concentrations in the cell, its N-terminal location dramatically reduced the occurrence of truncation and frameshift artifacts in selection experiments. We exploited this improvement to evolve a topologically redesigned chorismate mutase that is intrinsically disordered but already highly active for the conversion of chorismate to prephenate. After three rounds of mutagenesis and high-stringency selection, a robust and more nativelike variant was obtained that exhibited a catalytic efficiency (k(cat)/K(M) = 84000 M(-1) s(-1)) comparable to that of a natural dimeric chorismate mutase. Because of concomitant increases in catalyst yield, the level of intracellular prephenate production increased approximately 30-fold overall over the course of evolution.


Subject(s)
Archaeal Proteins/chemistry , Archaeal Proteins/metabolism , Chorismate Mutase/chemistry , Chorismate Mutase/metabolism , Directed Molecular Evolution/methods , Methanococcales/enzymology , Amino Acid Motifs , Amino Acid Sequence , Archaeal Proteins/genetics , Catalysis , Chorismate Mutase/genetics , Cyclohexanecarboxylic Acids/metabolism , Cyclohexenes/metabolism , Kinetics , Methanococcales/chemistry , Methanococcales/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Engineering
16.
J Mol Biol ; 399(4): 541-6, 2010 Jun 18.
Article in English | MEDLINE | ID: mdl-20433850

ABSTRACT

Consensus design is an appealing strategy for the stabilization of proteins. It exploits amino acid conservation in sets of homologous proteins to identify likely beneficial mutations. Nevertheless, its success depends on the phylogenetic diversity of the sequence set available. Here, we show that randomization of a single protein represents a reliable alternative source of sequence diversity that is essentially free of phylogenetic bias. A small number of functional protein sequences selected from binary-patterned libraries suffice as input for the consensus design of active enzymes that are easier to produce and substantially more stable than individual members of the starting data set. Although catalytic activity correlates less consistently with sequence conservation in these extensively randomized proteins, less extreme mutagenesis strategies might be adopted in practice to augment stability while maintaining function.


Subject(s)
Protein Engineering/methods , Proteins/chemistry , Proteins/genetics , Amino Acid Sequence , Base Sequence , Chorismate Mutase/chemistry , Chorismate Mutase/genetics , Chorismate Mutase/metabolism , Consensus Sequence , DNA, Bacterial/genetics , Enzyme Stability , Enzymes/chemistry , Enzymes/genetics , Enzymes/metabolism , Escherichia coli/enzymology , Escherichia coli/genetics , Models, Molecular , Molecular Sequence Data , Peptide Library , Phylogeny , Protein Conformation , Protein Stability , Proteins/metabolism , Sequence Alignment , Thermodynamics
17.
Protein Sci ; 19(5): 1000-10, 2010 May.
Article in English | MEDLINE | ID: mdl-20306491

ABSTRACT

Split proteins are versatile tools for detecting protein-protein interactions and studying protein folding. Here, we report a new, particularly small split enzyme, engineered from a thermostable chorismate mutase (CM). Upon dissecting the helical-bundle CM from Methanococcus jannaschii into a short N-terminal helix and a 3-helix segment and attaching an antiparallel leucine zipper dimerization domain to the individual fragments, we obtained a weakly active heterodimeric mutase. Using combinatorial mutagenesis and in vivo selection, we optimized the short linker sequences connecting the leucine zipper to the enzyme domain. One of the selected CMs was characterized in detail. It spontaneously assembles from the separately inactive fragments and exhibits wild-type like CM activity. Owing to the availability of a well characterized selection system, the simple 4-helix bundle topology, and the small size of the N-terminal helix, the heterodimeric CM could be a valuable scaffold for enzyme engineering efforts and as a split sensor for specifically oriented protein-protein interactions.


Subject(s)
Chorismate Mutase/chemistry , Chorismate Mutase/metabolism , Directed Molecular Evolution/methods , Protein Engineering/methods , Protein Subunits/metabolism , Amino Acid Sequence , Chorismate Mutase/genetics , Chorismate Mutase/isolation & purification , Combinatorial Chemistry Techniques/methods , Escherichia coli/genetics , Leucine Zippers , Methanococcus/enzymology , Methanococcus/genetics , Molecular Sequence Data , Mutagenesis , Protein Interaction Mapping , Protein Structure, Quaternary , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/isolation & purification , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sequence Alignment
18.
Biochim Biophys Acta ; 1804(4): 752-4, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19948253

ABSTRACT

The (13)C isotope effect for the conversion of prephenate to phenylpyruvate by the enzyme prephenate dehydratase from Methanocaldococcus jannaschii is 1.0334+/-0.0006. The size of this isotope effect suggests that the reaction is concerted. From the X-ray structure of a related enzyme, it appears that the only residue capable of acting as the general acid needed for removal of the hydroxyl group is threonine-172, which is contained in a conserved TRF motif. The more favorable entropy of activation for the enzyme-catalyzed process (25 eu larger than for the acid-catalyzed reaction) has been explained by a preorganized microenvironment that obviates the need for extensive solvent reorganization. This is consistent with forced planarity of the ring and side chain, which would place the leaving carboxyl and hydroxyl out of plane. Such distortion of the substrate may be a major contributor to catalysis.


Subject(s)
Archaeal Proteins/chemistry , Archaeal Proteins/metabolism , Methanococcales/enzymology , Prephenate Dehydratase/chemistry , Prephenate Dehydratase/metabolism , Archaeal Proteins/genetics , Carbon Isotopes , Catalysis , Catalytic Domain , Entropy , Enzyme Activation , Kinetics , Methanococcales/genetics , Prephenate Dehydratase/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Threonine/chemistry
19.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 65(Pt 10): 1048-52, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19851019

ABSTRACT

Chorismate mutase catalyzes a key step in the shikimate-biosynthetic pathway and hence is an essential enzyme in bacteria, plants and fungi. Mycobacterium tuberculosis contains two chorismate mutases, a secreted and an intracellular one, the latter of which (MtCM; Rv0948c; 90 amino-acid residues; 10 kDa) is the subject of this work. Here are reported the gene expression, purification and crystallization of MtCM alone and of its complex with another shikimate-pathway enzyme, DAHP synthase (MtDS; Rv2178c; 472 amino-acid residues; 52 kDa), which has been shown to enhance the catalytic efficiency of MtCM. The MtCM-MtDS complex represents the first noncovalent enzyme complex from the common shikimate pathway to be structurally characterized. Soaking experiments with a transition-state analogue are also reported. The crystals of MtCM and the MtCM-MtDS complex diffracted to 1.6 and 2.1 A resolution, respectively.


Subject(s)
3-Deoxy-7-Phosphoheptulonate Synthase/chemistry , Chorismate Mutase/chemistry , Mycobacterium tuberculosis/enzymology , Crystallization , Crystallography, X-Ray , Multienzyme Complexes/chemistry
20.
EMBO J ; 28(14): 2128-42, 2009 Jul 22.
Article in English | MEDLINE | ID: mdl-19556970

ABSTRACT

Chorismate mutase catalyzes a key step in the shikimate biosynthetic pathway towards phenylalanine and tyrosine. Curiously, the intracellular chorismate mutase of Mycobacterium tuberculosis (MtCM; Rv0948c) has poor activity and lacks prominent active-site residues. However, its catalytic efficiency increases >100-fold on addition of DAHP synthase (MtDS; Rv2178c), another shikimate-pathway enzyme. The 2.35 A crystal structure of the MtCM-MtDS complex bound to a transition-state analogue shows a central core formed by four MtDS subunits sandwiched between two MtCM dimers. Structural comparisons imply catalytic activation to be a consequence of the repositioning of MtCM active-site residues on binding to MtDS. The mutagenesis of the C-terminal extrusion of MtCM establishes conserved residues as part of the activation machinery. The chorismate-mutase activity of the complex, but not of MtCM alone, is inhibited synergistically by phenylalanine and tyrosine. The complex formation thus endows the shikimate pathway of M. tuberculosis with an important regulatory feature. Experimental evidence suggests that such non-covalent enzyme complexes comprising an AroQ(delta) subclass chorismate mutase like MtCM are abundant in the bacterial order Actinomycetales.


Subject(s)
3-Deoxy-7-Phosphoheptulonate Synthase/chemistry , Chorismate Mutase/chemistry , Mycobacterium tuberculosis/chemistry , Mycobacterium tuberculosis/enzymology , 3-Deoxy-7-Phosphoheptulonate Synthase/metabolism , Amino Acid Sequence , Catalytic Domain , Chorismate Mutase/genetics , Chorismate Mutase/metabolism , Cloning, Molecular , Corynebacterium glutamicum/enzymology , Crystallography, X-Ray , Enzyme Activation , Malates/chemistry , Models, Molecular , Molecular Sequence Data , Mycobacterium tuberculosis/metabolism , Phenylalanine/metabolism , Sequence Alignment , Shikimic Acid/metabolism , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL