Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 481
Filter
Add more filters

Publication year range
1.
Proc Natl Acad Sci U S A ; 121(32): e2400439121, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39074291

ABSTRACT

Protein engineering often targets binding pockets or active sites which are enriched in epistasis-nonadditive interactions between amino acid substitutions-and where the combined effects of multiple single substitutions are difficult to predict. Few existing sequence-fitness datasets capture epistasis at large scale, especially for enzyme catalysis, limiting the development and assessment of model-guided enzyme engineering approaches. We present here a combinatorially complete, 160,000-variant fitness landscape across four residues in the active site of an enzyme. Assaying the native reaction of a thermostable ß-subunit of tryptophan synthase (TrpB) in a nonnative environment yielded a landscape characterized by significant epistasis and many local optima. These effects prevent simulated directed evolution approaches from efficiently reaching the global optimum. There is nonetheless wide variability in the effectiveness of different directed evolution approaches, which together provide experimental benchmarks for computational and machine learning workflows. The most-fit TrpB variants contain a substitution that is nearly absent in natural TrpB sequences-a result that conservation-based predictions would not capture. Thus, although fitness prediction using evolutionary data can enrich in more-active variants, these approaches struggle to identify and differentiate among the most-active variants, even for this near-native function. Overall, this work presents a large-scale testing ground for model-guided enzyme engineering and suggests that efficient navigation of epistatic fitness landscapes can be improved by advances in both machine learning and physical modeling.


Subject(s)
Catalytic Domain , Epistasis, Genetic , Tryptophan Synthase , Catalytic Domain/genetics , Tryptophan Synthase/genetics , Tryptophan Synthase/metabolism , Tryptophan Synthase/chemistry , Protein Engineering/methods , Amino Acid Substitution , Models, Molecular
2.
Nat Chem Biol ; 20(8): 1086-1093, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38744987

ABSTRACT

Aromatic amino acids and their derivatives are diverse primary and secondary metabolites with critical roles in protein synthesis, cell structure and integrity, defense and signaling. All de novo aromatic amino acid production relies on a set of ancient and highly conserved chemistries. Here we introduce a new enzymatic transformation for L-tyrosine synthesis by demonstrating that the ß-subunit of tryptophan synthase-which natively couples indole and L-serine to form L-tryptophan-can act as a latent 'tyrosine synthase'. A single substitution of a near-universally conserved catalytic residue unlocks activity toward simple phenol analogs and yields exclusive para carbon-carbon bond formation to furnish L-tyrosines. Structural and mechanistic studies show how a new active-site water molecule orients phenols for a nonnative mechanism of alkylation, with additional directed evolution resulting in a net >30,000-fold rate enhancement. This new biocatalyst can be used to efficiently prepare valuable L-tyrosine analogs at gram scales and provides the missing chemistry for a conceptually different pathway to L-tyrosine.


Subject(s)
Tryptophan Synthase , Tyrosine , Tryptophan Synthase/metabolism , Tryptophan Synthase/chemistry , Tyrosine/chemistry , Tyrosine/metabolism , Catalytic Domain , Models, Molecular , Tyrosine Phenol-Lyase/metabolism , Tyrosine Phenol-Lyase/chemistry , Tyrosine Phenol-Lyase/genetics , Protein Subunits/chemistry , Protein Subunits/metabolism , Biocatalysis , Tryptophan/chemistry , Tryptophan/metabolism
3.
Proc Natl Acad Sci U S A ; 119(2)2022 01 11.
Article in English | MEDLINE | ID: mdl-34996869

ABSTRACT

NMR-assisted crystallography-the integrated application of solid-state NMR, X-ray crystallography, and first-principles computational chemistry-holds significant promise for mechanistic enzymology: by providing atomic-resolution characterization of stable intermediates in enzyme active sites, including hydrogen atom locations and tautomeric equilibria, NMR crystallography offers insight into both structure and chemical dynamics. Here, this integrated approach is used to characterize the tryptophan synthase α-aminoacrylate intermediate, a defining species for pyridoxal-5'-phosphate-dependent enzymes that catalyze ß-elimination and replacement reactions. For this intermediate, NMR-assisted crystallography is able to identify the protonation states of the ionizable sites on the cofactor, substrate, and catalytic side chains as well as the location and orientation of crystallographic waters within the active site. Most notable is the water molecule immediately adjacent to the substrate ß-carbon, which serves as a hydrogen bond donor to the ε-amino group of the acid-base catalytic residue ßLys87. From this analysis, a detailed three-dimensional picture of structure and reactivity emerges, highlighting the fate of the L-serine hydroxyl leaving group and the reaction pathway back to the preceding transition state. Reaction of the α-aminoacrylate intermediate with benzimidazole, an isostere of the natural substrate indole, shows benzimidazole bound in the active site and poised for, but unable to initiate, the subsequent bond formation step. When modeled into the benzimidazole position, indole is positioned with C3 in contact with the α-aminoacrylate Cß and aligned for nucleophilic attack. Here, the chemically detailed, three-dimensional structure from NMR-assisted crystallography is key to understanding why benzimidazole does not react, while indole does.


Subject(s)
Alanine/analogs & derivatives , Catalytic Domain , Crystallography, X-Ray/methods , Magnetic Resonance Spectroscopy/methods , Tryptophan Synthase/chemistry , Catalysis , Indoles , Magnetic Resonance Imaging , Nuclear Magnetic Resonance, Biomolecular , Pyridoxal Phosphate/metabolism , Tryptophan Synthase/metabolism
4.
Arch Microbiol ; 206(9): 390, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39222088

ABSTRACT

Essential amino acid, tryptophan which intake from food plays a critical role in numerous metabolic functions, exhibiting extensive biological functions and applications. Tryptophan is beneficial for the food sector by enhancing nutritional content and promoting the development of functional foods. A putative gene encoding tryptophan synthase was the first identified in Sphingobacterium soilsilvae Em02, a cellulosic bacterium making it inherently more environmentally friendly. The gene was cloned and expressed in exogenous host Escherichia coli, to elucidate its function. The recombinant tryptophan synthase with a molecular weight 42 KDa was expressed in soluble component. The enzymatic activity to tryptophan synthase in vivo was assessed using indole and L-serine and purified tryptophan synthase. The optimum enzymatic activity for tryptophan synthase was recorded at 50 ºC and pH 7.0, which was improved in the presence of metal ions Mg2+, Sr2+ and Mn2+, whereas Cu2+, Zn2+ and Co2+ proved to be inhibitory. Using site-directed mutagenesis, the consensus pattern HK-S-[GGGSN]-E-S in the tryptophan synthase was demonstrated with K100Q, S202A, G246A, E361A and S385A as the active sites. Tryptophan synthase has been demonstrated to possess the defining characteristics of the ß-subunits. The tryptophan synthase may eventually be useful for tryptophan production on a larger scale. Its diverse applications highlight the potential for improving both the quality and health benefits of food products, making it an essential component in advancing food science and technology.


Subject(s)
Escherichia coli , Mutagenesis, Site-Directed , Tryptophan Synthase , Tryptophan , Tryptophan Synthase/metabolism , Tryptophan Synthase/genetics , Tryptophan Synthase/chemistry , Tryptophan/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Sphingomonadaceae/enzymology , Sphingomonadaceae/genetics , Sphingomonadaceae/metabolism , Recombinant Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/chemistry , Catalytic Domain , Cloning, Molecular , Hydrogen-Ion Concentration , Indoles/metabolism , Catalysis , Serine/metabolism
5.
Faraday Discuss ; 252(0): 306-322, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-38910409

ABSTRACT

Enzymes exhibit diverse conformations, as represented in the free energy landscape (FEL). Such conformational diversity provides enzymes with the ability to evolve towards novel functions. The challenge lies in identifying mutations that enhance specific conformational changes, especially if located in distal sites from the active site cavity. The shortest path map (SPM) method, which we developed to address this challenge, constructs a graph based on the distances and correlated motions of residues observed in nanosecond timescale molecular dynamics (MD) simulations. We recently introduced a template based AlphaFold2 (tAF2) approach coupled with 10 nanosecond MD simulations to quickly estimate the conformational landscape of enzymes and assess how the FEL is shifted after mutation. In this study, we evaluate the potential of SPM when coupled with tAF2-MD in estimating conformational heterogeneity and identifying key conformationally-relevant positions. The selected model system is the beta subunit of tryptophan synthase (TrpB). We compare how the SPM pathways differ when integrating tAF2 with different MD simulation lengths from as short as 10 ns until 50 ns and considering two distinct Amber forcefield and water models (ff14SB/TIP3P versus ff19SB/OPC). The new methodology can more effectively capture the distal mutations found in laboratory evolution, thus showcasing the efficacy of tAF2-MD-SPM in rapidly estimating enzyme dynamics and identifying the key conformationally relevant hotspots for computational enzyme engineering.


Subject(s)
Biocatalysis , Molecular Dynamics Simulation , Protein Conformation , Tryptophan Synthase/chemistry , Tryptophan Synthase/metabolism , Catalytic Domain
6.
J Chem Inf Model ; 64(3): 983-1003, 2024 02 12.
Article in English | MEDLINE | ID: mdl-38291608

ABSTRACT

L-tryptophan (l-Trp), a vital amino acid for the survival of various organisms, is synthesized by the enzyme tryptophan synthase (TS) in organisms such as eubacteria, archaebacteria, protista, fungi, and plantae. TS, a pyridoxal 5'-phosphate (PLP)-dependent enzyme, comprises α and ß subunits that typically form an α2ß2 tetramer. The enzyme's activity is regulated by the conformational switching of its α and ß subunits between the open (T state) and closed (R state) conformations. Many microorganisms rely on TS for growth and replication, making the enzyme and the l-Trp biosynthetic pathway potential drug targets. For instance, Mycobacterium tuberculosis, Chlamydiae bacteria, Streptococcus pneumoniae, Francisella tularensis, Salmonella bacteria, and Cryptosporidium parasitic protozoa depend on l-Trp synthesis. Antibiotic-resistant salmonella strains have emerged, underscoring the need for novel drugs targeting the l-Trp biosynthetic pathway, especially for salmonella-related infections. A single amino acid mutation can significantly impact enzyme function, affecting stability, conformational dynamics, and active or allosteric sites. These changes influence interactions, catalytic activity, and protein-ligand/protein-protein interactions. This study focuses on the impact of mutating the ßGln114 residue on the catalytic and allosteric sites of TS. Extensive molecular dynamics simulations were conducted on E(PLP), E(AEX1), E(A-A), and E(C3) forms of TS using the WT, ßQ114A, and ßQ114N versions. The results show that both the ßQ114A and ßQ114N mutations increase protein backbone root mean square deviation fluctuations, destabilizing all TS forms. Conformational and hydrogen bond analyses suggest the significance of ßGln114 drifting away from cofactor/intermediates and forming hydrogen bonds with water molecules necessary for l-Trp biosynthesis. The ßQ114A mutation creates a gap between ßAla114 and cofactor/intermediates, hindering hydrogen bond formation due to short side chains and disrupting ß-sites. Conversely, the ßQ114N mutation positions ßAsn114 closer to cofactor/intermediates, forming hydrogen bonds with O3 of cofactors/intermediates and nearby water molecules, potentially disrupting the l-Trp biosynthetic mechanism.


Subject(s)
Cryptosporidiosis , Cryptosporidium , Tryptophan Synthase , Humans , Tryptophan Synthase/genetics , Tryptophan Synthase/chemistry , Tryptophan Synthase/metabolism , Catalytic Domain , Molecular Dynamics Simulation , Salmonella typhimurium/genetics , Cryptosporidium/metabolism , Protein Conformation , Amino Acids , Mutation , Water , Kinetics
7.
Molecules ; 29(4)2024 Feb 06.
Article in English | MEDLINE | ID: mdl-38398508

ABSTRACT

Tryptophan synthase (TRPS) is a complex enzyme responsible for tryptophan biosynthesis. It occurs in bacteria, plants, and fungi as an αßßα heterotetramer. Although encoded by independent genes in bacteria and plants, in fungi, TRPS is generated by a single gene that concurrently expresses the α and ß entities, which are linked by an elongated peculiar segment. We conducted 1 µs all-atom molecular dynamics simulations on Hemileia vastatrix TRPS to address two questions: (i) the role of the linker segment and (ii) the comparative mode of action. Since there is not an experimental structure, we started our simulations with homology modeling. Based on the results, it seems that TRPS makes use of an already-existing tunnel that can spontaneously move the indole moiety from the α catalytic pocket to the ß one. Such behavior was completely disrupted in the simulation without the linker. In light of these results and the αß dimer's low stability, the full-working TRPS single genes might be the result of a particular evolution. Considering the significant losses that Hemileia vastatrix causes to coffee plantations, our next course of action will be to use the TRPS to look for substances that can block tryptophan production and therefore control the disease.


Subject(s)
Basidiomycota , Molecular Dynamics Simulation , Tryptophan Synthase , Tryptophan Synthase/chemistry , Tryptophan Synthase/genetics , Tryptophan Synthase/metabolism , Tryptophan , Fungi/metabolism
8.
J Chem Phys ; 158(11): 115101, 2023 Mar 21.
Article in English | MEDLINE | ID: mdl-36948822

ABSTRACT

Tryptophan synthase (TRPS) is a bifunctional enzyme consisting of α- and ß-subunits that catalyzes the last two steps of L-tryptophan (L-Trp) biosynthesis. The first stage of the reaction at the ß-subunit is called ß-reaction stage I, which converts the ß-ligand from an internal aldimine [E(Ain)] to an α-aminoacrylate [E(A-A)] intermediate. The activity is known to increase 3-10-fold upon the binding of 3-indole-D-glycerol-3'-phosphate (IGP) at the α-subunit. The effect of α-ligand binding on ß-reaction stage I at the distal ß-active site is not well understood despite the abundant structural information available for TRPS. Here, we investigate the ß-reaction stage I by carrying out minimum-energy pathway searches based on a hybrid quantum mechanics/molecular mechanics (QM/MM) model. The free-energy differences along the pathway are also examined using QM/MM umbrella sampling simulations with QM calculations at the B3LYP-D3/aug-cc-pVDZ level of theory. Our simulations suggest that the sidechain orientation of ßD305 near the ß-ligand likely plays an essential role in the allosteric regulation: a hydrogen bond is formed between ßD305 and the ß-ligand in the absence of the α-ligand, prohibiting a smooth rotation of the hydroxyl group in the quinonoid intermediate, whereas the dihedral angle rotates smoothly after the hydrogen bond is switched from ßD305-ß-ligand to ßD305-ßR141. This switch could occur upon the IGP-binding at the α-subunit, as evidenced by the existing TRPS crystal structures.


Subject(s)
Tryptophan Synthase , Tryptophan Synthase/chemistry , Tryptophan Synthase/metabolism , Allosteric Regulation , Binding Sites , Ligands , Protein Conformation , Kinetics
9.
Proc Natl Acad Sci U S A ; 117(1): 346-354, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31871208

ABSTRACT

Tryptophan synthase (TS) is a heterotetrameric αßßα complex. It is characterized by the channeling of the reaction intermediate indole and the mutual activation of the α-subunit TrpA and the ß-subunit TrpB via a complex allosteric network. We have analyzed this allosteric network by means of ancestral sequence reconstruction (ASR), which is an in silico method to resurrect extinct ancestors of modern proteins. Previously, the sequences of TrpA and TrpB from the last bacterial common ancestor (LBCA) have been computed by means of ASR and characterized. LBCA-TS is similar to modern TS by forming a αßßα complex with indole channeling taking place. However, LBCA-TrpA allosterically decreases the activity of LBCA-TrpB, whereas, for example, the modern ncTrpA from Neptuniibacter caesariensis allosterically increases the activity of ncTrpB. To identify amino acid residues that are responsible for this inversion of the allosteric effect, all 6 evolutionary TrpA and TrpB intermediates that stepwise link LBCA-TS with ncTS were characterized. Remarkably, the switching from TrpB inhibition to TrpB activation by TrpA occurred between 2 successive TS intermediates. Sequence comparison of these 2 intermediates and iterative rounds of site-directed mutagenesis allowed us to identify 4 of 413 residues from TrpB that are crucial for its allosteric activation by TrpA. The effect of our mutational studies was rationalized by a community analysis based on molecular dynamics simulations. Our findings demonstrate that ancestral sequence reconstruction can efficiently identify residues contributing to allosteric signal propagation in multienzyme complexes.


Subject(s)
Bacterial Proteins/genetics , Computational Biology , Extinction, Biological , Protein Subunits/genetics , Tryptophan Synthase/genetics , Allosteric Regulation/genetics , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Oceanospirillaceae/genetics , Oceanospirillaceae/metabolism , Phylogeny , Protein Subunits/chemistry , Protein Subunits/metabolism , Sequence Alignment , Structural Homology, Protein , Tryptophan/biosynthesis , Tryptophan Synthase/chemistry , Tryptophan Synthase/metabolism
10.
Molecules ; 28(21)2023 Oct 26.
Article in English | MEDLINE | ID: mdl-37959692

ABSTRACT

Tryptophan synthetase (TSase), which functions as a tetramer, is a typical enzyme with a substrate channel effect, and shows excellent performance in the production of non-standard amino acids, histamine, and other biological derivatives. Based on previous work, we fused a mutant CE protein (colistin of E. coli, a polypeptide with antibacterial activity) sequence with the sequence of TSase to explore whether its catalytic activity could be enhanced, and we also analyzed whether the addition of a DNA scaffold was a feasible strategy. Here, dCE (CE protein without DNase activity) protein tags were constructed and fused to the TrapA and TrapB subunits of TSase, and the whole cell was used for the catalytic reaction. The results showed that after the dCE protein tag was fused to the TrapB subunit, its whole cell catalytic activity increased by 50%. Next, the two subunits were expressed separately, and the proteins were bound in vitro to ensure equimolar combination between the two subunits. After the dCE label was fused to TrapB, the activity of TSase assembled with TrapA also improved. A series of experiments revealed that the enzyme fused with dCE9 showed higher activity than the wild-type protein. In general, the activity of assembly TSase was optimal when the temperature was 50 °C and the pH was about 9.0. After a long temperature treatment, the enzyme maintained good activity. With the addition of exogenous nucleic acid, the activity of the enzyme increased. The maximum yield was 0.58 g/L, which was almost three times that of the wild-type TSase (0.21 g/L). The recombinant TSase constructed in this study with dCE fusion had the advantages of higher heat resistance and higher activity, and confirmed the feasibility of adding a nucleic acid scaffold, providing a new idea for the improvement of structurally similar enzymes.


Subject(s)
Nucleic Acids , Tryptophan Synthase , Tryptophan Synthase/chemistry , Tryptophan Synthase/genetics , Tryptophan Synthase/metabolism , Escherichia coli/metabolism , Amino Acids
11.
Biochemistry ; 61(18): 2025-2035, 2022 09 20.
Article in English | MEDLINE | ID: mdl-36084241

ABSTRACT

The regiospecific prenylation of an aromatic amino acid catalyzed by a dimethylallyl-l-tryptophan synthase (DMATS) is a key step in the biosynthesis of many fungal and bacterial natural products. DMATS enzymes share a common "ABBA" fold with divergent active site contours that direct alternative C-C, C-N, and C-O bond-forming trajectories. DMATS1 from Fusarium fujikuroi catalyzes the reverse N-prenylation of l-Trp by generating an allylic carbocation from dimethylallyl diphosphate (DMAPP) that then alkylates the indole nitrogen of l-Trp. DMATS1 stands out among the greater DMATS family because it exhibits unusually broad substrate specificity: it can utilize geranyl diphosphate (GPP) or l-Tyr as an alternative prenyl donor or acceptor, respectively; it can catalyze both forward and reverse prenylation, i.e., at C1 or C3 of DMAPP; and it can catalyze C-N and C-O bond-forming reactions. Here, we report the crystal structures of DMATS1 and its complexes with l-Trp or l-Tyr and unreactive thiolodiphosphate analogues of the prenyl donors DMAPP and GPP. Structures of ternary complexes mimic Michaelis complexes with actual substrates and illuminate active site features that govern prenylation regiochemistry. Comparison with CymD, a bacterial enzyme that catalyzes the reverse N-prenylation of l-Trp with DMAPP, indicates that bacterial and fungal DMATS enzymes share a conserved reaction mechanism. However, the narrower active site contour of CymD enforces narrower substrate specificity. Structure-function relationships established for DMATS enzymes will ultimately inform protein engineering experiments that will broaden the utility of these enzymes as useful tools for synthetic biology.


Subject(s)
Biological Products , Dimethylallyltranstransferase , Tryptophan Synthase , Catalysis , Dimethylallyltranstransferase/chemistry , Fusarium , Hemiterpenes , Indoles , Neoprene , Nitrogen , Organophosphorus Compounds , Prenylation , Substrate Specificity , Tryptophan/chemistry , Tryptophan Synthase/metabolism
12.
Bioessays ; 42(9): e2000092, 2020 09.
Article in English | MEDLINE | ID: mdl-32720327

ABSTRACT

A general framework by which dynamic interactions within a protein will promote the necessary series of structural changes, or "conformational cycle," required for function is proposed. It is suggested that the free-energy landscape of a protein is biased toward this conformational cycle. Fluctuations into higher energy, although thermally accessible, conformations drive the conformational cycle forward. The amino acid interaction network is defined as those intraprotein interactions that contribute most to the free-energy landscape. Some network connections are consistent in every structural state, while others periodically change their interaction strength according to the conformational cycle. It is reviewed here that structural transitions change these periodic network connections, which then predisposes the protein toward the next set of network changes, and hence the next structural change. These concepts are illustrated by recent work on tryptophan synthase. Disruption of these dynamic connections may lead to aberrant protein function and disease states.


Subject(s)
Tryptophan Synthase , Allosteric Regulation , Amino Acids , Catalysis , Protein Conformation , Tryptophan , Tryptophan Synthase/metabolism
13.
Proc Natl Acad Sci U S A ; 116(25): 12468-12477, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31097582

ABSTRACT

A striking difference between genital and ocular clinical isolates of Chlamydia trachomatis is that only the former express a functional tryptophan synthase and therefore can synthesize tryptophan by indole salvage. Ocular isolates uniformly cannot use indole due to inactivating mutations within tryptophan synthase, indicating a selection against maintaining this enzyme in the ocular environment. Here, we demonstrate that this selection occurs in two steps. First, specific indole derivatives, produced by the human gut microbiome and present in serum, rapidly induce expression of C. trachomatis tryptophan synthase, even under conditions of tryptophan sufficiency. We demonstrate that these indole derivatives function by acting as de-repressors of C. trachomatis TrpR. Second, trp operon de-repression is profoundly deleterious when infected cells are in an indole-deficient environment, because in the absence of indole, tryptophan synthase deaminates serine to pyruvate and ammonia. We have used biochemical and genetic approaches to demonstrate that expression of wild-type tryptophan synthase is required for the bactericidal production of ammonia. Pertinently, although these indole derivatives de-repress the trpRBA operon of C. trachomatis strains with trpA or trpB mutations, no ammonia is produced, and no deleterious effects are observed. Our studies demonstrate that tryptophan synthase can catalyze the ammonia-generating ß-elimination reaction within any live bacterium. Our results also likely explain previous observations demonstrating that the same indole derivatives inhibit the growth of other pathogenic bacterial species, and why high serum levels of these indole derivatives are favorable for the prognosis of diseased conditions associated with bacterial dysbiosis.


Subject(s)
Ammonia/metabolism , Chlamydia Infections/microbiology , Chlamydia trachomatis/metabolism , Eye/microbiology , Genitalia/microbiology , Tryptophan Synthase/metabolism , Chlamydia trachomatis/enzymology , Chlamydia trachomatis/genetics , Humans , Tryptophan/metabolism
14.
Chembiochem ; 22(1): 5-16, 2021 01 05.
Article in English | MEDLINE | ID: mdl-32677310

ABSTRACT

Tryptophan synthase (TrpS) has emerged as a paragon of noncanonical amino acid (ncAA) synthesis and is an ideal biocatalyst for synthetic and biological applications. TrpS catalyzes an irreversible, C-C bond-forming reaction between indole and serine to make l-tryptophan; native TrpS complexes possess fairly broad specificity for indole analogues, but are difficult to engineer to extend substrate scope or to confer other useful properties due to allosteric constraints and their heterodimeric structure. Directed evolution freed the catalytically relevant TrpS ß-subunit (TrpB) from allosteric regulation by its TrpA partner and has enabled dramatic expansion of the enzyme's substrate scope. This review examines the long and storied career of TrpS from the perspective of its application in ncAA synthesis and biocatalytic cascades.


Subject(s)
Amino Acids/biosynthesis , Tryptophan Synthase/metabolism , Amino Acids/chemistry , Biocatalysis , Tryptophan Synthase/chemistry
15.
Angew Chem Int Ed Engl ; 60(39): 21412-21417, 2021 09 20.
Article in English | MEDLINE | ID: mdl-34269506

ABSTRACT

The ß-subunit of tryptophan synthase (TrpB) catalyzes a PLP-mediated ß-substitution reaction between indole and serine to form L-Trp. A succession of TrpB protein engineering campaigns to expand the enzyme's nucleophile substrate range has enabled the biocatalytic production of diverse non-canonical amino acids (ncAAs). Here, we show that ketone-derived enolates can serve as nucleophiles in the TrpB reaction to achieve the asymmetric alkylation of ketones, an outstanding challenge in synthetic chemistry. We engineered TrpB by directed evolution to catalyze the asymmetric alkylation of propiophenone and 2-fluoroacetophenone with a high degree of selectivity. In reactions with propiophenone, preference for the opposite product diastereomer emerges over the course of evolution, demonstrating that full control over the stereochemistry at the new chiral center can be achieved. The addition of this new reaction to the TrpB platform is a crucial first step toward the development of efficient methods to synthesize non-canonical prolines and other chirally dense nitrogen heterocycles.


Subject(s)
Ketones/metabolism , Protein Engineering , Tryptophan Synthase/metabolism , Alkylation , Biocatalysis , Ketones/chemistry , Molecular Structure , Tryptophan Synthase/chemistry
16.
Plant J ; 98(2): 329-345, 2019 04.
Article in English | MEDLINE | ID: mdl-30604574

ABSTRACT

Primary metabolism plays an important role in plant growth and development, however the relationship between primary metabolism and the adaptive immune response is largely unknown. Here, we employed RNA interference (RNAi), virus-induced gene silencing (VIGS) technology, phytohormone profiling, genetic studies, and transcriptome and metabolome analysis to investigate the function of the tryptophan synthesis pathway in the resistance of cotton to V. dahliae. We found that knock-down of GbTSA1 (Tryptophan Synthase α) and GbTSB1 (tryptophan synthase ß) induced a spontaneous cell death phenotype in a salicylic acid (SA)-dependent manner and enhanced resistance to V. dahliae in cotton plants. Metabolome analysis showed that indole and indolic metabolites were highly accumulated in GbTSA1- or GbTSB1-silenced plants. Transcriptomic analysis showed that exogenous indole promotes the expression levels of genes involved in SA synthesis and the defense response. Similarly, indole application strongly enhanced cotton resistance to V. dahliae. These results suggested that metabolic intermediates in the Trp synthesis pathway may be a signal to activate SA synthesis. These results also provided a strategy to elicit plant defense responses by the application of indole.


Subject(s)
Cell Death , Gossypium/immunology , Gossypium/metabolism , Plant Immunity , Tryptophan Synthase/metabolism , Arabidopsis Proteins , Calcium-Binding Proteins , Disease Resistance/genetics , Disease Resistance/immunology , Gene Expression Profiling , Gene Expression Regulation, Plant , Gene Silencing , Gossypium/genetics , Metabolome , Plant Diseases , Plant Growth Regulators , Plant Proteins/genetics , Plant Proteins/metabolism , RNA Interference , Salicylic Acid/metabolism , Sequence Analysis , Transcriptome , Verticillium
17.
J Biomol NMR ; 74(6-7): 341-354, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32415580

ABSTRACT

Backbone assignments for the isolated α-subunit of Salmonella typhimurium tryptophan synthase (TS) are reported based on triple resonance solution-state NMR experiments on a uniformly 2H,13C,15N-labeled sample. From the backbone chemical shifts, secondary structure and random coil index order parameters (RCI-S2) are predicted. Titration with the 3-indole-D-glycerol 3'-phosphate analog, N-(4'-trifluoromethoxybenzenesulfonyl)-2-aminoethyl phosphate (F9), leads to chemical shift perturbations indicative of conformational changes from which an estimate of the dissociation constant is obtained. Comparisons of the backbone chemical-shifts, RCI-S2 values, and site-specific relaxation times with and without F9 reveal allosteric changes including modulation in secondary structures and loop rigidity induced upon ligand binding. A comparison is made to the X-ray crystal structure of the α-subunit in the full TS αßßα bi-enzyme complex and to two new X-ray crystal structures of the isolated TS α-subunit reported in this work.


Subject(s)
Nuclear Magnetic Resonance, Biomolecular/methods , Salmonella typhimurium/enzymology , Tryptophan Synthase/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Binding Sites , Catalysis , Crystallography, X-Ray , Models, Molecular , Molecular Dynamics Simulation , Nitrogen Isotopes , Protein Conformation , Protein Structure, Secondary , Protein Subunits/chemistry , Solutions , Tryptophan Synthase/metabolism
18.
Chembiochem ; 21(1-2): 80-83, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31513332

ABSTRACT

We report a simple, one-step enzymatic synthesis of the blue fluorescent noncanonical amino acid ß-(1-azulenyl)-l-alanine (AzAla). By using an engineered tryptophan synthase ß-subunit (TrpB), stereochemically pure AzAla can be synthesized at scale starting from commercially available azulene and l-serine. Mutation of a universally conserved catalytic glutamate in the active site to glycine has only a modest effect on native activity with indole but abolishes activity on azulene, suggesting that this glutamate activates azulene for nucleophilic attack by stabilization of the aromatic ion.


Subject(s)
Amino Acids/biosynthesis , Fluorescent Dyes/metabolism , Serine/biosynthesis , Tryptophan Synthase/metabolism , Amino Acids/chemistry , Biocatalysis , Fluorescent Dyes/chemistry , Models, Molecular , Molecular Structure , Serine/chemistry , Stereoisomerism , Tryptophan Synthase/chemistry
19.
Org Biomol Chem ; 18(22): 4189-4192, 2020 06 10.
Article in English | MEDLINE | ID: mdl-32452506

ABSTRACT

Isotopologs are powerful tools for investigating biological systems. We report a biosynthetic-cascade synthesis of Trp isotopologs starting from indole, glycine, and formaldehyde using the enzymes l-threonine aldolase and an engineered ß-subunit of tryptophan synthase. This modular route to Trp isotopologs is simple and inexpensive, enabling facile access to these compounds.


Subject(s)
Glycine Hydroxymethyltransferase/metabolism , Tryptophan Synthase/metabolism , Tryptophan/biosynthesis , Carbon Isotopes , Deuterium , Pyrococcus furiosus/enzymology , Tryptophan/chemistry
20.
Plant Mol Biol ; 99(3): 205-217, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30627860

ABSTRACT

KEY MESSAGE: We identified three dormant stages of Polygonatum kingianum and changes that occurred during dormancy transition in the following aspects including cell wall and hormones, as well as interaction among them. Polygonatum kingianum Coll.et Hemsl (P. kingianum) is an important traditional Chinese medicine, but the mechanism of its rhizome bud dormancy has not yet been studied systematically. In this study, three dormancy phases were induced under controlled conditions, and changes occurring during the transition were examined, focusing on phytohormones and the cell wall. As revealed by HPLC-MS (High Performance Liquid Chromatography-Mass Spectrometry) analysis, the endo- to non-dormancy transition was association with a reduced abscisic acid (ABA)/gibberellin (GA3) ratio, a decreased level of auxin (IAA) and an increased level of trans-zeatin (tZR). Transmission electron microscopy showed that plasmodesmata (PDs) and the cell wall of the bud underwent significant changes between endo- and eco-dormancy. A total of 95,462 differentially expressed genes (DEGs) were identified based on transcriptomics, and clustering and principal component analysis confirmed the different physiological statuses of the three types of bud samples. Changes in the abundance of transcripts associated with IAA, cytokinins (CTKs), GA, ABA, brassinolide (BR), jasmonic acid (JA), ethylene, salicylic acid (SA), PDs and cell wall-loosening factors were analysed during the bud dormancy transition in P. kingianum. Furthermore, nitrilase 4 (NIT4) and tryptophan synthase alpha chain (TSA1), which are related to IAA synthesis, were identified as hub genes of the co-expression network, and strong interactions between hormones and cell wall-related factors were observed. This research will provide a good model for chilling-treated rhizome bud dormancy in P. kingianum and cultivation of this plant.


Subject(s)
Cell Wall/genetics , Gene Expression Regulation, Plant , Gene Regulatory Networks , Indoleacetic Acids/metabolism , Plant Dormancy/genetics , Plant Growth Regulators/genetics , Polygonatum/genetics , Rhizome/genetics , Abscisic Acid/genetics , Abscisic Acid/metabolism , Brassinosteroids/metabolism , Cell Wall/metabolism , Cell Wall/ultrastructure , Cluster Analysis , Cyclopentanes/metabolism , Cytokinins/metabolism , Ethylenes/metabolism , Gene Expression Profiling , Gibberellins/genetics , Gibberellins/metabolism , Medicine, Chinese Traditional , Oxylipins/metabolism , Plant Dormancy/physiology , Plant Growth Regulators/metabolism , Plant Proteins/genetics , Plant Proteins/metabolism , Polygonatum/metabolism , Rhizome/metabolism , Salicylic Acid/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Steroids, Heterocyclic/metabolism , Tryptophan Synthase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL