Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.026
Filter
1.
Mar Drugs ; 22(8)2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39195464

ABSTRACT

Nonribosomal peptides (NRPs) are biosynthesized by nonribosomal peptide synthetases (NRPSs) and are widely distributed in both terrestrial and marine organisms. Many NRPs and their analogs are biologically active and serve as therapeutic agents. The adenylation (A) domain is a key catalytic domain that primarily controls the sequence of a product during the assembling of NRPs and thus plays a predominant role in the structural diversity of NRPs. Engineering of the A domain to alter substrate specificity is a potential strategy for obtaining novel NRPs for pharmaceutical studies. On the basis of introducing the catalytic mechanism and multiple functions of the A domains, this article systematically describes several representative NRPS engineering strategies targeting the A domain, including mutagenesis of substrate-specificity codes, substitution of condensation-adenylation bidomains, the entire A domain or its subdomains, domain insertion, and whole-module rearrangements.


Subject(s)
Peptide Synthases , Protein Engineering , Peptide Synthases/genetics , Peptide Synthases/metabolism , Peptide Synthases/chemistry , Substrate Specificity , Aquatic Organisms , Catalytic Domain , Animals
2.
Methods Enzymol ; 702: 75-87, 2024.
Article in English | MEDLINE | ID: mdl-39155121

ABSTRACT

NIS Synthetases are a widely distributed, novel superfamily of enzymes critical to stealth siderophore production-small molecules increasingly associated with virulence. Study of these enzymes for inhibition or utilization in biosynthesis of new antibiotics has been hindered by multiple kinetics assays utilizing different limiting reporters or relying on product dissociation as a precursor to signal. We present a label free, continuous readout assay optimized for NIS Synthetase systems utilizing an isothermal titration calorimetry instrument. This assay has been tested in an iterative system comparing multiple turnovers on a single substrate to a single bond formation event and is able to delineate these complex kinetics well. The ITC-based kinetic assay is the first label-free assay for the NIS field, which may allow for more detailed kinetic comparisons in the future, and may also have broader use for iterative enzymes in general.


Subject(s)
Calorimetry , Enzyme Assays , Peptide Synthases , Kinetics , Enzyme Assays/methods , Enzyme Assays/instrumentation , Calorimetry/methods , Peptide Synthases/metabolism , Peptide Synthases/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Siderophores/metabolism , Siderophores/chemistry , Substrate Specificity
3.
Methods Enzymol ; 702: 1-19, 2024.
Article in English | MEDLINE | ID: mdl-39155107

ABSTRACT

The biosynthesis of many bacterial siderophores employs a member of a family of ligases that have been defined as NRPS-independent siderophore (NIS) synthetases. These NIS synthetases use a molecule of ATP to produce an amide linkage between a carboxylate and an amine. Commonly used carboxylate substrates include citrate or α-ketoglutarate, or derivatives thereof, while the amines are often hydroxamate derivatives of lysine or ornithine, or their decarboxylated forms cadaverine and putrescine. Enzymes that employ three substrates to catalyze a reaction may proceed through alternate mechanisms. Some enzymes use sequential mechanisms in which all three substrates bind prior to any chemical steps. In such mechanisms, substrates can bind in a random, ordered, or mixed fashion. Alternately, other enzymes employ a ping-pong mechanism in which a chemical step occurs prior to the binding of all three substrates. Here we describe an enzyme assay that will distinguish among these different mechanisms for the NIS synthetase, using IucA, an enzyme involved in the production of aerobactin, as the model system.


Subject(s)
Peptide Synthases , Siderophores , Siderophores/metabolism , Siderophores/chemistry , Peptide Synthases/metabolism , Peptide Synthases/chemistry , Kinetics , Substrate Specificity , Enzyme Assays/methods , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Ketoglutaric Acids/metabolism , Ligases/metabolism , Ligases/chemistry
4.
Methods Enzymol ; 702: 89-119, 2024.
Article in English | MEDLINE | ID: mdl-39155122

ABSTRACT

Non-ribosomal peptide synthesis produces a wide range of bioactive peptide natural products and is reliant on a modular architecture based on repeating catalytic domains able to generate diverse peptide sequences. In this chapter we detail an in vitro biochemical assay to explore the substrate specificity of condensation domains, which are responsible for peptide elongation, from the biosynthetic machinery that produces from the siderophore fuscachelin. This assay removes the requirement to utilise the specificity of adjacent adenylation domains and allows the acceptance of a wide range of synthetic substrates to be explored.


Subject(s)
Siderophores , Substrate Specificity , Siderophores/chemistry , Siderophores/biosynthesis , Peptide Synthases/metabolism , Peptide Synthases/chemistry , Peptide Synthases/genetics , Peptides/chemistry , Peptides/metabolism , Peptide Biosynthesis, Nucleic Acid-Independent , Catalytic Domain
5.
Enzyme Microb Technol ; 180: 110481, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39047348

ABSTRACT

The thermal instability of γ-glutamylmethylamide synthetase (GMAS) from Methylovorus mays has imposed limitations on its industrial applications, affecting both stability and activity at reaction temperatures. In this study, disulfide bridges were introduced through a combination of directed evolution and rational design to enhance GMAS stability. Among the variants that we generated, M12 exhibited a 1.46-fold improvement in relative enzyme activity and a 6.23-fold increase in half-life at 40℃ compared to the wild-type GMAS. Employing variant M12 under optimal conditions, we achieved the production of 645.7 mM (112.49 g/L) L-theanine with a productivity of 29.3 mM/h, from 800 mM substrate in an ATP regeneration system. Our strategy significantly enhances the biosynthesis efficiency of L-theanine by preserving the structural stability of the enzyme during the catalysis process.


Subject(s)
Enzyme Stability , Glutamates , Peptide Synthases , Glutamates/metabolism , Glutamates/biosynthesis , Peptide Synthases/metabolism , Peptide Synthases/genetics , Peptide Synthases/chemistry , Directed Molecular Evolution , Kinetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , Temperature , Carbon-Nitrogen Ligases
6.
J Biol Chem ; 300(8): 107489, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38908753

ABSTRACT

Nonribosomal peptide synthetases (NRPSs) are responsible for the production of important biologically active peptides. The large, multidomain NRPSs operate through an assembly line strategy in which the growing peptide is tethered to carrier domains that deliver the intermediates to neighboring catalytic domains. While most NRPS domains catalyze standard chemistry of amino acid activation, peptide bond formation, and product release, some canonical NRPS catalytic domains promote unexpected chemistry. The paradigm monobactam antibiotic sulfazecin is produced through the activity of a terminal thioesterase domain of SulM, which catalyzes an unusual ß-lactam-forming reaction in which the nitrogen of the C-terminal N-sulfo-2,3-diaminopropionate residue attacks its thioester tether to release the monobactam product. We have determined the structure of the thioesterase domain as both a free-standing domain and a didomain complex with the upstream holo peptidyl-carrier domain. The position of variant lid helices results in an active site pocket that is quite constrained, a feature that is likely necessary to orient the substrate properly for ß-lactam formation. Modeling of a sulfazecin tripeptide into the active site identifies a plausible binding mode identifying potential interactions for the sulfamate and the peptide backbone with Arg2849 and Asn2819, respectively. The overall structure is similar to the ß-lactone-forming thioesterase domain that is responsible for similar ring closure in the production of obafluorin. We further use these insights to enable bioinformatic analysis to identify additional, uncharacterized ß-lactam-forming biosynthetic gene clusters by genome mining.


Subject(s)
Bacterial Proteins , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Protein Domains , Catalytic Domain , Thiolester Hydrolases/chemistry , Thiolester Hydrolases/metabolism , Thiolester Hydrolases/genetics , Peptide Synthases/chemistry , Peptide Synthases/metabolism , Peptide Synthases/genetics , Crystallography, X-Ray , Models, Molecular
7.
Faraday Discuss ; 252(0): 403-421, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-38832470

ABSTRACT

Peptide asparaginyl ligases (PALs) hold significant potential in protein bioconjugation due to their excellent kinetic properties and broad substrate compatibility. However, realizing their full potential in biocatalytic applications requires precise control of their activity. Inspired by nature, we aimed to compartmentalize a representative PAL, OaAEP1-C247A, within protein containers to create artificial organelles with substrate sorting capability. Two encapsulation approaches were explored using engineered lumazine synthases (AaLS). The initial strategy involved tagging the PAL with a super-positively charged GFP(+36) for encapsulation into the super-negatively charged AaLS-13 variant, but it resulted in undesired truncation of the enzyme. The second approach involved genetic fusion of the OaAEP1-C247A with a circularly permutated AaLS variant (cpAaLS) and its co-production with AaLS-13, which successfully enabled compartmentalization of the PAL within a patch-work protein cage. Although the caged PAL retained its activity, it was significantly reduced compared to the free enzyme (∼30-40-fold), likely caused by issues related to OaAEP1-C247A stability and folding. Nevertheless, these findings demonstrated the feasibility of the AaLS encapsulation approach and encourage further optimization in the design of peptide-ligating artificial organelles in E. coli, aiming for a more effective and stable system for protein modifications.


Subject(s)
Peptide Synthases , Peptide Synthases/metabolism , Peptide Synthases/chemistry , Peptides/chemistry , Peptides/metabolism , Multienzyme Complexes
8.
Curr Opin Chem Biol ; 81: 102494, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38936328

ABSTRACT

Reactive amino acid side chains play important roles in the binding of peptides to specific targets. In addition, their reactivity enables selective peptide conjugation and functionalization for pharmaceutical purposes. Diverse reactive amino acids are incorporated into nonribosomal peptides, which serve as a source for drug candidates. Notable examples include (poly)unsaturated (enamine, alkyne, and furyl) and halogenated residues, strained carbacycles (cyclopropyl and cyclopropanol), small heterocycles (oxirane and aziridine), and reactive N-N functionalities (hydrazones, diazo compounds, and diazeniumdiolates). Their biosynthesis requires diverse biocatalysts for sophisticated reaction mechanisms. Several avenues have been identified for their incorporation into peptides, the recruitment by adenylation domains or ligases, on-line modifications, and enzymatic tailoring reactions. Combined with protein engineering approaches, this knowledge provides new opportunities in synthetic biology and bioorthogonal chemistry.


Subject(s)
Amino Acids , Peptides , Amino Acids/metabolism , Amino Acids/chemistry , Peptides/metabolism , Peptides/chemistry , Peptide Biosynthesis, Nucleic Acid-Independent , Protein Engineering/methods , Peptide Synthases/metabolism , Peptide Synthases/chemistry
9.
Angew Chem Int Ed Engl ; 63(33): e202406360, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-38822735

ABSTRACT

Unnatural product (uNP) nonribosomal peptides promise to be a valuable source of pharmacophores for drug discovery. However, the extremely large size and complexity of the nonribosomal peptide synthetase (NRPS) enzymes pose formidable challenges to the production of such uNPs by combinatorial biosynthesis and synthetic biology. Here we report a new NRPS dissection strategy that facilitates the engineering and heterologous production of these NRPSs. This strategy divides NRPSs into "splitting units", each forming an enzyme subunit that contains catalytically independent modules. Functional collaboration between the subunits is then facilitated by artificially duplicating, at the N-terminus of the downstream subunit, the linker - thiolation domain - linker fragment that is resident at the C-terminus of the upstream subunit. Using the suggested split site that follows a conserved motif in the linker connecting the adenylation and the thiolation domains allows cognate or chimeric splitting unit pairs to achieve productivities that match, and in many cases surpass those of hybrid chimeric enzymes, and even those of intact NRPSs, upon production in a heterologous chassis. Our strategy provides facile options for the rational engineering of fungal NRPSs and for the combinatorial reprogramming of nonribosomal peptide production.


Subject(s)
Peptide Synthases , Protein Engineering , Peptide Synthases/metabolism , Peptide Synthases/chemistry , Peptide Synthases/genetics , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/metabolism
10.
Bioorg Med Chem ; 110: 117815, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38943807

ABSTRACT

The adenylation (A) domain of non-ribosomal peptide synthetases (NRPSs) catalyzes the adenylation reaction with substrate amino acids and ATP. Leveraging the distinct substrate specificity of A-domains, we previously developed photoaffinity probes for A-domains based on derivatization with a 5'-O-N-(aminoacyl)sulfamoyl adenosine (aminoacyl-AMS)-appended clickable benzophenone. Although our photoaffinity probes with different amino acid warheads enabled selective detection, visualization, and enrichment of target A-domains in proteomic environments, the effects of photoaffinity linkers have not been investigated. To explore the optimal benzophenone-based linker scaffold, we designed seven photoaffinity probes for the A-domains with different lengths, positions, and molecular shapes. Using probes 2-8 for the phenylalanine-activating A-domain of gramicidin S synthetase A (GrsA), we systematically investigated the binding affinity and labeling efficiency of the endogenous enzyme in a live producer cell. Our results indicated that the labeling efficiencies of probes 2-8 tended to depend on their binding affinities rather than on the linker length, flexibility, or position of the photoaffinity group. We also identified that probe 2 with a 4,4'-diaminobenzophenone linker exhibits the highest labeling efficiency for GrsA with fewer non-target labeling properties in live cells.


Subject(s)
Benzophenones , Peptide Synthases , Photoaffinity Labels , Benzophenones/chemistry , Benzophenones/chemical synthesis , Benzophenones/pharmacology , Benzophenones/metabolism , Photoaffinity Labels/chemistry , Photoaffinity Labels/chemical synthesis , Peptide Synthases/metabolism , Peptide Synthases/chemistry , Molecular Structure
11.
Sci Adv ; 10(25): eadm9404, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38896613

ABSTRACT

In the quest for new bioactive substances, nonribosomal peptide synthetases (NRPS) provide biodiversity by synthesizing nonproteinaceous peptides with high cellular activity. NRPS machinery consists of multiple modules, each catalyzing a unique series of chemical reactions. Incomplete understanding of the biophysical principles orchestrating these reaction arrays limits the exploitation of NRPSs in synthetic biology. Here, we use nuclear magnetic resonance (NMR) spectroscopy and mass spectrometry to solve the conundrum of how intermodular recognition is coupled with loaded carrier protein specificity in the tomaymycin NRPS. We discover an adaptor domain that directly recruits the loaded carrier protein from the initiation module to the elongation module and reveal its mechanism of action. The adaptor domain of the type found here has specificity rules that could potentially be exploited in the design of engineered NRPS machinery.


Subject(s)
Peptide Synthases , Peptide Synthases/metabolism , Peptide Synthases/chemistry , Substrate Specificity , Protein Domains , Protein Binding , Magnetic Resonance Spectroscopy/methods
12.
Structure ; 32(5): 520-522, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38701750

ABSTRACT

In a recent issue of Nature Chemical Biology, Folger et al. demonstrated a high-throughput approach for engineering peptide bond forming domains from non-ribosomal peptide synthesis. A non-ribosomal peptide synthetase module from surfactin biosynthesis was reprogrammed to accept a fatty acid substrate into peptide biosynthesis, thus illustrating the potential of this approach for generating novel bioactive peptides.


Subject(s)
Peptide Synthases , Protein Engineering , Peptide Synthases/metabolism , Peptide Synthases/chemistry , Peptide Synthases/genetics , Protein Engineering/methods , Peptides/metabolism , Peptides/chemistry
13.
Molecules ; 29(9)2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38731473

ABSTRACT

Chalkophomycin is a novel chalkophore with antibiotic activities isolated from Streptomyces sp. CB00271, while its potential in studying cellular copper homeostasis makes it an important probe and drug lead. The constellation of N-hydroxylpyrrole, 2H-oxazoline, diazeniumdiolate, and methoxypyrrolinone functional groups into one compact molecular architecture capable of coordinating cupric ions draws interest to unprecedented enzymology responsible for chalkophomycin biosynthesis. To elucidate the biosynthetic machinery for chalkophomycin production, the chm biosynthetic gene cluster from S. sp. CB00271 was identified, and its involvement in chalkophomycin biosynthesis was confirmed by gene replacement. The chm cluster was localized to a ~31 kb DNA region, consisting of 19 open reading frames that encode five nonribosomal peptide synthetases (ChmHIJLO), one modular polyketide synthase (ChmP), six tailoring enzymes (ChmFGMNQR), two regulatory proteins (ChmAB), and four resistance proteins (ChmA'CDE). A model for chalkophomycin biosynthesis is proposed based on functional assignments from sequence analysis and structure modelling, and is further supported by analogy to over 100 chm-type gene clusters in public databases. Our studies thus set the stage to fully investigate chalkophomycin biosynthesis and to engineer chalkophomycin analogues through a synthetic biology approach.


Subject(s)
Multigene Family , Peptide Synthases , Polyketide Synthases , Streptomyces , Streptomyces/genetics , Streptomyces/enzymology , Streptomyces/metabolism , Polyketide Synthases/genetics , Polyketide Synthases/metabolism , Polyketide Synthases/chemistry , Peptide Synthases/metabolism , Peptide Synthases/genetics , Peptide Synthases/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry
14.
Science ; 383(6689): eadg4320, 2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38513038

ABSTRACT

Many clinically used drugs are derived from or inspired by bacterial natural products that often are produced through nonribosomal peptide synthetases (NRPSs), megasynthetases that activate and join individual amino acids in an assembly line fashion. In this work, we describe a detailed phylogenetic analysis of several bacterial NRPSs that led to the identification of yet undescribed recombination sites within the thiolation (T) domain that can be used for NRPS engineering. We then developed an evolution-inspired "eXchange Unit between T domains" (XUT) approach, which allows the assembly of NRPS fragments over a broad range of GC contents, protein similarities, and extender unit specificities, as demonstrated for the specific production of a proteasome inhibitor designed and assembled from five different NRPS fragments.


Subject(s)
Bacterial Proteins , Evolution, Molecular , Peptide Synthases , Protein Engineering , Peptide Synthases/chemistry , Peptide Synthases/classification , Peptide Synthases/genetics , Phylogeny , Amino Acid Sequence/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/classification , Bacterial Proteins/genetics , Sequence Analysis, Protein
15.
Nat Prod Rep ; 41(7): 1180-1205, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38488017

ABSTRACT

Covering: 1997 to July 2023The adenylation reaction has been a subject of scientific intrigue since it was first recognized as essential to many biological processes, including the homeostasis and pathogenicity of some bacteria and the activation of amino acids for protein synthesis in mammals. Several foundational studies on adenylation (A) domains have facilitated an improved understanding of their molecular structures and biochemical properties, in particular work on nonribosomal peptide synthetases (NRPSs). In NRPS pathways, A domains activate their respective acyl substrates for incorporation into a growing peptidyl chain, and many nonribosomal peptides are bioactive. From a natural product drug discovery perspective, improving existing bioinformatics platforms to predict unique NRPS products more accurately from genomic data is desirable. Here, we summarize characterization efforts of A domains primarily from NRPS pathways from July 1997 up to July 2023, covering protein structure elucidation, in vitro assay development, and in silico tools for improved predictions.


Subject(s)
Computational Biology , Peptide Synthases , Peptide Synthases/metabolism , Peptide Synthases/chemistry , Computational Biology/methods , Molecular Structure , Biological Products/metabolism , Biological Products/chemistry
16.
Angew Chem Int Ed Engl ; 63(20): e202317753, 2024 05 13.
Article in English | MEDLINE | ID: mdl-38488324

ABSTRACT

In multi-domain nonribosomal peptide synthetases (NRPSs) the order of domains and their catalytic specificities dictate the structure of the peptide product. Peptidyl-carrier proteins (PCPs) bind activated amino acids and channel elongating peptidyl intermediates along the protein template. To this end, fine-tuned interactions with the catalytic domains and large-scale PCP translocations are necessary. Despite crystal structure snapshots of several PCP-domain interactions, the conformational dynamics under catalytic conditions in solution remain poorly understood. We report a FRET reporter of gramicidin S synthetase 1 (GrsA; with A-PCP-E domains) to study for the first time the interaction between PCP and adenylation (A) domain in the presence of an epimerization (E) domain, a competing downstream partner for the PCP. Bulk FRET measurements showed that upon PCP aminoacylation a conformational shift towards PCP binding to the A domain occurs, indicating the E domain acts on its PCP substrate out of a disfavored conformational equilibrium. Furthermore, the A domain was found to preferably bind the D-Phe-S-Ppant-PCP stereoisomer, suggesting it helps in establishing the stereoisomeric mixture in favor of the D-aminoacyl moiety. These observations surprisingly show that the conformational logic can deviate from the order of domains and thus reveal new principles in the multi-domain interplay of NRPSs.


Subject(s)
Fluorescence Resonance Energy Transfer , Peptide Synthases , Peptide Synthases/chemistry , Peptide Synthases/metabolism , Carrier Proteins/chemistry , Carrier Proteins/metabolism
17.
Structure ; 32(4): 440-452.e4, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38340732

ABSTRACT

Nonribosomal peptide synthetases (NRPSs) are large multidomain enzymes for the synthesis of a variety of bioactive peptides in a modular and pipelined fashion. Here, we investigated how the condensation (C) domain and the adenylation (A) domain cooperate with each other for the efficient catalytic activity in microcystin NRPS modules. We solved two crystal structures of the microcystin NRPS modules, representing two different conformations in the NRPS catalytic cycle. Our data reveal that the dynamic interaction between the C and the A domains in these modules is mediated by the conserved "RXGR" motif, and this interaction is important for the adenylation activity. Furthermore, the "RXGR" motif-mediated dynamic interaction and its functional regulation are prevalent in different NRPSs modules possessing both the A and the C domains. This study provides new insights into the catalytic mechanism of NRPSs and their engineering strategy for synthetic peptides with different structures and properties.


Subject(s)
Microcystins , Peptide Synthases , Peptide Synthases/chemistry , Molecular Conformation , Peptides
18.
Nat Chem Biol ; 20(6): 761-769, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38308044

ABSTRACT

Engineered biosynthetic assembly lines could revolutionize the sustainable production of bioactive natural product analogs. Although yeast display is a proven, powerful tool for altering the substrate specificity of gatekeeper adenylation domains in nonribosomal peptide synthetases (NRPSs), comparable strategies for other components of these megaenzymes have not been described. Here we report a high-throughput approach for engineering condensation (C) domains responsible for peptide elongation. We show that a 120-kDa NRPS module, displayed in functional form on yeast, can productively interact with an upstream module, provided in solution, to produce amide products tethered to the yeast surface. Using this system to screen a large C-domain library, we reprogrammed a surfactin synthetase module to accept a fatty acid donor, increasing catalytic efficiency for this noncanonical substrate >40-fold. Because C domains can function as selectivity filters in NRPSs, this methodology should facilitate the precision engineering of these molecular assembly lines.


Subject(s)
Peptide Synthases , Peptide Synthases/metabolism , Peptide Synthases/genetics , Peptide Synthases/chemistry , Substrate Specificity , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , Protein Engineering/methods , High-Throughput Screening Assays , Protein Domains
19.
Nat Chem ; 16(2): 259-268, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38049653

ABSTRACT

Many peptide-derived natural products are produced by non-ribosomal peptide synthetases (NRPSs) in an assembly-line fashion. Each amino acid is coupled to a designated peptidyl carrier protein (PCP) through two distinct reactions catalysed sequentially by the single active site of the adenylation domain (A-domain). Accumulating evidence suggests that large-amplitude structural changes occur in different NRPS states; yet how these molecular machines orchestrate such biochemical sequences has remained elusive. Here, using single-molecule Förster resonance energy transfer, we show that the A-domain of gramicidin S synthetase I adopts structurally extended and functionally obligatory conformations for alternating between adenylation and thioester-formation structures during enzymatic cycles. Complementary biochemical, computational and small-angle X-ray scattering studies reveal interconversion among these three conformations as intrinsic and hierarchical where intra-A-domain organizations propagate to remodel inter-A-PCP didomain configurations during catalysis. The tight kinetic coupling between structural transitions and enzymatic transformations is quantified, and how the gramicidin S synthetase I A-domain utilizes its inherent conformational dynamics to drive directional biosynthesis with a flexibly linked PCP domain is revealed.


Subject(s)
Gramicidin , Peptide Synthases , Protein Structure, Tertiary , Peptide Synthases/chemistry , Catalytic Domain
20.
Biochim Biophys Acta Proteins Proteom ; 1872(1): 140972, 2024 01 01.
Article in English | MEDLINE | ID: mdl-37951518

ABSTRACT

Non-ribosomal peptide synthetases (NRPSs) generate chemically complex compounds and their modular architecture suggests that changing their domain organization can predictably alter their products. Ebony, a small three-domain NRPS, catalyzes the formation of ß-alanine containing amides from biogenic amines. To examine the necessity of interdomain interactions, we modeled and docked domains of Ebony to reveal potential interfaces between them. Testing the same domain combinations in vitro showed that 8 % of activity was preserved after Ebony was dissected into a di-domain and a detached C-terminal domain, suggesting that sufficient interaction was maintained after dissection. Our work creates a model to identify domain interfaces necessary for catalysis, an important step toward utilizing Ebony as a combinatorial engineering platform for novel amides.


Subject(s)
Amides , Peptide Synthases , Peptide Synthases/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL