Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 804
Filter
1.
Commun Biol ; 7(1): 1029, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39169227

ABSTRACT

Several bacterial flagellins are O-glycosylated with nonulosonic acids on surface-exposed Serine/Threonine residues by Maf glycosyltransferases. The Clostridium botulinum Maf glycosyltransferase (CbMaf) displays considerable donor substrate promiscuity, enabling flagellin O-glycosylation with N-acetyl neuraminic acid (Neu5Ac) and 3-deoxy-D-manno-octulosonic acid in the absence of the native nonulosonic acid, a legionaminic acid derivative. Here, we have explored the sequence/structure attributes of the acceptor substrate, flagellin, required by CbMaf glycosyltransferase for glycosylation with Neu5Ac and KDO, by co-expressing C. botulinum flagellin constructs with CbMaf glycosyltransferase in an E. coli strain producing cytidine-5'-monophosphate (CMP)-activated Neu5Ac, and employing intact mass spectrometry analysis and sialic acid-specific flagellin biotinylation as readouts. We found that CbMaf was able to glycosylate mini-flagellin constructs containing shortened alpha-helical secondary structural scaffolds and reduced surface-accessible loop regions, but not non-cognate flagellin. Our experiments indicated that CbMaf glycosyltransferase recognizes individual Ser/Thr residues in their local surface-accessible conformations, in turn, supported in place by the secondary structural scaffold. Further, CbMaf glycosyltransferase also robustly glycosylated chimeric proteins constructed by grafting cognate mini-flagellin sequences onto an unrelated beta-sandwich protein. Our recombinant engineering experiments highlight the potential of CbMaf glycosyltransferase in future glycoengineering applications, especially for the neo-O-sialylation of proteins, employing E. coli strains expressing CMP-Neu5Ac (and not CMP-KDO).


Subject(s)
Clostridium botulinum , Flagellin , Glycosyltransferases , Substrate Specificity , Glycosyltransferases/metabolism , Glycosyltransferases/genetics , Glycosyltransferases/chemistry , Flagellin/metabolism , Flagellin/genetics , Flagellin/chemistry , Clostridium botulinum/enzymology , Clostridium botulinum/metabolism , Clostridium botulinum/genetics , Glycosylation , Escherichia coli/genetics , Escherichia coli/metabolism , Sugar Acids/metabolism , Protein Engineering , N-Acetylneuraminic Acid/metabolism , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , Sialic Acids
2.
Proc Natl Acad Sci U S A ; 121(35): e2321204121, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39172782

ABSTRACT

Upon DNA damage, numerous proteins are targeted for ubiquitin-dependent proteasomal degradation, which is an integral part of the DNA repair program. Although details of the ubiquitination processes have been intensively studied, little is known about whether and how the 26S proteasome is regulated in the DNA damage response (DDR). Here, we show that human Rpn10/PSMD4, one of the three ubiquitin receptors of the 26S proteasome, is rapidly phosphorylated in response to different types of DNA damage. The phosphorylation occurs at Rpn10-Ser266 within a conserved SQ motif recognized by ATM/ATR/DNA-PK. Blockade of S266 phosphorylation attenuates homologous recombination-mediated DNA repair and sensitizes cells to genotoxic insults. In vitro and in cellulo experiments indicate that phosphorylation of S266, located in the flexible linker between the two ubiquitin-interacting motifs (UIMs) of Rpn10, alters the configuration of UIMs, and actually reduces ubiquitin chain (substrate) binding. As a result, essential DDR proteins such as BRCA1 are spared from premature degradation and allowed sufficient time to engage in DNA repair, a scenario supported by proximity labeling and quantitative proteomic studies. These findings reveal an inherent self-limiting mechanism of the proteasome that, by controlling substrate recognition through Rpn10 phosphorylation, fine-tunes protein degradation for optimal responses under stress.


Subject(s)
DNA Damage , DNA Repair , Proteasome Endopeptidase Complex , Proteasome Endopeptidase Complex/metabolism , Humans , Phosphorylation , Ubiquitin/metabolism , BRCA1 Protein/metabolism , Substrate Specificity , Ubiquitination , RNA-Binding Proteins
3.
Carbohydr Polym ; 343: 122474, 2024 Nov 01.
Article in English | MEDLINE | ID: mdl-39174099

ABSTRACT

Alginate is one of the most important marine colloidal polysaccharides, and its oligosaccharides have been proven to possess diverse biological functions. Alginate lyases could specifically degrade alginate and therefore serve as desirable tools for the research and development of alginate. In this report, a novel catalytic domain, which demonstrated no significant sequence similarity with all previously defined functional domains, was verified to exhibit a random endo-acting lyase activity to alginate. The action pattern analysis revealed that the heterologously expressed protein, named Aly44A, preferred to degrade polyM. Its minimum substrates and the minimum products were identified as unsaturated alginate trisaccharides and disaccharides, respectively. Based on the sequence novelty of Aly44A and its homologs, a new polysaccharide lyase family (PL44) was proposed. The discovery of the novel enzyme and polysaccharide lyase family provided a new entrance for the gene-mining and acquiring of alginate lyases, and would facilitate to the utilization of alginate and its oligosaccharides.


Subject(s)
Alginates , Polysaccharide-Lyases , Polysaccharide-Lyases/metabolism , Polysaccharide-Lyases/chemistry , Polysaccharide-Lyases/genetics , Alginates/chemistry , Alginates/metabolism , Substrate Specificity , Catalytic Domain , Oligosaccharides/chemistry , Oligosaccharides/metabolism , Amino Acid Sequence , Hexuronic Acids/chemistry , Hexuronic Acids/metabolism
4.
ACS Synth Biol ; 13(8): 2600-2610, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39110689

ABSTRACT

The engineering of enzymatic activity generally involves alteration of the protein primary sequences, which introduce structural changes that give rise to functional improvements. Mechanical forces have been used to interrogate protein biophysics, leading to deep mechanistic insights in single-molecule studies. Here, we use simple DNA springs to apply small pulling forces to perturb the active site of a thermostable alcohol dehydrogenase. Methods were developed to enable the study of different spring lengths and spring orientations under bulk catalysis conditions. Tension applied across the active site expanded the binding pocket volume and shifted the preference of the enzyme for longer chain-length substrates, which could be tuned by altering the spring length and the resultant applied force. The substrate specificity changes did not occur when the DNA spring was either severed or rotated by ∼90°. These findings demonstrate an alternative approach in protein engineering, where active site architectures can be dynamically and reversibly remodeled using applied mechanical forces.


Subject(s)
Alcohol Dehydrogenase , Biocatalysis , Catalytic Domain , DNA , Protein Engineering , Protein Engineering/methods , Alcohol Dehydrogenase/genetics , Alcohol Dehydrogenase/metabolism , Alcohol Dehydrogenase/chemistry , DNA/metabolism , DNA/chemistry , DNA/genetics , Substrate Specificity
5.
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
6.
Org Biomol Chem ; 22(33): 6713-6717, 2024 08 22.
Article in English | MEDLINE | ID: mdl-39119636

ABSTRACT

Penicillin-binding protein-type thioesterases (PBP-type TEs) catalyze head-to-tail macrolactamization in bacterial nonribosomal peptide biosynthesis. Here the scope of FlkO, a new PBP-type TE in cyclofaulknamycin biosynthesis, was thoroughly evaluated. The preference for small residues at the substrate C-terminus was consistent with the decreased volume of its putative substrate-binding pocket.


Subject(s)
Penicillin-Binding Proteins , Penicillin-Binding Proteins/metabolism , Penicillin-Binding Proteins/chemistry , Streptomyces/enzymology , Streptomyces/metabolism , Thiolester Hydrolases/metabolism , Thiolester Hydrolases/chemistry , Substrate Specificity
7.
J Agric Food Chem ; 72(33): 18585-18593, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39133835

ABSTRACT

d-Tagatose is a highly promising functional sweetener known for its various physiological functions. In this study, a novel tagatose 4-epimerase from Thermoprotei archaeon (Thar-T4Ease), with the ability to convert d-fructose to d-tagatose, was discovered through a combination of structure similarity search and sequence-based protein clustering. The recombinant Thar-T4Ease exhibited optimal activity at pH 8.5 and 85 °C, in the presence of 1 mM Ni2+. Its kcat and kcat/Km values toward d-fructose were measured to be 248.5 min-1 and 2.117 mM-1·min-1, respectively. Notably, Thar-T4Ease exhibited remarkable thermostability, with a t1/2 value of 198 h at 80 °C. Moreover, it achieved a conversion ratio of 18.9% using 100 g/L d-fructose as the substrate. Finally, based on sequence and structure analysis, crucial residues for the catalytic activity of Thar-T4Ease were identified by molecular docking and site-directed mutagenesis. This research expands the repertoire of enzymes with C4-epimerization activity and opens up new possibilities for the cost-effective production of d-tagatose from d-fructose.


Subject(s)
Enzyme Stability , Hexoses , Molecular Docking Simulation , Hexoses/chemistry , Hexoses/metabolism , Kinetics , Archaeal Proteins/genetics , Archaeal Proteins/chemistry , Archaeal Proteins/metabolism , Fructose/chemistry , Fructose/metabolism , Carbohydrate Epimerases/genetics , Carbohydrate Epimerases/chemistry , Carbohydrate Epimerases/metabolism , Hydrogen-Ion Concentration , Substrate Specificity , Hot Temperature , Amino Acid Sequence , Racemases and Epimerases/genetics , Racemases and Epimerases/chemistry , Racemases and Epimerases/metabolism
8.
Nat Commun ; 15(1): 7134, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39164235

ABSTRACT

The mechanism by which RNAP selects cognate substrates and discriminates between deoxy and ribonucleotides is of fundamental importance to the fidelity of transcription. Here, we present cryo-EM structures of human mitochondrial transcription elongation complexes that reveal substrate ATP bound in Entry and Insertion Sites. In the Entry Site, the substrate binds along the O helix of the fingers domain of mtRNAP but does not interact with the templating DNA base. Interactions between RNAP and the triphosphate moiety of the NTP in the Entry Site ensure discrimination against nucleosides and their diphosphate and monophosphate derivatives but not against non-cognate rNTPs and dNTPs. Closing of the fingers domain over the catalytic site results in delivery of both the templating DNA base and the substrate into the Insertion Site and recruitment of the catalytic magnesium ions. The cryo-EM data also reveal a conformation adopted by mtRNAP to reject a non-cognate substrate from its active site. Our findings establish a structural basis for substrate binding and suggest a unified mechanism of NTP selection for single-subunit RNAPs.


Subject(s)
Catalytic Domain , Cryoelectron Microscopy , DNA-Directed RNA Polymerases , Mitochondria , Humans , DNA-Directed RNA Polymerases/metabolism , DNA-Directed RNA Polymerases/chemistry , Substrate Specificity , Mitochondria/metabolism , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/chemistry , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/chemistry , Models, Molecular , Protein Binding , Binding Sites
9.
Sci Rep ; 14(1): 17892, 2024 08 02.
Article in English | MEDLINE | ID: mdl-39095391

ABSTRACT

Proteins possessing double active sites have the potential to revolutionise enzyme design strategies. This study extensively explored an enzyme that contains both a natural active site (NAS) and an engineered active site (EAS), focusing on understanding its structural and functional properties. Metadynamics simulations were employed to investigate how substrates interacted with their respective active sites. The results revealed that both the NAS and EAS exhibited similar minimum energy states, indicating comparable binding affinities. However, it became apparent that the EAS had a weaker binding site for the substrate due to its smaller pocket and constrained conformation. Interestingly, the EAS also displayed dynamic behaviour, with the substrate observed to move outside the pocket, suggesting the possibility of substrate translocation. To gain further insights, steered molecular dynamics (SMD) simulations were conducted to study the conformational changes of the substrate and its interactions with catalytic residues. Notably, the substrate adopted distinct conformations, including near-attack conformations, in both the EAS and NAS. Nevertheless, the NAS demonstrated superior binding minima for the substrate compared to the EAS, reinforcing the observation that the engineered active site was less favourable for substrate binding due to its limitations. The QM/MM (Quantum mechanics and molecular mechanics) analyses highlight the energy disparity between NAS and EAS. Specifically, EAS exhibited elevated energy levels due to its engineered active site being located on the surface. This positioning exposes the substrate to solvents and water molecules, adding to the energy challenge. Consequently, the engineered enzyme did not provide a significant advantage in substrate binding over the single active site protein. Further, the investigation of internal channels and tunnels within the protein shed light on the pathways facilitating transport between the two active sites. By unravelling the complex dynamics and functional characteristics of this double-active site protein, this study offers valuable insights into novel strategies of enzyme engineering. These findings establish a solid foundation for future research endeavours aimed at harnessing the potential of double-active site proteins in diverse biotechnological applications.


Subject(s)
Catalytic Domain , Molecular Dynamics Simulation , Protein Engineering , Protein Engineering/methods , Enzymes/chemistry , Enzymes/metabolism , Substrate Specificity , Protein Conformation , Binding Sites , Protein Binding
10.
J Agric Food Chem ; 72(32): 18100-18109, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39090787

ABSTRACT

Inulin has found commercial applications in the pharmaceutical, nutraceutical, and food industries due to its beneficial health effects. The enzymatic biosynthesis of microbial inulin has garnered increasing attention. In this study, molecular modification was applied to Lactobacillus mulieris UMB7800 inulosucrase, an enzyme that specifically produces high-molecular weight inulin, to enhance its catalytic activity and thermostability. Among the 18 variable regions, R5 was identified as a crucial region significantly impacting enzymatic activity by replacing it with more conserved sequences. Site-directed mutagenesis combined with saturated mutagenesis revealed that the mutant A250 V increased activity by 68%. Additionally, after screening candidate mutants by rational design, four single-point mutants, S344D, H434P, E526D, and G531P, were shown to enhance thermostability. The final combinational mutant, M5, exhibited a 66% increase in activity and a 5-fold enhancement in half-life at 55 °C. These findings are significant for understanding the catalytic activity and thermostability of inulosucrase and are promising for the development of microbial inulin biosynthesis platforms.


Subject(s)
Bacterial Proteins , Enzyme Stability , Hexosyltransferases , Inulin , Lactobacillus , Mutagenesis, Site-Directed , Inulin/metabolism , Inulin/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Hexosyltransferases/genetics , Hexosyltransferases/metabolism , Hexosyltransferases/chemistry , Lactobacillus/enzymology , Lactobacillus/genetics , Lactobacillus/metabolism , Kinetics , Hot Temperature , Protein Engineering , Substrate Specificity
11.
Sci Rep ; 14(1): 18420, 2024 08 08.
Article in English | MEDLINE | ID: mdl-39117724

ABSTRACT

A zinc metallopeptidase neurolysin (Nln) processes diverse bioactive peptides to regulate signaling in the mammalian nervous system. To understand how Nln interacts with various peptides with dissimilar sequences, we determined crystal structures of Nln in complex with diverse peptides including dynorphins, angiotensin, neurotensin, and bradykinin. The structures show that Nln binds these peptides in a large dumbbell-shaped interior cavity constricted at the active site, making minimal structural changes to accommodate different peptide sequences. The structures also show that Nln readily binds similar peptides with distinct registers, which can determine whether the peptide serves as a substrate or a competitive inhibitor. We analyzed the activities and binding of Nln toward various forms of dynorphin A peptides, which highlights the promiscuous nature of peptide binding and shows how dynorphin A (1-13) potently inhibits the Nln activity while dynorphin A (1-8) is efficiently cleaved. Our work provides insights into the broad substrate specificity of Nln and may aid in the future design of small molecule modulators for Nln.


Subject(s)
Dynorphins , Neurotensin , Humans , Substrate Specificity , Dynorphins/chemistry , Dynorphins/metabolism , Neurotensin/chemistry , Neurotensin/metabolism , Metalloendopeptidases/metabolism , Metalloendopeptidases/chemistry , Metalloendopeptidases/antagonists & inhibitors , Protein Binding , Crystallography, X-Ray , Models, Molecular , Catalytic Domain , Bradykinin/chemistry , Bradykinin/metabolism , Angiotensins/metabolism , Angiotensins/chemistry , Amino Acid Sequence
12.
Appl Microbiol Biotechnol ; 108(1): 436, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39126499

ABSTRACT

Microbial non-phosphorylative oxidative pathways present promising potential in the biosynthesis of platform chemicals from the hemicellulosic fraction of lignocellulose. An L-arabinonate dehydratase from Rhizobium leguminosarum bv. trifolii catalyzes the rate-limiting step in the non-phosphorylative oxidative pathways, that is, converts sugar acid to 2-dehydro-3-deoxy sugar acid. We have shown earlier that the enzyme forms a dimer of dimers, in which the C-terminal histidine residue from one monomer participates in the formation of the active site of an adjacent monomer. The histidine appears to be conserved across the sequences of sugar acid dehydratases. To study the role of the C-terminus, five variants (H579A, H579F, H579L, H579Q, and H579W) were produced. All variants showed decreased activity for the tested sugar acid substrates, except the variant H579L on D-fuconate, which showed about 20% increase in activity. The reaction kinetic data showed that the substrate preference was slightly modified in H579L compared to the wild-type enzyme, demonstrating that the alternation of the substrate preference of sugar acid dehydratases is possible. In addition, a crystal structure of H579L was determined at 2.4 Å with a product analog 2-oxobutyrate. This is the first enzyme-ligand complex structure from an IlvD/EDD superfamily enzyme. The binding of 2-oxobutyrate suggests how the substrate would bind into the active site in the orientation, which could lead to the dehydration reaction. KEY POINTS: • Mutation of the last histidine at the C-terminus changed the catalytic activity of L-arabinonate dehydratase from R. leguminosarum bv. trifolii against various C5/C6 sugar acids. • The variant H579L of L-arabinonate dehydratase showed an alteration of substrate preferences compared with the wild type. • The first enzyme-ligand complex crystal structure of an IlvD/EDD superfamily enzyme was solved.


Subject(s)
Hydro-Lyases , Rhizobium leguminosarum , Hydro-Lyases/metabolism , Hydro-Lyases/genetics , Hydro-Lyases/chemistry , Substrate Specificity , Rhizobium leguminosarum/enzymology , Rhizobium leguminosarum/genetics , Kinetics , Catalytic Domain , Sugar Acids/metabolism , Histidine/metabolism , Histidine/chemistry , Histidine/genetics , Protein Multimerization , Models, Molecular , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism
13.
Protein Sci ; 33(9): e5144, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39150221

ABSTRACT

MpaG' is an S-adenosyl-L-methionine (SAM)-dependent methyltransferase involved in the compartmentalized biosynthesis of mycophenolic acid (MPA), a first-line immunosuppressive drug for organ transplantations and autoimmune diseases. MpaG' catalyzes the 5-O-methylation of three precursors in MPA biosynthesis including demethylmycophenolic acid (DMMPA), 4-farnesyl-3,5-dihydroxy-6-methylphthalide (FDHMP), and an intermediate containing three fewer carbon atoms compared to FDHMP (FDHMP-3C) with different catalytic efficiencies. Here, we report the crystal structures of S-adenosyl-L-homocysteine (SAH)/DMMPA-bound MpaG', SAH/FDHMP-3C-bound MpaG', and SAH/FDHMP-bound MpaG' to understand the catalytic mechanism of MpaG' and structural basis for its substrate flexibility. Structural and biochemical analyses reveal that MpaG' utilizes the catalytic dyad H306-E362 to deprotonate the C5 hydroxyl group of the substrates for the following methylation. The three substrates with differently modified farnesyl moieties are well accommodated in a large semi-open substrate binding pocket with the orientation of their phthalide moiety almost identical. Based on the structure-directed mutagenesis, a single mutant MpaG'Q267A is engineered with significantly improved catalytic efficiency for all three substrates. This study expands the mechanistic understanding and the pocket engineering strategy for O-methyltransferases involved in fungal natural product biosynthesis. Our research also highlights the potential of O-methyltransferases to modify diverse substrates by protein design and engineering.


Subject(s)
Methyltransferases , Mycophenolic Acid , Mycophenolic Acid/chemistry , Mycophenolic Acid/metabolism , Methyltransferases/chemistry , Methyltransferases/metabolism , Methyltransferases/genetics , Substrate Specificity , Crystallography, X-Ray , Models, Molecular , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Fungal Proteins/genetics , Catalytic Domain
14.
Protein Sci ; 33(9): e5139, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39150063

ABSTRACT

The main protease from coronaviruses and the 3C protease from enteroviruses play a crucial role in processing viral polyproteins, making them attractive targets for the development of antiviral agents. In this study, we employed a combinatorial chemistry approach-HyCoSuL-to compare the substrate specificity profiles of the main and 3C proteases from alphacoronaviruses, betacoronaviruses, and enteroviruses. The obtained data demonstrate that coronavirus Mpros exhibit overlapping substrate specificity in all binding pockets, whereas the 3Cpro from enterovirus displays slightly different preferences toward natural and unnatural amino acids at the P4-P2 positions. However, chemical tools such as substrates, inhibitors, and activity-based probes developed for SARS-CoV-2 Mpro can be successfully applied to investigate the activity of the Mpro from other coronaviruses as well as the 3Cpro from enteroviruses. Our study provides a structural framework for the development of broad-spectrum antiviral compounds.


Subject(s)
Antiviral Agents , Coronavirus 3C Proteases , Enterovirus , SARS-CoV-2 , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Substrate Specificity , Enterovirus/enzymology , Enterovirus/drug effects , SARS-CoV-2/drug effects , SARS-CoV-2/enzymology , Coronavirus 3C Proteases/antagonists & inhibitors , Coronavirus 3C Proteases/metabolism , Coronavirus 3C Proteases/chemistry , Humans , Coronavirus/enzymology , Coronavirus/drug effects
15.
World J Microbiol Biotechnol ; 40(10): 303, 2024 Aug 17.
Article in English | MEDLINE | ID: mdl-39153119

ABSTRACT

Dye-decolorizing peroxidases (DyPs) belong to a novel superfamily of heme peroxidases that can oxidize recalcitrant compounds. In the current study, the GlDyP2 gene from Ganoderma lucidum was heterologously expressed in Escherichia coli, and the enzymatic properties of the recombinant GlDyP2 protein were investigated. The GlDyP2 protein could oxidize not only the typical peroxidase substrate ABTS but also two lignin substrates, namely guaiacol and 2,6-dimethoxy phenol (DMP). For the ABTS substrate, the optimum pH and temperature of GlDyP2 were 4.0 and 35 °C, respectively. The pH stability and thermal stability of GlDyP2 were also measured; the results showed that GlDyP2 could function normally in the acidic environment, with a T50 value of 51 °C. Moreover, compared to untreated controls, the activity of GlDyP2 was inhibited by 1.60 mM of Mg2+, Ni2+, Mn2+, and ethanol; 0.16 mM of Cu2+, Zn2+, methanol, isopropyl alcohol, and Na2EDTA·2H2O; and 0.016 mM of Fe2+ and SDS. The kinetic constants of recombinant GlDyP2 for oxidizing ABTS, Reactive Blue 19, guaiacol, and DMP were determined; the results showed that the recombination GlDyP2 exhibited the strongest affinity and the most remarkable catalytic efficiency towards guaiacol in the selected substrates. GlDyP2 also exhibited decolorization and detoxification capabilities towards several dyes, including Reactive Blue 19, Reactive Brilliant Blue X-BR, Reactive Black 5, Methyl Orange, Trypan Blue, and Malachite Green. In conclusion, GlDyP2 has good application potential for treating dye wastewater.


Subject(s)
Coloring Agents , Enzyme Stability , Escherichia coli , Guaiacol , Recombinant Proteins , Reishi , Temperature , Coloring Agents/metabolism , Coloring Agents/chemistry , Reishi/genetics , Reishi/enzymology , Reishi/metabolism , Hydrogen-Ion Concentration , Escherichia coli/genetics , Escherichia coli/metabolism , Recombinant Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/chemistry , Guaiacol/metabolism , Guaiacol/analogs & derivatives , Biodegradation, Environmental , Kinetics , Benzothiazoles/metabolism , Substrate Specificity , Lignin/metabolism , Oxidation-Reduction , Peroxidase/genetics , Peroxidase/metabolism , Peroxidase/chemistry , Fungal Proteins/genetics , Fungal Proteins/metabolism , Fungal Proteins/chemistry , Peroxidases/genetics , Peroxidases/metabolism , Peroxidases/chemistry , Water Pollutants, Chemical/metabolism , Azo Compounds/metabolism , Wastewater/microbiology , Wastewater/chemistry , Sulfonic Acids/metabolism , Anthraquinones , Rosaniline Dyes
16.
Biochim Biophys Acta Gen Subj ; 1868(10): 130687, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39097174

ABSTRACT

Human glycosyltransferases (GTs) play crucial roles in glycan biosynthesis, exhibiting diverse domain architectures. This study explores the functional diversity of "add-on" domains within human GTs, using data from the AlphaFold Protein Structure Database. Among 215 annotated human GTs, 74 contain one or more add-on domains in addition to their catalytic domain. These domains include lectin folds, fibronectin type III, and thioredoxin-like domains and contribute to substrate specificity, oligomerization, and consequent enzymatic activity. Notably, certain GTs possess dual enzymatic functions due to catalytic add-on domains. The analysis highlights the importance of add-on domains in enzyme functionality and disease implications, such as congenital disorders of glycosylation. This comprehensive overview enhances our understanding of GT domain organization, providing insights into glycosylation mechanisms and potential therapeutic targets.


Subject(s)
Catalytic Domain , Glycosyltransferases , Protein Domains , Humans , Glycosyltransferases/metabolism , Glycosyltransferases/chemistry , Substrate Specificity , Glycosylation , Models, Molecular , Polysaccharides/metabolism , Polysaccharides/chemistry
17.
Nat Commun ; 15(1): 7068, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39152129

ABSTRACT

Laboratory evolution studies have demonstrated that parallel evolutionary trajectories can lead to genetically distinct enzymes with high activity towards a non-preferred substrate. However, it is unknown whether such enzymes have convergent conformational dynamics and mechanistic features. To address this question, we use as a model the wild-type Homo sapiens kynureninase (HsKYNase), which is of great interest for cancer immunotherapy. Earlier, we isolated HsKYNase_66 through an unusual evolutionary trajectory, having a 410-fold increase in the kcat/KM for kynurenine (KYN) and reverse substrate selectivity relative to HsKYNase. Here, by following a different evolutionary trajectory we generate a genetically distinct variant, HsKYNase_93D9, that exhibits KYN catalytic activity comparable to that of HsKYNase_66, but instead it is a "generalist" that accepts 3'-hydroxykynurenine (OH-KYN) with the same proficiency. Pre-steady-state kinetic analysis reveals that while the evolution of HsKYNase_66 is accompanied by a change in the rate-determining step of the reactions, HsKYNase_93D9 retains the same catalytic mechanism as HsKYNase. HDX-MS shows that the conformational dynamics of the two enzymes are markedly different and distinct from ortholog prokaryotic enzymes with high KYN activity. Our work provides a mechanistic framework for understanding the relationship between evolutionary mechanisms and phenotypic traits of evolved generalist and specialist enzyme species.


Subject(s)
Evolution, Molecular , Hydrolases , Kynurenine , Substrate Specificity , Hydrolases/chemistry , Hydrolases/metabolism , Hydrolases/genetics , Humans , Kynurenine/metabolism , Kynurenine/chemistry , Kinetics , Protein Conformation
18.
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
19.
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
20.
Elife ; 132024 Aug 20.
Article in English | MEDLINE | ID: mdl-39163210

ABSTRACT

1-Naphthylamine (1NA), which is harmful to human and aquatic animals, has been used widely in the manufacturing of dyes, pesticides, and rubber antioxidants. Nevertheless, little is known about its environmental behavior and no bacteria have been reported to use it as the growth substrate. Herein, we describe a pathway for 1NA degradation in the isolate Pseudomonas sp. strain JS3066, determine the structure and mechanism of the enzyme NpaA1 that catalyzes the initial reaction, and reveal how the pathway evolved. From genetic and enzymatic analysis, a five gene-cluster encoding a dioxygenase system was determined to be responsible for the initial steps in 1NA degradation through glutamylation of 1NA. The γ-glutamylated 1NA was subsequently oxidized to 1,2-dihydroxynaphthalene which was further degraded by the well-established pathway of naphthalene degradation via catechol. A glutamine synthetase-like (GS-like) enzyme (NpaA1) initiates 1NA glutamylation, and this enzyme exhibits a broad substrate selectivity toward a variety of anilines and naphthylamine derivatives. Structural analysis revealed that the aromatic residues in the 1NA entry tunnel and the V201 site in the large substrate-binding pocket significantly influence NpaA1's substrate preferences. The findings enhance understanding of degrading polycyclic aromatic amines, and will also enable the application of bioremediation at naphthylamine contaminated sites.


Subject(s)
1-Naphthylamine , Pseudomonas , Pseudomonas/enzymology , Pseudomonas/genetics , Pseudomonas/metabolism , Substrate Specificity , 1-Naphthylamine/analogs & derivatives , 1-Naphthylamine/metabolism , Biodegradation, Environmental , Dioxygenases/metabolism , Dioxygenases/genetics , Dioxygenases/chemistry , Metabolic Networks and Pathways , Multigene Family , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL