Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34.051
Filter
1.
J Nat Prod ; 87(8): 2014-2020, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39142023

ABSTRACT

A high throughput screen performed to identify catalytic inhibitors of the oncogenic fusion form of cAMP-dependent protein kinase A catalytic subunit alpha (J-PKAcα) found an individual fraction from an organic extract of the marine soft coral Acrozoanthus australiae as active. Bioassay-guided isolation led to the identification of a 2-amino adenine alkaloid acroamine A (1), the first secondary metabolite discovered from this genus and previously reported as a synthetic product. As a naturally occurring protein kinase inhibitor, to unambiguously assign its chemical structure using modern spectroscopic and spectrometric techniques, five N-methylated derivatives acroamines A1-A5 (2-6) were semisynthesized. Three additional brominated congeners A6-A8 (7-9) were also semisynthesized to investigate the structure-activity relationship of the nine compounds as J-PKAcα inhibitors. Compounds 1-9 were tested for J-PKAcα and wild-type PKA inhibitory activities, which were observed exclusively in acroamine A (1) and its brominated analogs (7-9) achieving moderate potency (IC50 2-50 µM) while none of the N-methylated analogs exhibited kinase inhibition.


Subject(s)
Alkaloids , Anthozoa , Cyclic AMP-Dependent Protein Kinases , Animals , Anthozoa/chemistry , Molecular Structure , Alkaloids/pharmacology , Alkaloids/chemistry , Alkaloids/isolation & purification , Structure-Activity Relationship , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Adenine/pharmacology , Adenine/analogs & derivatives , Adenine/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Catalytic Domain
2.
Biochemistry ; 63(16): 2040-2050, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39088332

ABSTRACT

Hydrogen-bonding (H-bonding) interactions in metalloprotein active sites can critically regulate enzyme function. Changes in the protein structure triggered by interplay with substrates, products, and partner proteins are often translated to the metallocofactor by way of specific changes in H-bond networks connected to the active site. However, the complexities of metalloprotein architecture and mechanism often preclude our ability to define the precise molecular interactions giving rise to these intricate regulatory pathways. To address this shortcoming, we have developed conformationally switchable artificial metalloproteins (swArMs) in which allosteric Gln-binding triggers protein conformational changes that impact the microenvironment surrounding an installed metallocofactor. Herein, we report a combined structural, spectroscopic, and computational approach to enhance the conformation-dependent changes in H-bond interactions surrounding the metallocofactor site of a swArM. Structure-informed molecular dynamics simulations were employed to predict point mutations that could enhance active site H-bond interactions preferentially in the Gln-bound holo-conformation of the swArM. Testing our predictions via the unique infrared spectral signals associated with the metallocofactor site, we have identified three key residues capable of imparting conformational control over the metallocofactor microenvironment. The resultant swArMs not only model biologically relevant structural regulation but also provide an enhanced Gln-responsive biological probe to be leveraged in future biosensing applications.


Subject(s)
Hydrogen Bonding , Metalloproteins , Molecular Dynamics Simulation , Protein Conformation , Metalloproteins/chemistry , Metalloproteins/metabolism , Metalloproteins/genetics , Catalytic Domain
3.
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
4.
Appl Microbiol Biotechnol ; 108(1): 441, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39145831

ABSTRACT

Considering the structure of the bacterial GH15 family glucoamylase (GA), Thermoplasma trehalase Tvn1315 may be composed of a ß-sandwich domain (BD) and a catalytic domain (CD). Tvn1315 BD weakly binds to insoluble ß-glucans, such as cellulose, and helps fold CD. To determine how aromatic residues contribute to proper folding and enzyme activity, we performed alanine scanning for 32 aromatic residues in the BD. The study did not identify a single residue involved in glucan binding. However, several aromatic residues were found to be involved in BD or CD folding and in modulating the activity of the full-length enzyme. Among those aromatic residue mutations, the W43A mutation led to reduced solubility of the BD and full-length protein and resulted in a full-length enzyme with significantly lower activity. The activity of W43F and W43Y was significantly higher than that of W43A. In addition, Ala substitutions of Tyr83, Tyr113, and Tyr17 led to a reduction in trehalase activity, but Phe substitutions of these residues could be tolerated, as these mutants maintained activities similar to WT activity. Thus, these aromatic residues in BD may interact with CD and modulate enzyme activity. KEY POINTS: • Aromatic residues in the BD are involved in BD and CD folding. • Aromatic residues in the BD near the CD active site modulate enzyme activity. • BD interacts with CD and closely modulates enzyme activity.


Subject(s)
Catalytic Domain , Protein Folding , Trehalase , Trehalase/genetics , Trehalase/metabolism , Trehalase/chemistry , Amino Acids, Aromatic/metabolism , Amino Acid Substitution
5.
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
6.
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
7.
BMC Res Notes ; 17(1): 216, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095914

ABSTRACT

OBJECTIVE: Proteasomes are conserved proteases crucial for proteostasis in eukaryotes and are promising drug targets for protozoan parasites. Yet, the proteasomes of Entamoeba histolytica remain understudied. The study's objective was to analyse the differences in the substrate binding pockets of amoeba proteasomes from those of host, and computational modelling of ß5 catalytic subunit, with the goal of finding selective inhibitors. RESULTS: Comparative sequence analysis revealed differences in substrate binding sites of E. histolytica proteasomes, especially in the S1 and S3 pockets of the catalytic beta subunits, implying differences in substrate preference and susceptibility to inhibitors from host proteasomes. This was strongly supported by significantly lower sensitivity to MG132 mediated inhibition of amoebic proteasome ß5 subunit's chymotryptic activity compared to human proteasomes, also reflected in lower sensitivity of E. histolytica to MG132 for inhibition of proliferation. Computational models of ß4 and ß5 subunits, and a docked ß4-ß5 model revealed a binding pocket between ß4-ß5, similar to that of Leishmania tarentolae. Selective inhibitors for visceral leishmaniasis, LXE408 and compound 8, docked well to this pocket. This functional and sequence-based analysis predicts differences between amoebic and host proteasomes that can be utilized to develop rationally designed, selective inhibitors against E. histolytica.


Subject(s)
Entamoeba histolytica , Proteasome Endopeptidase Complex , Entamoeba histolytica/enzymology , Entamoeba histolytica/metabolism , Proteasome Endopeptidase Complex/metabolism , Humans , Binding Sites , Leupeptins/pharmacology , Substrate Specificity , Protozoan Proteins/metabolism , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Proteasome Inhibitors/pharmacology , Molecular Docking Simulation , Amino Acid Sequence , Catalytic Domain , Protein Binding , Models, Molecular
8.
Sci Adv ; 10(32): eado5504, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39121211

ABSTRACT

Phosphoryl transfer is a fundamental reaction in cellular signaling and metabolism that requires Mg2+ as an essential cofactor. While the primary function of Mg2+ is electrostatic activation of substrates, such as ATP, the full spectrum of catalytic mechanisms exerted by Mg2+ is not known. In this study, we integrate structural biology methods, molecular dynamic (MD) simulations, phylogeny, and enzymology assays to provide molecular insights into Mg2+-dependent structural reorganization in the active site of the metabolic enzyme adenylate kinase. Our results demonstrate that Mg2+ induces a conformational rearrangement of the substrates (ATP and ADP), resulting in a 30° adjustment of the angle essential for reversible phosphoryl transfer, thereby optimizing it for catalysis. MD simulations revealed transitions between conformational substates that link the fluctuation of the angle to large-scale enzyme dynamics. The findings contribute detailed insight into Mg2+ activation of enzymes and may be relevant for reversible and irreversible phosphoryl transfer reactions.


Subject(s)
Adenylate Kinase , Catalytic Domain , Magnesium , Molecular Dynamics Simulation , Magnesium/metabolism , Magnesium/chemistry , Adenylate Kinase/metabolism , Adenylate Kinase/chemistry , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/chemistry , Protein Conformation , Adenosine Diphosphate/metabolism , Adenosine Diphosphate/chemistry
9.
Nat Commun ; 15(1): 6673, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39107302

ABSTRACT

Allosteric regulation of inosine 5'-monophosphate dehydrogenase (IMPDH), an essential enzyme of purine metabolism, contributes to the homeostasis of adenine and guanine nucleotides. However, the precise molecular mechanism of IMPDH regulation in bacteria remains unclear. Using biochemical and cryo-EM approaches, we reveal the intricate molecular mechanism of the IMPDH allosteric regulation in mycobacteria. The enzyme is inhibited by both GTP and (p)ppGpp, which bind to the regulatory CBS domains and, via interactions with basic residues in hinge regions, lock the catalytic core domains in a compressed conformation. This results in occlusion of inosine monophosphate (IMP) substrate binding to the active site and, ultimately, inhibition of the enzyme. The GTP and (p)ppGpp allosteric effectors bind to their dedicated sites but stabilize the compressed octamer by a common mechanism. Inhibition is relieved by the competitive displacement of GTP or (p)ppGpp by ATP allowing IMP-induced enzyme expansion. The structural knowledge and mechanistic understanding presented here open up new possibilities for the development of allosteric inhibitors with antibacterial potential.


Subject(s)
Guanosine Triphosphate , IMP Dehydrogenase , IMP Dehydrogenase/metabolism , IMP Dehydrogenase/chemistry , IMP Dehydrogenase/antagonists & inhibitors , Allosteric Regulation , Guanosine Triphosphate/metabolism , Cryoelectron Microscopy , Catalytic Domain , Models, Molecular , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Guanosine Pentaphosphate/metabolism , Inosine Monophosphate/metabolism , Inosine Monophosphate/chemistry , Protein Binding , Adenosine Triphosphate/metabolism , Mycobacterium smegmatis/enzymology , Mycobacterium smegmatis/metabolism
10.
Molecules ; 29(15)2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39124857

ABSTRACT

The superfamily of acid proteases has two catalytic aspartates for proteolysis of their peptide substrates. Here, we show a minimal structural scaffold, the structural catalytic core (SCC), which is conserved within each family of acid proteases, but varies between families, and thus can serve as a structural marker of four individual protease families. The SCC is a dimer of several structural blocks, such as the DD-link, D-loop, and G-loop, around two catalytic aspartates in each protease subunit or an individual chain. A dimer made of two (D-loop + DD-link) structural elements makes a DD-zone, and the D-loop + G-loop combination makes a psi-loop. These structural markers are useful for protein comparison, structure identification, protein family separation, and protein engineering.


Subject(s)
Catalytic Domain , Models, Molecular , Peptide Hydrolases/chemistry , Peptide Hydrolases/metabolism , Amino Acid Sequence , Protein Conformation
11.
Int J Mol Sci ; 25(15)2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39125622

ABSTRACT

Bacteria are known to be constantly adapting to become resistant to antibiotics. Currently, efficient antibacterial compounds are still available; however, it is only a matter of time until these compounds also become inefficient. Ribonucleases are the enzymes responsible for the maturation and degradation of RNA molecules, and many of them are essential for microbial survival. Members of the PNPase and RNase II families of exoribonucleases have been implicated in virulence in many pathogens and, as such, are valid targets for the development of new antibacterials. In this paper, we describe the use of virtual high-throughput screening (vHTS) to identify chemical compounds predicted to bind to the active sites within the known structures of RNase II and PNPase from Escherichia coli. The subsequent in vitro screening identified compounds that inhibited the activity of these exoribonucleases, with some also affecting cell viability, thereby providing proof of principle for utilizing the known structures of these enzymes in the pursuit of new antibacterials.


Subject(s)
Anti-Bacterial Agents , Enzyme Inhibitors , Escherichia coli , Exoribonucleases , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Exoribonucleases/antagonists & inhibitors , Exoribonucleases/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Escherichia coli/drug effects , Escherichia coli/enzymology , Catalytic Domain , High-Throughput Screening Assays/methods , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/antagonists & inhibitors , Bacteria/drug effects , Bacteria/enzymology
12.
Int J Mol Sci ; 25(15)2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39125865

ABSTRACT

Inflammation is a protective stress response triggered by external stimuli, with 5-lipoxygenase (5LOX) playing a pivotal role as a potent mediator of the leukotriene (Lts) inflammatory pathway. Nordihydroguaiaretic acid (NDGA) functions as a natural orthosteric inhibitor of 5LOX, while 3-acetyl-11-keto-ß-boswellic acid (AKBA) acts as a natural allosteric inhibitor targeting 5LOX. However, the precise mechanisms of inhibition have remained unclear. In this study, Gaussian accelerated molecular dynamics (GaMD) simulation was employed to elucidate the inhibitory mechanisms of NDGA and AKBA on 5LOX. It was found that the orthosteric inhibitor NDGA was tightly bound in the protein's active pocket, occupying the active site and inhibiting the catalytic activity of the 5LOX enzyme through competitive inhibition. The binding of the allosteric inhibitor AKBA induced significant changes at the distal active site, leading to a conformational shift of residues 168-173 from a loop to an α-helix and significant negative correlated motions between residues 285-290 and 375-400, reducing the distance between these segments. In the simulation, the volume of the active cavity in the stable conformation of the protein was reduced, hindering the substrate's entry into the active cavity and, thereby, inhibiting protein activity through allosteric effects. Ultimately, Markov state models (MSM) were used to identify and classify the metastable states of proteins, revealing the transition times between different conformational states. In summary, this study provides theoretical insights into the inhibition mechanisms of 5LOX by AKBA and NDGA, offering new perspectives for the development of novel inhibitors specifically targeting 5LOX, with potential implications for anti-inflammatory drug development.


Subject(s)
Arachidonate 5-Lipoxygenase , Lipoxygenase Inhibitors , Markov Chains , Molecular Dynamics Simulation , Arachidonate 5-Lipoxygenase/metabolism , Arachidonate 5-Lipoxygenase/chemistry , Lipoxygenase Inhibitors/pharmacology , Lipoxygenase Inhibitors/chemistry , Humans , Catalytic Domain , Protein Binding , Masoprocol/pharmacology , Masoprocol/chemistry , Protein Conformation
13.
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
14.
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
15.
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
16.
Nat Commun ; 15(1): 7348, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39187482

ABSTRACT

Annotating active sites in enzymes is crucial for advancing multiple fields including drug discovery, disease research, enzyme engineering, and synthetic biology. Despite the development of numerous automated annotation algorithms, a significant trade-off between speed and accuracy limits their large-scale practical applications. We introduce EasIFA, an enzyme active site annotation algorithm that fuses latent enzyme representations from the Protein Language Model and 3D structural encoder, and then aligns protein-level information with the knowledge of enzymatic reactions using a multi-modal cross-attention framework. EasIFA outperforms BLASTp with a 10-fold speed increase and improved recall, precision, f1 score, and MCC by 7.57%, 13.08%, 9.68%, and 0.1012, respectively. It also surpasses empirical-rule-based algorithm and other state-of-the-art deep learning annotation method based on PSSM features, achieving a speed increase ranging from 650 to 1400 times while enhancing annotation quality. This makes EasIFA a suitable replacement for conventional tools in both industrial and academic settings. EasIFA can also effectively transfer knowledge gained from coarsely annotated enzyme databases to smaller, high-precision datasets, highlighting its ability to model sparse and high-quality databases. Additionally, EasIFA shows potential as a catalytic site monitoring tool for designing enzymes with desired functions beyond their natural distribution.


Subject(s)
Algorithms , Catalytic Domain , Deep Learning , Enzymes , Enzymes/metabolism , Enzymes/chemistry , Databases, Protein , Molecular Sequence Annotation/methods , Computational Biology/methods
17.
Proc Natl Acad Sci U S A ; 121(36): e2318527121, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39190355

ABSTRACT

Catalysis and translocation of multisubunit DNA-directed RNA polymerases underlie all cellular mRNA synthesis. RNA polymerase II (Pol II) synthesizes eukaryotic pre-mRNAs from a DNA template strand buried in its active site. Structural details of catalysis at near-atomic resolution and precise arrangement of key active site components have been elusive. Here, we present the free-electron laser (FEL) structures of a matched ATP-bound Pol II and the hyperactive Rpb1 T834P bridge helix (BH) mutant at the highest resolution to date. The radiation-damage-free FEL structures reveal the full active site interaction network, including the trigger loop (TL) in the closed conformation, bonafide occupancy of both site A and B Mg2+, and, more importantly, a putative third (site C) Mg2+ analogous to that described for some DNA polymerases but not observed previously for cellular RNA polymerases. Molecular dynamics (MD) simulations of the structures indicate that the third Mg2+ is coordinated and stabilized at its observed position. TL residues provide half of the substrate binding pocket while multiple TL/BH interactions induce conformational changes that could allow translocation upon substrate hydrolysis. Consistent with TL/BH communication, a FEL structure and MD simulations of the T834P mutant reveal rearrangement of some active site interactions supporting potential plasticity in active site function and long-distance effects on both the width of the central channel and TL conformation, likely underlying its increased elongation rate at the expense of fidelity.


Subject(s)
Catalytic Domain , Magnesium , Molecular Dynamics Simulation , RNA Polymerase II , Transcription, Genetic , RNA Polymerase II/metabolism , RNA Polymerase II/chemistry , RNA Polymerase II/genetics , Magnesium/metabolism , Magnesium/chemistry , Lasers , Protein Conformation , Electrons , Protein Binding , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/chemistry , Binding Sites
18.
Proc Natl Acad Sci U S A ; 121(34): e2314999121, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39133844

ABSTRACT

Mutations in protein active sites can dramatically improve function. The active site, however, is densely packed and extremely sensitive to mutations. Therefore, some mutations may only be tolerated in combination with others in a phenomenon known as epistasis. Epistasis reduces the likelihood of obtaining improved functional variants and dramatically slows natural and lab evolutionary processes. Research has shed light on the molecular origins of epistasis and its role in shaping evolutionary trajectories and outcomes. In addition, sequence- and AI-based strategies that infer epistatic relationships from mutational patterns in natural or experimental evolution data have been used to design functional protein variants. In recent years, combinations of such approaches and atomistic design calculations have successfully predicted highly functional combinatorial mutations in active sites. These were used to design thousands of functional active-site variants, demonstrating that, while our understanding of epistasis remains incomplete, some of the determinants that are critical for accurate design are now sufficiently understood. We conclude that the space of active-site variants that has been explored by evolution may be expanded dramatically to enhance natural activities or discover new ones. Furthermore, design opens the way to systematically exploring sequence and structure space and mutational impacts on function, deepening our understanding and control over protein activity.


Subject(s)
Epistasis, Genetic , Mutation , Evolution, Molecular , Proteins/genetics , Proteins/chemistry , Proteins/metabolism , Catalytic Domain , Protein Engineering/methods
19.
Phys Chem Chem Phys ; 26(34): 22413-22422, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39140173

ABSTRACT

Acetylcholine (Ach) is a common neurotransmitter in the central nervous system (CNS) and peripheral nervous system (PNS). It is one of the neurotransmitters in the autonomic nervous system and the main neurotransmitter in all autonomic ganglia. Experiments have confirmed that electromagnetic waves can affect the synthesis of animal neurotransmitters, but the microscopic effects of electromagnetic waves in the terahertz (THz) frequency band are still unclear. Based on density functional theory (DFT) and molecular dynamics (MD) simulation methods, this paper studies the effect of THz electromagnetic waves on the binding of choline to choline acetyltransferase (ChAT). By emitting THz waves that resonate with the characteristic vibration mode of choline near the active site, it was found that THz waves with a frequency of 45.3 THz affected the binding of choline to ChAT, especially the binding of the active site histidine His324 to choline. The main evidence is that under the action of THz waves, the binding free energy of choline to histidine His324 and ChAT at the active site was significantly reduced compared to noE, which may have a potential impact on the enzymatic synthesis of Ach. It is expected to achieve the purpose of regulating the synthesis of the neurotransmitter Ach under the action of THz waves and treat certain nervous system diseases.


Subject(s)
Choline O-Acetyltransferase , Choline , Molecular Dynamics Simulation , Choline O-Acetyltransferase/metabolism , Choline O-Acetyltransferase/chemistry , Choline/metabolism , Choline/chemistry , Terahertz Radiation , Catalytic Domain , Protein Binding , Density Functional Theory , Binding Sites , Acetylcholine/metabolism , Acetylcholine/chemistry , Thermodynamics
20.
Proc Natl Acad Sci U S A ; 121(34): e2400912121, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39145930

ABSTRACT

Myo-inositol-1-phosphate synthase (MIPS) catalyzes the NAD+-dependent isomerization of glucose-6-phosphate (G6P) into inositol-1-phosphate (IMP), controlling the rate-limiting step of the inositol pathway. Previous structural studies focused on the detailed molecular mechanism, neglecting large-scale conformational changes that drive the function of this 240 kDa homotetrameric complex. In this study, we identified the active, endogenous MIPS in cell extracts from the thermophilic fungus Thermochaetoides thermophila. By resolving the native structure at 2.48 Å (FSC = 0.143), we revealed a fully populated active site. Utilizing 3D variability analysis, we uncovered conformational states of MIPS, enabling us to directly visualize an order-to-disorder transition at its catalytic center. An acyclic intermediate of G6P occupied the active site in two out of the three conformational states, indicating a catalytic mechanism where electrostatic stabilization of high-energy intermediates plays a crucial role. Examination of all isomerases with known structures revealed similar fluctuations in secondary structure within their active sites. Based on these findings, we established a conformational selection model that governs substrate binding and eventually inositol availability. In particular, the ground state of MIPS demonstrates structural configurations regardless of substrate binding, a pattern observed across various isomerases. These findings contribute to the understanding of MIPS structure-based function, serving as a template for future studies targeting regulation and potential therapeutic applications.


Subject(s)
Catalytic Domain , Inositol , Myo-Inositol-1-Phosphate Synthase , Myo-Inositol-1-Phosphate Synthase/metabolism , Myo-Inositol-1-Phosphate Synthase/genetics , Myo-Inositol-1-Phosphate Synthase/chemistry , Inositol/metabolism , Inositol/chemistry , Inositol Phosphates/metabolism , Glucose-6-Phosphate/metabolism , Glucose-6-Phosphate/chemistry , Models, Molecular , Protein Conformation , Fungal Proteins/metabolism , Fungal Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...