Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.228
Filter
Add more filters

Publication year range
1.
Cell ; 170(1): 172-184.e11, 2017 Jun 29.
Article in English | MEDLINE | ID: mdl-28648660

ABSTRACT

Membrane scission is essential for intracellular trafficking. While BAR domain proteins such as endophilin have been reported in dynamin-independent scission of tubular membrane necks, the cutting mechanism has yet to be deciphered. Here, we combine a theoretical model, in vitro, and in vivo experiments revealing how protein scaffolds may cut tubular membranes. We demonstrate that the protein scaffold bound to the underlying tube creates a frictional barrier for lipid diffusion; tube elongation thus builds local membrane tension until the membrane undergoes scission through lysis. We call this mechanism friction-driven scission (FDS). In cells, motors pull tubes, particularly during endocytosis. Through reconstitution, we show that motors not only can pull out and extend protein-scaffolded tubes but also can cut them by FDS. FDS is generic, operating even in the absence of amphipathic helices in the BAR domain, and could in principle apply to any high-friction protein and membrane assembly.


Subject(s)
Endocytosis , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Acyltransferases/chemistry , Acyltransferases/metabolism , Animals , Biomechanical Phenomena , Friction , Humans , Lipid Metabolism , Protein Domains , Rats
2.
Nature ; 625(7993): 74-78, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38110574

ABSTRACT

Enzymes are recognized as exceptional catalysts for achieving high stereoselectivities1-3, but their ability to control the reactivity and stereoinduction of free radicals lags behind that of chemical catalysts4. Thiamine diphosphate (ThDP)-dependent enzymes5 are well-characterized systems that inspired the development of N-heterocyclic carbenes (NHCs)6-8 but have not yet been proved viable in asymmetric radical transformations. There is a lack of a biocompatible and general radical-generation mechanism, as nature prefers to avoid radicals that may be harmful to biological systems9. Here we repurpose a ThDP-dependent lyase as a stereoselective radical acyl transferase (RAT) through protein engineering and combination with organophotoredox catalysis10. Enzyme-bound ThDP-derived ketyl radicals are selectively generated through single-electron oxidation by a photoexcited organic dye and then cross-coupled with prochiral alkyl radicals with high enantioselectivity. Diverse chiral ketones are prepared from aldehydes and redox-active esters (35 examples, up to 97% enantiomeric excess (e.e.)) by this method. Mechanistic studies reveal that this previously elusive dual-enzyme catalysis/photocatalysis directs radicals with the unique ThDP cofactor and evolvable active site. This work not only expands the repertoire of biocatalysis but also provides a unique strategy for controlling radicals with enzymes, complementing existing chemical tools.


Subject(s)
Acyltransferases , Biocatalysis , Light , Lyases , Acylation , Acyltransferases/chemistry , Acyltransferases/metabolism , Aldehydes/metabolism , Biocatalysis/radiation effects , Catalytic Domain , Free Radicals/metabolism , Ketones/metabolism , Lyases/chemistry , Lyases/metabolism , Oxidation-Reduction , Protein Engineering , Stereoisomerism , Thiamine Pyrophosphate/metabolism
3.
Nature ; 626(8000): 852-858, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38326608

ABSTRACT

Bile acids (BAs) are steroid detergents in bile that contribute to the absorption of fats and fat-soluble vitamins while shaping the gut microbiome because of their antimicrobial properties1-4. Here we identify the enzyme responsible for a mechanism of BA metabolism by the gut microbiota involving amino acid conjugation to the acyl-site of BAs, thus producing a diverse suite of microbially conjugated bile acids (MCBAs). We show that this transformation is mediated by acyltransferase activity of bile salt hydrolase (bile salt hydrolase/transferase, BSH/T). Clostridium perfringens BSH/T rapidly performed acyl transfer when provided various amino acids and taurocholate, glycocholate or cholate, with an optimum at pH 5.3. Amino acid conjugation by C. perfringens BSH/T was diverse, including all proteinaceous amino acids except proline and aspartate. MCBA production was widespread among gut bacteria, with strain-specific amino acid use. Species with similar BSH/T amino acid sequences had similar conjugation profiles and several bsh/t alleles correlated with increased conjugation diversity. Tertiary structure mapping of BSH/T followed by mutagenesis experiments showed that active site structure affects amino acid selectivity. These MCBA products had antimicrobial properties, where greater amino acid hydrophobicity showed greater antimicrobial activity. Inhibitory concentrations of MCBAs reached those measured natively in the mammalian gut. MCBAs fed to mice entered enterohepatic circulation, in which liver and gallbladder concentrations varied depending on the conjugated amino acid. Quantifying MCBAs in human faecal samples showed that they reach concentrations equal to or greater than secondary and primary BAs and were reduced after bariatric surgery, thus supporting MCBAs as a significant component of the BA pool that can be altered by changes in gastrointestinal physiology. In conclusion, the inherent acyltransferase activity of BSH/T greatly diversifies BA chemistry, creating a set of previously underappreciated metabolites with the potential to affect the microbiome and human health.


Subject(s)
Acyltransferases , Amidohydrolases , Bile Acids and Salts , Clostridium perfringens , Gastrointestinal Microbiome , Animals , Humans , Mice , Acyltransferases/chemistry , Acyltransferases/metabolism , Alleles , Amidohydrolases/chemistry , Amidohydrolases/metabolism , Amino Acids/metabolism , Anti-Infective Agents/metabolism , Anti-Infective Agents/pharmacology , Bariatric Surgery , Bile Acids and Salts/chemistry , Bile Acids and Salts/metabolism , Catalytic Domain , Clostridium perfringens/enzymology , Clostridium perfringens/metabolism , Feces/chemistry , Gallbladder/metabolism , Gastrointestinal Microbiome/physiology , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Liver/metabolism , Taurocholic Acid/metabolism
4.
Cell ; 149(1): 137-45, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22464326

ABSTRACT

Functioning as key players in cellular regulation of membrane curvature, BAR domain proteins bend bilayers and recruit interaction partners through poorly understood mechanisms. Using electron cryomicroscopy, we present reconstructions of full-length endophilin and its N-terminal N-BAR domain in their membrane-bound state. Endophilin lattices expose large areas of membrane surface and are held together by promiscuous interactions between endophilin's amphipathic N-terminal helices. Coarse-grained molecular dynamics simulations reveal that endophilin lattices are highly dynamic and that the N-terminal helices are required for formation of a stable and regular scaffold. Furthermore, endophilin accommodates different curvatures through a quantized addition or removal of endophilin dimers, which in some cases causes dimerization of endophilin's SH3 domains, suggesting that the spatial presentation of SH3 domains, rather than affinity, governs the recruitment of downstream interaction partners.


Subject(s)
Acyltransferases/metabolism , Acyltransferases/chemistry , Acyltransferases/genetics , Acyltransferases/ultrastructure , Animals , Cell Membrane/chemistry , Cell Membrane/metabolism , Cryoelectron Microscopy , Models, Molecular , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Protein Interaction Domains and Motifs , Protein Structure, Tertiary , Rats
5.
Nature ; 581(7808): 323-328, 2020 05.
Article in English | MEDLINE | ID: mdl-32433611

ABSTRACT

Triacylglycerols store metabolic energy in organisms and have industrial uses as foods and fuels. Excessive accumulation of triacylglycerols in humans causes obesity and is associated with metabolic diseases1. Triacylglycerol synthesis is catalysed by acyl-CoA diacylglycerol acyltransferase (DGAT) enzymes2-4, the structures and catalytic mechanisms of which remain unknown. Here we determined the structure of dimeric human DGAT1, a member of the membrane-bound O-acyltransferase (MBOAT) family, by cryo-electron microscopy at approximately 3.0 Å resolution. DGAT1 forms a homodimer through N-terminal segments and a hydrophobic interface, with putative active sites within the membrane region. A structure obtained with oleoyl-CoA substrate resolved at approximately 3.2 Å shows that the CoA moiety binds DGAT1 on the cytosolic side and the acyl group lies deep within a hydrophobic channel, positioning the acyl-CoA thioester bond near an invariant catalytic histidine residue. The reaction centre is located inside a large cavity, which opens laterally to the membrane bilayer, providing lipid access to the active site. A lipid-like density-possibly representing an acyl-acceptor molecule-is located within the reaction centre, orthogonal to acyl-CoA. Insights provided by the DGAT1 structures, together with mutagenesis and functional studies, provide the basis for a model of the catalysis of triacylglycerol synthesis by DGAT.


Subject(s)
Biocatalysis , Cryoelectron Microscopy , Diacylglycerol O-Acyltransferase/metabolism , Diacylglycerol O-Acyltransferase/ultrastructure , Triglycerides/biosynthesis , Acyl Coenzyme A/chemistry , Acyl Coenzyme A/metabolism , Acyl Coenzyme A/ultrastructure , Acyltransferases/chemistry , Acyltransferases/metabolism , Catalytic Domain , Cell Membrane/chemistry , Cell Membrane/metabolism , Diacylglycerol O-Acyltransferase/chemistry , Histidine/chemistry , Histidine/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Protein Multimerization , Substrate Specificity
6.
Nature ; 573(7772): 139-143, 2019 09.
Article in English | MEDLINE | ID: mdl-31462771

ABSTRACT

Signal transducer and activator of transcription 3 (STAT3) has a critical role in regulating cell fate, inflammation and immunity1,2. Cytokines and growth factors activate STAT3 through kinase-mediated tyrosine phosphorylation and dimerization3,4. It remains unknown whether other factors promote STAT3 activation through different mechanisms. Here we show that STAT3 is post-translationally S-palmitoylated at the SRC homology 2 (SH2) domain, which promotes the dimerization and transcriptional activation of STAT3. Fatty acids can directly activate STAT3 by enhancing its palmitoylation, in synergy with cytokine stimulation. We further identified ZDHHC19 as a palmitoyl acyltransferase that regulates STAT3. Cytokine stimulation increases STAT3 palmitoylation by promoting the association between ZDHHC19 and STAT3, which is mediated by the SH3 domain of GRB2. Silencing ZDHHC19 blocks STAT3 palmitoylation and dimerization, and impairs the cytokine- and fatty-acid-induced activation of STAT3. ZDHHC19 is frequently amplified in multiple human cancers, including in 39% of lung squamous cell carcinomas. High levels of ZDHHC19 correlate with high levels of nuclear STAT3 in patient samples. In addition, knockout of ZDHHC19 in lung squamous cell carcinoma cells significantly blocks STAT3 activity, and inhibits the fatty-acid-induced formation of tumour spheres as well as tumorigenesis induced by high-fat diets in an in vivo mouse model. Our studies reveal that fatty-acid- and ZDHHC19-mediated palmitoylation are signals that regulate STAT3, which provides evidence linking the deregulation of palmitoylation to inflammation and cancer.


Subject(s)
Acyltransferases/metabolism , Fatty Acids/metabolism , Lipoylation , Lung Neoplasms/metabolism , STAT3 Transcription Factor/metabolism , Acyltransferases/antagonists & inhibitors , Acyltransferases/chemistry , Acyltransferases/deficiency , Animals , Carcinogenesis , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Conserved Sequence , Cysteine/metabolism , Disease Models, Animal , Heterografts , Humans , Inflammation/metabolism , Inflammation/pathology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Lung Neoplasms/pathology , Mice , Mice, SCID , Neoplasm Transplantation , Phosphorylation , Protein Multimerization , STAT3 Transcription Factor/chemistry , Signal Transduction , src Homology Domains
7.
Genes Dev ; 31(2): 88-100, 2017 01 15.
Article in English | MEDLINE | ID: mdl-28202538

ABSTRACT

The eukaryotic RNA exosome is an essential and conserved protein complex that can degrade or process RNA substrates in the 3'-to-5' direction. Since its discovery nearly two decades ago, studies have focused on determining how the exosome, along with associated cofactors, achieves the demanding task of targeting particular RNAs for degradation and/or processing in both the nucleus and cytoplasm. In this review, we highlight recent advances that have illuminated roles for the RNA exosome and its cofactors in specific biological pathways, alongside studies that attempted to dissect these activities through structural and biochemical characterization of nuclear and cytoplasmic RNA exosome complexes.


Subject(s)
Exosomes/metabolism , Homeostasis/genetics , RNA/metabolism , Acyltransferases/chemistry , Acyltransferases/metabolism , Animals , Catalytic Domain , Exosomes/chemistry , Humans , RNA Transport , Serine Endopeptidases/chemistry , Serine Endopeptidases/metabolism
8.
J Biol Chem ; 299(4): 103069, 2023 04.
Article in English | MEDLINE | ID: mdl-36841477

ABSTRACT

Transferases are ubiquitous across all known life. While much work has been done to understand and describe these essential enzymes, there have been minimal efforts to exert tight and reversible control over their activity for various biotechnological applications. Here, we apply a rational, computation-guided methodology to design and test a transferase-class enzyme allosterically regulated by light-oxygen-voltage 2 sensing domain. We utilize computational techniques to determine the intrinsic allosteric networks within N-acyltransferase (Orf11/∗Dbv8) and identify potential allosteric sites on the protein's surface. We insert light-oxygen-voltage 2 sensing domain at the predicted allosteric site, exerting reversible control over enzymatic activity. We demonstrate blue-light regulation of N-acyltransferase (Orf11/∗Dbv8) function. Our study for the first time demonstrates optogenetic regulation of a transferase-class enzyme as a proof-of-concept for controllable transferase design. This successful design opens the door for many future applications in metabolic engineering and cellular programming.


Subject(s)
Acyltransferases , Recombinant Fusion Proteins , Acyltransferases/chemistry , Acyltransferases/genetics , Allosteric Regulation , Allosteric Site , Light , Oxygen , Protein Domains , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Enzyme Activation/radiation effects
9.
Biochem Biophys Res Commun ; 718: 150080, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38735137

ABSTRACT

Catalytic promiscuity of enzymes plays a pivotal role in driving the evolution of plant specialized metabolism. Chalcone synthase (CHS) catalyzes the production of 2',4,4',6'-tetrahydroxychalcone (THC), a common precursor of plant flavonoids, from p-coumaroyl-coenzyme A (-CoA) and three malonyl-CoA molecules. CHS has promiscuous product specificity, producing a significant amount of p-coumaroyltriacetic lactone (CTAL) in vitro. However, mechanistic aspects of this CHS promiscuity remain to be clarified. Here, we show that the product specificity of soybean CHS (GmCHS1) is altered by CoA, a reaction product, which selectively inhibits THC production (IC50, 67 µM) and enhances CTAL production. We determined the structure of a ternary GmCHS1/CoA/naringenin complex, in which CoA is bound to the CoA-binding tunnel via interactions with Lys55, Arg58, and Lys268. Replacement of these residues by alanine resulted in an enhanced THC/CTAL production ratio, suggesting the role of these residues in the CoA-mediated alteration of product specificity. In the ternary complex, a mobile loop ("the K-loop"), which contains Lys268, was in a "closed conformation" placing over the CoA-binding tunnel, whereas in the apo and binary complex structures, the K-loop was in an "open conformation" and remote from the tunnel. We propose that the production of THC involves a transition of the K-loop conformation between the open and closed states, whereas synthesis of CTAL is independent of it. In the presence of CoA, an enzyme conformer with the closed K-loop conformation becomes increasingly dominant, hampering the transition of K-loop conformations to result in decreased THC production and increased CTAL production.


Subject(s)
Acyltransferases , Glycine max , Acyltransferases/chemistry , Acyltransferases/metabolism , Acyltransferases/genetics , Glycine max/enzymology , Substrate Specificity , Coenzyme A/metabolism , Coenzyme A/chemistry , Models, Molecular , Protein Conformation , Chalcones/chemistry , Chalcones/metabolism , Plant Proteins/chemistry , Plant Proteins/metabolism , Plant Proteins/genetics
10.
Nature ; 562(7726): 286-290, 2018 10.
Article in English | MEDLINE | ID: mdl-30283133

ABSTRACT

Membrane-bound O-acyltransferases (MBOATs) are a superfamily of integral transmembrane enzymes that are found in all kingdoms of life1. In bacteria, MBOATs modify protective cell-surface polymers. In vertebrates, some MBOAT enzymes-such as acyl-coenzyme A:cholesterol acyltransferase and diacylglycerol acyltransferase 1-are responsible for lipid biosynthesis or phospholipid remodelling2,3. Other MBOATs, including porcupine, hedgehog acyltransferase and ghrelin acyltransferase, catalyse essential lipid modifications of secreted proteins such as Wnt, hedgehog and ghrelin, respectively4-10. Although many MBOAT proteins are important drug targets, little is known about their molecular architecture and functional mechanisms. Here we present crystal structures of DltB, an MBOAT responsible for the D-alanylation of cell-wall teichoic acid in Gram-positive bacteria11-16, both alone and in complex with the D-alanyl donor protein DltC. DltB contains a ring of 11 peripheral transmembrane helices, which shield a highly conserved extracellular structural funnel extending into the middle of the lipid bilayer. The conserved catalytic histidine residue is located at the bottom of this funnel and is connected to the intracellular DltC through a narrow tunnel. Mutation of either the catalytic histidine or the DltC-binding site of DltB abolishes the D-alanylation of lipoteichoic acid and sensitizes the Gram-positive bacterium Bacillus subtilis to cell-wall stress, which suggests cross-membrane catalysis involving the tunnel. Structure-guided sequence comparison among DltB and vertebrate MBOATs reveals a conserved structural core and suggests that MBOATs from different organisms have similar catalytic mechanisms. Our structures provide a template for understanding structure-function relationships in MBOATs and for developing therapeutic MBOAT inhibitors.


Subject(s)
Acyltransferases/chemistry , Acyltransferases/metabolism , Lipid Bilayers/metabolism , Acyltransferases/genetics , Amino Acid Sequence , Animals , Bacillus subtilis/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Biocatalysis , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cell Wall/metabolism , Conserved Sequence , Crystallography, X-Ray , Histidine/genetics , Histidine/metabolism , Lipid Bilayers/chemistry , Lipopolysaccharides/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Models, Molecular , Mutation , Protein Binding , Structure-Activity Relationship , Teichoic Acids/metabolism
11.
Proc Natl Acad Sci U S A ; 118(36)2021 09 07.
Article in English | MEDLINE | ID: mdl-34480000

ABSTRACT

The O-acetylation of exopolysaccharides, including the essential bacterial cell wall polymer peptidoglycan, confers resistance to their lysis by exogenous hydrolases. Like the enzymes catalyzing the O-acetylation of exopolysaccharides in the Golgi of animals and fungi, peptidoglycan O-acetyltransferase A (OatA) is predicted to be an integral membrane protein comprised of a membrane-spanning acyltransferase-3 (AT-3) domain and an extracytoplasmic domain; for OatA, these domains are located in the N- and C-terminal regions of the enzyme, respectively. The recombinant C-terminal domain (OatAC) has been characterized as an SGNH acetyltransferase, but nothing was known about the function of the N-terminal AT-3 domain (OatAN) or its homologs associated with other acyltransferases. We report herein the experimental determination of the topology of Staphylococcus aureus OatAN, which differs markedly from that predicted in silico. We present the biochemical characterization of OatAN as part of recombinant OatA and demonstrate that acetyl-CoA serves as the substrate for OatAN Using in situ and in vitro assays, we characterized 35 engineered OatA variants which identified a catalytic triad of Tyr-His-Glu residues. We trapped an acetyl group from acetyl-CoA on the catalytic Tyr residue that is located on an extracytoplasmic loop of OatAN Further enzymatic characterization revealed that O-acetyl-Tyr represents the substrate for OatAC We propose a model for OatA action involving the translocation of acetyl groups from acetyl-CoA across the cytoplasmic membrane by OatAN and their subsequent intramolecular transfer to OatAC for the O-acetylation of peptidoglycan via the concerted action of catalytic Tyr and Ser residues.


Subject(s)
Acetyltransferases/metabolism , Acyltransferases/metabolism , Bacterial Proteins/metabolism , Peptidoglycan/metabolism , Staphylococcus aureus/enzymology , Acetyl Coenzyme A/metabolism , Acetylation , Acetyltransferases/chemistry , Acyltransferases/chemistry , Amino Acid Sequence , Bacterial Proteins/chemistry , Catalytic Domain , Cell Wall/enzymology , Muramidase/metabolism , Substrate Specificity
12.
Int J Mol Sci ; 25(11)2024 May 22.
Article in English | MEDLINE | ID: mdl-38891840

ABSTRACT

Chalcone synthase (CHS) and chalcone isomerase (CHI) catalyze the first two committed steps of the flavonoid pathway that plays a pivotal role in the growth and reproduction of land plants, including UV protection, pigmentation, symbiotic nitrogen fixation, and pathogen resistance. Based on the obtained X-ray crystal structures of CHS, CHI, and chalcone isomerase-like protein (CHIL) from the same monocotyledon, Panicum virgatum, along with the results of the steady-state kinetics, spectroscopic/thermodynamic analyses, intermolecular interactions, and their effect on each catalytic step are proposed. In addition, PvCHI's unique activity for both naringenin chalcone and isoliquiritigenin was analyzed, and the observed hierarchical activity for those type-I and -II substrates was explained with the intrinsic characteristics of the enzyme and two substrates. The structure of PvCHS complexed with naringenin supports uncompetitive inhibition. PvCHS displays intrinsic catalytic promiscuity, evident from the formation of p-coumaroyltriacetic acid lactone (CTAL) in addition to naringenin chalcone. In the presence of PvCHIL, conversion of p-coumaroyl-CoA to naringenin through PvCHS and PvCHI displayed ~400-fold increased Vmax with reduced formation of CTAL by 70%. Supporting this model, molecular docking, ITC (Isothermal Titration Calorimetry), and FRET (Fluorescence Resonance Energy Transfer) indicated that both PvCHI and PvCHIL interact with PvCHS in a non-competitive manner, indicating the plausible allosteric effect of naringenin on CHS. Significantly, the presence of naringenin increased the affinity between PvCHS and PvCHIL, whereas naringenin chalcone decreased the affinity, indicating a plausible feedback mechanism to minimize spontaneous incorrect stereoisomers. These are the first findings from a three-body system from the same species, indicating the importance of the macromolecular assembly of CHS-CHI-CHIL in determining the amount and type of flavonoids produced in plant cells.


Subject(s)
Acyltransferases , Intramolecular Lyases , Intramolecular Lyases/metabolism , Intramolecular Lyases/chemistry , Acyltransferases/metabolism , Acyltransferases/chemistry , Plant Proteins/metabolism , Plant Proteins/chemistry , Flavonoids/metabolism , Flavonoids/chemistry , Kinetics , Flavanones/chemistry , Flavanones/metabolism , Chalcones/chemistry , Chalcones/metabolism , Substrate Specificity , Crystallography, X-Ray , Molecular Docking Simulation , Models, Molecular , Protein Binding , Protein Conformation
13.
Biochemistry ; 62(1): 17-21, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36512613

ABSTRACT

Acyltransferase (AT) recognizes its cognate acyl carrier protein (ACP) for functional transfer of an acyl unit in polyketide biosynthesis. However, structural characterization of AT-ACP complexes is limited because of the weak and transient interactions between them. In the biosynthesis of macrolactam polyketide vicenistatin, the trans-acting loading AT VinK transfers a dipeptidyl unit from the stand-alone ACP VinL to the ACP domain (VinP1ACPL) of the loading module of modular polyketide synthase VinP1. Although the previously determined structure of the VinK-VinL complex clearly illustrates the VinL recognition mechanism of VinK, how VinK recognizes VinP1ACPL remains unclear. Here, the crystal structure of a covalent VinK-VinP1ACPL complex formed with a pantetheine-type cross-linking probe is reported at 3.0 Å resolution. The structure of the VinK-VinP1ACPL complex provides detailed insights into the transient interactions between VinK and VinP1ACPL. The importance of residues in the binding interface was confirmed by site-directed mutational analyses. The binding interface between VinK and VinP1ACPL is similar to that between VinK and VinL, although some of the interface residues are different. However, the ACP orientation and interaction mode observed in the VinK-VinP1ACPL complex are different from those observed in other AT-ACP complexes such as the disorazole trans-AT-ACP complex and cis-AT-ACP complexes of modular polyketide synthases. Thus, AT-ACP binding interface interactions are different in each type of AT-ACP pair.


Subject(s)
Polyketide Synthases , Polyketides , Polyketide Synthases/chemistry , Acyltransferases/chemistry , Acyl Carrier Protein/metabolism
14.
Biochemistry ; 62(11): 1589-1593, 2023 06 06.
Article in English | MEDLINE | ID: mdl-37184546

ABSTRACT

Fragment antigen-binding domains of antibodies (Fabs) are powerful probes of structure-function relationships of assembly line polyketide synthases (PKSs). We report the discovery and characterization of Fabs interrogating the structure and function of the ketosynthase-acyltransferase (KS-AT) core of Module 2 of the 6-deoxyerythronolide B synthase (DEBS). Two Fabs (AC2 and BB1) were identified to potently inhibit the catalytic activity of Module 2. Both AC2 and BB1 were found to modulate ACP-mediated reactions catalyzed by this module, albeit by distinct mechanisms. AC2 primarily affects the rate (kcat), whereas BB1 increases the KM of an ACP-mediated reaction. A third Fab, AA5, binds to the KS-AT fragment of DEBS Module 2 without altering either parameter; it is phenotypically reminiscent of a previously characterized Fab, 1B2, shown to principally recognize the N-terminal helical docking domain of DEBS Module 3. Crystal structures of AA5 and 1B2 bound to the KS-AT fragment of Module 2 were solved to 2.70 and 2.65 Å resolution, respectively, and revealed entirely distinct recognition features of the two antibodies. The new tools and insights reported here pave the way toward advancing our understanding of the structure-function relationships of DEBS Module 2, arguably the most well-studied module of an assembly line PKS.


Subject(s)
Erythromycin , Polyketide Synthases , Polyketide Synthases/chemistry , Acyltransferases/chemistry , Antibodies
15.
J Biol Chem ; 298(8): 102203, 2022 08.
Article in English | MEDLINE | ID: mdl-35764173

ABSTRACT

Lipoic acid is a sulfur-containing cofactor indispensable for the function of several metabolic enzymes. In microorganisms, lipoic acid can be salvaged from the surroundings by lipoate protein ligase A (LplA), an ATP-dependent enzyme. Alternatively, it can be synthesized by the sequential actions of lipoate protein ligase B (LipB) and lipoyl synthase (LipA). LipB takes up the octanoyl chain from C8-acyl carrier protein (C8-ACP), a byproduct of the type II fatty acid synthesis pathway, and transfers it to a conserved lysine of the lipoyl domain of a dehydrogenase. However, the molecular basis of its substrate recognition is still not fully understood. Using Escherichia coli LipB as a model enzyme, we show here that the octanoyl-transferase mainly recognizes the 4'-phosphopantetheine-tethered acyl-chain of its donor substrate and weakly binds the apo-acyl carrier protein. We demonstrate LipB can accept octanoate from its own ACP and noncognate ACPs, as well as C8-CoA. Furthermore, our 1H saturation transfer difference and 31P NMR studies demonstrate the binding of adenosine, as well as the phosphopantetheine arm of CoA to LipB, akin to binding to LplA. Finally, we show a conserved 71RGG73 loop, analogous to the lipoate-binding loop of LplA, is required for full LipB activity. Collectively, our studies highlight commonalities between LipB and LplA in their mechanism of substrate recognition. This knowledge could be of significance in the treatment of mitochondrial fatty acid synthesis related disorders.


Subject(s)
Acyltransferases/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/enzymology , Acyl Carrier Protein/metabolism , Acyltransferases/metabolism , Coenzyme A/metabolism , Escherichia coli/chemistry , Escherichia coli Proteins/metabolism , Ligases/metabolism , Pantetheine/analogs & derivatives , Thioctic Acid/metabolism
16.
J Am Chem Soc ; 145(48): 26308-26317, 2023 12 06.
Article in English | MEDLINE | ID: mdl-37983668

ABSTRACT

Friedel-Crafts acylation (FCA) is a highly beneficial approach in organic chemistry for creating the important C-C bonds that are necessary for building intricate frameworks between aromatic substrates and an acyl group. However, there are few reports about enzyme catalyzed FCA reactions. In this study, 4-acyl-5-aminoimidazole alkaloids (AAIAs), streptimidazoles A-C (1-3), and the enantiopure (+)-nocarimidazole C (4) as well as their ribosides, streptimidazolesides A-D (5-8), were identified from the fermentation broth of Streptomyces sp. OUCMDZ-944 or heterologous S. coelicolor M1154 mutant. The biosynthetic gene cluster (smz) was identified, and the biosynthetic pathway of AAIAs was elucidated for the first time. In vivo and in vitro studies proved the catalytic activity of the four essential genes smzB, -C, -E, and -F for AAIAs biosynthesis and clarified the biosynthetic process of the alkaloids. The ligase SmzE activates fatty acyl groups and connects them to the acyl carrier protein (ACP) holo-SmzF. Then, the acyl group is transferred onto the key residue Cys49 of SmzB, a new Friedel-Crafts acyltransferase (FCase). Subsequently, the FCA reaction between the acyl groups and 5-aminoimidazole ribonucleotide (AIR) occurs to generate the key intermediate AAIA-nucleotides catalyzed by SmzB. Finally, the hydrolase SmzC catalyzes the N-glycosidic bond cleavage of the intermediates to form AAIAs. Structural simulation, molecular modeling, and mutational analysis of SmzB showed that Tyr26, Cys49, and Tyr93 are the key catalytic residues in the C-C bond formation of the acyl chain of AAIAs, providing mechanistic insights into the enzymatic FCA reaction.


Subject(s)
Acyltransferases , Imidazoles , Acyltransferases/chemistry , Acyl Carrier Protein/chemistry , Catalysis
17.
J Am Chem Soc ; 145(16): 8822-8832, 2023 04 26.
Article in English | MEDLINE | ID: mdl-37057992

ABSTRACT

Modular polyketide synthases (PKSs) are polymerases that employ α-carboxyacyl-CoAs as extender substrates. This enzyme family contains several catalytic modules, where each module is responsible for a single round of polyketide chain extension. Although PKS modules typically use malonyl-CoA or methylmalonyl-CoA for chain elongation, many other malonyl-CoA analogues are used to diversify polyketide structures in nature. Previously, we developed a method to alter an extension substrate of a given module by exchanging an acyltransferase (AT) domain while maintaining protein folding. Here, we report in vitro polyketide biosynthesis by 13 PKSs (the wild-type PKS and 12 AT-exchanged PKSs with unusual ATs) and 14 extender substrates. Our ∼200 in vitro reactions resulted in 13 structurally different polyketides, including several polyketides that have not been reported. In some cases, AT-exchanged PKSs produced target polyketides by >100-fold compared to the wild-type PKS. These data also indicate that most unusual AT domains do not incorporate malonyl-CoA and methylmalonyl-CoA but incorporate various rare extender substrates that are equal to in size or slightly larger than natural substrates. We developed a computational workflow to predict the approximate AT substrate range based on active site volumes to support the selection of ATs. These results greatly enhance our understanding of rare AT domains and demonstrate the benefit of using the proposed PKS engineering strategy to produce novel chemicals in vitro.


Subject(s)
Polyketide Synthases , Polyketides , Polyketide Synthases/metabolism , Acyltransferases/chemistry , Catalytic Domain , Polyketides/metabolism , Substrate Specificity
18.
Biochemistry ; 61(10): 922-932, 2022 05 17.
Article in English | MEDLINE | ID: mdl-35507417

ABSTRACT

The biosynthesis of wax esters and triglycerides in bacteria is accomplished through the action of the wax ester synthase/acyl-coenzyme A:diacylglycerol acyltransferase (WS/DGAT or wax ester synthase). A hallmark of these enzymes is the broad substrate profile that accepts alcohols, diglycerides, and fatty acyl-CoAs of various carbon chain lengths and degrees of branching. These enzymes have a broad biotechnological potential due to their role in producing high-value lipids or simple fuels similar to biodiesel through biosynthetic routes. Recently, a crystal structure was solved for the wax ester synthase from Marinobacter aquaeolei VT8 (Maqu_0168), providing a much clearer picture of the architecture of this enzyme and enabling a more precise analysis of the important structural features of the protein. In this work, we used the structure to canvas amino acids lining the proposed substrate-binding pockets and tested the effects of exchanging specific residues on the substrate profiles. We also developed an approach to better probe the residues that alter fatty acyl-CoA selectivity, which has proven more difficult to investigate. Our findings provide an improved blueprint for future efforts to understand how these enzymes position substrates for catalysis and to tailor or improve these enzymes in future biosynthetic schemes.


Subject(s)
Acyltransferases , Waxes , Acyl Coenzyme A/metabolism , Acyltransferases/chemistry , Diacylglycerol O-Acyltransferase/metabolism , Esters/chemistry , Substrate Specificity , Waxes/metabolism
19.
Biochemistry ; 61(13): 1273-1285, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35730892

ABSTRACT

Glycosylphosphatidylinositol (GPI) anchoring of proteins is a eukaryotic, post-translational modification catalyzed by GPI transamidase (GPI-T). The Saccharomyces cerevisiae GPI-T is composed of five membrane-bound subunits: Gpi8, Gaa1, Gpi16, Gpi17, and Gab1. GPI-T has been recalcitrant to in vitro structure and function studies because of its complexity and membrane-solubility. Furthermore, a reliable, quantitative, in vitro assay for this important post-translational modification has remained elusive despite its discovery more than three decades ago.Three recent reports describe the structure of GPI-T from S. cerevisiae and humans, shedding critical light on this important enzyme and offering insight into the functions of its different subunits. Here, we present the purification and characterization of a truncated soluble GPI-T heterotrimer complex (Gpi823-306, Gaa150-343, and Gpi1620-551) without transmembrane domains. Using this simplified heterotrimer, we report the first quantitative method to measure GPI-T activity in vitro and demonstrate that this soluble, minimalistic GPI-T retains transamidase activity. These results contribute to our understanding of how this enzyme is organized and functions, and provide a method to screen potential GPI-T inhibitors.


Subject(s)
Acyltransferases , Saccharomyces cerevisiae Proteins , Acyltransferases/chemistry , Acyltransferases/metabolism , Glycosylphosphatidylinositols , Humans , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae Proteins/metabolism
20.
J Biol Chem ; 296: 100140, 2021.
Article in English | MEDLINE | ID: mdl-33268381

ABSTRACT

Endophilin plays key roles during endocytosis of cellular receptors, including generating membrane curvature to drive internalization. Electrostatic interactions between endophilin's BIN/Amphiphysin/Rvs domain and anionic membrane lipids have been considered the major driving force in curvature generation. However, the SH3 domain of endophilin also interacts with the proline-rich third intracellular loop (TIL) of various G-protein-coupled receptors (GPCRs), and it is unclear whether this interaction has a direct role in generating membrane curvature during endocytosis. To examine this, we designed model membranes with a membrane density of 1400 receptors per µm2 represented by a covalently conjugated TIL region from the ß1-adrenergic receptor. We observed that TIL recruits endophilin to membranes composed of 95 mol% of zwitterionic lipids via the SH3 domain. More importantly, endophilin recruited via TIL tubulates vesicles and gets sorted onto highly curved membrane tubules. These observations indicate that the cellular membrane bending and curvature sensing activities of endophilin can be facilitated through detection of the TIL of activated GPCRs in addition to binding to anionic lipids. Furthermore, we show that TIL electrostatically interacts with membranes composed of anionic lipids. Therefore, anionic lipids can modulate TIL/SH3 domain binding. Overall, our findings imply that an interplay between TIL, charged membrane lipids, BAR domain, and SH3 domain could exist in the biological system and that these components may act in coordination to regulate the internalization of cellular receptors.


Subject(s)
Acyltransferases/metabolism , Cell Membrane/metabolism , Endocytosis , Lipids/chemistry , Proline-Rich Protein Domains , Receptors, Adrenergic, beta/metabolism , Acyltransferases/chemistry , Acyltransferases/genetics , Humans , Protein Interaction Domains and Motifs , Protein Transport , Receptors, Adrenergic, beta/genetics , src Homology Domains
SELECTION OF CITATIONS
SEARCH DETAIL