Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 508
Filtrar
1.
J Am Soc Mass Spectrom ; 34(11): 2567-2574, 2023 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-37812744

RESUMO

Several analytical challenges make it difficult to accurately measure coenzyme A (CoA) metaboforms, including insufficient stability and a lack of available metabolite standards. Consequently, our understanding of CoA biology and the modulation of human diseases may be nascent. CoA's serve as lipid precursors, energy intermediates, and mediators of post-translational modifications of proteins. Here, we present a liquid chromatography-mass spectrometry (LC-MS) approach to measure malonyl-CoA, acetyl-CoA, and succinyl-CoA in complex biological samples. Additionally, we evaluated workflows to increase sample stability. We used reference standards to optimize CoA assay sensitivity and test CoA metabolite stability as a function of the reconstitution solvent. We show that using glass instead of plastic sample vials decreases CoA signal loss and improves the sample stability. We identify additives that improve CoA stability and facilitate accurate analysis of CoA species across large sample sets. We apply our optimized workflow to biological samples of skeletal muscle cells cultured under hypoxic and normoxia conditions. Together, our workflow improves the detection and identification of CoA species through targeted analysis in complex biological samples.


Assuntos
Acil Coenzima A , Malonil Coenzima A , Humanos , Malonil Coenzima A/metabolismo , Acetilcoenzima A/metabolismo , Acil Coenzima A/química , Acil Coenzima A/metabolismo , Células Musculares/química , Células Musculares/metabolismo
2.
Curr Opin Struct Biol ; 82: 102671, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37542911

RESUMO

The crotonase fold is generated by a framework of four repeats of a ßßα-unit, extended by two helical regions. The active site of crotonase superfamily (CS) enzymes is located at the N-terminal end of the helix of the third repeat, typically being covered by a C-terminal helix. A major subset of CS-enzymes catalyzes acyl-CoA-dependent reactions, allowing for a diverse range of acyl-tail modifications. Most of these enzymes occur as trimers or hexamers (dimers of trimers), but monomeric forms are also observed. A common feature of the active sites of CS-enzymes is an oxyanion hole, formed by two peptide-NH hydrogen bond donors, which stabilises the negatively charged thioester oxygen atom of the reaction intermediate. Structural properties and possible use of these enzymes for biotechnological applications are discussed.


Assuntos
Acil Coenzima A , Enoil-CoA Hidratase , Enoil-CoA Hidratase/química , Enoil-CoA Hidratase/metabolismo , Acil Coenzima A/química , Acil Coenzima A/metabolismo , Domínio Catalítico , Sítios de Ligação , Cristalografia por Raios X
3.
Drug Metab Pharmacokinet ; 52: 100509, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37515836

RESUMO

Although acyl-CoA conjugates are known to have higher reactivity than acyl glucuronides, few studies have been conducted to evaluate the risk of the conjugates. In the present study, we aimed to develop a trapping assay for acyl-CoA conjugates using trapping reagents we have developed previously. It was revealed that Cys-Dan, which has both a thiol and an amino group, was the most effective in forming stable adducts containing an amide bond after intramolecular acyl migration. Additionally, we also developed a hepatocyte-based trapping assay in the present study to overcome the shortcomings of liver microsomes. Although liver microsomes are commonly used as enzyme sources in trapping assays, they lack some of the enzymes required for drug metabolism and detoxification systems. In human hepatocytes, our three trapping reagents, CysGlu-Dan, Dap-Dan and Cys-Dan, captured CYP-dependent reactive metabolites, reactive acyl glucuronides, and reactive acyl-CoA conjugates, respectively. The work suggests that the trapping assay with the reagents in hepatocytes is useful to evaluate the risk of reactive metabolites in drug discovery.


Assuntos
Acil Coenzima A , Glucuronídeos , Humanos , Acil Coenzima A/química , Acil Coenzima A/metabolismo , Acil Coenzima A/farmacologia , Indicadores e Reagentes/metabolismo , Glucuronídeos/metabolismo , Microssomos Hepáticos/metabolismo , Compostos de Sulfidrila/metabolismo , Compostos de Sulfidrila/farmacologia
4.
Int J Biol Macromol ; 226: 608-617, 2023 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-36521700

RESUMO

Vanillin (3-methoxy-4-hydroxybenzaldehyde) is one of the most important flavoring substances used in the cosmetic and food industries. Feruloyl-CoA hydratase/lyase (FCHL) is an enzyme that catalyzes the production of vanillin from feruloyl-CoA. In this study, we report kinetic parameters and biochemical properties of FCHL from Sphingomonas paucimobilis SYK-6 (SpFCHL). Also, the crystal structures of an apo-form of SpFCHL and two complexed forms with acetyl-CoA and vanillin/CoA was present. Comparing the apo structure to its complexed forms of SpFCHL, a gate loop with an "open and closed" role was observed at the entrance of the substrate-binding site. With vanillin and CoA complexed to SpFCHL, we captured a conformational change in the feruloyl moiety-binding pocket that repositions the catalytic SpFCHLE146 and other key residues. This binding pocket does not tightly fit the vanillin structure, suggesting substrate promiscuity of this enzyme. This observation is in good agreement with assay results for phenylpropanoid-CoAs and indicates important physicochemical properties of the substrate for the hydratase/lyase reaction mechanism. In addition, we showed that various phenolic aldehydes could be produced using the 4CL-FCHL biosynthesis platform.


Assuntos
Liases , Aldeídos , Acil Coenzima A/química
5.
Chem Res Toxicol ; 35(10): 1777-1788, 2022 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-36200746

RESUMO

Glucuronidation and CoA (coenzyme A) conjugation are common pathways for the elimination of carboxylic acid-containing drug molecules. In some instances, these biotransformations have been associated with toxicity (such as idiosyncratic hepatic injury, renal impairment, hemolytic anemia, gastrointestinal inflammation, and bladder cancer) attributed to, in part, the propensity of acyl glucuronides and acyl CoA thioesters to covalently modify biological macromolecules such as proteins and DNA. It is to be noted that, while acyl glucuronidation and CoA conjugation are indeed implicated in adverse effects, there are many safe drugs in the market that are cleared by these reactive pathways. It is therefore important that new molecular entities with carboxylic acid groups are evaluated for toxicity in a manner that is not unreasonably risk-averse. In the absence of truly predictable methods, therefore, the general approach is to apply a set of end points to generate a weight-of-evidence evaluation. In practice, the focus is to identify structural liabilities and provide structure-activity recommendations early in the program, at a stage where an attempt to improve reactive metabolism does not deoptimize other critical drug-quality criteria. This review will present a high-level overview of the chemistry of glucuronidation and CoA conjugation and provide a discussion of the possible mechanisms of adverse effects that have been associated with these pathways, as well as how such potential hazards are addressed while delivering a new chemical entity for clinical evaluation.


Assuntos
Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Glucuronídeos , Acil Coenzima A/química , Acil Coenzima A/metabolismo , Ácidos Carboxílicos/química , Coenzima A , Glucuronídeos/metabolismo , Humanos , Proteínas/metabolismo
6.
Curr Opin Struct Biol ; 77: 102463, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36183446

RESUMO

S-acylation is a reversible posttranslational modification, where a long-chain fatty acid is attached to a protein through a thioester linkage. Being the most abundant form of lipidation in humans, a family of twenty-three human zDHHC integral membrane enzymes catalyze this reaction. Previous structures of the apo and lipid bound zDHHCs shed light into the molecular details of the active site and binding pocket. Here, we delve further into the details of fatty acyl-CoA recognition by zDHHC acyltransferases using insights from the recent structure. We additionally review indirect evidence that suggests acyl-CoAs do not diffuse freely in the cytosol, but are channeled into specific pathways, and comment on the suggested mechanisms for fatty acyl-CoA compartmentalization and intracellular transport, to finally speculate about the potential mechanisms that underlie fatty acyl-CoA delivery to zDHHC enzymes.


Assuntos
Acetiltransferases , Acil Coenzima A , Aciltransferases , Humanos , Acetiltransferases/metabolismo , Acil Coenzima A/química , Acil Coenzima A/metabolismo , Acilação , Processamento de Proteína Pós-Traducional , Aciltransferases/química , Aciltransferases/metabolismo
7.
Proc Natl Acad Sci U S A ; 119(7)2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-35140179

RESUMO

S-acylation, also known as palmitoylation, is the most abundant form of protein lipidation in humans. This reversible posttranslational modification, which targets thousands of proteins, is catalyzed by 23 members of the DHHC family of integral membrane enzymes. DHHC enzymes use fatty acyl-CoA as the ubiquitous fatty acyl donor and become autoacylated at a catalytic cysteine; this intermediate subsequently transfers the fatty acyl group to a cysteine in the target protein. Protein S-acylation intersects with almost all areas of human physiology, and several DHHC enzymes are considered as possible therapeutic targets against diseases such as cancer. These efforts would greatly benefit from a detailed understanding of the molecular basis for this crucial enzymatic reaction. Here, we combine X-ray crystallography with all-atom molecular dynamics simulations to elucidate the structure of the precatalytic complex of human DHHC20 in complex with palmitoyl CoA. The resulting structure reveals that the fatty acyl chain inserts into a hydrophobic pocket within the transmembrane spanning region of the protein, whereas the CoA headgroup is recognized by the cytosolic domain through polar and ionic interactions. Biochemical experiments corroborate the predictions from our structural model. We show, using both computational and experimental analyses, that palmitoyl CoA acts as a bivalent ligand where the interaction of the DHHC enzyme with both the fatty acyl chain and the CoA headgroup is important for catalytic chemistry to proceed. This bivalency explains how, in the presence of high concentrations of free CoA under physiological conditions, DHHC enzymes can efficiently use palmitoyl CoA as a substrate for autoacylation.


Assuntos
Acil Coenzima A/química , Acil Coenzima A/metabolismo , Aciltransferases/metabolismo , Aciltransferases/genética , Domínio Catalítico , Membrana Celular/enzimologia , Regulação Enzimológica da Expressão Gênica , Humanos , Lipoilação , Modelos Moleculares , Simulação de Dinâmica Molecular , Mutação , Ligação Proteica , Conformação Proteica , Domínios Proteicos
8.
Xenobiotica ; 52(1): 16-25, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35084285

RESUMO

Some drugs with carboxylic acid moieties can potentially cause rare but severe hepatotoxicity. The reactive chemical species generated by drug metabolism are thought to be one reason for this event. Although the phase II conjugation metabolism of carboxylic acids generally renders a compound more polar and inactive, it is also responsible for the formation of reactive metabolites.This study aimed to provide a new approach towards the risk assessment of carboxylic acids in the aspect of reactive acyl CoA metabolites.Although acyl CoA metabolites have been concerned, it is difficult to detect them because of their instability. We investigated the trapping agents for acyl CoA metabolites. We found that cysteine is a good trapping agent and developed an assay method for the reactivity of acyl CoA metabolites. We evaluated 17 drugs with carboxylic acid moieties, all drugs concerned with hepatotoxicity displayed reactive potential. With consideration of the exposure of each parent drug, the correlation between drug labels and the calculated risk of carboxylic drugs was improved.These evaluations can be conducted without radiochemical reagents or the authentic standards of metabolites. We believe that the method will be beneficial for drug discovery.


Assuntos
Acil Coenzima A , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Acil Coenzima A/química , Acil Coenzima A/metabolismo , Ácidos Carboxílicos/metabolismo , Cisteína , Humanos , Medição de Risco
9.
Biochem Biophys Res Commun ; 594: 8-14, 2022 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-35066379

RESUMO

Lignin is a highly complex phenolic polymer which is essential for plants, but also makes it difficult for industrial processing. Engineering lignin by introducing relatively labile linkages into the lignin backbone can render it more amenable to chemical depolymerization. It has been reported that introducing a feruloyl-coenzyme A monolignol transferase from Angelica sinensis (AsFMT) into poplar could incorporate monolignol ferulate conjugates (ML-FAs) into lignin polymers, suggesting a promising way to manipulate plants for readily deconstructing. FMT catalyzes a reaction between monolignols and feruloyl-CoA to produce ML-FAs and free CoA-SH. However, the mechanisms of substrate specificity and catalytic process of FMT remains poorly understood. Here we report the structure of AsFMT, which adopts a typical fold of BAHD acyltransferase family. Structural comparisons with other BAHD homologs reveal several unique structural features of AsFMT, different from those of the BAHD homologs. Further molecular docking studies showed that T375 in AsFMT may function as an oxyanion hole to stabilize the reaction intermediate and also proposed a role of H278 in the binding of the nucleophilic hydroxyl group of monolignols. Together, this study provides important structural insights into the reactions catalyzed by AsFMT and will shed light on its future application in lignin engineering.


Assuntos
Acil Coenzima A/química , Aldeído Oxirredutases/química , Angelica/enzimologia , Oxirredutases/química , Catálise , Domínio Catalítico , Cristalografia por Raios X , Lignina/química , Simulação de Acoplamento Molecular , Ligação Proteica , Conformação Proteica , Domínios Proteicos , Especificidade por Substrato , Transferases/metabolismo , Ultracentrifugação
10.
J Biol Chem ; 298(1): 101522, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34952003

RESUMO

Actinobacterial 2-hydroxyacyl-CoA lyase reversibly catalyzes the thiamine diphosphate-dependent cleavage of 2-hydroxyisobutyryl-CoA to formyl-CoA and acetone. This enzyme has great potential for use in synthetic one-carbon assimilation pathways for sustainable production of chemicals, but lacks details of substrate binding and reaction mechanism for biochemical reengineering. We determined crystal structures of the tetrameric enzyme in the closed conformation with bound substrate, covalent postcleavage intermediate, and products, shedding light on active site architecture and substrate interactions. Together with molecular dynamics simulations of the covalent precleavage complex, the complete catalytic cycle is structurally portrayed, revealing a proton transfer from the substrate acyl Cß hydroxyl to residue E493 that returns it subsequently to the postcleavage Cα-carbanion intermediate. Kinetic parameters obtained for mutants E493A, E493Q, and E493K confirm the catalytic role of E493 in the WT enzyme. However, the 10- and 50-fold reduction in lyase activity in the E493A and E493Q mutants, respectively, compared with WT suggests that water molecules may contribute to proton transfer. The putative catalytic glutamate is located on a short α-helix close to the active site. This structural feature appears to be conserved in related lyases, such as human 2-hydroxyacyl-CoA lyase 2. Interestingly, a unique feature of the actinobacterial 2-hydroxyacyl-CoA lyase is a large C-terminal lid domain that, together with active site residues L127 and I492, restricts substrate size to ≤C5 2-hydroxyacyl residues. These details about the catalytic mechanism and determinants of substrate specificity pave the ground for designing tailored catalysts for acyloin condensations for one-carbon and short-chain substrates in biotechnological applications.


Assuntos
Acil Coenzima A , Liases , Acil Coenzima A/química , Acil Coenzima A/metabolismo , Carbono , Catálise , Domínio Catalítico , Cristalografia por Raios X , Humanos , Liases/química , Liases/metabolismo , Prótons , Relação Estrutura-Atividade , Especificidade por Substrato
11.
FEBS J ; 289(4): 1023-1042, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34601806

RESUMO

Anaerobic toluene degradation proceeds by fumarate addition to produce (R)-benzylsuccinate as first intermediate, which is further degraded via ß-oxidation by five enzymes encoded in the conserved bbs operon. This study characterizes two enzymes of this pathway, (E)-benzylidenesuccinyl-CoA hydratase (BbsH), and (S,R)-2-(α-hydroxybenzyl)succinyl-CoA dehydrogenase (BbsCD) from Thauera aromatica. BbsH, a member of the enoyl-CoA hydratase family, converts (E)-benzylidenesuccinyl-CoA to 2-(α-hydroxybenzyl)succinyl-CoA and was subsequently used in a coupled enzyme assay with BbsCD, which belongs to the short-chain dehydrogenases/reductase (SDR) family. The BbsCD crystal structure shows a C2-symmetric heterotetramer consisting of BbsC2 and BbsD2 dimers. BbsD subunits are catalytically active and capable of binding NAD+ and substrate, whereas BbsC subunits represent built-in pseudoenzyme moieties lacking all motifs of the SDR family required for substrate binding or catalysis. Molecular modeling studies predict that the active site of BbsD is specific for conversion of the (S,R)-diastereomer of 2-(α-hydroxybenzyl)succinyl-CoA to (S)-2-benzoylsuccinyl-CoA by hydride transfer to the re-face of nicotinamide adenine dinucleotide (NAD)+ . Furthermore, BbsC subunits are not engaged in substrate binding and merely serve as scaffold for the BbsD dimer. BbsCD represents a novel clade of related enzymes within the SDR family, which adopt a heterotetrameric architecture and catalyze the ß-oxidation of aromatic succinate adducts.


Assuntos
Redutases-Desidrogenases de Cadeia Curta/metabolismo , Thauera/enzimologia , Tolueno/metabolismo , Acil Coenzima A/biossíntese , Acil Coenzima A/química , Biocatálise , Modelos Moleculares , Estrutura Molecular , Succinatos/química , Succinatos/metabolismo , Compostos de Sulfidrila/química , Compostos de Sulfidrila/metabolismo , Tolueno/química
12.
Nat Commun ; 12(1): 6869, 2021 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-34824256

RESUMO

As the major component of cell membranes, phosphatidylcholine (PC) is synthesized de novo in the Kennedy pathway and then undergoes extensive deacylation-reacylation remodeling via Lands' cycle. The re-acylation is catalyzed by lysophosphatidylcholine acyltransferase (LPCAT) and among the four LPCAT members in human, the LPCAT3 preferentially introduces polyunsaturated acyl onto the sn-2 position of lysophosphatidylcholine, thereby modulating the membrane fluidity and membrane protein functions therein. Combining the x-ray crystallography and the cryo-electron microscopy, we determined the structures of LPCAT3 in apo-, acyl donor-bound, and acyl receptor-bound states. A reaction chamber was revealed in the LPCAT3 structure where the lysophosphatidylcholine and arachidonoyl-CoA were positioned in two tunnels connected near to the catalytic center. A side pocket was found expanding the tunnel for the arachidonoyl CoA and holding the main body of arachidonoyl. The structural and functional analysis provides the basis for the re-acylation of lysophosphatidylcholine and the substrate preference during the reactions.


Assuntos
1-Acilglicerofosfocolina O-Aciltransferase/química , Fosfolipídeos/química , 1-Acilglicerofosfocolina O-Aciltransferase/metabolismo , Acil Coenzima A/química , Acil Coenzima A/metabolismo , Acilação , Animais , Domínio Catalítico , Galinhas , Microscopia Crioeletrônica , Cristalografia por Raios X , Lisofosfatidilcolinas/química , Lisofosfatidilcolinas/metabolismo , Modelos Moleculares , Fosfolipídeos/metabolismo , Multimerização Proteica , Relação Estrutura-Atividade , Especificidade por Substrato
13.
Sci Rep ; 11(1): 20842, 2021 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-34675283

RESUMO

3-Hydroxy-3-methylglutaryl-coenzyme A reductase (HMGR) is a crucial enzyme in the ergosterol biosynthesis pathway. The aim of this study was to obtain, purify, characterize, and overexpress five point mutations in highly conserved regions of the catalytic domain of Candida glabrata HMGR (CgHMGR) to explore the function of key amino acid residues in enzymatic activity. Glutamic acid (Glu) was substituted by glutamine in the E680Q mutant (at the dimerization site), Glu by glutamine in E711Q (at the substrate binding site), aspartic acid by alanine in D805A, and methionine by arginine in M807R (the latter two at the cofactor binding site). A double mutation, E680Q-M807R, was included. Regarding recombinant and wild-type CgHMGR, in vitro enzymatic activity was significantly lower for the former, as was the in silico binding energy of simvastatin, alpha-asarone and the HMG-CoA substrate. E711Q displayed the lowest enzymatic activity and binding energy, suggesting the importance of Glu711 (in the substrate binding site). The double mutant CgHMGR E680Q-M807R exhibited the second lowest enzymatic activity. Based on the values of the kinetic parameters KM and Vmax, the mutated amino acids appear to participate in binding. The current findings provide insights into the role of residues in the catalytic site of CgHMGR.


Assuntos
Acil Coenzima A/genética , Candida glabrata/genética , Proteínas Fúngicas/genética , Mutação Puntual , Acil Coenzima A/química , Acil Coenzima A/metabolismo , Sítios de Ligação , Candida glabrata/química , Candida glabrata/metabolismo , Domínio Catalítico , Proteínas Fúngicas/química , Proteínas Fúngicas/metabolismo , Modelos Moleculares , Filogenia , Especificidade por Substrato
14.
Biochem J ; 478(15): 3047-3062, 2021 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-34338286

RESUMO

Activation of enzymes by monovalent cations (M+) is a widespread phenomenon in biology. Despite this, there are few structure-based studies describing the underlying molecular details. Thiolases are a ubiquitous and highly conserved family of enzymes containing both K+-activated and K+-independent members. Guided by structures of naturally occurring K+-activated thiolases, we have used a structure-based approach to engineer K+-activation into a K+-independent thiolase. To our knowledge, this is the first demonstration of engineering K+-activation into an enzyme, showing the malleability of proteins to accommodate M+ ions as allosteric regulators. We show that a few protein structural features encode K+-activation in this class of enzyme. Specifically, two residues near the substrate-binding site are sufficient for K+-activation: A tyrosine residue is required to complete the K+ coordination sphere, and a glutamate residue provides a compensating charge for the bound K+ ion. Further to these, a distal residue is important for positioning a K+-coordinating water molecule that forms a direct hydrogen bond to the substrate. The stability of a cation-π interaction between a positively charged residue and the substrate is determined by the conformation of the loop surrounding the substrate-binding site. Our results suggest that this cation-π interaction effectively overrides K+-activation, and is, therefore, destabilised in K+-activated thiolases. Evolutionary conservation of these amino acids provides a promising signature sequence for predicting K+-activation in thiolases. Together, our structural, biochemical and bioinformatic work provide important mechanistic insights into how enzymes can be allosterically activated by M+ ions.


Assuntos
Acetil-CoA C-Acetiltransferase/metabolismo , Proteínas de Bactérias/metabolismo , Cátions Monovalentes/metabolismo , Ativação Enzimática , Potássio/metabolismo , Zoogloea/isolamento & purificação , Acetilcoenzima A/química , Acetilcoenzima A/metabolismo , Acetil-CoA C-Acetiltransferase/química , Acetil-CoA C-Acetiltransferase/genética , Acil Coenzima A/química , Acil Coenzima A/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Biocatálise , Cátions Monovalentes/química , Cristalografia por Raios X , Cinética , Modelos Moleculares , Mutação , Potássio/química , Ligação Proteica , Conformação Proteica , Engenharia de Proteínas , Multimerização Proteica , Especificidade por Substrato , Zoogloea/enzimologia , Zoogloea/genética
15.
Angew Chem Int Ed Engl ; 60(25): 13996-14004, 2021 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-33837610

RESUMO

Despite increasing awareness of the biological impacts of long-chain fatty acyl-CoA esters (LCACoAs), our knowledge about the subcellular distribution and regulatory functions of these acyl-CoA molecules is limited by a lack of methods for detecting LCACoAs in living cells. Here, we report development of a genetically encoded fluorescent sensor that enables ratiometric quantification of LCACoAs in living cells and subcellular compartments. We demonstrate how this FadR-cpYFP fusion "LACSer sensor" undergoes LCACoA-induced conformational changes reflected in easily detectable fluorescence responses, and show proof-of-concept for real-time monitoring of LCACoAs in human cells. Subsequently, we applied LACSer in scientific studies investigating how disruption of ACSL enzymes differentially reduces cytosolic and mitochondrial LCACoA levels, and show how genetic disruption of an acyl-CoA binding protein (ACBP) alters mitochondrial accumulation of LCACoAs.


Assuntos
Acil Coenzima A/metabolismo , Técnicas Biossensoriais , Ésteres/metabolismo , Fluorescência , Acil Coenzima A/química , Ésteres/química , Células HEK293 , Humanos
16.
FEBS J ; 288(19): 5768-5780, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33843134

RESUMO

Mycophenolic acid (MPA) is a fungal natural product and first-line immunosuppressive drug for organ transplantations and autoimmune diseases. In the compartmentalized biosynthesis of MPA, the acyl-coenzyme A (CoA) hydrolase MpaH' located in peroxisomes catalyzes the highly specific hydrolysis of MPA-CoA to produce the final product MPA. The strict substrate specificity of MpaH' not only averts undesired hydrolysis of various cellular acyl-CoAs, but also prevents MPA-CoA from further peroxisomal ß-oxidation catabolism. To elucidate the structural basis for this important property, in this study, we solve the crystal structures of the substrate-free form of MpaH' and the MpaH'S139A mutant in complex with the product MPA. The MpaH' structure reveals a canonical α/ß-hydrolase fold with an unusually large cap domain and a rare location of the acidic residue D163 of catalytic triad after strand ß6. MpaH' also forms an atypical dimer with the unique C-terminal helices α13 and α14 arming the cap domain of the other protomer and indirectly participating in the substrate binding. With these characteristics, we propose that MpaH' and its homologs form a new subfamily of α/ß hydrolase fold protein. The crystal structure of MpaH'S139A /MPA complex and the modeled structure of MpaH'/MPA-CoA, together with the structure-guided mutagenesis analysis and isothermal titration calorimetry (ITC) measurements, provide important mechanistic insights into the high substrate specificity of MpaH'.


Assuntos
Acil Coenzima A/química , Hidrolases/ultraestrutura , Ácido Micofenólico/metabolismo , Peroxissomos/ultraestrutura , Sequência de Aminoácidos/genética , Domínio Catalítico/genética , Hidrolases/química , Hidrolases/genética , Ácido Micofenólico/química , Penicillium/genética , Penicillium/ultraestrutura , Peroxissomos/enzimologia , Estrutura Secundária de Proteína/genética , Especificidade por Substrato/genética
17.
Arch Biochem Biophys ; 701: 108793, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33587905

RESUMO

We have undertaken a spectral deconvolution of the three FADs of EtfAB/bcd to the spectral changes seen in the course of reduction, including the spectrally distinct anionic and neutral semiquinone states of electron-transferring and bcd flavins. We also demonstrate that, unlike similar systems, no charge-transfer complex is observed on titration of the reduced M. elsdenii EtfAB with NAD+. Finally, and significantly, we find that removal of the et FAD from EtfAB results in an uncrossing of the half-potentials of the bifurcating FAD that remains in the protein, as reflected in the accumulation of substantial FAD•- in the course of reductive titrations of the depleted EtfAB with sodium dithionite.


Assuntos
Acil Coenzima A/química , Proteínas de Bactérias/química , Megasphaera elsdenii/enzimologia , NADH NADPH Oxirredutases/química , NAD/química , Acil Coenzima A/genética , Proteínas de Bactérias/genética , Megasphaera elsdenii/genética , NAD/genética , NADH NADPH Oxirredutases/genética , Oxirredução
18.
Nat Struct Mol Biol ; 28(3): 249-257, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33558762

RESUMO

Human serine palmitoyltransferase (SPT) complex catalyzes the initial and rate-limiting step in the de novo biosynthesis of all sphingolipids. ORMDLs regulate SPT function, with human ORMDL3 being related to asthma. Here we report three high-resolution cryo-EM structures: the human SPT complex, composed of SPTLC1, SPTLC2 and SPTssa; the SPT-ORMDL3 complex; and the SPT-ORMDL3 complex bound to two substrates, PLP-L-serine (PLS) and a non-reactive palmitoyl-CoA analogue. SPTLC1 and SPTLC2 form a dimer of heterodimers as the catalytic core. SPTssa participates in acyl-CoA coordination, thereby stimulating the SPT activity and regulating the substrate selectivity. ORMDL3 is located in the center of the complex, serving to stabilize the SPT assembly. Our structural and biochemical analyses provide a molecular basis for the assembly and substrate selectivity of the SPT and SPT-ORMDL3 complexes, and lay a foundation for mechanistic understanding of sphingolipid homeostasis and for related therapeutic drug development.


Assuntos
Microscopia Crioeletrônica , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Complexos Multiproteicos , Serina C-Palmitoiltransferase/química , Serina C-Palmitoiltransferase/metabolismo , Acil Coenzima A/química , Acil Coenzima A/metabolismo , Acil Coenzima A/ultraestrutura , Sítios de Ligação , Biocatálise , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/ultraestrutura , Modelos Moleculares , Complexos Multiproteicos/química , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Complexos Multiproteicos/ultraestrutura , Mutação , Fosfato de Piridoxal/química , Fosfato de Piridoxal/metabolismo , Reprodutibilidade dos Testes , Serina/química , Serina/metabolismo , Serina C-Palmitoiltransferase/genética , Serina C-Palmitoiltransferase/ultraestrutura , Especificidade por Substrato
19.
Biochemistry ; 60(5): 365-372, 2021 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-33482062

RESUMO

LnmK stereospecifically accepts (2R)-methylmalonyl-CoA, generating propionyl-S-acyl carrier protein to support polyketide biosynthesis. LnmK and its homologues are the only known enzymes that carry out a decarboxylation (DC) and acyl transfer (AT) reaction in the same active site as revealed by structure-function studies. Substrate-assisted catalysis powers LnmK, as decarboxylation of (2R)-methylmalonyl-CoA generates an enolate capable of deprotonating active site Tyr62, and the Tyr62 phenolate subsequently attacks propionyl-CoA leading to a propionyl-O-LnmK acyl-enzyme intermediate. Due to the inherent reactivity of LnmK and methylmalonyl-CoA, a substrate-bound structure could not be obtained. To gain insight into substrate specificity, stereospecificity, and catalytic mechanism, we determined the structures of LnmK with bound substrate analogues that bear malonyl-thioester isosteres where the carboxylate is represented by a nitro or sulfonate group. The nitro-bearing malonyl-thioester isosteres bind in the nitronate form, with specific hydrogen bonds that allow modeling of the (2R)-methylmalonyl-CoA substrate and rationalization of stereospecificity. The sulfonate isosteres bind in multiple conformations, suggesting the large active site of LnmK allows multiple binding modes. Considering the smaller malonyl group has more conformational freedom than the methylmalonyl group, we hypothesized the active site can entropically screen against catalysis with the smaller malonyl-CoA substrate. Indeed, our kinetic analysis reveals malonyl-CoA is accepted at 1% of the rate of methylmalonyl-CoA. This study represents another example of how our nitro- and sulfonate-bearing methylmalonyl-thioester isosteres are of use for elucidating enzyme-substrate binding interactions and revealing insights into catalytic mechanism. Synthesis of a larger panel of analogues presents an opportunity to study enzymes with complicated structure-function relationships such as acyl-CoA carboxylases, trans-carboxytransferases, malonyltransferases, and ß-ketoacylsynthases.


Assuntos
Aciltransferases/química , Carboxiliases/química , Proteína de Transporte de Acila/metabolismo , Acil Coenzima A/química , Carbono-Carbono Ligases/química , Catálise , Domínio Catalítico , Malonil Coenzima A/metabolismo , Streptomyces/metabolismo , Streptomyces coelicolor/metabolismo , Especificidade por Substrato
20.
FEBS J ; 288(3): 1008-1026, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32329961

RESUMO

The flavin-based electron bifurcation (FBEB) system from Acidaminococcus fermentans is composed of the electron transfer flavoprotein (EtfAB) and butyryl-CoA dehydrogenase (Bcd). α-FAD binds to domain II of the A-subunit of EtfAB, ß-FAD to the B-subunit of EtfAB and δ-FAD to Bcd. NADH reduces ß-FAD to ß-FADH- , which bifurcates one electron to the high potential α-FAD•- semiquinone followed by the other to the low potential ferredoxin (Fd). As deduced from crystal structures, upon interaction of EtfAB with Bcd, the formed α-FADH- approaches δ-FAD by rotation of domain II, yielding δ-FAD•- . Repetition of this process leads to a second reduced ferredoxin (Fd- ) and δ-FADH- , which reduces crotonyl-CoA to butyryl-CoA. In this study, we measured the redox properties of the components EtfAB, EtfaB (Etf without α-FAD), Bcd, and Fd, as well as of the complexes EtfaB:Bcd, EtfAB:Bcd, EtfaB:Fd, and EftAB:Fd. In agreement with the structural studies, we have shown for the first time that the interaction of EtfAB with Bcd drastically decreases the midpoint reduction potential of α-FAD to be within the same range of that of ß-FAD and to destabilize the semiquinone of α-FAD. This finding clearly explains that these interactions facilitate the passing of electrons from ß-FADH- via α-FAD•- to the final electron acceptor δ-FAD•- on Bcd. The interactions modulate the semiquinone stability of δ-FAD in an opposite way by having a greater semiquinone stability than in free Bcd.


Assuntos
Acidaminococcus/metabolismo , Proteínas de Bactérias/metabolismo , Benzoquinonas/metabolismo , Butiril-CoA Desidrogenase/metabolismo , Flavoproteínas Transferidoras de Elétrons/metabolismo , Flavinas/metabolismo , Acil Coenzima A/química , Acil Coenzima A/metabolismo , Proteínas de Bactérias/química , Benzoquinonas/química , Butiril-CoA Desidrogenase/química , Transporte de Elétrons , Flavoproteínas Transferidoras de Elétrons/química , Elétrons , Ferredoxinas/química , Ferredoxinas/metabolismo , Flavina-Adenina Dinucleotídeo/química , Flavina-Adenina Dinucleotídeo/metabolismo , Modelos Biológicos , Oxirredução , Ligação Proteica , Espectrofotometria
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...