Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 124
Filtrar
1.
J Agric Food Chem ; 72(30): 16900-16910, 2024 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-39016109

RESUMO

S-Adenosylmethionine (SAM) is a crucial metabolic intermediate playing irreplaceable roles in organismal activities. However, the synthesis of SAM by methionine adenosyltransferase (MAT) is hindered by low conversion due to severe product inhibition. Herein structure-guided semirational engineering was conducted on MAT from Escherichia coli (EcMAT) to mitigate the product inhibitory effect. Compared with the wild-type EcMAT, the best variant E56Q/Q105R exhibited an 8.13-fold increase in half maximal inhibitory concentration and a 4.46-fold increase in conversion (150 mM ATP and l-methionine), leading to a SAM titer of 47.02 g/L. Another variant, E56N/Q105R, showed superior thermostability with an impressive 85.30-fold increase in half-life (50 °C) value. Furthermore, molecular dynamics (MD) simulation results demonstrate that the alleviation in product inhibitory effect could be attributed to facilitated product release. This study offers molecular insights into the mitigated product inhibition, and provides valuable guidance for engineering MAT toward enhanced catalytic performance.


Assuntos
Escherichia coli , Metionina Adenosiltransferase , S-Adenosilmetionina , Metionina Adenosiltransferase/genética , Metionina Adenosiltransferase/metabolismo , Metionina Adenosiltransferase/química , S-Adenosilmetionina/metabolismo , S-Adenosilmetionina/química , Escherichia coli/genética , Escherichia coli/metabolismo , Engenharia de Proteínas , Cinética , Simulação de Dinâmica Molecular , Estabilidade Enzimática , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/química
3.
J Am Chem Soc ; 146(27): 18722-18729, 2024 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-38943667

RESUMO

Methylation, a widely occurring natural modification serving diverse regulatory and structural functions, is carried out by a myriad of S-adenosyl-l-methionine (AdoMet)-dependent methyltransferases (MTases). The AdoMet cofactor is produced from l-methionine (Met) and ATP by a family of multimeric methionine adenosyltransferases (MAT). To advance mechanistic and functional studies, strategies for repurposing the MAT and MTase reactions to accept extended versions of the transferable group from the corresponding precursors have been exploited. Here, we used structure-guided engineering of mouse MAT2A to enable biocatalytic production of an extended AdoMet analogue, Ado-6-azide, from a synthetic methionine analogue, S-(6-azidohex-2-ynyl)-l-homocysteine (N3-Met). Three engineered MAT2A variants showed catalytic proficiency with the extended analogues and supported DNA derivatization in cascade reactions with M.TaqI and an engineered variant of mouse DNMT1 both in the absence and presence of competing Met. We then installed two of the engineered variants as MAT2A-DNMT1 cascades in mouse embryonic stem cells by using CRISPR-Cas genome editing. The resulting cell lines maintained normal viability and DNA methylation levels and showed Dnmt1-dependent DNA modification with extended azide tags upon exposure to N3-Met in the presence of physiological levels of Met. This for the first time demonstrates a genetically stable system for biosynthetic production of an extended AdoMet analogue, which enables mild metabolic labeling of a DNMT-specific methylome in live mammalian cells.


Assuntos
Metilação de DNA , Metionina Adenosiltransferase , Metionina Adenosiltransferase/metabolismo , Metionina Adenosiltransferase/genética , Metionina Adenosiltransferase/química , Animais , Camundongos , Engenharia de Proteínas , Epigenoma , S-Adenosilmetionina/metabolismo , S-Adenosilmetionina/química , DNA (Citosina-5-)-Metiltransferase 1/metabolismo , DNA (Citosina-5-)-Metiltransferase 1/genética , Humanos
4.
Drug Dev Res ; 85(1): e22122, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37819020

RESUMO

The use of cell growth-based assays to identify inhibitory compounds is straightforward and inexpensive, but is also inherently insensitive and somewhat nonspecific. To overcome these limitations and develop a sensitive, specific cell-based assay, two different approaches were combined. To address the sensitivity limitation, different fluorescent proteins have been introduced into a bacterial expression system to serve as growth reporters. To overcome the lack of specificity, these protein reporters have been incorporated into a plasmid in which they are paired with different orthologs of an essential target enzyme, in this case l-methionine S-adenosyltransferase (MAT, AdoMet synthetase). Screening compounds that serve as specific inhibitors will reduce the growth of only a subset of strains, because these strains are identical, except for which target ortholog they carry. Screening several such strains in parallel not only reveals potential inhibitors but the strains also serve as specificity controls for one another. The present study makes use of an existing Escherichia coli strain that carries a deletion of metK, the gene for MAT. Transformation with these plasmids leads to a complemented strain that no longer requires externally supplied S-adenosylmethionine for growth, but its growth is now dependent on the activity of the introduced MAT ortholog. The resulting fluorescent strains provide a platform to screen chemical compound libraries and identify species-selective inhibitors of AdoMet synthetases. A pilot study of several chemical libraries using this platform identified new lead compounds that are ortholog-selective inhibitors of this enzyme family, some of which target the protozoal human pathogen Cryptosporidium parvum.


Assuntos
Criptosporidiose , Cryptosporidium , Humanos , Metionina Adenosiltransferase/genética , Metionina Adenosiltransferase/química , Metionina Adenosiltransferase/metabolismo , S-Adenosilmetionina/metabolismo , Projetos Piloto , Cryptosporidium/metabolismo , Escherichia coli/genética
5.
J Agric Food Chem ; 71(42): 15692-15700, 2023 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-37846083

RESUMO

S-Adenosylmethionine (SAM) acts as a methyl donor in living organisms, and S-adenosylmethionine synthetase (MetK) is an essential enzyme for cells, as it synthesizes SAM from methionine and adenosine triphosphate (ATP). This study determined the crystal structures of the apo form and adenosine/triphosphate complex form of MetK from Corynebacterium glutamicum (CgMetK). Results showed that CgMetK has an allosteric inhibitor binding site for the SAM product in the vicinity of the active site and is inhibited by SAM both competitively and noncompetitively. Through structure-guided protein engineering, the CgMetKE68A variant was developed that exhibited an almost complete release of inhibition by SAM with rather enhanced enzyme activity. The crystal structure of the CgMetKE68A variant revealed that the formation of a new hydrogen bond between Tyr66 and Glu102 by the E68A mutation disrupted the allosteric SAM binding site and also improved the protein thermal stability by strengthening the tetramerization of the enzyme.


Assuntos
Corynebacterium glutamicum , Metionina Adenosiltransferase , Metionina Adenosiltransferase/genética , Metionina Adenosiltransferase/química , Metionina Adenosiltransferase/metabolismo , Corynebacterium glutamicum/genética , Corynebacterium glutamicum/metabolismo , Metionina/metabolismo , S-Adenosilmetionina/metabolismo , Trifosfato de Adenosina/metabolismo
6.
Protein Sci ; 31(7): e4352, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35762725

RESUMO

Homomers are prevalent in bacterial proteomes, particularly among core metabolic enzymes. Homomerization is often key to function and regulation, and interfaces that facilitate the formation of homomeric enzymes are subject to intense evolutionary change. However, our understanding of the molecular mechanisms that drive evolutionary variation in homomeric complexes is still lacking. How is the diversification of protein interfaces linked to variation in functional regulation and structural integrity of homomeric complexes? To address this question, we studied quaternary structure evolution of bacterial methionine S-adenosyltransferases (MATs)-dihedral homotetramers formed along a large and conserved dimeric interface harboring two active sites, and a small, recently evolved, interdimeric interface. Here, we show that diversity in the physicochemical properties of small interfaces is directly linked to variability in the kinetic stability of MAT quaternary complexes and in modes of their functional regulation. Specifically, hydrophobic interactions within the small interface of Escherichia coli MAT render the functional homotetramer kinetically stable yet impose severe aggregation constraints on complex assembly. These constraints are alleviated by electrostatic interactions that accelerate dimer-dimer assembly. In contrast, Neisseria gonorrhoeae MAT adopts a nonfunctional dimeric state due to the low hydrophobicity of its small interface and the high flexibility of its active site loops, which perturbs small interface integrity. Remarkably, in the presence of methionine and ATP, N. gonorrhoeae MAT undergoes substrate-induced assembly into a functional tetrameric state. We suggest that evolution acts on the interdimeric interfaces of MATs to tailor the regulation of their activity and stability to unique organismal needs.


Assuntos
Metionina Adenosiltransferase , Proteínas , Domínio Catalítico , Escherichia coli/metabolismo , Metionina , Metionina Adenosiltransferase/química , Metionina Adenosiltransferase/genética , Metionina Adenosiltransferase/metabolismo , Modelos Moleculares , Proteínas/química , Relação Estrutura-Atividade
7.
Chembiochem ; 23(1): e202100437, 2022 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-34606675

RESUMO

Methylation and demethylation of DNA, RNA and proteins constitutes a major regulatory mechanism in epigenetic processes. Investigations would benefit from the ability to install photo-cleavable groups at methyltransferase target sites that block interactions with reader proteins until removed by non-damaging light in the visible spectrum. Engineered methionine adenosyltransferases (MATs) have been exploited in cascade reactions with methyltransferases (MTases) to modify biomolecules with non-natural groups, including first evidence for accepting photo-cleavable groups. We show that an engineered MAT from Methanocaldococcus jannaschii (PC-MjMAT) is 308-fold more efficient at converting ortho-nitrobenzyl-(ONB)-homocysteine than the wildtype enzyme. PC-MjMAT is active over a broad range of temperatures and compatible with MTases from mesophilic organisms. We solved the crystal structures of wildtype and PC-MjMAT in complex with AdoONB and a red-shifted derivative thereof. These structures reveal that aromatic stacking interactions within the ligands are key to accommodating the photocaging groups in PC-MjMAT. The enlargement of the binding pocket eliminates steric clashes to enable AdoMet analogue binding. Importantly, PC-MjMAT exhibits remarkable activity on methionine analogues with red-shifted ONB-derivatives enabling photo-deprotection of modified DNA by visible light.


Assuntos
DNA/química , Luz , Metionina Adenosiltransferase/química , RNA/química , DNA/genética , DNA/metabolismo , Methanocaldococcus/enzimologia , Metionina Adenosiltransferase/genética , Metionina Adenosiltransferase/metabolismo , Estrutura Molecular , Processos Fotoquímicos , Engenharia de Proteínas , RNA/genética , RNA/metabolismo
8.
FEBS Open Bio ; 12(1): 130-145, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34655277

RESUMO

Methionine adenosyltransferase (MAT) catalyzes the biosynthesis of S-adenosyl methionine from l-methionine and ATP. MAT enzymes are ancient, believed to share a common ancestor, and are highly conserved in all three domains of life. However, the sequences of archaeal MATs show considerable divergence compared with their bacterial and eukaryotic counterparts. Furthermore, the structural significance and functional significance of this sequence divergence are not well understood. In the present study, we employed structural analysis and ancestral sequence reconstruction to investigate archaeal MAT divergence. We observed that the dimer interface containing the active site (which is usually well conserved) diverged considerably between the bacterial/eukaryotic MATs and archaeal MAT. A detailed investigation of the available structures supports the sequence analysis outcome: The protein domains and subdomains of bacterial and eukaryotic MAT are more similar than those of archaea. Finally, we resurrected archaeal MAT ancestors. Interestingly, archaeal MAT ancestors show substrate specificity, which is lost during evolution. This observation supports the hypothesis of a common MAT ancestor for the three domains of life. In conclusion, we have demonstrated that archaeal MAT is an ideal system for studying an enzyme family that evolved differently in one domain compared with others while maintaining the same catalytic activity.


Assuntos
Archaea , Metionina Adenosiltransferase , Archaea/genética , Archaea/metabolismo , Domínio Catalítico , Metionina , Metionina Adenosiltransferase/química , Metionina Adenosiltransferase/genética , Metionina Adenosiltransferase/metabolismo , S-Adenosilmetionina/química
9.
Int J Mol Sci ; 22(24)2021 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-34948004

RESUMO

Catalytic MATα1 subunits associate into kinetically distinct homo-dimers (MAT III) and homo-tetramers (MAT I) that synthesize S-adenosylmethionine in the adult liver. Pathological reductions in S-adenosylmethionine levels correlate with MAT III accumulation; thus, it is important to know the determinants of dimer-dimer associations. Here, polar interactions (<3.5 Å) at the rat MAT I dimer-dimer interface were disrupted by site-directed mutagenesis. Heterologous expression rendered decreased soluble mutant MATα1 levels that appeared mostly as dimers. Substitutions at the B1-B2 or B3-C1 ß-strand loops, or changes in charge on helix α2 located behind, induced either MAT III or MAT I accumulation. Notably, double mutants combining neutral changes on helix α2 with substitutions at either ß-strand loop further increased MAT III content. Mutations had negligible impact on secondary or tertiary protein structure, but induced changes of 5-10 °C in thermal stability. All mutants preserved tripolyphosphatase activity, although AdoMet synthesis was only detected in single mutants. Kinetic parameters were altered in all purified proteins, their AdoMet synthesis Vmax and methionine affinities correlating with the association state induced by the corresponding mutations. In conclusion, polar interactions control MATα1 tetramerization and kinetics, diverse effects being induced by changes on opposite ß-sheet loops putatively leading to subtle variations in central domain ß-sheet orientation.


Assuntos
Substituição de Aminoácidos , Metionina Adenosiltransferase/química , Metionina Adenosiltransferase/metabolismo , Animais , Metionina Adenosiltransferase/genética , Modelos Moleculares , Mutagênese Sítio-Dirigida , Multimerização Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Ratos
10.
J Am Chem Soc ; 143(43): 18325-18330, 2021 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-34668717

RESUMO

Human methionine adenosyltransferase MAT2A provides S-adenosyl-l-methionine (AdoMet) for methyl-transfer reactions. Epigenetic methylations influence expression patterns in development and in cancer. Transition-state analysis and kinetic studies have described the mechanism of AdoMet and triphosphate formation at the catalytic site. Hydrolysis of triphosphate to pyrophosphate and phosphate by MAT2A is required for product release and proceeds through a second chemical transition state. Crystal structures of MAT2A with analogues of AdoMet and pyrophosphate were obtained in the presence of Mg2+, Al3+, and F-. MgF3- is trapped as a PO3- mimic in a structure with malonate filling the pyrophosphate site. NMR demonstrates that MgF3- and AlF30 are bound by MAT2A as mimics of the departing phosphoryl group. Crystallographic analysis reveals a planar MgF3- acting to mimic a phosphoryl (PO3-) leaving group. The modeled transition state with PO3- has the phosphorus atom sandwiched symmetrically and equidistant (approximately 2 Å) between a pyrophosphate oxygen and the water nucleophile. A catalytic site arginine directs the nucleophilic water to the phosphoryl leaving group. The catalytic geometry of the transition-state reconstruction predicts a loose transition state with characteristics of symmetric nucleophilic displacement.


Assuntos
Biocatálise , Metionina Adenosiltransferase/metabolismo , Polifosfatos/metabolismo , Domínio Catalítico , Cristalografia por Raios X , Humanos , Hidrólise , Metionina Adenosiltransferase/química , Modelos Químicos , Polifosfatos/química , Ligação Proteica , Água/metabolismo
11.
PLoS Comput Biol ; 17(6): e1009107, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34133419

RESUMO

We describe an approach for integrating distance restraints from Double Electron-Electron Resonance (DEER) spectroscopy into Rosetta with the purpose of modeling alternative protein conformations from an initial experimental structure. Fundamental to this approach is a multilateration algorithm that harnesses sets of interconnected spin label pairs to identify optimal rotamer ensembles at each residue that fit the DEER decay in the time domain. Benchmarked relative to data analysis packages, the algorithm yields comparable distance distributions with the advantage that fitting the DEER decay and rotamer ensemble optimization are coupled. We demonstrate this approach by modeling the protonation-dependent transition of the multidrug transporter PfMATE to an inward facing conformation with a deviation to the experimental structure of less than 2Å Cα RMSD. By decreasing spin label rotamer entropy, this approach engenders more accurate Rosetta models that are also more closely clustered, thus setting the stage for more robust modeling of protein conformational changes.


Assuntos
Algoritmos , Modelos Moleculares , Conformação Proteica , Bacteriófago T4/enzimologia , Biologia Computacional , Espectroscopia de Ressonância de Spin Eletrônica/estatística & dados numéricos , Metionina Adenosiltransferase/química , Simulação de Dinâmica Molecular/estatística & dados numéricos , Proteínas Associadas à Resistência a Múltiplos Medicamentos/química , Muramidase/química , Pyrococcus furiosus/enzimologia , Software , Marcadores de Spin
12.
J Biol Chem ; 296: 100270, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33428944

RESUMO

RNA methylations of varied RNA species (mRNA, tRNA, rRNA, non-coding RNA) generate a range of modified nucleotides, including N6-methyladenosine. Here we study the enzymology of three human RNA methyltransferases that methylate the adenosine amino group in diverse contexts, when it is: the first transcribed nucleotide after the mRNA cap (PCIF1), at position 1832 of 18S rRNA (MettL5-Trm112 complex), and within a hairpin in the 3' UTR of the S-adenosyl-l-methionine synthetase (MettL16). Among these three enzymes, the catalytic efficiency ranges from PCIF1, with the fastest turnover rate of >230 h-1 µM-1 on mRNA cap analog, down to MettL16, which has the lowest rate of ∼3 h-1 µM-1 acting on an RNA hairpin. Both PCIF1 and MettL5 have a binding affinity (Km) of ∼1 µM or less for both substrates of SAM and RNA, whereas MettL16 has significantly lower binding affinities for both (Km >0.4 mM for SAM and ∼10 µM for RNA). The three enzymes are active over a wide pH range (∼5.4-9.4) and have different preferences for ionic strength. Sodium chloride at 200 mM markedly diminished methylation activity of MettL5-Trm112 complex, whereas MettL16 had higher activity in the range of 200 to 500 mM NaCl. Zinc ion inhibited activities of all three enzymes. Together, these results illustrate the diversity of RNA adenosine methyltransferases in their enzymatic mechanisms and substrate specificities and underline the need for assay optimization in their study.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Metiltransferases/genética , Proteínas Nucleares/genética , RNA Ribossômico 18S/genética , Proteínas Adaptadoras de Transdução de Sinal/química , Adenosina/genética , Humanos , Metionina Adenosiltransferase/química , Metionina Adenosiltransferase/genética , Metilação , Metiltransferases/química , Complexos Multiproteicos/química , Complexos Multiproteicos/genética , Proteínas Nucleares/química , S-Adenosilmetionina/metabolismo , Especificidade por Substrato
13.
Bioorg Med Chem Lett ; 30(19): 127442, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32730944

RESUMO

Active or allosteric site arginines can form diverse interactions with ligands including different types of cation-π interactions, H-bond interactions and non-bond, non-canonical interactions. This provides many opportunities for creative structure-based drug design to improve potency, introduce novelty, and modulate MoA (mode of action), and even to achieve selectivity. This digest will use some recent drug targets of interest as examples to illustrate different types of interactions and how these interactions impact on potency, MoA, and selectivity.


Assuntos
Arginina/química , Proteína de Ligação a CREB/metabolismo , Inibidores Enzimáticos/metabolismo , Metionina Adenosiltransferase/metabolismo , Compostos Orgânicos/metabolismo , Complexo Repressor Polycomb 2/metabolismo , Sítio Alostérico , Antineoplásicos/química , Antineoplásicos/metabolismo , Proteína de Ligação a CREB/química , Domínio Catalítico , Desenho de Fármacos , Inibidores Enzimáticos/química , Humanos , Ligantes , Metionina Adenosiltransferase/química , Compostos Orgânicos/química , Complexo Repressor Polycomb 2/química , Ligação Proteica
14.
Acta Crystallogr D Struct Biol ; 76(Pt 6): 594-607, 2020 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-32496220

RESUMO

Methionine adenosyltransferase (MAT) deficiency, characterized by isolated persistent hypermethioninemia (IPH), is caused by mutations in the MAT1A gene encoding MATαl, one of the major hepatic enzymes. Most of the associated hypermethioninemic conditions are inherited as autosomal recessive traits; however, dominant inheritance of hypermethioninemia is caused by an Arg264His (R264H) mutation. This mutation has been confirmed in a screening programme of newborns as the most common mutation in babies with IPH. Arg264 makes an inter-subunit salt bridge located at the dimer interface where the active site assembles. Here, it is demonstrated that the R264H mutation results in greatly reduced MAT activity, while retaining its ability to dimerize, indicating that the lower activity arises from alteration at the active site. The first crystallographic structure of the apo form of the wild-type MATαl enzyme is provided, which shows a tetrameric assembly in which two compact dimers combine to form a catalytic tetramer. In contrast, the crystal structure of the MATαl R264H mutant reveals a weaker dimeric assembly, suggesting that the mutation lowers the affinity for dimer-dimer interaction. The formation of a hetero-oligomer with the regulatory MATßV1 subunit or incubation with a quinolone-based compound (SCR0911) results in the near-full recovery of the enzymatic activity of the pathogenic mutation R264H, opening a clear avenue for a therapeutic solution based on chemical interventions that help to correct the defect of the enzyme in its ability to metabolize methionine.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/genética , Glicina N-Metiltransferase/deficiência , Padrões de Herança , Metionina Adenosiltransferase/química , Domínio Catalítico , Glicina N-Metiltransferase/genética , Humanos , Metionina Adenosiltransferase/genética , Mutação , Multimerização Proteica
15.
PLoS Comput Biol ; 16(5): e1007904, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32453784

RESUMO

S-adenosylmethionine (SAM) is one of the most important enzyme substrates. It is vital for the function of various proteins, including large group of methyltransferases (MTs). Intriguingly, some bacterial and eukaryotic MTs, while catalysing the same reaction, possess significantly different topologies, with the former being a knotted one. Here, we conducted a comprehensive analysis of SAM conformational space and factors that affect its vastness. We investigated SAM in two forms: free in water (via NMR studies and explicit solvent simulations) and bound to proteins (based on all data available in the PDB and on all-atom molecular dynamics simulations in water). We identified structural descriptors-angles which show the major differences in SAM conformation between unknotted and knotted methyltransferases. Moreover, we report that this is caused mainly by a characteristic for knotted MTs compact binding site formed by the knot and the presence of adenine-binding loop. Additionally, we elucidate conformational restrictions imposed on SAM molecules by other protein groups in comparison to conformational space in water.


Assuntos
Sítios de Ligação , Metionina Adenosiltransferase/química , S-Adenosilmetionina/química , Adenina/química , Motivos de Aminoácidos , Biologia Computacional/métodos , Simulação por Computador , Bases de Dados de Proteínas , Glicina/química , Espectroscopia de Ressonância Magnética , Simulação de Dinâmica Molecular , Análise de Componente Principal , Ligação Proteica , Domínios Proteicos , Dobramento de Proteína , Solventes , Temperatura , Água/química , tRNA Metiltransferases/química
16.
Int J Biol Macromol ; 151: 554-565, 2020 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-32057875

RESUMO

S-adenosylmethionine synthases (MATs) are responsible for production of S-adenosylmethionine, the cofactor essential for various methylation reactions, production of polyamines and phytohormone ethylene, etc. Plants have two distinct MAT types (I and II). This work presents the structural analysis of MATs from Arabidopsis thaliana (AtMAT1 and AtMAT2, both type I) and Medicago truncatula (MtMAT3a, type II), which, unlike most MATs from other domains of life, are dimers where three-domain subunits are sandwiched flat with one another. Although MAT types are very similar, their subunits are differently oriented within the dimer. Structural snapshots along the enzymatic reaction reveal the exact conformation of precatalytic methionine in the active site and show a binding niche, characteristic only for plant MATs, that may serve as a lock of the gate loop. Nevertheless, plants, in contrary to mammals, lack the MAT regulatory subunit, and the regulation of plant MAT activity is still puzzling. Our structures open a possibility of an allosteric activity regulation of type I plant MATs by linear compounds, like polyamines, which would tighten the relationship between S-adenosylmethionine and polyamine biosynthesis.


Assuntos
Arabidopsis/enzimologia , Medicago truncatula/enzimologia , Metionina Adenosiltransferase/química , Modelos Moleculares , Conformação Proteica , Sequência de Aminoácidos , Domínio Catalítico , Isoenzimas , Ligantes , Ligação Proteica , Multimerização Proteica , S-Adenosilmetionina/química , S-Adenosilmetionina/metabolismo
17.
J Struct Biol ; 210(1): 107462, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-31962159

RESUMO

Methionine adenosyltransferases catalyse the biosynthesis of S-adenosylmethionine, the primary methyl group donor in biochemical reactions, through the condensation of methionine and ATP. Here, we report the structural analysis of the Pyrococcus furiosus methionine adenosyltransferase (PfMAT) captured in the unliganded, substrate- and product-bound states. The conformational changes taking place during the enzymatic catalytic cycle are allosterically propagated by amino acid residues conserved in the archaeal orthologues to induce an asymmetric dimer structure. The distinct occupancy of the active sites within a PfMAT dimer is consistent with a half-site reactivity that is mediated by a product-induced negative cooperativity. The structures of intermediate states of PfMAT reported here suggest a distinct molecular mechanism for S-adenosylmethionine synthesis in Archaea, likely consequence of the evolutionary pressure to achieve protein stability under extreme conditions.


Assuntos
Metionina Adenosiltransferase/metabolismo , Pyrococcus furiosus/enzimologia , Sítios de Ligação , Catálise , Cristalografia por Raios X/métodos , Metionina Adenosiltransferase/química , Conformação Proteica , Pyrococcus furiosus/metabolismo
18.
J Mol Biol ; 431(24): 4796-4816, 2019 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-31520601

RESUMO

Methionine S-adenosyltransferases (MATs) are predominantly homotetramers, comprised of dimers of dimers. The larger, highly conserved intradimeric interface harbors two active sites, making the dimer the obligatory functional unit. However, functionality of the smaller, more diverged, and recently evolved interdimeric interface is largely unknown. Here, we show that the interdimeric interface of Ureaplasmaurealiticum MAT has evolved to control the catalytic activity and structural integrity of the homotetramer in response to product accumulation. When all four active sites are occupied with the product, S-adenosylmethionine (SAM), binding of four additional SAM molecules to the interdimeric interface prompts a ∼45° shift in the dimer orientation and a concomitant ∼60% increase in the interface area. This rearrangement inhibits the enzymatic activity by locking the flexible active site loops in a closed state and renders the tetramer resistant to proteolytic degradation. Our findings suggest that the interdimeric interface of MATs is subject to rapid evolutionary changes that tailor the molecular properties of the entire homotetramer to the specific needs of the organism.


Assuntos
Metionina Adenosiltransferase/química , Multimerização Proteica , Ureaplasma/enzimologia , Sítios de Ligação , Espectrometria de Massas , Metionina Adenosiltransferase/metabolismo , Modelos Moleculares , Conformação Proteica , Estabilidade Proteica , Proteólise , S-Adenosilmetionina/química , S-Adenosilmetionina/metabolismo , Relação Estrutura-Atividade
19.
Enzyme Microb Technol ; 129: 109355, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31307578

RESUMO

S-adenosylmethionine synthetase (MAT) catalyzes the synthesis of S-adenosylmethionine (SAM) from ATP and L-methionine. SAM is the major methyl donor for more than 100 transmethylation reactions. It is also a common cosubstrate involved in transsulfuration and aminopropylation. However, product inhibition largely restrains the activity of MAT and limits the enzymatic synthesis of SAM. In this research, the product inhibition of MAT from Escherichia coli was reduced via semi-rational modification. A triple variant (Variant III, I303 V/I65 V/L186 V) showed a 42-fold increase in Ki,ATP and a 2.08-fold increase in specific activity when compared to wild-type MAT. Its Ki,ATP was 0.42 mM and specific acitivity was 3.78 ±0.19 U/mg. Increased Ki,ATP means reduced product inhibition which enhances SAM accumulation. The SAM produced by Variant III could reach to 3.27 mM while SAM produced by wild-type MAT was 1.62 mM in the presence of 10 mM substrates. When the residue in 104th of Variant III was further optimized by site-saturated mutagenesis, the specific activity of Variant IV (I303 V/I65 V/L186 V/N104 K) reached to 6.02 ±0.22 U/mg at 37 °C, though the SAM concentration decreased to 2.68 mM with 10 mM substrates. Analysis of protein 3D structure suggests that changes in hydrogen bonds or other ligand interactions around active site may account for the variety of product inhibition and enzyme activity. The Variant III and Variant IV with reduced inhibition and improved enzyme activity in the study would be more suitable candidates for SAM production in the future.


Assuntos
Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimologia , Metionina Adenosiltransferase/química , Metionina Adenosiltransferase/metabolismo , Catálise , Domínio Catalítico , Escherichia coli/química , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Cinética , Metionina/metabolismo , Metionina Adenosiltransferase/genética , Modelos Moleculares , S-Adenosilmetionina/metabolismo
20.
Acta Crystallogr F Struct Biol Commun ; 75(Pt 4): 290-298, 2019 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-30950830

RESUMO

S-Adenosyl-L-methionine (AdoMet), the primary methyl donor in most biological methylation reactions, is produced from ATP and methionine in a multistep reaction catalyzed by AdoMet synthetase. The diversity of group-transfer reactions that involve AdoMet places this compound at a key crossroads in amino-acid, nucleic acid and lipid metabolism, and disruption of its synthesis has adverse consequences for all forms of life. The family of AdoMet synthetases is highly conserved, and structures of this enzyme have been determined from organisms ranging from bacteria to humans. Here, the structure of an AdoMet synthetase from the infectious parasite Cryptosporidium parvum has been determined as part of an effort to identify structural differences in this enzyme family that can guide the development of species-selective inhibitors. This enzyme form has a less extensive subunit interface than some previously determined structures, and contains some key structural differences from the human enzyme in an allosteric site, presenting an opportunity for the design of selective inhibitors against the AdoMet synthetase from this organism.


Assuntos
Cryptosporidium parvum/enzimologia , Metionina Adenosiltransferase/química , Regulação Alostérica , Sequência de Aminoácidos , Cristalização , Humanos , Modelos Moleculares , Multimerização Proteica , Homologia de Sequência de Aminoácidos , Homologia Estrutural de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA