Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
1.
Cell ; 159(5): 1188-1199, 2014 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-25416954

RESUMO

Glutamine is the primary metabolite of nitrogen assimilation from inorganic nitrogen sources in microorganisms and plants. The ability to monitor cellular nitrogen status is pivotal for maintaining metabolic homeostasis and sustaining growth. The present study identifies a glutamine-sensing mechanism common in the entire plant kingdom except Brassicaceae. The plastid-localized PII signaling protein controls, in a glutamine-dependent manner, the key enzyme of the ornithine synthesis pathway, N-acetyl-l-glutamate kinase (NAGK), that leads to arginine and polyamine formation. Crystal structures reveal that the plant-specific C-terminal extension of PII, which we term the Q loop, forms a low-affinity glutamine-binding site. Glutamine binding alters PII conformation, promoting interaction and activation of NAGK. The binding motif is highly conserved in plants except Brassicaceae. A functional Q loop restores glutamine sensing in a recombinant Arabidopsis thaliana PII protein, demonstrating the modular concept of the glutamine-sensing mechanism adopted by PII proteins during the evolution of plant chloroplasts.


Assuntos
Glutamina/metabolismo , Plantas/metabolismo , Sequência de Aminoácidos , Chlamydomonas reinhardtii/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Nitrogênio/metabolismo , Proteínas PII Reguladoras de Nitrogênio/metabolismo , Fosfotransferases (Aceptor do Grupo Carboxila)/química , Fosfotransferases (Aceptor do Grupo Carboxila)/metabolismo , Plantas/classificação , Alinhamento de Sequência
2.
Microb Cell Fact ; 17(1): 147, 2018 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-30227873

RESUMO

BACKGROUND: Microbial biosynthesis of natural products holds promise for preclinical studies and treating diseases. For instance, pinocembrin is a natural flavonoid with important pharmacologic characteristics and is widely used in preclinical studies. However, high yield of natural products production is often limited by the intracellular cofactor level, including adenosine triphosphate (ATP). To address this challenge, tailored modification of ATP concentration in Escherichia coli was applied in efficient pinocembrin production. RESULTS: In the present study, a clustered regularly interspaced short palindromic repeats (CRISPR) interference system was performed for screening several ATP-related candidate genes, where metK and proB showed its potential to improve ATP level and increased pinocembrin production. Subsequently, the repression efficiency of metK and proB were optimized to achieve the appropriate levels of ATP and enhancing the pinocembrin production, which allowed the pinocembrin titer increased to 102.02 mg/L. Coupled with the malonyl-CoA engineering and optimization of culture and induction condition, a final pinocembrin titer of 165.31 mg/L was achieved, which is 10.2-fold higher than control strains. CONCLUSIONS: Our results introduce a strategy to approach the efficient biosynthesis of pinocembrin via ATP level strengthen using CRISPR interference. Furthermore coupled with the malonyl-CoA engineering and induction condition have been optimized for pinocembrin production. The results and engineering strategies demonstrated here would hold promise for the ATP level improvement of other flavonoids by CRISPRi system, thereby facilitating other flavonoids production.


Assuntos
Trifosfato de Adenosina/metabolismo , Flavanonas/biossíntese , Engenharia Metabólica/métodos , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Escherichia coli , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Engenharia Genética , Metionina Adenosiltransferase/química , Metionina Adenosiltransferase/genética , Fosfotransferases (Aceptor do Grupo Carboxila)/química , Fosfotransferases (Aceptor do Grupo Carboxila)/genética
3.
Appl Microbiol Biotechnol ; 101(9): 3485-3492, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28341883

RESUMO

N-acetylglutamate kinase (NAGK) catalyzes the phosphorylation of N-acetylglutamate. In many bacteria, NAGK catalysis is the rate controlling step in the L-arginine biosynthesis pathway from glutamate to L-arginine and is allosterically inhibited by L-arginine. Many data show that conformational dynamics of NAGKs are essential for their function. The demonstration of the conformational mechanism provides a potential way to improve the yield of arginine. Due to the lack of NAGK catalysis step in arginine synthesis route of mammals, the elucidation of the dynamic mechanism can also provide a way to design a new antivirus drug. This paper reviews how the dynamics affect the activity of NAGKs and are controlled by the effectors. X-ray crystallography and modeling data have shown that in NAGKs, the structural elements required for inhibitor and substrate binding, catalysis and product release, are highly mobile. It is possible to eliminate the inhibition of the arginine and/or block the synthesis of arginine by disturbing the flexibility of the NAGKs. Amino acid kinase family is thought to share some common dynamic features; the flexible structural elements of NAGKs have been identified, but the details of the dynamics and the signal transfer pathways are yet to be elucidated.


Assuntos
Glutamatos/metabolismo , Fosfotransferases (Aceptor do Grupo Carboxila)/química , Fosfotransferases (Aceptor do Grupo Carboxila)/metabolismo , Regulação Alostérica , Bactérias/enzimologia , Cristalografia por Raios X , Modelos Moleculares , Fosforilação , Conformação Proteica
4.
Proc Natl Acad Sci U S A ; 111(29): 10532-7, 2014 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-25002480

RESUMO

Extracellular fatty acid incorporation into the phospholipids of Staphylococcus aureus occurs via fatty acid phosphorylation. We show that fatty acid kinase (Fak) is composed of two dissociable protein subunits encoded by separate genes. FakA provides the ATP binding domain and interacts with two distinct FakB proteins to produce acyl-phosphate. The FakBs are fatty acid binding proteins that exchange bound fatty acid/acyl-phosphate with fatty acid/acyl-phosphate presented in detergent micelles or liposomes. The ΔfakA and ΔfakB1 ΔfakB2 strains were unable to incorporate extracellular fatty acids into phospholipid. FakB1 selectively bound saturated fatty acids whereas FakB2 preferred unsaturated fatty acids. Affymetrix array showed a global perturbation in the expression of virulence genes in the ΔfakA strain. The severe deficiency in α-hemolysin protein secretion in ΔfakA and ΔfakB1 ΔfakB2 mutants coupled with quantitative mRNA measurements showed that fatty acid kinase activity was required to support virulence factor transcription. These data reveal the function of two conserved gene families, their essential role in the incorporation of host fatty acids by Gram-positive pathogens, and connects fatty acid kinase to the regulation of virulence factor transcription in S. aureus.


Assuntos
Proteínas de Bactérias/metabolismo , Ácidos Graxos/metabolismo , Interações Hospedeiro-Patógeno , Fosfotransferases (Aceptor do Grupo Carboxila)/metabolismo , Staphylococcus aureus/enzimologia , Sequência de Aminoácidos , Proteínas de Bactérias/química , Dados de Sequência Molecular , Fosfolipídeos/metabolismo , Fosfotransferases (Aceptor do Grupo Carboxila)/química , Staphylococcus aureus/patogenicidade , Especificidade por Substrato , Transcrição Gênica , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
5.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 8): 1640-8, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26249345

RESUMO

Kinases are ubiquitous enzymes that are pivotal to many biochemical processes. There are contrasting views on the phosphoryl-transfer mechanism in propionate kinase, an enzyme that reversibly transfers a phosphoryl group from propionyl phosphate to ADP in the final step of non-oxidative catabolism of L-threonine to propionate. Here, X-ray crystal structures of propionate- and nucleotide-bound Salmonella typhimurium propionate kinase are reported at 1.8-2.0 Å resolution. Although the mode of nucleotide binding is comparable to those of other members of the ASKHA superfamily, propionate is bound at a distinct site deeper in the hydrophobic pocket defining the active site. The propionate carboxyl is at a distance of ∼ 5 Å from the γ-phosphate of the nucleotide, supporting a direct in-line transfer mechanism. The phosphoryl-transfer reaction is likely to occur via an associative SN2-like transition state that involves a pentagonal bipyramidal structure with the axial positions occupied by the nucleophile of the substrate and the O atom between the ß- and the γ-phosphates, respectively. The proximity of the strictly conserved His175 and Arg236 to the carboxyl group of the propionate and the γ-phosphate of ATP suggests their involvement in catalysis. Moreover, ligand binding does not induce global domain movement as reported in some other members of the ASKHA superfamily. Instead, residues Arg86, Asp143 and Pro116-Leu117-His118 that define the active-site pocket move towards the substrate and expel water molecules from the active site. The role of Ala88, previously proposed to be the residue determining substrate specificity, was examined by determining the crystal structures of the propionate-bound Ala88 mutants A88V and A88G. Kinetic analysis and structural data are consistent with a significant role of Ala88 in substrate-specificity determination. The active-site pocket-defining residues Arg86, Asp143 and the Pro116-Leu117-His118 segment are also likely to contribute to substrate specificity.


Assuntos
Nucleotídeos/metabolismo , Fosfotransferases (Aceptor do Grupo Carboxila)/química , Fosfotransferases (Aceptor do Grupo Carboxila)/metabolismo , Propionatos/metabolismo , Salmonella typhimurium/enzimologia , Sítios de Ligação , Cristalografia por Raios X , Cinética , Modelos Moleculares , Ligação Proteica , Salmonella typhimurium/química , Salmonella typhimurium/metabolismo , Especificidade por Substrato
6.
Biochim Biophys Acta ; 1834(10): 2036-44, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23747922

RESUMO

Short-chain fatty acids (SCFAs) play a major role in carbon cycle and can be utilized as a source of carbon and energy by bacteria. Salmonella typhimurium propionate kinase (StTdcD) catalyzes reversible transfer of the γ-phosphate of ATP to propionate during l-threonine degradation to propionate. Kinetic analysis revealed that StTdcD possesses broad ligand specificity and could be activated by various SCFAs (propionate>acetate≈butyrate), nucleotides (ATP≈GTP>CTP≈TTP; dATP>dGTP>dCTP) and metal ions (Mg(2+)≈Mn(2+)>Co(2+)). Inhibition of StTdcD by tricarboxylic acid (TCA) cycle intermediates such as citrate, succinate, α-ketoglutarate and malate suggests that the enzyme could be under plausible feedback regulation. Crystal structures of StTdcD bound to PO4 (phosphate), AMP, ATP, Ap4 (adenosine tetraphosphate), GMP, GDP, GTP, CMP and CTP revealed that binding of nucleotide mainly involves hydrophobic interactions with the base moiety and could account for the broad biochemical specificity observed between the enzyme and nucleotides. Modeling and site-directed mutagenesis studies suggest Ala88 to be an important residue involved in determining the rate of catalysis with SCFA substrates. Molecular dynamics simulations on monomeric and dimeric forms of StTdcD revealed plausible open and closed states, and also suggested role for dimerization in stabilizing segment 235-290 involved in interfacial interactions and ligand binding. Observation of an ethylene glycol molecule bound sufficiently close to the γ-phosphate in StTdcD complexes with triphosphate nucleotides supports direct in-line phosphoryl transfer.


Assuntos
Alanina/química , Proteínas de Bactérias/química , Nucleotídeos/química , Fosfotransferases (Aceptor do Grupo Carboxila)/química , Propionatos/química , Salmonella typhimurium/química , Alanina/genética , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Cristalografia por Raios X , Ensaios Enzimáticos , Etilenoglicol/química , Interações Hidrofóbicas e Hidrofílicas , Cinética , Magnésio/química , Manganês/química , Simulação de Dinâmica Molecular , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fosfotransferases (Aceptor do Grupo Carboxila)/genética , Multimerização Proteica , Salmonella typhimurium/enzimologia , Alinhamento de Sequência , Especificidade por Substrato
7.
Protein Expr Purif ; 91(1): 61-8, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23867361

RESUMO

Streptococcus pyogenes (group A Streptococcus, GAS) is an important human pathogen causing mild superficial infections of skin and mucous membranes, but also life-threatening systemic diseases. S. pyogenes and other prokaryotic organisms use the arginine deiminase system (ADS) for survival in acidic environments. In this study, the arginine deiminase (AD), and carbamate kinase (CK) from S. pyogenes M49 strain 591 were heterologously expressed in Escherichia coli DH5α, purified, and kinetically characterized. AD and CK from S. pyogenes M49 share high amino acid sequence similarity with the respective enzymes from Lactococcus lactis subsp. lactis IL1403 (45.6% and 53.5% identical amino acids) and Enterococcus faecalis V583 (66.8% and 66.8% identical amino acids). We found that the arginine deiminase of S. pyogenes is not allosterically regulated by the intermediates and products of the arginine degradation (e.g., ATP, citrulline, carbamoyl phosphate). The Km and Vmax values for arginine were 1.13±0.12mM (mean±SD) and 1.51±0.07µmol/min/mg protein. The carbamate kinase is inhibited by ATP but unaffected by arginine and citrulline. The Km and Vmax values for ADP were 0.72±0.08mM and 1.10±0.10µmol/min/mg protein and the Km for carbamoyl phosphate was 0.65±0.07mM. The optimum pH and temperature for both enzymes were 6.5 and 37°C, respectively.


Assuntos
Hidrolases/metabolismo , Fosfotransferases (Aceptor do Grupo Carboxila)/metabolismo , Proteínas Recombinantes/metabolismo , Streptococcus pyogenes/enzimologia , Regulação Alostérica , Sequência de Aminoácidos , Escherichia coli/genética , Genes Bacterianos , Concentração de Íons de Hidrogênio , Hidrolases/química , Hidrolases/genética , Cinética , Dados de Sequência Molecular , Óperon , Fosfotransferases (Aceptor do Grupo Carboxila)/química , Fosfotransferases (Aceptor do Grupo Carboxila)/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Alinhamento de Sequência , Streptococcus pyogenes/genética , Temperatura
8.
Eukaryot Cell ; 11(10): 1249-56, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22903977

RESUMO

Acetate kinase (ACK) catalyzes the reversible synthesis of acetyl phosphate by transfer of the γ-phosphate of ATP to acetate. Here we report the first biochemical and kinetic characterization of a eukaryotic ACK, that from the protist Entamoeba histolytica. Our characterization revealed that this protist ACK is the only known member of the ASKHA structural superfamily, which includes acetate kinase, hexokinase, and other sugar kinases, to utilize inorganic pyrophosphate (PP(i))/inorganic phosphate (P(i)) as the sole phosphoryl donor/acceptor. Detection of ACK activity in E. histolytica cell extracts in the direction of acetate/PP(i) formation but not in the direction of acetyl phosphate/P(i) formation suggests that the physiological direction of the reaction is toward acetate/PP(i) production. Kinetic parameters determined for each direction of the reaction are consistent with this observation. The E. histolytica PP(i)-forming ACK follows a sequential mechanism, supporting a direct in-line phosphoryl transfer mechanism as previously reported for the well-characterized Methanosarcina thermophila ATP-dependent ACK. Characterizations of enzyme variants altered in the putative acetate/acetyl phosphate binding pocket suggested that acetyl phosphate binding is not mediated solely through a hydrophobic interaction but also through the phosphoryl group, as for the M. thermophila ACK. However, there are key differences in the roles of certain active site residues between the two enzymes. The absence of known ACK partner enzymes raises the possibility that ACK is part of a novel pathway in Entamoeba.


Assuntos
Difosfatos/metabolismo , Entamoeba histolytica/enzimologia , Fosfotransferases (Aceptor do Grupo Carboxila)/metabolismo , Proteínas de Protozoários/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Domínio Catalítico , Entamoeba histolytica/genética , Dados de Sequência Molecular , Organofosfatos/metabolismo , Fosfotransferases (Aceptor do Grupo Carboxila)/química , Fosfotransferases (Aceptor do Grupo Carboxila)/genética , Proteínas de Protozoários/química , Proteínas de Protozoários/genética
9.
J Bacteriol ; 194(11): 2791-801, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22447897

RESUMO

In many microorganisms, the first step of arginine biosynthesis is catalyzed by the classical N-acetylglutamate synthase (NAGS), an enzyme composed of N-terminal amino acid kinase (AAK) and C-terminal histone acetyltransferase (GNAT) domains that bind the feedback inhibitor arginine and the substrates, respectively. In NAGS, three AAK domain dimers are interlinked by their N-terminal helices, conforming a hexameric ring, whereas each GNAT domain sits on the AAK domain of an adjacent dimer. The arginine inhibition of Pseudomonas aeruginosa NAGS was strongly hampered, abolished, or even reverted to modest activation by changes in the length/sequence of the short linker connecting both domains, supporting a crucial role of this linker in arginine regulation. Linker cleavage or recombinant domain production allowed the isolation of each NAGS domain. The AAK domain was hexameric and inactive, whereas the GNAT domain was monomeric/dimeric and catalytically active although with ∼50-fold-increased and ∼3-fold-decreased K(m)(glutamate) and k(cat) values, respectively, with arginine not influencing its activity. The deletion of N-terminal residues 1 to 12 dissociated NAGS into active dimers, catalyzing the reaction with substrate kinetics and arginine insensitivity identical to those for the GNAT domain. Therefore, the interaction between the AAK and GNAT domains from different dimers modulates GNAT domain activity, whereas the hexameric architecture appears to be essential for arginine inhibition. We proved the closeness of the AAK domains of NAGS and N-acetylglutamate kinase (NAGK), the enzyme that catalyzes the next arginine biosynthesis step, shedding light on the origin of classical NAGS, by showing that a double mutation (M26K L240K) in the isolated NAGS AAK domain elicited NAGK activity.


Assuntos
Aminoácido N-Acetiltransferase/metabolismo , Fosfotransferases (Aceptor do Grupo Carboxila)/metabolismo , Pseudomonas aeruginosa/enzimologia , Aminoácido N-Acetiltransferase/química , Aminoácido N-Acetiltransferase/genética , Arginina/metabolismo , Catálise , Dimerização , Cinética , Dados de Sequência Molecular , Fosfotransferases (Aceptor do Grupo Carboxila)/química , Fosfotransferases (Aceptor do Grupo Carboxila)/genética , Estrutura Terciária de Proteína , Pseudomonas aeruginosa/química , Pseudomonas aeruginosa/genética
10.
Biochem Biophys Res Commun ; 420(3): 692-7, 2012 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-22452987

RESUMO

The de novo biosynthesis of arginine in microorganisms and plants is accomplished via several enzymatic steps. The enzyme N-acetyl glutamate kinase (NAGK) catalyzes the phosphorylation of the γ-COO(-) group of N-acetyl-L-glutamate (NAG) by adenosine triphosphate (ATP) which is the second rate limiting step in arginine biosynthesis pathway. Here we report the crystal structure of putative N-acetyl glutamate kinase (NAGK) from Thermus thermophilus HB8 (TtNAGK) determined at 1.92Šresolution. The structural analysis of TtNAGK suggests that the dimeric quaternary state of the enzyme and arginine insensitive nature are similar to mesophilic Escherichia coli NAGK. These features are significantly different from its thermophilic homolog Thermatoga maritima NAGK which is hexameric and arginine-sensitive. TtNAGK is devoid of its substrates but contains two sulfates at the active site. Very interestingly the active site of the enzyme adopts a conformation which is not completely open or closed and likely represents an intermediate stage in the catalytic cycle unlike its structural homologs, which all exist either in the open or closed conformation. Engineering arginine biosynthesis pathway enzymes for the production of l-arginine is an important industrial application. The structural comparison of TtNAGK with EcNAGK revealed the structural basis of thermostability of TtNAGK and this information could be very useful to generate mutants of NAGK with increased overall stability.


Assuntos
Fosfotransferases (Aceptor do Grupo Carboxila)/química , Thermus thermophilus/enzimologia , Arginina/química , Arginina/farmacologia , Domínio Catalítico , Estabilidade Enzimática , Retroalimentação Fisiológica , Temperatura Alta , Fosfotransferases (Aceptor do Grupo Carboxila)/antagonistas & inibidores , Fosfotransferases (Aceptor do Grupo Carboxila)/genética , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
11.
Amino Acids ; 43(1): 255-66, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21901472

RESUMO

N-acetyl-L-glutamate kinase (EC 2.7.2.8) is first committed in the specific L-arginine pathway of Corynebacterium sp. A limited increase of L-arginine production for the argB overexpression in the engineering C. creantum SYPA-CCB strain indicated that L-arginine feedback inhibition plays an influence on the L-arginine production. In this study, we have performed site-directed mutagenesis of the key enzyme (NAGK) and the three mutations (E19R, H26E and H268D) exhibited the increase of I0.5R efficiently. Thereby, the multi-mutated NAGKM3 (including E19R/H26E/H268D) was generated and its I0.5R of L-arginine of the mutant was increased remarkably, whereas the NAGK enzyme activities did not declined. To get a feedback-resistant and robust L-arginine producer, the engineered strains SYPA-CCBM3 were constructed. Introducing the argBM3 gene enabled the NAGK enzyme activity insensitive to the intracellular arginine concentrations resulted in an enhanced arginine biosynthesis flux and decreased formation of by-products. The L-arginine synthesis was largely enhanced due to the overexpression of the argBM3, which is resistant to feedback resistant by L-arginine. Thus L-arginine production could reach 45.6 g/l, about 41.7% higher compared with the initial strain. This is an example of up-modulation of the flux through the L-arginine metabolic pathway by deregulating the key enzyme of the pathway.


Assuntos
Arginina/metabolismo , Corynebacterium/genética , Corynebacterium/metabolismo , Regulação Bacteriana da Expressão Gênica , Fosfotransferases (Aceptor do Grupo Carboxila)/genética , Fosfotransferases (Aceptor do Grupo Carboxila)/metabolismo , Sequência de Aminoácidos , Retroalimentação Fisiológica , Engenharia Genética , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação , Fosfotransferases (Aceptor do Grupo Carboxila)/química , Estrutura Secundária de Proteína , Alinhamento de Sequência
12.
PLoS Comput Biol ; 7(9): e1002201, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21980279

RESUMO

Oligomerization is a functional requirement for many proteins. The interfacial interactions and the overall packing geometry of the individual monomers are viewed as important determinants of the thermodynamic stability and allosteric regulation of oligomers. The present study focuses on the role of the interfacial interactions and overall contact topology in the dynamic features acquired in the oligomeric state. To this aim, the collective dynamics of enzymes belonging to the amino acid kinase family both in dimeric and hexameric forms are examined by means of an elastic network model, and the softest collective motions (i.e., lowest frequency or global modes of motions) favored by the overall architecture are analyzed. Notably, the lowest-frequency modes accessible to the individual subunits in the absence of multimerization are conserved to a large extent in the oligomer, suggesting that the oligomer takes advantage of the intrinsic dynamics of the individual monomers. At the same time, oligomerization stiffens the interfacial regions of the monomers and confers new cooperative modes that exploit the rigid-body translational and rotational degrees of freedom of the intact monomers. The present study sheds light on the mechanism of cooperative inhibition of hexameric N-acetyl-L-glutamate kinase by arginine and on the allosteric regulation of UMP kinases. It also highlights the significance of the particular quaternary design in selectively determining the oligomer dynamics congruent with required ligand-binding and allosteric activities.


Assuntos
Fosfotransferases/química , Fosfotransferases/metabolismo , Regulação Alostérica , Sítio Alostérico , Aminoácidos/metabolismo , Domínio Catalítico , Biologia Computacional , Simulação por Computador , Dimerização , Modelos Moleculares , Simulação de Dinâmica Molecular , Núcleosídeo-Fosfato Quinase/química , Núcleosídeo-Fosfato Quinase/metabolismo , Fosfotransferases (Aceptor do Grupo Carboxila)/química , Fosfotransferases (Aceptor do Grupo Carboxila)/metabolismo , Ligação Proteica , Multimerização Proteica , Estrutura Quaternária de Proteína , Especificidade por Substrato , Termodinâmica
13.
Biosci Biotechnol Biochem ; 76(3): 454-61, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22451384

RESUMO

γ-Glutamyl kinase (GK) is the rate-limiting enzyme in proline synthesis in microorganisms. Most microbial GKs contain an N-terminal kinase domain and a C-terminal pseudouridine synthase and archaeosine transglycosylase (PUA) domain. In contrast, higher eukaryotes possess a bifunctional Δ(1)-pyrroline-5-carboxylate synthetase, which consists of a PUA-free GK domain and a γ-glutamyl phosphate reductase (GPR) domain. Here, to examine the role of the C-terminal region, including the PUA domain of Saccharomyces cerevisiae GK, we constructed a variety of truncated yeast GK and GK/GPR fusion proteins from which the C-terminal region was deleted. A complementation test in Escherichia coli and S. cerevisiae and enzymatic analysis of recombinant proteins revealed that a 67-residue linker sequence between a 255-residue kinase domain and a 106-residue PUA domain is essential for GK activity. It also appeared that 67 or more residues of the C-terminal region, not the PUA domain itself, are required for the full display of GK activity. Further, the GK/GPR fusion protein was functional in E. coli, but decreased stability and Mg-binding ability as compared to wild-type GK. These results suggest that the C-terminal region of S. cerevisiae GK is involved in the folding and/or the stability of the kinase domain.


Assuntos
Fosfotransferases (Aceptor do Grupo Carboxila)/química , Fosfotransferases (Aceptor do Grupo Carboxila)/metabolismo , Saccharomyces cerevisiae/enzimologia , Sequência de Aminoácidos , Escherichia coli/genética , Dados de Sequência Molecular , Fosfotransferases (Aceptor do Grupo Carboxila)/genética , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Saccharomyces cerevisiae/genética , Deleção de Sequência
14.
Curr Microbiol ; 64(2): 164-72, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22101454

RESUMO

Arginine biosynthesis in Corynebacterium glutamicum proceeds via a pathway that is controlled by arginine through feedback inhibition of NAGK, the enzyme that converts N-acetyl-L-glutamate (NAG) to N-acety-L-glutamy-L-phosphate. In this study, the gene argB encoding NAGK from C. glutamicum ATCC 13032 was site-directed, and the L-arginine-binding sites of feedback inhibition in Cglu_NAGK are described. The N-helix and C-terminal residues were first deleted, and the results indicated that they are both necessary for Cglu_NAGK, whereas, the complete N-helix deletion (the front 28 residues) abolished the L-arginine inhibition. Further, we study here the impact on these functions of 12 site-directed mutations affecting seven residues of Cglu_NAGK, chosen on the basis of homology structural alignment. The E19R, H26E, and H268N variants could increase the I0.5 (R) 50-60 fold, and the G287D and R209A mutants could increase the I0.5 (R) 30-40 fold. The E281A mutagenesis resulted in the substrate kinetics being greatly influenced. The W23A variant had a lower specific enzyme activity. These results explained that the five amino acid residues (E19, H26, R209, H268, and G287) located in or near N-helix are all essential for the formation of arginine inhibition.


Assuntos
Arginina/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Corynebacterium glutamicum/enzimologia , Retroalimentação Fisiológica , Fosfotransferases (Aceptor do Grupo Carboxila)/química , Fosfotransferases (Aceptor do Grupo Carboxila)/genética , Sequência de Aminoácidos , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Corynebacterium glutamicum/classificação , Corynebacterium glutamicum/genética , Corynebacterium glutamicum/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fosfotransferases (Aceptor do Grupo Carboxila)/metabolismo , Filogenia , Ligação Proteica , Alinhamento de Sequência
15.
PLoS Comput Biol ; 6(4): e1000738, 2010 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-20386738

RESUMO

N-acetyl-L-glutamate kinase (NAGK) is the structural paradigm for examining the catalytic mechanisms and dynamics of amino acid kinase family members. Given that the slow conformational dynamics of the NAGK (at the microseconds time scale or slower) may be rate-limiting, it is of importance to assess the mechanisms of the most cooperative modes of motion intrinsically accessible to this enzyme. Here, we present the results from normal mode analysis using an elastic network model representation, which shows that the conformational mechanisms for substrate binding by NAGK strongly correlate with the intrinsic dynamics of the enzyme in the unbound form. We further analyzed the potential mechanisms of allosteric signalling within NAGK using a Markov model for network communication. Comparative analysis of the dynamics of family members strongly suggests that the low-frequency modes of motion and the associated intramolecular couplings that establish signal transduction are highly conserved among family members, in support of the paradigm sequence-->structure-->dynamics-->function.


Assuntos
Biologia Computacional/métodos , Simulação de Dinâmica Molecular , Fosfotransferases (Aceptor do Grupo Carboxila)/química , Regulação Alostérica , Anisotropia , Domínio Catalítico , Cadeias de Markov , Distribuição Normal , Fosfotransferases (Aceptor do Grupo Carboxila)/metabolismo , Transdução de Sinais , Relação Estrutura-Atividade
16.
Sci Rep ; 11(1): 12535, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-34131190

RESUMO

PII proteins constitute a widespread signal transduction superfamily in the prokaryotic world. The canonical PII signal proteins sense metabolic state of the cells by binding the metabolite molecules ATP, ADP and 2-oxoglutarate. Depending on bound effector molecule, PII proteins interact with and modulate the activity of multiple target proteins. To investigate the complexity of interactions of PII with target proteins, analytical methods that do not disrupt the native cellular context are required. To this purpose, split luciferase proteins have been used to develop a novel complementation reporter called NanoLuc Binary Technology (NanoBiT). The luciferase NanoLuc is divided in two subunits: a 18 kDa polypeptide termed "Large BiT" and a 1.3 kDa peptide termed "Small BiT", which only weakly associate. When fused to proteins of interest, they reconstitute an active luciferase when the proteins of interest interact. Therefore, we set out to develop a new NanoBiT sensor based on the interaction of PII protein from Synechocystis sp. PCC6803 with PII-interacting protein X (PipX) and N-acetyl-L-glutamate kinase (NAGK). The novel NanoBiT sensor showed unprecedented sensitivity, which made it possible to detect even weak and transient interactions between PII variants and their interacting partners, thereby shedding new light in PII signalling processes.


Assuntos
Proteínas de Bactérias/química , Técnicas Biossensoriais , Proteínas PII Reguladoras de Nitrogênio/isolamento & purificação , Fosfotransferases (Aceptor do Grupo Carboxila)/química , Difosfato de Adenosina/química , Trifosfato de Adenosina/química , Ácidos Cetoglutáricos/química , Nanotecnologia , Proteínas PII Reguladoras de Nitrogênio/química , Synechococcus/química
17.
Artigo em Inglês | MEDLINE | ID: mdl-20383005

RESUMO

Carbamate kinase catalyzes the reversible conversion of carbamoyl phosphate and ADP to ATP and ammonium carbamate, which is hydrolyzed to ammonia and carbonate. The three-dimensional structure of carbamate kinase from the human parasite Giardia lamblia (glCK) has been determined at 3 A resolution. The crystals belonged to the monoclinic space group P2(1), with unit-cell parameters a = 69.77, b = 85.41, c = 102.1 A, beta = 106.8 degrees . The structure was refined to a final R factor of 0.227. The essentiality of glCK together with its absence in humans makes the enzyme an attractive candidate for anti-Giardia drug development. Steady-state kinetic rate constants have been determined. The k(cat) for ATP formation is 319 +/- 9 s(-1). The K(m) values for carbamoyl phosphate and ADP are 85 +/- 6 and 70 +/- 5 microM, respectively. The structure suggests that three invariant lysine residues (Lys131, Lys216 and Lys278) may be involved in the binding of substrates and phosphoryl transfer. The structure of glCK reveals that a glycerol molecule binds in the likely carbamoyl phosphate-binding site.


Assuntos
Giardia lamblia/enzimologia , Fosfotransferases (Aceptor do Grupo Carboxila)/química , Carbamoil-Fosfato/química , Carbamoil-Fosfato/metabolismo , Domínio Catalítico , Cristalografia por Raios X , Glicerol/química , Glicerol/metabolismo , Modelos Moleculares , Fosfotransferases (Aceptor do Grupo Carboxila)/isolamento & purificação , Fosfotransferases (Aceptor do Grupo Carboxila)/metabolismo , Estrutura Quaternária de Proteína , Homologia Estrutural de Proteína
18.
Eukaryot Cell ; 8(3): 262-70, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19114501

RESUMO

We report characterization of the gene encoding putative transcription factor PRO1, identified in transcriptional profiling studies as being downregulated in the chestnut blight fungus Cryphonectria parasitica in response to infection by virulence-attenuating hypoviruses. Sequence analysis confirmed that pro1 encodes a Zn(II)(2)Cys(6) binuclear cluster DNA binding protein with significant sequence similarity to the pro1 gene product that controls fruiting body development in Sordaria macrospora. Targeted disruption of the C. parasitica pro1 gene resulted in two phenotypic changes that also accompany hypovirus infection, a significant reduction in asexual sporulation that could be reversed by exposure to high light intensity, and loss of female fertility. The pro1 disruption mutant, however, retained full virulence. Although hypovirus CHV1-EP713 infection was established in the pro1 disruption mutant, infected colonies continually produced virus-free sectors, suggesting that PRO1 is required for stable maintenance of hypovirus infection. These results complement the recent characterization of the hypovirus-responsive homologue of the Saccharomyces cerevisiae Ste12 C(2)H(2) zinc finger transcription factor gene, cpst12, which was shown to be required for C. parasitica female fertility and virulence.


Assuntos
Ascomicetos/enzimologia , Ascomicetos/virologia , Proteínas Fúngicas/metabolismo , Fosfotransferases (Aceptor do Grupo Carboxila)/metabolismo , Doenças das Plantas/microbiologia , Vírus de RNA/fisiologia , Reprodução Assexuada , Aesculus/microbiologia , Sequência de Aminoácidos , Ascomicetos/patogenicidade , Ascomicetos/fisiologia , Proteínas Fúngicas/química , Proteínas Fúngicas/genética , Dados de Sequência Molecular , Fosfotransferases (Aceptor do Grupo Carboxila)/química , Fosfotransferases (Aceptor do Grupo Carboxila)/genética , Alinhamento de Sequência , Esporos Fúngicos/enzimologia , Esporos Fúngicos/genética , Esporos Fúngicos/fisiologia , Esporos Fúngicos/virologia , Virulência
19.
FEBS J ; 287(3): 465-482, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31287617

RESUMO

During evolution, several algae and plants became heterotrophic and lost photosynthesis; however, in most cases, a nonphotosynthetic plastid was maintained. Among these organisms, the colourless alga Polytomella parva is a special case, as its plastid is devoid of any DNA, but is maintained for specific metabolic tasks carried out by nuclear encoded enzymes. This makes P. parva attractive to study molecular events underlying the transition from autotrophic to heterotrophic lifestyle. Here we characterize metabolic adaptation strategies of P. parva in comparison to the closely related photosynthetic alga Chlamydomonas reinhardtii with a focus on the role of plastid-localized PII signalling protein. Polytomella parva accumulates significantly higher amounts of most TCA cycle intermediates as well as glutamate, aspartate and arginine, the latter being specific for the colourless plastid. Correlating with the altered metabolite status, the carbon/nitrogen sensory PII signalling protein and its regulatory target N-acetyl-l-glutamate-kinase (NAGK; the controlling enzyme of arginine biosynthesis) show unique features: They have co-evolved into a stable hetero-oligomeric complex, irrespective of effector molecules. The PII signalling protein, so far known as a transiently interacting signalling protein, appears as a permanent subunit of the enzyme NAGK. NAGK requires PII to properly sense the feedback inhibitor arginine, and moreover, PII tunes arginine-inhibition in response to glutamine. No other PII effector molecules interfere, indicating that the PII-NAGK system in P. parva has lost the ability to estimate the cellular energy and carbon status but has specialized to provide an entirely glutamine-dependent arginine feedback control, highlighting the evolutionary plasticity of PII signalling system.


Assuntos
Clorofíceas/metabolismo , Evolução Molecular , Proteínas PII Reguladoras de Nitrogênio/metabolismo , Fosfotransferases (Aceptor do Grupo Carboxila)/metabolismo , Proteínas de Plantas/metabolismo , Arginina/metabolismo , Chlamydomonas reinhardtii/metabolismo , Clorofíceas/genética , Retroalimentação Fisiológica , Proteínas PII Reguladoras de Nitrogênio/química , Proteínas PII Reguladoras de Nitrogênio/genética , Fosfotransferases (Aceptor do Grupo Carboxila)/química , Fosfotransferases (Aceptor do Grupo Carboxila)/genética , Proteínas de Plantas/química , Proteínas de Plantas/genética , Ligação Proteica , Multimerização Proteica
20.
J Bacteriol ; 191(8): 2521-9, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19201797

RESUMO

The enzymatic transfer of phosphoryl groups is central to the control of many cellular processes. One of the phosphoryl transfer mechanisms, that of acetate kinase, is not completely understood. Besides better understanding of the mechanism of acetate kinase, knowledge of the structure of butyrate kinase 2 (Buk2) will aid in the interpretation of active-site structure and provide information on the structural basis of substrate specificity. The gene buk2 from Thermotoga maritima encodes a member of the ASKHA (acetate and sugar kinases/heat shock cognate/actin) superfamily of phosphotransferases. The encoded protein Buk2 catalyzes the phosphorylation of butyrate and isobutyrate. We have determined the 2.5-A crystal structure of Buk2 complexed with (beta,gamma-methylene) adenosine 5'-triphosphate. Buk2 folds like an open-shelled clam, with each of the two domains representing one of the two shells. In the open active-site cleft between the N- and C-terminal domains, the active-site residues consist of two histidines, two arginines, and a cluster of hydrophobic residues. The ATP binding region of Buk2 in the C-terminal domain consists of abundant glycines for nucleotide binding, and the ATP binding motif is similar to those of other members of the ASKHA superfamily. The enzyme exists as an octamer, in which four disulfide bonds form between intermolecular cysteines. Sequence alignment and structure superposition identify the simplicity of the monomeric Buk2 structure, a probable substrate binding site, the key residues in catalyzing phosphoryl transfer, and the substrate specificity differences among Buk2, acetate, and propionate kinases. The possible enzyme mechanisms are discussed.


Assuntos
Proteínas de Bactérias/química , Fosfotransferases (Aceptor do Grupo Carboxila)/química , Thermotoga maritima/enzimologia , Sítios de Ligação , Domínio Catalítico , Cristalografia por Raios X , Modelos Moleculares , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa