Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Biol Inorg Chem ; 24(6): 841-848, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31218442

RESUMO

Ribonucleotide reductase (RNR) has been extensively probed as a target enzyme in the search for selective antibiotics. Here we report on the mechanism of inhibition of nine compounds, serving as representative examples of three different inhibitor classes previously identified by us to efficiently inhibit RNR. The interaction between the inhibitors and Pseudomonas aeruginosa RNR was elucidated using a combination of electron paramagnetic resonance spectroscopy and thermal shift analysis. All nine inhibitors were found to efficiently quench the tyrosyl radical present in RNR, required for catalysis. Three different mechanisms of radical quenching were identified, and shown to depend on reduction potential of the assay solution and quaternary structure of the protein complex. These results form a good foundation for further development of P. aeruginosa selective antibiotics. Moreover, this study underscores the complex nature of RNR inhibition and the need for detailed spectroscopic studies to unravel the mechanism of RNR inhibitors.


Assuntos
Radicais Livres/química , Radicais Livres/metabolismo , Pseudomonas aeruginosa/enzimologia , Ribonucleotídeo Redutases/metabolismo , Tirosina/química , Tirosina/metabolismo , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo , Ribonucleotídeo Redutases/genética
2.
J Biol Chem ; 293(41): 15889-15900, 2018 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-30166338

RESUMO

Class I ribonucleotide reductase (RNR) consists of a catalytic subunit (NrdA) and a radical-generating subunit (NrdB) that together catalyze reduction of ribonucleotides to their corresponding deoxyribonucleotides. NrdB from the firmicute Facklamia ignava is a unique fusion protein with N-terminal add-ons of a glutaredoxin (Grx) domain followed by an ATP-binding domain, the ATP cone. Grx, usually encoded separately from the RNR operon, is a known RNR reductant. We show that the fused Grx domain functions as an efficient reductant of the F. ignava class I RNR via the common dithiol mechanism and, interestingly, also via a monothiol mechanism, although less efficiently. To our knowledge, a Grx that uses both of these two reaction mechanisms has not previously been observed with a native substrate. The ATP cone is in most RNRs an N-terminal domain of the catalytic subunit. It is an allosteric on/off switch promoting ribonucleotide reduction in the presence of ATP and inhibiting RNR activity in the presence of dATP. We found that dATP bound to the ATP cone of F. ignava NrdB promotes formation of tetramers that cannot form active complexes with NrdA. The ATP cone bound two dATP molecules but only one ATP molecule. F. ignava NrdB contains the recently identified radical-generating cofactor MnIII/MnIV We show that NrdA from F. ignava can form a catalytically competent RNR with the MnIII/MnIV-containing NrdB from the flavobacterium Leeuwenhoekiella blandensis In conclusion, F. ignava NrdB is fused with a Grx functioning as an RNR reductant and an ATP cone serving as an on/off switch.


Assuntos
Glutarredoxinas/metabolismo , Ribonucleotídeo Redutases/metabolismo , Aerococcaceae/química , Catálise , Nucleotídeos de Desoxiadenina/metabolismo , Flavobacteriaceae/química , Transferência Genética Horizontal , Glutarredoxinas/química , Glutarredoxinas/genética , Oxirredução , Ligação Proteica , Domínios Proteicos , Multimerização Proteica/efeitos dos fármacos , Ribonucleotídeo Redutases/genética
3.
Elife ; 72018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29388911

RESUMO

Ribonucleotide reductases (RNRs) are key enzymes in DNA metabolism, with allosteric mechanisms controlling substrate specificity and overall activity. In RNRs, the activity master-switch, the ATP-cone, has been found exclusively in the catalytic subunit. In two class I RNR subclasses whose catalytic subunit lacks the ATP-cone, we discovered ATP-cones in the radical-generating subunit. The ATP-cone in the Leeuwenhoekiella blandensis radical-generating subunit regulates activity via quaternary structure induced by binding of nucleotides. ATP induces enzymatically competent dimers, whereas dATP induces non-productive tetramers, resulting in different holoenzymes. The tetramer forms by interactions between ATP-cones, shown by a 2.45 Å crystal structure. We also present evidence for an MnIIIMnIV metal center. In summary, lack of an ATP-cone domain in the catalytic subunit was compensated by transfer of the domain to the radical-generating subunit. To our knowledge, this represents the first observation of transfer of an allosteric domain between components of the same enzyme complex.


Assuntos
Trifosfato de Adenosina/metabolismo , Flavobacteriaceae/enzimologia , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Ribonucleotídeo Redutases/química , Ribonucleotídeo Redutases/metabolismo , Regulação Alostérica , Cristalografia por Raios X , Conformação Proteica , Multimerização Proteica
4.
J Biol Chem ; 292(46): 19044-19054, 2017 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-28972190

RESUMO

Ribonucleotide reductases (RNRs) catalyze the reduction of ribonucleotides to the corresponding deoxyribonucleotides, used in DNA synthesis and repair. Two different mechanisms help deliver the required electrons to the RNR active site. Formate can be used as reductant directly in the active site, or glutaredoxins or thioredoxins reduce a C-terminal cysteine pair, which then delivers the electrons to the active site. Here, we characterized a novel cysteine-rich C-terminal domain (CRD), which is present in most class II RNRs found in microbes. The NrdJd-type RNR from the bacterium Stackebrandtia nassauensis was used as a model enzyme. We show that the CRD is involved in both higher oligomeric state formation and electron transfer to the active site. The CRD-dependent formation of high oligomers, such as tetramers and hexamers, was induced by addition of dATP or dGTP, but not of dTTP or dCTP. The electron transfer was mediated by an array of six cysteine residues at the very C-terminal end, which also coordinated a zinc atom. The electron transfer can also occur between subunits, depending on the enzyme's oligomeric state. An investigation of the native reductant of the system revealed no interaction with glutaredoxins or thioredoxins, indicating that this class II RNR uses a different electron source. Our results indicate that the CRD has a crucial role in catalytic turnover and a potentially new terminal reduction mechanism and suggest that the CRD is important for the activities of many class II RNRs.


Assuntos
Actinomycetales/química , Proteínas de Bactérias/química , Cisteína/química , Ribonucleotídeo Redutases/química , Dedos de Zinco , Actinomycetales/genética , Actinomycetales/metabolismo , Regulação Alostérica , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Domínio Catalítico , Cristalografia por Raios X , Cisteína/genética , Cisteína/metabolismo , Transporte de Elétrons , Modelos Moleculares , Oxirredução , Filogenia , Domínios Proteicos , Multimerização Proteica , Ribonucleotídeo Redutases/genética , Ribonucleotídeo Redutases/metabolismo
5.
Mol Oncol ; 10(9): 1375-1386, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27511871

RESUMO

Ribonucleotide Reductase (RNR) is the sole enzyme that catalyzes the reduction of ribonucleotides into deoxyribonucleotides. Even though RNR is a recognized target for antiproliferative molecules, and the main target of the approved drug hydroxyurea, few new leads targeted to this enzyme have been developed. We have evaluated a recently identified set of RNR inhibitors with respect to inhibition of the human enzyme and cellular toxicity. One compound, NSC73735, is particularly interesting; it is specific for leukemia cells and is the first identified compound that hinders oligomerization of the mammalian large RNR subunit. Similar to hydroxyurea, it caused a disruption of the cell cycle distribution of cultured HL-60 cells. In contrast to hydroxyurea, the disruption was reversible, indicating higher specificity. NSC73735 thus defines a potential lead candidate for RNR-targeted anticancer drugs, as well as a chemical probe with better selectivity for RNR inhibition than hydroxyurea.


Assuntos
Desoxirribonucleosídeos/farmacologia , Inibidores Enzimáticos/farmacologia , Ribonucleotídeo Redutases/antagonistas & inibidores , Bioensaio , Ciclo Celular/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/química , Citometria de Fluxo , Regulação da Expressão Gênica/efeitos dos fármacos , Células HL-60 , Humanos , Hidroxiureia/farmacologia , Estrutura Quaternária de Proteína , Subunidades Proteicas/antagonistas & inibidores , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Ribonucleotídeo Redutases/química , Ribonucleotídeo Redutases/metabolismo , Temperatura
6.
PLoS One ; 10(7): e0134293, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26225432

RESUMO

The opportunistic pathogen Pseudomonas aeruginosa can grow under both aerobic and anaerobic conditions. Its flexibility with respect to oxygen load is reflected by the fact that its genome encodes all three existing classes of ribonucleotides reductase (RNR): the oxygen-dependent class I RNR, the oxygen-indifferent class II RNR, and the oxygen-sensitive class III RNR. The P. aeruginosa class II RNR is expressed as two separate polypeptides (NrdJa and NrdJb), a unique example of a split RNR enzyme in a free-living organism. A split class II RNR is also found in a few closely related γ-Proteobacteria. We have characterized the P. aeruginosa class II RNR and show that both subunits are required for formation of a biologically functional enzyme that can sustain vitamin B12-dependent growth. Binding of the B12 coenzyme as well as substrate and allosteric effectors resides in the NrdJa subunit, whereas the NrdJb subunit mediates efficient reductive dithiol exchange during catalysis. A combination of activity assays and activity-independent methods like surface plasmon resonance and gas phase electrophoretic macromolecule analysis suggests that the enzymatically active form of the enzyme is a (NrdJa-NrdJb)2 homodimer of heterodimers, and a combination of hydrogen-deuterium exchange experiments and molecular modeling suggests a plausible region in NrdJa that interacts with NrdJb. Our detailed characterization of the split NrdJ from P. aeruginosa provides insight into the biochemical function of a unique enzyme known to have central roles in biofilm formation and anaerobic growth.


Assuntos
Pseudomonas aeruginosa/enzimologia , Ribonucleotídeo Redutases/metabolismo , Ligação Proteica
7.
J Biol Chem ; 290(28): 17339-48, 2015 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-25971975

RESUMO

Ribonucleotide reductase (RNR) catalyzes the reduction of ribonucleotides to the corresponding deoxyribonucleotides, which are used as building blocks for DNA replication and repair. This process is tightly regulated via two allosteric sites, the specificity site (s-site) and the overall activity site (a-site). The a-site resides in an N-terminal ATP cone domain that binds dATP or ATP and functions as an on/off switch, whereas the composite s-site binds ATP, dATP, dTTP, or dGTP and determines which substrate to reduce. There are three classes of RNRs, and class I RNRs consist of different combinations of α and ß subunits. In eukaryotic and Escherichia coli class I RNRs, dATP inhibits enzyme activity through the formation of inactive α6 and α4ß4 complexes, respectively. Here we show that the Pseudomonas aeruginosa class I RNR has a duplicated ATP cone domain and represents a third mechanism of overall activity regulation. Each α polypeptide binds three dATP molecules, and the N-terminal ATP cone is critical for binding two of the dATPs because a truncated protein lacking this cone could only bind dATP to its s-site. ATP activates the enzyme solely by preventing dATP from binding. The dATP-induced inactive form is an α4 complex, which can interact with ß2 to form a non-productive α4ß2 complex. Other allosteric effectors induce a mixture of α2 and α4 forms, with the former being able to interact with ß2 to form active α2ß2 complexes. The unique features of the P. aeruginosa RNR are interesting both from evolutionary and drug discovery perspectives.


Assuntos
Proteínas de Bactérias/metabolismo , Pseudomonas aeruginosa/enzimologia , Ribonucleotídeo Redutases/metabolismo , Trifosfato de Adenosina/metabolismo , Regulação Alostérica , Sítio Alostérico , Sequência de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Nucleotídeos de Desoxiadenina/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Cinética , Dados de Sequência Molecular , Estrutura Quaternária de Proteína , Subunidades Proteicas , Pseudomonas aeruginosa/genética , Ribonucleotídeo Redutases/química , Ribonucleotídeo Redutases/genética , Deleção de Sequência
8.
Nano Lett ; 13(4): 1393-8, 2013 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-23520995

RESUMO

In the present study, we introduce a novel method for in vivo imaging of the biodistribution of single wall carbon nanotubes (SWNTs) labeled with recombinant thermo-stable Luciola cruciata luciferase (LcL). In addition, we highlight a new application for green fluorescent proteins in which they are utilized as imaging moieties for SWNTs. Carbon nanotubes show great positive potential compared to other drug nanocarriers with respect to loading capacity, cell internalization, and biodegradability. We have also studied the effect of binding mode (chemical conjugation and physical adsorption) on the chemiluminescence activity, decay rate, and half-life. We have shown that through proper chemical conjugation of LcL to CNTs, LcL remained biologically active for the catalysis of d-luciferin in the presence of ATP to release detectable amounts of photons for in vivo imaging. Chemiluminescence of LcL allows imaging of CNTs and their cargo in nonsuperficial locations at an organ resolution with no need of an excitation source. Loading LcL-CNTs with the antitumor antibiotic doxorubicin did not alter their biological activity for imaging. In vivo imaging of LcL-CNTs has been carried out using "IVIS spectrum" showing the uptake of LcL-CNTs by different organs in mice. We believe that the LcL-CNT system is an advanced powerful tool for in vivo imaging and therefore a step toward the advancement of the nanomedicine field.


Assuntos
Diagnóstico por Imagem , Doxorrubicina/química , Luciferases/química , Nanotubos de Carbono/química , Animais , Estabilidade Enzimática , Vaga-Lumes/enzimologia , Camundongos , Coloração e Rotulagem , Temperatura
9.
J Biol Chem ; 288(12): 8198-8208, 2013 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-23372162

RESUMO

Ribonucleotide reductases (RNRs) catalyze the only pathway for de novo synthesis of deoxyribonucleotides needed for DNA replication and repair. The vast majority of eukaryotes encodes only a class I RNR, but interestingly some eukaryotes, including the social amoeba Dictyostelium discoideum, encode both a class I and a class II RNR. The amino acid sequence of the D. discoideum class I RNR is similar to other eukaryotic RNRs, whereas that of its class II RNR is most similar to the monomeric class II RNRs found in Lactobacillus spp. and a few other bacteria. Here we report the first study of RNRs in a eukaryotic organism that encodes class I and class II RNRs. Both classes of RNR genes were expressed in D. discoideum cells, although the class I transcripts were more abundant and strongly enriched during mid-development compared with the class II transcript. The quaternary structure, allosteric regulation, and properties of the diiron-oxo/radical cofactor of D. discoideum class I RNR are similar to those of the mammalian RNRs. Inhibition of D. discoideum class I RNR by hydroxyurea resulted in a 90% reduction in spore formation and decreased the germination viability of the surviving spores by 75%. Class II RNR could not compensate for class I inhibition during development, and an excess of vitamin B12 coenzyme, which is essential for class II activity, did not improve spore formation. We suggest that class I is the principal RNR during D. discoideum development and growth and is important for spore formation, possibly by providing dNTPs for mitochondrial replication.


Assuntos
Dictyostelium/enzimologia , Proteínas de Protozoários/metabolismo , Ribonucleotídeo Redutases/metabolismo , Regulação Alostérica , Complexos de Coordenação/química , Cistina Difosfato/química , Dictyostelium/genética , Dictyostelium/fisiologia , Inibidores Enzimáticos/farmacologia , Radicais Livres/química , Expressão Gênica , Regulação Enzimológica da Expressão Gênica , Guanosina Difosfato/química , Ferro/química , Cinética , Filogenia , Proteínas de Protozoários/genética , Ribonucleotídeo Redutases/antagonistas & inibidores , Ribonucleotídeo Redutases/química , Ribonucleotídeo Redutases/genética , Espectrofotometria Ultravioleta , Esporos de Protozoários/enzimologia , Esporos de Protozoários/genética , Tirosina/química
10.
Crit Rev Biochem Mol Biol ; 47(1): 50-63, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22050358

RESUMO

Ribonucleotide reductase (RNR) is the only source for de novo production of the four deoxyribonucleoside triphosphate (dNTP) building blocks needed for DNA synthesis and repair. It is crucial that these dNTP pools are carefully balanced, since mutation rates increase when dNTP levels are either unbalanced or elevated. RNR is the major player in this homeostasis, and with its four different substrates, four different allosteric effectors and two different effector binding sites, it has one of the most sophisticated allosteric regulations known today. In the past few years, the structures of RNRs from several bacteria, yeast and man have been determined in the presence of allosteric effectors and substrates, revealing new information about the mechanisms behind the allosteric regulation. A common theme for all studied RNRs is a flexible loop that mediates modulatory effects from the allosteric specificity site (s-site) to the catalytic site for discrimination between the four substrates. Much less is known about the allosteric activity site (a-site), which functions as an on-off switch for the enzyme's overall activity by binding ATP (activator) or dATP (inhibitor). The two nucleotides induce formation of different enzyme oligomers, and a recent structure of a dATP-inhibited α(6)ß(2) complex from yeast suggested how its subunits interacted non-productively. Interestingly, the oligomers formed and the details of their allosteric regulation differ between eukaryotes and Escherichia coli. Nevertheless, these differences serve a common purpose in an essential enzyme whose allosteric regulation might date back to the era when the molecular mechanisms behind the central dogma evolved.


Assuntos
Sítio Alostérico/fisiologia , DNA/biossíntese , Desoxirribonucleotídeos/metabolismo , Taxa de Mutação , Ribonucleotídeo Redutases/metabolismo , Trifosfato de Adenosina/metabolismo , Regulação Alostérica/fisiologia , Domínio Catalítico/fisiologia , Nucleotídeos de Desoxiadenina/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Lactobacillus leichmannii/genética , Lactobacillus leichmannii/metabolismo , Modelos Moleculares , Conformação Proteica , Ribonucleotídeo Redutases/química , Leveduras/genética , Leveduras/metabolismo
11.
J Biol Chem ; 286(38): 33053-60, 2011 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-21832039

RESUMO

Bacillus anthracis is a severe mammalian pathogen encoding a class Ib ribonucleotide reductase (RNR). RNR is a universal enzyme that provides the four essential deoxyribonucleotides needed for DNA replication and repair. Almost all Bacillus spp. encode both class Ib and class III RNR operons, but the B. anthracis class III operon was reported to encode a pseudogene, and conceivably class Ib RNR is necessary for spore germination and proliferation of B. anthracis upon infection. The class Ib RNR operon in B. anthracis encodes genes for the catalytic NrdE protein, the tyrosyl radical metalloprotein NrdF, and the flavodoxin protein NrdI. The tyrosyl radical in NrdF is stabilized by an adjacent Mn(2)(III) site (Mn-NrdF) formed by the action of the NrdI protein or by a Fe(2)(III) site (Fe-NrdF) formed spontaneously from Fe(2+) and O(2). In this study, we show that the properties of B. anthracis Mn-NrdF and Fe-NrdF are in general similar for interaction with NrdE and NrdI. Intriguingly, the enzyme activity of Mn-NrdF was approximately an order of magnitude higher than that of Fe-NrdF in the presence of the class Ib-specific physiological reductant NrdH, strongly suggesting that the Mn-NrdF form is important in the life cycle of B. anthracis. Whether the Fe-NrdF form only exists in vitro or whether the NrdF protein in B. anthracis is a true cambialistic enzyme that can work with either manganese or iron remains to be established.


Assuntos
Bacillus anthracis/enzimologia , Proteínas de Bactérias/metabolismo , Manganês/metabolismo , Ribonucleotídeo Redutases/metabolismo , Apoproteínas/metabolismo , Flavodoxina/metabolismo , Holoenzimas/metabolismo , Ferro/metabolismo , Ligação Proteica , Estrutura Quaternária de Proteína , Espectrofotometria Ultravioleta , Ressonância de Plasmônio de Superfície
12.
Nucleic Acids Res ; 39(4): 1381-9, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20972217

RESUMO

Ribonucleotide reductase (RNR) is a critical enzyme of nucleotide metabolism, synthesizing precursors for DNA replication and repair. In prokaryotic genomes, RNR genes are commonly targeted by mobile genetic elements, including free standing and intron-encoded homing endonucleases and inteins. Here, we describe a unique molecular solution to assemble a functional product from the RNR large subunit gene, nrdA that has been fragmented into two smaller genes by the insertion of mobE, a mobile endonuclease. We show that unique sequences that originated during the mobE insertion and that are present as C- and N-terminal tails on the split NrdA-a and NrdA-b polypeptides, are absolutely essential for enzymatic activity. Our data are consistent with the tails functioning as protein interaction domains to assemble the tetrameric (NrdA-a/NrdA-b)(2) large subunit necessary for a functional RNR holoenzyme. The tails represent a solution distinct from RNA and protein splicing or programmed DNA rearrangements to restore function from a fragmented coding region and may represent a general mechanism to neutralize fragmentation of essential genes by mobile genetic elements.


Assuntos
Sequências Repetitivas Dispersas , Ribonucleotídeo Redutases/química , Ribonucleotídeo Redutases/genética , Bacteriófagos/enzimologia , Domínio Catalítico , Dimerização , Holoenzimas/genética , Mutação , Domínios e Motivos de Interação entre Proteínas , Ribonucleotídeo Redutases/metabolismo
13.
Protein Eng Des Sel ; 23(8): 633-41, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20534631

RESUMO

Ribonucleotide reductase (RNR) synthesizes deoxyribonucleotides for DNA replication and repair and is controlled by sophisticated allosteric regulation involving differential affinity of nucleotides for regulatory sites. We have developed a robust and sensitive method for coupling biotinylated RNRs to surface plasmon resonance streptavidin biosensor chips via a 30.5 A linker. In comprehensive studies on three RNRs effector nucleotides strengthened holoenzyme interactions, whereas substrate had no effect on subunit interactions. The RNRs differed in their response to the negative allosteric effector dATP that binds to an ATP-cone domain. A tight RNR complex was formed in Escherichia coli class Ia RNR with a functional ATP cone. No strengthening of subunit interactions was observed in the class Ib RNR from the human pathogen Bacillus anthracis that lacks the ATP cone. A moderate strengthening was seen in the atypical Aeromonas hydrophila phage 1 class Ia RNR that has a split catalytic subunit and a non-functional ATP cone with remnant dATP-mediated regulatory features. We also successfully immobilized a functional catalytic NrdA subunit of the E.coli enzyme, facilitating study of nucleotide interactions. Our surface plasmon resonance methodology has the potential to provide biological insight into nucleotide-mediated regulation of any RNR, and can be used for high-throughput screening of potential RNR inhibitors.


Assuntos
Proteínas de Bactérias/metabolismo , Técnicas Biossensoriais , Enzimas Imobilizadas/metabolismo , Ribonucleotídeo Redutases/metabolismo , Ressonância de Plasmônio de Superfície , Trifosfato de Adenosina/metabolismo , Regulação Alostérica , Bacillus anthracis/enzimologia , Bacillus anthracis/genética , Bacillus anthracis/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Sítios de Ligação , Biotina/química , Biotina/metabolismo , Enzimas Imobilizadas/química , Enzimas Imobilizadas/genética , Escherichia coli/enzimologia , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Ligação Proteica , Ribonucleosídeo Difosfato Redutase/química , Ribonucleosídeo Difosfato Redutase/genética , Ribonucleosídeo Difosfato Redutase/metabolismo , Ribonucleotídeo Redutases/química , Ribonucleotídeo Redutases/genética , Estreptavidina/química , Estreptavidina/metabolismo , Especificidade por Substrato
14.
J Biol Chem ; 283(51): 35310-8, 2008 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-18835811

RESUMO

Ribonucleotide reductase (RNR) is a key enzyme for the synthesis of the four DNA building blocks. Class Ia RNRs contain two subunits, denoted R1 (alpha) and R2 (beta). These enzymes are regulated via two nucleotide-binding allosteric sites on the R1 subunit, termed the specificity and overall activity sites. The specificity site binds ATP, dATP, dTTP, or dGTP and determines the substrate to be reduced, whereas the overall activity site binds dATP (inhibitor) or ATP. By using gas-phase electrophoretic mobility macromolecule analysis and enzyme assays, we found that the Escherichia coli class Ia RNR formed an inhibited alpha(4)beta(4) complex in the presence of dATP and an active alpha(2)beta(2) complex in the presence of ATP (main substrate: CDP), dTTP (substrate: GDP) or dGTP (substrate: ADP). The R1-R2 interaction was 30-50 times stronger in the alpha(4)beta(4) complex than in the alpha(2)beta(2) complex, which was in equilibrium with free alpha(2) and beta(2) subunits. Studies of a known E. coli R1 mutant (H59A) showed that deficient dATP inhibition correlated with reduced ability to form alpha(4)beta(4) complexes. ATP could also induce the formation of a generally inhibited alpha(4)beta(4) complex in the E. coli RNR but only when used in combination with high concentrations of the specificity site effectors, dTTP/dGTP. Both allosteric sites are therefore important for alpha(4)beta(4) formation and overall activity regulation. The E. coli RNR differs from the mammalian enzyme, which is stimulated by ATP also in combination with dGTP/dTTP and forms active and inactive alpha(6)beta(2) complexes.


Assuntos
Sítio Alostérico/fisiologia , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimologia , Nucleotídeos/metabolismo , Ribonucleotídeo Redutases/metabolismo , Regulação Alostérica/fisiologia , Proteínas de Escherichia coli/genética , Estrutura Quaternária de Proteína/fisiologia , Especificidade por Substrato/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...