Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros

Intervalo de año de publicación
1.
Nature ; 563(7731): 416-420, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30429545

RESUMEN

Ribonucleotide reductase (RNR) catalyses the only known de novo pathway for the production of all four deoxyribonucleotides that are required for DNA synthesis1,2. It is essential for all organisms that use DNA as their genetic material and is a current drug target3,4. Since the discovery that iron is required for function in the aerobic, class I RNR found in all eukaryotes and many bacteria, a dinuclear metal site has been viewed as necessary to generate and stabilize the catalytic radical that is essential for RNR activity5-7. Here we describe a group of RNR proteins in Mollicutes-including Mycoplasma pathogens-that possess a metal-independent stable radical residing on a modified tyrosyl residue. Structural, biochemical and spectroscopic characterization reveal a stable 3,4-dihydroxyphenylalanine (DOPA) radical species that directly supports ribonucleotide reduction in vitro and in vivo. This observation overturns the presumed requirement for a dinuclear metal site in aerobic ribonucleotide reductase. The metal-independent radical requires new mechanisms for radical generation and stabilization, processes that are targeted by RNR inhibitors. It is possible that this RNR variant provides an advantage under metal starvation induced by the immune system. Organisms that encode this type of RNR-some of which are developing resistance to antibiotics-are involved in diseases of the respiratory, urinary and genital tracts. Further characterization of this RNR family and its mechanism of cofactor generation will provide insight into new enzymatic chemistry and be of value in devising strategies to combat the pathogens that utilize it. We propose that this RNR subclass is denoted class Ie.


Asunto(s)
Dihidroxifenilalanina/química , Dihidroxifenilalanina/metabolismo , Metales , Mycoplasma/metabolismo , Ribonucleótidos/metabolismo , Secuencia de Aminoácidos , Escherichia coli/enzimología , Escherichia coli/genética , Escherichia coli/metabolismo , Sistema Inmunológico/metabolismo , Hierro/metabolismo , Metales/metabolismo , Modelos Moleculares , Mycoplasma/efectos de los fármacos , Mycoplasma/enzimología , Mycoplasma/genética , Operón/genética , Oxidación-Reducción , Ribonucleótido Reductasas/química , Ribonucleótido Reductasas/metabolismo , Ribonucleótidos/química , Tirosina/química , Tirosina/metabolismo
2.
Biochemistry ; 61(2): 92-106, 2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-34941255

RESUMEN

Ribonucleotide reductase (RNR) is an essential enzyme with a complex mechanism of allosteric regulation found in nearly all living organisms. Class I RNRs are composed of two proteins, a large α-subunit (R1) and a smaller ß-subunit (R2) that exist as homodimers, that combine to form an active heterotetramer. Aquifex aeolicus is a hyperthermophilic bacterium with an unusual RNR encoding a 346-residue intein in the DNA sequence encoding its R2 subunit. We present the first structures of the A. aeolicus R1 and R2 (AaR1 and AaR2, respectively) proteins as well as the biophysical and biochemical characterization of active and inactive A. aeolicus RNR. While the active oligomeric state and activity regulation of A. aeolicus RNR are similar to those of other characterized RNRs, the X-ray crystal structures also reveal distinct features and adaptations. Specifically, AaR1 contains a ß-hairpin hook structure at the dimer interface, which has an interesting π-stacking interaction absent in other members of the NrdAh subclass, and its ATP cone houses two ATP molecules. We determined structures of two AaR2 proteins: one purified from a construct lacking the intein (AaR2) and a second purified from a construct including the intein sequence (AaR2_genomic). These structures in the context of metal content analysis and activity data indicate that AaR2_genomic displays much higher iron occupancy and activity compared to AaR2, suggesting that the intein is important for facilitating complete iron incorporation, particularly in the Fe2 site of the mature R2 protein, which may be important for the survival of A. aeolicus in low-oxygen environments.


Asunto(s)
Proteínas Bacterianas/química , Ribonucleótido Reductasas/química , Regulación Alostérica , Aquifex/química , Aquifex/metabolismo , Proteínas Bacterianas/metabolismo , Cristalografía por Rayos X , Modelos Moleculares , Conformación Proteica , Multimerización de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Ribonucleótido Reductasas/metabolismo
3.
J Biol Chem ; 295(46): 15576-15587, 2020 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-32883811

RESUMEN

Ribonucleotide reductase (RNR) is a central enzyme for the synthesis of DNA building blocks. Most aerobic organisms, including nearly all eukaryotes, have class I RNRs consisting of R1 and R2 subunits. The catalytic R1 subunit contains an overall activity site that can allosterically turn the enzyme on or off by the binding of ATP or dATP, respectively. The mechanism behind the ability to turn the enzyme off via the R1 subunit involves the formation of different types of R1 oligomers in most studied species and R1-R2 octamers in Escherichia coli To better understand the distribution of different oligomerization mechanisms, we characterized the enzyme from Clostridium botulinum, which belongs to a subclass of class I RNRs not studied before. The recombinantly expressed enzyme was analyzed by size-exclusion chromatography, gas-phase electrophoretic mobility macromolecular analysis, EM, X-ray crystallography, and enzyme assays. Interestingly, it shares the ability of the E. coli RNR to form inhibited R1-R2 octamers in the presence of dATP but, unlike the E. coli enzyme, cannot be turned off by combinations of ATP and dGTP/dTTP. A phylogenetic analysis of class I RNRs suggests that activity regulation is not ancestral but was gained after the first subclasses diverged and that RNR subclasses with inhibition mechanisms involving R1 oligomerization belong to a clade separated from the two subclasses forming R1-R2 octamers. These results give further insight into activity regulation in class I RNRs as an evolutionarily dynamic process.


Asunto(s)
Proteínas Bacterianas/metabolismo , Clostridium botulinum/enzimología , Ribonucleótido Reductasas/metabolismo , Proteínas Bacterianas/clasificación , Dominio Catalítico , Cristalografía por Rayos X , Nucleótidos de Desoxiadenina/química , Dimerización , Escherichia coli/metabolismo , Filogenia , Estructura Cuaternaria de Proteína , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Ribonucleótido Reductasas/clasificación
4.
J Biol Chem ; 293(41): 15889-15900, 2018 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-30166338

RESUMEN

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.


Asunto(s)
Glutarredoxinas/metabolismo , Ribonucleótido Reductasas/metabolismo , Aerococcaceae/química , Catálisis , Nucleótidos de Desoxiadenina/metabolismo , Flavobacteriaceae/química , Transferencia de Gen Horizontal , Glutarredoxinas/química , Glutarredoxinas/genética , Oxidación-Reducción , Unión Proteica , Dominios Proteicos , Multimerización de Proteína/efectos de los fármacos , Ribonucleótido Reductasas/genética
5.
J Biol Inorg Chem ; 24(6): 841-848, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31218442

RESUMEN

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.


Asunto(s)
Radicales Libres/química , Radicales Libres/metabolismo , Pseudomonas aeruginosa/enzimología , Ribonucleótido Reductasas/metabolismo , Tirosina/química , Tirosina/metabolismo , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo , Ribonucleótido Reductasas/genética
6.
J Biol Inorg Chem ; 24(6): 863-877, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31414238

RESUMEN

Outside of the photosynthetic machinery, high-valent manganese cofactors are rare in biology. It was proposed that a recently discovered subclass of ribonucleotide reductase (RNR), class Id, is dependent on a Mn2(IV,III) cofactor for catalysis. Class I RNRs consist of a substrate-binding component (NrdA) and a metal-containing radical-generating component (NrdB). Herein we utilize a combination of EPR spectroscopy and enzyme assays to underscore the enzymatic relevance of the Mn2(IV,III) cofactor in class Id NrdB from Facklamia ignava. Once formed, the Mn2(IV,III) cofactor confers enzyme activity that correlates well with cofactor quantity. Moreover, we present the X-ray structure of the apo- and aerobically Mn-loaded forms of the homologous class Id NrdB from Leeuwenhoekiella blandensis, revealing a dimanganese centre typical of the subclass, with a tyrosine residue maintained at distance from the metal centre and a lysine residue projected towards the metals. Structural comparison of the apo- and metal-loaded forms of the protein reveals a refolding of the loop containing the conserved lysine and an unusual shift in the orientation of helices within a monomer, leading to the opening of a channel towards the metal site. Such major conformational changes have not been observed in NrdB proteins before. Finally, in vitro reconstitution experiments reveal that the high-valent manganese cofactor is not formed spontaneously from oxygen, but can be generated from at least two different reduced oxygen species, i.e. H2O2 and superoxide (O 2·- ). Considering the observed differences in the efficiency of these two activating reagents, we propose that the physiologically relevant mechanism involves superoxide.


Asunto(s)
Manganeso/metabolismo , Ribonucleótido Reductasas/metabolismo , Aerococcaceae/metabolismo , Cristalografía por Rayos X , Espectroscopía de Resonancia por Spin del Electrón , Flavobacteriaceae/metabolismo , Radicales Libres/metabolismo , Peróxido de Hidrógeno/metabolismo , Oxidación-Reducción , Ribonucleótido Reductasas/química , Ribonucleótido Reductasas/genética , Superóxidos/metabolismo
7.
J Biol Chem ; 292(46): 19044-19054, 2017 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-28972190

RESUMEN

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.


Asunto(s)
Actinomycetales/química , Proteínas Bacterianas/química , Cisteína/química , Ribonucleótido Reductasas/química , Dedos de Zinc , Actinomycetales/genética , Actinomycetales/metabolismo , Regulación Alostérica , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Dominio Catalítico , Cristalografía por Rayos X , Cisteína/genética , Cisteína/metabolismo , Transporte de Electrón , Modelos Moleculares , Oxidación-Reducción , Filogenia , Dominios Proteicos , Multimerización de Proteína , Ribonucleótido Reductasas/genética , Ribonucleótido Reductasas/metabolismo
8.
J Biol Chem ; 290(28): 17339-48, 2015 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-25971975

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/metabolismo , Pseudomonas aeruginosa/enzimología , Ribonucleótido Reductasas/metabolismo , Adenosina Trifosfato/metabolismo , Regulación Alostérica , Sitio Alostérico , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Nucleótidos de Desoxiadenina/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Cinética , Datos de Secuencia Molecular , Estructura Cuaternaria de Proteína , Subunidades de Proteína , Pseudomonas aeruginosa/genética , Ribonucleótido Reductasas/química , Ribonucleótido Reductasas/genética , Eliminación de Secuencia
9.
J Biol Chem ; 289(46): 31940-31949, 2014 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-25262022

RESUMEN

Ribonucleotide reductases (RNRs) catalyze the conversion of ribonucleotides to deoxyribonucleotides, and represent the only de novo pathway to provide DNA building blocks. Three different classes of RNR are known, denoted I-III. Class I RNRs are heteromeric proteins built up by α and ß subunits and are further divided into different subclasses, partly based on the metal content of the ß-subunit. In subclass Ib RNR the ß-subunit is denoted NrdF, and harbors a manganese-tyrosyl radical cofactor. The generation of this cofactor is dependent on a flavodoxin-like maturase denoted NrdI, responsible for the formation of an active oxygen species suggested to be either a superoxide or a hydroperoxide. Herein we report on the magnetic properties of the manganese-tyrosyl radical cofactor of Bacillus anthracis NrdF and the redox properties of B. anthracis NrdI. The tyrosyl radical in NrdF is stabilized through its interaction with a ferromagnetically coupled manganese dimer. Moreover, we show through a combination of redox titration and protein electrochemistry that in contrast to hitherto characterized NrdIs, the B. anthracis NrdI is stable in its semiquinone form (NrdIsq) with a difference in electrochemical potential of ∼110 mV between the hydroquinone and semiquinone state. The under anaerobic conditions stable NrdIsq is fully capable of generating the oxidized, tyrosyl radical-containing form of Mn-NrdF when exposed to oxygen. This latter observation strongly supports that a superoxide radical is involved in the maturation mechanism, and contradicts the participation of a peroxide species. Additionally, EPR spectra on whole cells revealed that a significant fraction of NrdI resides in its semiquinone form in vivo, underscoring that NrdIsq is catalytically relevant.


Asunto(s)
Bacillus anthracis/enzimología , Quinonas/química , Ribonucleósido Difosfato Reductasa/química , Ribonucleósido Difosfato Reductasa/genética , Superóxidos/química , Antibacterianos/química , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Catálisis , Electrodos , Espectroscopía de Resonancia por Spin del Electrón , Radicales Libres , Magnetismo , Manganeso/química , Metales/química , Oxidación-Reducción , Oxígeno/química , Ribonucleótido Reductasas/química , Ribonucleótido Reductasas/genética , Espectrofotometría Ultravioleta
10.
Proc Natl Acad Sci U S A ; 109(25): 9798-803, 2012 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-22665797

RESUMEN

Ribonucleotide reductase (RNR) catalyzes reduction of the four different ribonucleotides to their corresponding deoxyribonucleotides and is the rate-limiting enzyme in DNA synthesis. RNR is a well-established target for the antiproliferative drugs Gemzar and Hydrea, for antisense therapy, and in combination chemotherapies. Surprisingly, few novel drugs that target RNR have emerged, partly because RNR activity assays are laboratory-intense and exclude high-throughput methodologies. Here, we present a previously undescribed PCR-based assay for RNR activity measurements in microplate format. We validated the approach by screening a diverse library of 1,364 compounds for inhibitors of class I RNR from the opportunistic pathogen Pseudomonas aeruginosa, and we identified 27 inhibitors with IC(50) values from ∼200 nM to 30 µM. Interestingly, a majority of the identified inhibitors have been found inactive in human cell lines as well as in anticancer and in vivo tumor tests as reported by the PubChem BioAssay database. Four of the RNR inhibitors inhibited growth of P. aeruginosa, and two were also found to affect the transcription of RNR genes and to decrease the cellular deoxyribonucleotide pools. This unique PCR-based assay works with any RNR enzyme and any substrate nucleotide, and thus opens the door to high-throughput screening for RNR inhibitors in drug discovery.


Asunto(s)
Antibacterianos/farmacología , Inhibidores Enzimáticos/farmacología , Ribonucleótido Reductasas/antagonistas & inhibidores , Concentración 50 Inhibidora , Pseudomonas aeruginosa/enzimología
11.
Crit Rev Biochem Mol Biol ; 47(1): 50-63, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22050358

RESUMEN

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.


Asunto(s)
Sitio Alostérico/fisiología , ADN/biosíntesis , Desoxirribonucleótidos/metabolismo , Tasa de Mutación , Ribonucleótido Reductasas/metabolismo , Adenosina Trifosfato/metabolismo , Regulación Alostérica/fisiología , Dominio Catalítico/fisiología , Nucleótidos de Desoxiadenina/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Lactobacillus leichmannii/genética , Lactobacillus leichmannii/metabolismo , Modelos Moleculares , Conformación Proteica , Ribonucleótido Reductasas/química , Levaduras/genética , Levaduras/metabolismo
12.
J Biol Chem ; 288(12): 8198-8208, 2013 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-23372162

RESUMEN

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.


Asunto(s)
Dictyostelium/enzimología , Proteínas Protozoarias/metabolismo , Ribonucleótido Reductasas/metabolismo , Regulación Alostérica , Complejos de Coordinación/química , Citidina Difosfato/química , Dictyostelium/genética , Dictyostelium/fisiología , Inhibidores Enzimáticos/farmacología , Radicales Libres/química , Expresión Génica , Regulación Enzimológica de la Expresión Génica , Guanosina Difosfato/química , Hierro/química , Cinética , Filogenia , Proteínas Protozoarias/genética , Ribonucleótido Reductasas/antagonistas & inhibidores , Ribonucleótido Reductasas/química , Ribonucleótido Reductasas/genética , Espectrofotometría Ultravioleta , Esporas Protozoarias/enzimología , Esporas Protozoarias/genética , Tirosina/química
13.
J Biol Chem ; 287(47): 39686-97, 2012 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-23012357

RESUMEN

Bacillus anthracis is the causative agent of anthrax, which is associated with a high mortality rate. Like several medically important bacteria, B. anthracis lacks glutathione but encodes many genes annotated as thioredoxins, thioredoxin reductases, and glutaredoxin-like proteins. We have cloned, expressed, and characterized three potential thioredoxins, two potential thioredoxin reductases, and three glutaredoxin-like proteins. Of these, thioredoxin 1 (Trx1) and NrdH reduced insulin, 5,5'-dithiobis-(2-nitrobenzoic acid) (DTNB), and the manganese-containing type Ib ribonucleotide reductase (RNR) from B. anthracis in the presence of NADPH and thioredoxin reductase 1 (TR1), whereas thioredoxin 2 (Trx2) could only reduce DTNB. Potential TR2 was verified as an FAD-containing protein reducible by dithiothreitol but not by NAD(P)H. The recently discovered monothiol bacillithiol did not work as a reductant for RNR, either directly or via any of the redoxins. The catalytic efficiency of Trx1 was 3 and 20 times higher than that of Trx2 and NrdH, respectively, as substrates for TR1. Additionally, the catalytic efficiency of Trx1 as an electron donor for RNR was 7-fold higher than that of NrdH. In extracts of B. anthracis, Trx1 was responsible for almost all of the disulfide reductase activity, whereas Western blots showed that the level of Trx1 was 15 and 60 times higher than that of Trx2 and NrdH, respectively. Our findings demonstrate that the most important general disulfide reductase system in B. anthracis is TR1/Trx1 and that Trx1 is the physiologically relevant electron donor for RNR. This information may provide a basis for the development of novel antimicrobial therapies targeting this severe pathogen.


Asunto(s)
Bacillus anthracis/metabolismo , Proteínas Bacterianas/metabolismo , Ribonucleótido Reductasas/metabolismo , Tiorredoxina Reductasa 1/metabolismo , Tiorredoxinas/metabolismo , Bacillus anthracis/genética , Proteínas Bacterianas/genética , Electrones , NADP/genética , NADP/metabolismo , Oxidación-Reducción , Ribonucleótido Reductasas/genética , Tiorredoxina Reductasa 1/genética , Tiorredoxinas/genética
14.
J Biol Chem ; 287(24): 20565-75, 2012 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-22535960

RESUMEN

In the postgenomic era, bioinformatic analysis of sequence similarity is an immensely powerful tool to gain insight into evolution and protein function. Over long evolutionary distances, however, sequence-based methods fail as the similarities become too low for phylogenetic analysis. Macromolecular structure generally appears better conserved than sequence, but clear models for how structure evolves over time are lacking. The exponential growth of three-dimensional structural information may allow novel structure-based methods to drastically extend the evolutionary time scales amenable to phylogenetics and functional classification of proteins. To this end, we analyzed 80 structures from the functionally diverse ferritin-like superfamily. Using evolutionary networks, we demonstrate that structural comparisons can delineate and discover groups of proteins beyond the "twilight zone" where sequence similarity does not allow evolutionary analysis, suggesting that considerable and useful evolutionary signal is preserved in three-dimensional structures.


Asunto(s)
Evolución Molecular , Ferritinas/química , Ferritinas/clasificación , Modelos Moleculares , Filogenia , Ferritinas/genética , Estructura Terciaria de Proteína
15.
Nucleic Acids Res ; 39(4): 1381-9, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20972217

RESUMEN

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.


Asunto(s)
Secuencias Repetitivas Esparcidas , Ribonucleótido Reductasas/química , Ribonucleótido Reductasas/genética , Bacteriófagos/enzimología , Dominio Catalítico , Dimerización , Holoenzimas/genética , Mutación , Dominios y Motivos de Interacción de Proteínas , Ribonucleótido Reductasas/metabolismo
16.
Science ; 382(6666): 109-113, 2023 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-37797025

RESUMEN

Aerobic ribonucleotide reductases (RNRs) initiate synthesis of DNA building blocks by generating a free radical within the R2 subunit; the radical is subsequently shuttled to the catalytic R1 subunit through proton-coupled electron transfer (PCET). We present a high-resolution room temperature structure of the class Ie R2 protein radical captured by x-ray free electron laser serial femtosecond crystallography. The structure reveals conformational reorganization to shield the radical and connect it to the translocation path, with structural changes propagating to the surface where the protein interacts with the catalytic R1 subunit. Restructuring of the hydrogen bond network, including a notably short O-O interaction of 2.41 angstroms, likely tunes and gates the radical during PCET. These structural results help explain radical handling and mobilization in RNR and have general implications for radical transfer in proteins.


Asunto(s)
Proteínas Bacterianas , Entomoplasmataceae , Ribonucleótido Reductasas , Transporte de Electrón , Protones , Ribonucleótido Reductasas/química , Cristalografía por Rayos X/métodos , Entomoplasmataceae/enzimología , Dominio Catalítico , Proteínas Bacterianas/química
17.
J Biol Chem ; 286(38): 33053-60, 2011 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-21832039

RESUMEN

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.


Asunto(s)
Bacillus anthracis/enzimología , Proteínas Bacterianas/metabolismo , Manganeso/metabolismo , Ribonucleótido Reductasas/metabolismo , Apoproteínas/metabolismo , Flavodoxina/metabolismo , Holoenzimas/metabolismo , Hierro/metabolismo , Unión Proteica , Estructura Cuaternaria de Proteína , Espectrofotometría Ultravioleta , Resonancia por Plasmón de Superficie
18.
Nat Commun ; 13(1): 2700, 2022 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-35577776

RESUMEN

Ribonucleotide reductase (RNR) is an essential enzyme that catalyzes the synthesis of DNA building blocks in virtually all living cells. NrdR, an RNR-specific repressor, controls the transcription of RNR genes and, often, its own, in most bacteria and some archaea. NrdR senses the concentration of nucleotides through its ATP-cone, an evolutionarily mobile domain that also regulates the enzymatic activity of many RNRs, while a Zn-ribbon domain mediates binding to NrdR boxes upstream of and overlapping the transcription start site of RNR genes. Here, we combine biochemical and cryo-EM studies of NrdR from Streptomyces coelicolor to show, at atomic resolution, how NrdR binds to DNA. The suggested mechanism involves an initial dodecamer loaded with two ATP molecules that cannot bind to DNA. When dATP concentrations increase, an octamer forms that is loaded with one molecule each of dATP and ATP per monomer. A tetramer derived from this octamer then binds to DNA and represses transcription of RNR. In many bacteria - including well-known pathogens such as Mycobacterium tuberculosis - NrdR simultaneously controls multiple RNRs and hence DNA synthesis, making it an excellent target for novel antibiotics development.


Asunto(s)
Ribonucleótido Reductasas , Streptomyces coelicolor , Adenosina Trifosfato/metabolismo , Microscopía por Crioelectrón , Regulación Bacteriana de la Expresión Génica , Nucleótidos/química , Ribonucleótido Reductasas/genética , Ribonucleótido Reductasas/metabolismo , Streptomyces coelicolor/metabolismo
19.
Infect Immun ; 79(7): 2663-9, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21502590

RESUMEN

The roles of different ribonucleotide reductases (RNRs) in bacterial pathogenesis have not been studied systematically. In this work we analyzed the importance of the different Pseudomonas aeruginosa RNRs in pathogenesis using the Drosophila melanogaster host-pathogen interaction model. P. aeruginosa codes for three different RNRs with different environmental requirements. Class II and III RNR chromosomal mutants exhibited reduced virulence in this model. Translational reporter fusions of RNR gene nrdA, nrdJ, or nrdD to the green fluorescent protein were constructed to measure the expression of each class during the infection process. Analysis of the P. aeruginosa infection by flow cytometry revealed increased expression of nrdJ and nrdD and decreased nrdA expression during the infection process. Expression of each RNR class fits with the pathogenicities of the chromosomal deletion mutants. An extended understanding of the pathogenicity and physiology of P. aeruginosa will be important for the development of novel drugs against infections in cystic fibrosis patients.


Asunto(s)
Drosophila melanogaster/microbiología , Interacciones Huésped-Patógeno , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/patogenicidad , Ribonucleótido Reductasas/genética , Animales , Proteínas Bacterianas , Citometría de Flujo , Regulación Bacteriana de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Mutación , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/enzimología , Ribonucleótido Reductasas/metabolismo , Eliminación de Secuencia
20.
Front Mol Biosci ; 8: 713608, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34381817

RESUMEN

The essential enzyme ribonucleotide reductase (RNR) is highly regulated both at the level of overall activity and substrate specificity. Studies of class I, aerobic RNRs have shown that overall activity is downregulated by the binding of dATP to a small domain known as the ATP-cone often found at the N-terminus of RNR subunits, causing oligomerization that prevents formation of a necessary α2ß2 complex between the catalytic (α2) and radical generating (ß2) subunits. In some relatively rare organisms with RNRs of the subclass NrdAi, the ATP-cone is found at the N-terminus of the ß subunit rather than more commonly the α subunit. Binding of dATP to the ATP-cone in ß results in formation of an unusual ß4 tetramer. However, the structural basis for how the formation of the active complex is hindered by such oligomerization has not been studied. Here we analyse the low-resolution three-dimensional structures of the separate subunits of an RNR from subclass NrdAi, as well as the α4ß4 octamer that forms in the presence of dATP. The results reveal a type of oligomer not previously seen for any class of RNR and suggest a mechanism for how binding of dATP to the ATP-cone switches off catalysis by sterically preventing formation of the asymmetrical α2ß2 complex.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA