Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 103(26): 9850-4, 2006 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-16777966

RESUMO

Ribonucleotide reductase (RNR) from Chlamydia trachomatis is a class I RNR composed of proteins R1 and R2. In protein R2, the tyrosine residue harboring the radical that is necessary for catalysis in other class I RNRs is replaced by a phenylalanine. Active C. trachomatis RNR instead uses the Fe(III)-Fe(IV) state of the iron cluster in R2 as an initiator of catalysis. The paramagnetic Fe(III)-Fe(IV) state, identified by (57)Fe substitution, becomes electron spin resonance detectable in samples that are frozen during conditions of ongoing catalysis. Its amount depends on the conditions for catalysis, such as incubation temperature and the R1/R2 ratio. The results link induction of the Fe(III)-Fe(IV) state with enzyme activity of chlamydial RNR. Based on these observations, a reaction scheme is proposed for the iron site. This scheme includes (i) an activation cycle involving reduction and an oxygen reaction in R2 and (ii) a catalysis cycle involving substrate binding and turnover in R1.


Assuntos
Proteínas de Bactérias/química , Chlamydia trachomatis/enzimologia , Ferro/química , Ribonucleotídeo Redutases/química , Tirosina/química , Catálise , Espectroscopia de Ressonância de Spin Eletrônica , Radicais Livres/química , Oxirredução
2.
Eur J Biochem ; 268(16): 4527-36, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11502214

RESUMO

Unregulated transcription of protein-encoding genes in vitro is dependent on 12-subunit core RNA polymerase II and five general transcription factors; TATA binding protein (TBP), transcription factor (TF)IIB, TFIIE, TFIIF, and TFIIH. Here we describe cloning of the mouse cDNAs encoding TFIIB and the small and large TFIIE and TFIIF subunits. The cDNAs have been used to express the corresponding proteins in recombinant form in Escherichia coli and in Sf21 insect cells, and all proteins have been purified to > 90% homogeneity. We have also purified a recombinant His6-tagged mouse TBP to near homogeneity and show that it is active in both a reconstituted mouse in vitro transcription system and a TBP-dependent in vitro transcription system from Saccharomyces cerevisiae. The more complex general transcription factors, TFIIH and RNA polymerase II, were purified more than 1000-fold and to near homogeneity, respectively, from tissue cultured mouse cells. When combined, the purified factors were sufficient to initiate transcription from different promoters in vitro. Functional studies of the S-phase-specific mouse ribonucleotide reductase R2 promoter using both the highly purified system described here (a mouse cell nuclear extract in vitro transcription system) and in vivo R2-promoter reporter gene assays together identify an NF-Y interacting promoter proximal CCAAT-box as being essential for high-level expression from the R2 promoter.


Assuntos
Proteínas de Ligação a DNA/fisiologia , RNA Polimerase II/metabolismo , Proteínas de Saccharomyces cerevisiae , Fatores Associados à Proteína de Ligação a TATA , Fator de Transcrição TFIID , Fatores de Transcrição TFII , Fatores de Transcrição/fisiologia , Transcrição Gênica , Animais , Clonagem Molecular , Camundongos , Regiões Promotoras Genéticas , Proteínas Recombinantes/isolamento & purificação , Ribonucleotídeo Redutases/genética , Proteína de Ligação a TATA-Box , Fator de Transcrição TFIIB , Fator de Transcrição TFIIH
3.
J Biol Chem ; 276(44): 40647-51, 2001 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-11517226

RESUMO

Recently, a homologue of the small subunit of mammalian ribonucleotide reductase (RNR) was discovered, called p53R2. Unlike the well characterized S phase-specific RNR R2 protein, the new form was induced in response to DNA damage by the p53 protein. Because the R2 protein is specifically degraded in late mitosis and absent in G0/G1 cells, the induction of the p53R2 protein may explain how resting cells can obtain deoxyribonucleotides for DNA repair. However, no direct demonstration of RNR activity of the p53R2 protein was presented and furthermore, no corresponding RNR large subunit was identified. In this study we show that recombinant, highly purified human and mouse p53R2 proteins contain an iron-tyrosyl free radical center, and both proteins form an active RNR complex with the human and mouse R1 proteins. UV irradiation of serum-starved, G0/G1-enriched mouse fibroblasts, stably transformed with an R1 promoter-luciferase reporter gene construct, caused a 3-fold increase in luciferase activity 24 h after irradiation, paralleled by an increase in the levels of R1 protein. Taken together, our data indicate that the R1 protein can function as the normal partner of the p53R2 protein and that an R1-p53R2 complex can supply resting cells with deoxyribonucleotides for DNA repair.


Assuntos
Proteínas de Ciclo Celular , Divisão Celular , Dano ao DNA , Ribonucleotídeo Redutases/metabolismo , Sequência de Aminoácidos , Animais , Eletroforese em Gel de Poliacrilamida , Humanos , Técnicas In Vitro , Camundongos , Dados de Sequência Molecular , Ligação Proteica , RNA Mensageiro/genética , Ribonucleotídeo Redutases/química , Ribonucleotídeo Redutases/genética , Homologia de Sequência de Aminoácidos
4.
EMBO J ; 20(13): 3544-53, 2001 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-11432841

RESUMO

The evolutionarily conserved protein kinases Mec1 and Rad53 are required for checkpoint response and growth. Here we show that their role in growth is to remove the ribonucleotide reductase inhibitor Sml1 to ensure DNA replication. Sml1 protein levels fluctuate during the cell cycle, being lowest during S phase. The disappearance of Sml1 protein in S phase is due to post-transcriptional regulation and is associated with protein phosphorylation. Both phosphorylation and diminution of Sml1 require MEC1 and RAD53. More over, failure to remove Sml1 in mec1 and rad53 mutants results in incomplete DNA replication, defective mitochondrial DNA propagation, decreased dNTP levels and cell death. Interestingly, similar regulation of Sml1 also occurs after DNA damage. In this case, the regulation requires MEC1 and RAD53, as well as other checkpoint genes. Therefore, Sml1 is a new target of the DNA damage checkpoint and its removal is a conserved function of Mec1 and Rad53 during growth and after damage.


Assuntos
Proteínas de Ciclo Celular , Dano ao DNA , Reparo do DNA , Replicação do DNA , Inibidores Enzimáticos , Proteínas Fúngicas/metabolismo , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases , Ribonucleotídeo Redutases/antagonistas & inibidores , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae/fisiologia , Quinase do Ponto de Checagem 2 , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/efeitos da radiação , Replicação do DNA/efeitos dos fármacos , Replicação do DNA/efeitos da radiação , DNA Fúngico/efeitos dos fármacos , DNA Fúngico/genética , DNA Fúngico/efeitos da radiação , Raios gama , Genótipo , Hidroxiureia/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crescimento & desenvolvimento , Raios Ultravioleta
5.
Proc Natl Acad Sci U S A ; 98(11): 6412-6, 2001 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-11353848

RESUMO

The drugs in clinical use against African sleeping sickness are toxic, costly, or inefficient. We show that Trypanosoma brucei, which causes this disease, has very low levels of CTP, which are due to a limited capacity for de novo synthesis and the lack of salvage pathways. The CTP synthetase inhibitors 6-diazo-5-oxo-l-norleucine (DON) and alpha-amino-3-chloro-4,5-dihydro-5-isoxazoleacetic acid (acivicin) reduced the parasite CTP levels even further and inhibited trypanosome proliferation in vitro and in T. brucei-infected mice. In mammalian cells, DON mainly inhibits de novo purine biosynthesis, a pathway lacking in trypanosomes. We could rescue DON-treated human and mouse fibroblasts by the addition of the purine base hypoxanthine to the growth medium. For treatment of sleeping sickness, we propose the use of CTP synthetase inhibitors alone or in combination with appropriate nucleosides or bases.


Assuntos
Carbono-Nitrogênio Ligases/antagonistas & inibidores , Diazo-Oxo-Norleucina/farmacologia , Inibidores Enzimáticos/farmacologia , Isoxazóis/farmacologia , Tripanossomicidas/farmacologia , Trypanosoma brucei brucei/enzimologia , Tripanossomíase Africana/tratamento farmacológico , Trifosfato de Adenosina/metabolismo , Animais , Células Cultivadas , Citidina/farmacologia , Citidina Trifosfato/biossíntese , Citidina Trifosfato/metabolismo , Diazo-Oxo-Norleucina/uso terapêutico , Inibidores Enzimáticos/uso terapêutico , Fibroblastos/citologia , Guanina/farmacologia , Guanosina Trifosfato/metabolismo , Humanos , Hipoxantinas/farmacologia , Líquido Intracelular , Isoxazóis/uso terapêutico , Camundongos , Camundongos Endogâmicos BALB C , Tripanossomicidas/uso terapêutico , Trypanosoma brucei brucei/efeitos dos fármacos , Trypanosoma brucei brucei/crescimento & desenvolvimento , Tripanossomíase Africana/sangue , Tripanossomíase Africana/parasitologia , Uridina Trifosfato/metabolismo
6.
Mol Cell Biol ; 20(23): 9076-83, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11074005

RESUMO

In budding yeast, MEC1 and RAD53 are essential for cell growth. Previously we reported that mec1 or rad53 lethality is suppressed by removal of Sml1, a protein that binds to the large subunit of ribonucleotide reductase (Rnr1) and inhibits RNR activity. To understand further the relationship between this suppression and the Sml1-Rnr1 interaction, we randomly mutagenized the SML1 open reading frame. Seven mutations were identified that did not affect protein expression levels but relieved mec1 and rad53 inviability. Interestingly, all seven mutations abolish the Sml1 interaction with Rnr1, suggesting that this interaction causes the lethality observed in mec1 and rad53 strains. The mutant residues all cluster within the 33 C-terminal amino acids of the 104-amino-acid-long Sml1 protein. Four of these residues reside within an alpha-helical structure that was revealed by nuclear magnetic resonance studies. Moreover, deletions encompassing the N-terminal half of Sml1 do not interfere with its RNR inhibitory activity. Finally, the seven sml1 mutations also disrupt the interaction with yeast Rnr3 and human R1, suggesting a conserved binding mechanism between Sml1 and the large subunit of RNR from different species.


Assuntos
Proteínas de Ciclo Celular , Inibidores Enzimáticos , Proteínas Fúngicas/genética , Proteínas Quinases/genética , Proteínas Serina-Treonina Quinases , Ribonucleotídeo Redutases/antagonistas & inibidores , Proteínas de Saccharomyces cerevisiae , Supressão Genética , Quinase do Ponto de Checagem 2 , Cromossomos Fúngicos , Análise Mutacional de DNA , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Mutação de Sentido Incorreto , Ligação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Soluções , Especificidade da Espécie , Fator Trefoil-2 , Técnicas do Sistema de Duplo-Híbrido
7.
J Virol ; 74(24): 11557-65, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11090153

RESUMO

Ribonucleotide reductase (RNR) is an essential enzyme for the de novo synthesis of both cellular and viral DNA and catalyzes the conversion of ribonucleoside diphosphates into the corresponding deoxyribonucleoside diphosphates. The enzyme consists of two nonidentical subunits, termed R1 and R2, whose expression is very low in resting cells and maximal in S-phase cells. Here we show that murine cytomegalovirus (MCMV) replication depends on ribonucleotide reduction since it is prevented by the RNR inhibitor hydroxyurea. MCMV infection of quiescent fibroblasts markedly induces both mRNA and protein corresponding to the cellular R2 subunit, whereas expression of the cellular R1 subunit does not appear to be up-regulated. The increase in R2 gene expression is due to an increase in gene transcription, since the activity of a reporter gene driven by the mouse R2 promoter is induced following virus infection. Cotransfection experiments revealed that expression of the viral immediate-early 1 protein was sufficient to mediate the increase in R2 promoter activity. It was found that the viral gene M45, encoding a putative homologue of the R1 subunit, is expressed 24 and 48 h after infection. Meanwhile, we observed an expansion of the deoxyribonucleoside triphosphate pool between 24 and 48 h after infection; however, neither CDP reduction nor viral replication was inhibited by treatment with 10 mM thymidine. These findings indicate the induction of an RNR activity with an altered allosteric regulation compared to the mouse RNR following MCMV infection and suggest that the virus R1 homologue may complex with the induced cellular R2 protein to reconstitute a new RNR activity.


Assuntos
Citomegalovirus/fisiologia , Fibroblastos/virologia , Ribonucleotídeo Redutases/fisiologia , Replicação Viral , Animais , Camundongos
8.
J Biol Chem ; 275(42): 33021-6, 2000 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-10884394

RESUMO

We compared the allosteric regulation and effector binding properties of wild type R1 protein and R1 protein with a mutation in the "activity site" (D57N) of mouse ribonucleotide reductase. Wild type R1 had two effector-binding sites per polypeptide chain: one site (activity site) for dATP and ATP, with dATP-inhibiting and ATP-stimulating catalytic activity; and a second site (specificity site) for dATP, ATP, dTTP, and dGTP, directing substrate specificity. Binding of dATP to the specificity site had a 20-fold higher affinity than to the activity site. In all these respects, mouse R1 resembles Escherichia coli R1. Results with D57N were complicated by the instability of the protein, but two major changes were apparent. First, enzyme activity was stimulated by both dATP and ATP, suggesting that D57N no longer distinguished between the two nucleotides. Second, the two binding sites for dATP both had the same low affinity for the nucleotide, similar to that of the activity site of wild type R1. Thus the mutation in the activity site had decreased the affinity for dATP at the specificity site, demonstrating the interaction between the two sites.


Assuntos
Ribonucleotídeo Redutases/química , Ribonucleotídeo Redutases/metabolismo , Trifosfato de Adenosina/metabolismo , Regulação Alostérica , Sítio Alostérico , Substituição de Aminoácidos , Animais , Sítios de Ligação , Ligação Competitiva , Catálise , Nucleotídeos de Desoxiadenina/farmacologia , Desoxirribonucleotídeos/metabolismo , Desoxirribonucleotídeos/farmacologia , Cinética , Camundongos , Mutagênese Sítio-Dirigida , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
9.
J Biol Chem ; 275(23): 17747-53, 2000 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-10747958

RESUMO

Ribonucleotide reductase (RNR) plays a central role in the formation and control of the optimal levels of deoxyribonucleoside triphosphates, which are required for DNA replication and DNA repair processes. Mammalian RNRs are composed of two nonidentical subunits, proteins R1 and R2. The levels of the limiting R2 protein control overall RNR activity during the mammalian cell cycle, being undetectable in G(1) phase and increasing in S phase. We show that in proliferating mammalian cells, the transcription of the R2 gene, once activated in the beginning of S phase, reaches its maximum 6-7 h later and then declines. Surprisingly, DNA damage and replication blocks neither increase nor prolong the R2 promoter activity in S phase. Instead, the cell cycle activity of the mammalian enzyme is controlled by an S phase/DNA damage-specific stabilization of the R2 protein, which is effective until cells pass into mitosis.


Assuntos
Ciclo Celular/fisiologia , Divisão Celular/fisiologia , Dano ao DNA , Reparo do DNA , Replicação do DNA , Ribonucleotídeo Redutases/genética , Ribonucleotídeo Redutases/metabolismo , Células 3T3 , Animais , Afidicolina/farmacologia , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/efeitos da radiação , Divisão Celular/efeitos dos fármacos , Divisão Celular/efeitos da radiação , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/efeitos da radiação , Replicação do DNA/efeitos dos fármacos , Replicação do DNA/efeitos da radiação , Fase G1 , Regulação Enzimológica da Expressão Gênica , Hidroxiureia/farmacologia , Camundongos , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/efeitos da radiação , Proteínas Recombinantes/metabolismo , Fase S , Transcrição Gênica , Transfecção , Raios Ultravioleta
10.
Proc Natl Acad Sci U S A ; 97(6): 2474-9, 2000 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-10716984

RESUMO

Ribonucleotide reductase (RNR) catalyzes the de novo synthesis of deoxyribonucleotides. Eukaryotes have an alpha(2)beta(2) form of RNR consisting of two homodimeric subunits, proteins R1 (alpha(2)) and R2 (beta(2)). The R1 protein is the business end of the enzyme containing the active site and the binding sites for allosteric effectors. The R2 protein is a radical storage device containing an iron center-generated tyrosyl free radical. Previous work has identified an RNR protein in yeast, Rnr4p, which is homologous to other R2 proteins but lacks a number of conserved amino acid residues involved in iron binding. Using highly purified recombinant yeast RNR proteins, we demonstrate that the crucial role of Rnr4p (beta') is to fold correctly and stabilize the radical-storing Rnr2p by forming a stable 1:1 Rnr2p/Rnr4p complex. This complex sediments at 5.6 S as a betabeta' heterodimer in a sucrose gradient. In the presence of Rnr1p, both polypeptides of the Rnr2p/Rnr4p heterodimer cosediment at 9.7 S as expected for an alpha(2)betabeta' heterotetramer, where Rnr4p plays an important role in the interaction between the alpha(2) and the betabeta ' subunits. The specific activity of the Rnr2p complexed with Rnr4p is 2,250 nmol deoxycytidine 5'-diphosphate formed per min per mg, whereas the homodimer of Rnr2p shows no activity. This difference in activity may be a consequence of the different conformations of the inactive homodimeric Rnr2p and the active Rnr4p-bound form, as shown by CD spectroscopy. Taken together, our results show that the Rnr2p/Rnr4p heterodimer is the active form of the yeast RNR small subunit.


Assuntos
Proteínas Fúngicas/química , Ferro/química , Ribonucleotídeo Redutases/química , Western Blotting , Centrifugação com Gradiente de Concentração , Dicroísmo Circular , Relação Dose-Resposta a Droga , Espectroscopia de Ressonância de Spin Eletrônica , Escherichia coli/metabolismo , Radicais Livres/química , Ligação Proteica , Conformação Proteica , Dobramento de Proteína , Isoformas de Proteínas , Proteínas Recombinantes/química , Ribonucleotídeo Redutases/isolamento & purificação , Temperatura , Fatores de Tempo
11.
J Biol Chem ; 274(51): 36679-83, 1999 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-10593972

RESUMO

Ribonucleotide reductase (RNR) catalyzes the reduction of ribonucleotides to deoxyribonucleotides; this step is rate-limiting in DNA precursor synthesis. A number of regulatory mechanisms ensure optimal deoxyribonucleotide pools, which are essential for cell viability. The best studied mechanisms are transcriptional regulation of the RNR genes during the cell cycle and in the response to DNA damage, and the allosteric regulation of ribonucleotide reductase by nucleoside triphosphates. Recently, another mode of RNR regulation has been hypothesized in yeast. A novel protein, Sml1, was shown to bind to the Rnr1 protein of the yeast ribonucleotide reductase; this interaction was proposed to inhibit ribonucleotide reductase activity when DNA synthesis is not required (Zhao, X., Muller, E.G.D., and Rothstein, R. (1998) Mol. Cell 2, 329-340). Here, we use highly purified recombinant proteins to directly demonstrate that the Sml1 protein is a strong inhibitor of yeast RNR. The Sml1p specifically binds to the yeast Rnr1p in a 1:1 ratio with a dissociation constant of 0.4 microM. Interestingly, Sml1p also specifically binds to the mouse ribonucleotide reductase R1 protein. However, the inhibition observed in an in vitro mouse ribonucleotide reductase assay is less pronounced than the inhibition in yeast and probably occurs via a different mechanism.


Assuntos
Replicação do DNA , Inibidores Enzimáticos , Proteínas Fúngicas/metabolismo , Ribonucleotídeo Redutases/antagonistas & inibidores , Ribonucleotídeo Redutases/metabolismo , Proteínas de Saccharomyces cerevisiae , Animais , Dano ao DNA , DNA Fúngico , Camundongos , Saccharomyces cerevisiae , Especificidade por Substrato
12.
J Med Chem ; 42(17): 3243-50, 1999 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-10464011

RESUMO

Continuing our studies on ribonucleotide reductase (RNR) mechanism-based inhibitors, we have now prepared the diphosphates (DP) of 2'-O-allyl-1-beta-D-arabinofuranosyl-uracil and -cytosine and 2'-O-allyl-9-beta-D-arabinofuranosyl-adenine and evaluated their inhibitory activity against recombinant murine RNR. 2'-O-Allyl-araUDP proved to be inhibitory to RNR at an IC(50) of 100 microM, whereas 2'-O-allyl-araCDP was only marginally active (IC(50) 1 mM) and 2'-O-allyl-araADP was completely inactive. The susceptibility of the parent nucleosides to phosphorylation by thymidine kinase and 2'-deoxycytidine kinase was also investigated, and all nucleosides proved to be poor substrates for the above-cited kinases. Moreover, prodrugs of 2'-O-allyl-araU and -araC monophosphates, namely 2'-O-allyl-5'-(phenylethoxy-L-alanyl phosphate)-araU and -araC, were prepared and tested against tumor cell proliferation but proved to be inactive. A molecular modeling study has been conducted in order to explain our results. The data confirm that for both the natural and analogue nucleoside diphosphates, the principal determinant interaction with the active site of RNR is with the diphosphate group, which forms strong hydrogen bonds with Glu623, Thr624, Ser625, and Thr209. Our findings indicate that the poor phosphorylation may represent an explanation for the lack of marked in vitro cytostatic activity of the test compounds.


Assuntos
Antineoplásicos/síntese química , Arabinofuranosiluracila/síntese química , Citarabina/síntese química , Ribonucleotídeo Redutases/antagonistas & inibidores , Vidarabina/síntese química , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Arabinofuranosiluracila/química , Arabinofuranosiluracila/farmacologia , Citarabina/química , Citarabina/farmacologia , Desoxicitidina Quinase/química , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Camundongos , Modelos Moleculares , Fosforilação , Pró-Fármacos/síntese química , Pró-Fármacos/química , Pró-Fármacos/farmacologia , Proteínas Recombinantes/antagonistas & inibidores , Relação Estrutura-Atividade , Timidina Quinase/química , Células Tumorais Cultivadas , Vidarabina/química , Vidarabina/farmacologia
13.
Nucleosides Nucleotides ; 18(4-5): 1007-8, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10432732

RESUMO

The diphosphates of a series of 2'-O-allyl-1-beta-D-arabinofuranosyl derivatives, previously obtained by us, have been prepared and tested for their inhibitory activity in an in vitro assay using R1 and R2 subunits of the purified recombinant mouse ribonucleotide reductase (RNR). 2'-O-Allyl-araU diphosphate proved to be inhibitory, with an IC50 of 100 microM. The 5'-phosphoramidate pronucleotide of 2'-O-allyl-araU was also prepared and tested for inhibition of tumor cell proliferation.


Assuntos
Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Nucleosídeos/síntese química , Nucleosídeos/farmacologia , Ribonucleotídeo Redutases/antagonistas & inibidores , Animais , Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Divisão Celular/efeitos dos fármacos , Camundongos , Compostos Organofosforados/química
14.
J Biol Chem ; 274(34): 23746-51, 1999 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-10446134

RESUMO

Ribonucleotide reductase catalyzes all de novo synthesis of deoxyribonucleotides. The mammalian enzyme consists of two non-identical subunits, the R1 and R2 proteins, each inactive alone. The R1 subunit contains the active site, whereas the R2 protein harbors a binuclear iron center and a tyrosyl free radical essential for catalysis. It has been proposed that the radical properties of the R2 subunit are transferred approximately 35 A to the active site of the R1 protein, through a coupled electron/proton transfer along a conserved hydrogen-bonded chain, i.e. a radical transfer pathway (RTP). To gain a better insight into the properties and requirements of the proposed RTP, we have used site-directed mutagenesis to replace the conserved tyrosine 370 in the mouse R2 protein with tryptophan or phenylalanine. This residue is located close to the flexible C terminus, known to be essential for binding to the R1 protein. Our results strongly indicate that Tyr(370) links the RTP between the R1 and R2 proteins. Interruption of the hydrogen-bonded chain in Y370F inactivates the enzyme complex. Alteration of the same chain in Y370W slows down the RTP, resulting in a 58 times lower specific activity compared with the native R2 protein and a loss of the free radical during catalysis.


Assuntos
Ribonucleotídeo Redutases/química , Animais , Catálise , Radicais Livres , Ferro , Camundongos , Mutagênese Sítio-Dirigida , Relação Estrutura-Atividade , Tirosina
15.
J Biol Chem ; 274(25): 17696-704, 1999 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-10364210

RESUMO

The ferrous iron/oxygen reconstitution reaction in protein R2 of mouse and Escherichia coli ribonucleotide reductase (RNR) leads to the formation of a stable protein-linked tyrosyl radical and a mu-oxo-bridged diferric iron center, both necessary for enzyme activity. We have studied the reconstitution reaction in three protein R2 mutants Y177W, Y177F, and Y177C of mouse RNR to investigate if other residues at the site of the radical forming Tyr-177 can harbor free radicals. In Y177W we observed for the first time the formation of a tryptophan radical in protein R2 of mouse RNR with a lifetime of several minutes at room temperature. We assign it to an oxidized neutral tryptophan radical on Trp-177, based on selective deuteration and EPR and electron nuclear double resonance spectroscopy in H2O and D2O solution. The reconstitution reaction at 22 degrees C in both Y177F and Y177C leads to the formation of a so-called intermediate X which has previously been assigned to an oxo (hydroxo)-bridged Fe(III)/Fe(IV) cluster. Surprisingly, in both mutants that do not have successor radicals as Trp. in Y177W, this cluster exists on a much longer time scale (several seconds) at room temperature than has been reported for X in E. coli Y122F or native mouse protein R2. All three mouse R2 mutants were enzymatically inactive, indicating that only a tyrosyl radical at position 177 has the capability to take part in the reduction of substrates.


Assuntos
Escherichia coli/enzimologia , Ferro/química , Oxigênio/química , Ribonucleotídeo Redutases/química , Triptofano/química , Tirosina/química , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Espectroscopia de Ressonância de Spin Eletrônica , Radicais Livres/química , Cinética , Espectrometria de Massas , Camundongos , Estrutura Molecular , Mutação , Ribonucleotídeo Redutases/genética , Espectrofotometria
16.
Biochemistry ; 38(12): 3610-4, 1999 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-10090747

RESUMO

8-Oxo-7,8-dihydroguanine- (8-oxoguanine-) containing nucleotides are generated in the cellular nucleotide pool by the action of oxygen radicals produced during normal cellular metabolism. We examined the interconversion and metabolic fate of 8-oxoguanine-containing ribonucleotides in mammalian cells. (1) 8-OxoGTP can be generated not only by direct oxidation of GTP but also by phosphorylation of 8-oxoGDP by nucleotide diphosphate kinase, and the 8-oxoGTP thus formed can serve as a substrate for RNA polymerase II to induce transcription errors. (2) MTH1 protein carrying intrinsic 8-oxo-dGTPase activity has the potential to hydrolyze 8-oxoGTP to 8-oxoGMP, thus preventing misincorporation of 8-oxoguanine into RNA. 8-OxoGMP, the degradation product, cannot be reutilized, since guanylate kinase, which has the potential to phosphorylate both GMP and dGMP, is inactive on 8-oxoGMP. (3) Ribonucleotide reductase, which catalyzes reduction of four naturally occurring ribonucleoside diphosphates, cannot convert 8-oxoguanine-containing ribonucleotide to the deoxyribonucleotide. This step appears to serve as a gatekeeper to prevent formation of mutagenic substrates for DNA synthesis from oxidized ribonucleotides.


Assuntos
Guanina/análogos & derivados , Guanosina Trifosfato/análogos & derivados , Ribonucleotídeo Redutases/metabolismo , Animais , Bovinos , Células Cultivadas , Guanina/metabolismo , Guanina/farmacologia , Nucleotídeos de Guanina/metabolismo , Guanosina Trifosfato/metabolismo , Guanosina Trifosfato/farmacologia , Humanos , Células Jurkat , Mamíferos , RNA/biossíntese , RNA/efeitos dos fármacos , RNA/metabolismo , Especificidade por Substrato , Suínos
17.
J Biol Chem ; 273(51): 34098-104, 1998 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-9852067

RESUMO

Trypanosoma brucei is the causative agent for African sleeping sickness. We have made in vitro and in vivo studies on the allosteric regulation of the trypanosome ribonucleotide reductase, a key enzyme in the production of dNTPs needed for DNA synthesis. Results with the isolated recombinant trypanosome ribonucleotide reductase showed that dATP specifically directs pyrimidine ribonucleotide reduction instead of being a general negative effector as in other related ribonucleotide reductases, whereas dTTP and dGTP directed GDP and ADP reduction, respectively. Pool measurements of NDPs, NTPs, and dNTPs in the cultivated bloodstream form of trypanosomes exposed to deoxyribonucleosides or inhibited by hydroxyurea confirmed our in vitro allosteric regulation model of ribonucleotide reductase. Interestingly, the trypanosomes had extremely low CDP and CTP pools, whereas the dCTP pool was comparable with that of other dNTPs. The trypanosome ribonucleotide reductase seems adapted to this situation by having a high affinity for the CDP/UDP-specific effector dATP and a high catalytic efficiency, Kcat/Km, for CDP reduction. Thymidine and deoxyadenosine were readily taken up and phosphorylated to dTTP and dATP, respectively, the latter in a nonsaturating manner. This uncontrolled uptake of deoxyadenosine strongly inhibited trypanosome proliferation, a valuable observation in the search for new trypanocidal nucleoside analogues.


Assuntos
Desoxirribonucleotídeos/metabolismo , Ribonucleotídeo Redutases/metabolismo , Ribonucleotídeos/metabolismo , Trypanosoma brucei brucei/enzimologia , Regulação Alostérica , Animais , Desoxirribonucleotídeos/farmacologia , Cinética , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Trypanosoma brucei brucei/isolamento & purificação , Tripanossomíase Africana/sangue , Tripanossomíase Africana/parasitologia
18.
J Biol Chem ; 273(45): 29816-21, 1998 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-9792697

RESUMO

Ribonucleotide reductase is essential for DNA synthesis. In mammalian cells, the enzyme consists of two non-identical subunits, proteins R1 and R2. The expression of the mouse R1 and R2 genes is strictly correlated to S phase. Using promoter-reporter gene constructs, we have defined a region of the TATA-less mouse ribonucleotide reductase R1 gene promoter that correlates reporter gene expression to S phase. This is demonstrated in stably transformed cells both synchronized by serum starvation and separated by centrifugal elutriation, suggesting that the R1 gene expression during the cell cycle is mainly regulated at the transcriptional level. The region contains four protein-binding DNA elements, beta (nucleotides -189 to -167), alpha (-98 to -76), Inr (-4 to +16), and gamma (+34 to +61), together regulating promoter activity. The nearly identical upstream elements, alpha and beta, each form three DNA-protein complexes in gel shift assays. We have identified YY1 as a component in at least one of the complexes using supershift antibodies and a yeast one-hybrid screening of a mouse cDNA library using the alpha element as a target. Transient transfection assays demonstrate that the alpha and beta elements are mainly important for the R1 promoter strength and suggest that YY1 functions as an activator.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Regiões Promotoras Genéticas , Ribonucleotídeo Redutases/genética , Fatores de Transcrição/metabolismo , Animais , Sequência de Bases , Ciclo Celular , Pegada de DNA , Primers do DNA , Fatores de Ligação de DNA Eritroide Específicos , Regulação da Expressão Gênica , Camundongos , Ligação Proteica , Processamento Pós-Transcricional do RNA , TATA Box , Transcrição Gênica , Células Tumorais Cultivadas , Fator de Transcrição YY1
19.
J Biol Chem ; 273(34): 21463-72, 1998 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-9705274

RESUMO

Class I ribonucleotide reductases consist of two subunits, R1 and R2. The active site is located in R1; active R2 contains a diferric center and a tyrosyl free radical (Tyr.), both essential for enzymatic activity. The proposed mechanism for the enzymatic reaction includes the transport of a reducing equivalent, i.e. electron or hydrogen radical, across a 35-A distance between Tyr. in R2 and the active site in R1, which are connected by a hydrogen-bonded chain of conserved, catalytically essential amino acid residues. Asp266 and Trp103 in mouse R2 are part of this radical transfer pathway. The diferric/Tyr. site in R2 is reconstituted spontaneously by mixing iron-free apoR2 with Fe(II) and O2. The reconstitution reaction requires the delivery of an external reducing equivalent to form the diferric/Tyr. site. Reconstitution kinetics were investigated in mouse apo-wild type R2 and the three mutants D266A, W103Y, and W103F by rapid freeze-quench electron paramagnetic resonance with >/=4 Fe(II)/R2 at various reaction temperatures. The kinetics of Tyr. formation in D266A and W103Y is on average 20 times slower than in wild type R2. More strikingly, Tyr. formation is completely suppressed in W103F. No change in the reconstitution kinetics was found starting from Fe(II)-preloaded proteins, which shows that the mutations do not affect the rate of iron binding. Our results are consistent with a reaction mechanism using Asp266 and Trp103 for delivery of the external reducing equivalent. Further, the results with W103F suggest that an intact hydrogen-bonded chain is crucial for the reaction, indicating that the external reducing equivalent is a H. Finally, the formation of Tyr. is not the slowest step of the reaction as it is in Escherichia coli R2, consistent with a stronger interaction between Tyr. and the iron center in mouse R2. A new electron paramagnetic resonance visible intermediate named mouse X, strikingly similar to species X found in E. coli R2, was detected only in small amounts under certain conditions. We propose that it may be an intermediate in a side reaction leading to a diferric center without forming the neighboring Tyr.


Assuntos
Ribonucleotídeo Redutases/metabolismo , Tirosina/metabolismo , Animais , Sítios de Ligação , Dimerização , Radicais Livres , Cinética , Magnetismo , Camundongos , Modelos Químicos , Mutagênese Sítio-Dirigida , Ribonucleotídeo Redutases/genética , Temperatura
20.
J Lipid Res ; 38(10): 2111-24, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9374133

RESUMO

Guinea pig apolipoprotein C-II (apoC-II) lacks four amino acid residues in the amino-terminal, lipid-binding part compared to apoC-II from other mammalian species (Andersson et al. 1991. J. Biol. Chem. 266: 4074-4080). To explore whether this structural difference explains the low ability of guinea pig plasma to activate lipoprotein lipase in vitro, we have expressed guinea pig apoC-II in Escherichia coli and have constructed an insertion mutant with the four missing amino acid residues compared to human apoC-II. With a synthetic emulsion of long-chain triacylglycerols, both the wild-type guinea pig apoC-II and the insertion mutant stimulated lipoprotein lipase similar to human apoC-II, but with chylomicrons from an apoC-II-deficient patient, 5- to 10-fold more of both wild-type guinea pig apoC-II and the insertion mutant were needed. Studies of tryptophane fluorescence indicated a slight difference in how guinea pig apoC-II interacted with liposomes, and presumably with lipoproteins, as compared to human apoC-II. The level of apoC-II (11.5 +/- 5.4 microg/ml) was lower in guinea pig compared to human plasma, and most of guinea pig apoC-II was on HDL-like particles. These had decreased ability to donate apoC-II to lipid emulsions compared to human HDL. Some guinea pig apoC-II was associated with LDL which, as demonstrated by surface plasmon resonance, had higher affinity for lipoprotein lipase than human LDL, and inhibited rather than stimulated the lipase reaction in vitro. We conclude that while guinea pig apoC-II is fully competent to stimulate lipoprotein lipase, the sum of several different factors explains the low ability of guinea pig plasma to accomplish stimulation.


Assuntos
Apolipoproteínas C/genética , Apolipoproteínas C/metabolismo , Lipoproteínas HDL/sangue , Lipoproteínas LDL/sangue , Animais , Apolipoproteína C-II , Apolipoproteínas C/sangue , Apolipoproteínas C/farmacologia , Centrifugação com Gradiente de Concentração , Cromatografia Líquida de Alta Pressão , Quilomícrons/metabolismo , Escherichia coli/genética , Emulsões Gordurosas Intravenosas/metabolismo , Expressão Gênica , Cobaias , Humanos , Cinética , Lipase Lipoproteica/metabolismo , Lipoproteínas/sangue , Lipoproteínas/isolamento & purificação , Lipoproteínas HDL/química , Lipoproteínas LDL/química , Masculino , Mutagênese Insercional , Ligação Proteica , Proteínas Recombinantes/biossíntese , Deleção de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA