Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 195
Filtrar
1.
Biochemistry ; 51(45): 9094-103, 2012 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-23050701

RESUMO

Pseudomonas aeruginosa possesses an unusual pathway for 5'-methylthioadenosine (MTA) metabolism involving deamination to 5'-methylthioinosine (MTI) followed by N-ribosyl phosphorolysis to hypoxanthine and 5-methylthio-α-d-ribose 1-phosphate. The specific MTI phosphorylase of P. aeruginosa has been reported [Guan, R., Ho, M. C., Almo, S. C., and Schramm, V. L. (2011) Biochemistry 50, 1247-1254], and here we characterize MTA deaminase from P. aeruginosa (PaMTADA). Genomic analysis indicated the PA3170 locus to be a candidate for MTA deaminase (MTADA). Protein encoded by PA3170 was expressed and shown to deaminate MTA with 40-fold greater catalytic efficiency for MTA than for adenosine. The k(cat)/K(m) value of 1.6 × 10(7) M(-1) s(-1) for MTA is the highest catalytic efficiency known for an MTA deaminase. 5'-Methylthiocoformycin (MTCF) is a 4.8 pM transition state analogue for PaMTADA but causes no significant inhibition of human adenosine deaminase or MTA phosphorylase. MTCF is permeable to P. aeruginosa and exhibits an IC(50) of 3 nM on cellular PaMTADA activity. PaMTADA is the only activity in P. aeruginosa extracts to act on MTA. MTA and 5-methylthio-α-d-ribose are involved in quorum sensing pathways; thus, PaMTADA is a potential target for quorum sensing. The crystal structure of PaMTADA in complex with MTCF shows the transition state mimic 8(R)-hydroxyl group in contact with a catalytic site Zn(2+), the 5'-methylthio group in a hydrophobic pocket, and the transition state mimic of the diazepine ring in contact with a catalytic site Glu.


Assuntos
Desoxiadenosinas/metabolismo , Nucleosídeo Desaminases/metabolismo , Pseudomonas aeruginosa/enzimologia , Percepção de Quorum , Tionucleosídeos/metabolismo , Adenosina Desaminase/metabolismo , Sequência de Aminoácidos , Coformicina/análogos & derivados , Coformicina/farmacologia , Cristalografia por Raios X , Humanos , Ligases , Metiltioinosina/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Nucleosídeo Desaminases/antagonistas & inibidores , Alinhamento de Sequência , Especificidade por Substrato
2.
Annu Rev Immunol ; 26: 317-53, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18304004

RESUMO

All retroviruses, including HIV-1, display species-specific patterns of infection. The impaired growth of these retroviruses in foreign and sometimes even in their natural hosts often stems from the action of potent host-encoded "viral restriction factors" that form important protective components of the innate immune system. The discovery of APOBEC3G and related cytidine deaminases as one class of host restriction factors and of the action of HIV-1 Vif as a specific APOBEC3G antagonist have stimulated intense scientific interest. This Vif-APOBEC3G axis now forms a very attractive target for development of an entirely new class of anti-HIV drugs. In this review, we summarize current understanding of the mechanism of action of the APOBEC3 family of enzymes, their intriguing regulation within cells, the impact of these enzymes on viral evolution and disease progression, and their roles in controlling not only the replication of exogenous retroviruses but also the retrotransposition of endogenous retroelements.


Assuntos
Imunidade Inata/fisiologia , Nucleosídeo Desaminases/metabolismo , Retroviridae/fisiologia , Desaminase APOBEC-3G , Animais , Citidina Desaminase/antagonistas & inibidores , Citidina Desaminase/metabolismo , Produtos do Gene vif/fisiologia , Humanos , Modelos Biológicos , Nucleosídeo Desaminases/antagonistas & inibidores , Retroviridae/genética , Replicação Viral
3.
Biochemistry ; 46(47): 13638-47, 2007 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-17973496

RESUMO

To estimate the relative importance of alternate routes of spontaneous degradation of DNA and the rate enhancements produced by enzymes catalyzing these reactions, rate constants and thermodynamic activation parameters for the degradation of 2'-deoxynucleosides at 25 degrees C were determined by extrapolation of rates observed in the temperature range between 90 and 200 degrees C in neutral phosphate buffer. Rates of deamination of 2'-deoxycytidine, 1-methylcytosine, and cytidine were found to be identical within experimental error (t1/2 approximately 20 years, 37 degrees C). Rate constants for deamination of 2'-deoxyadenosine and 2'-deoxyguanosine, which could not be determined directly because of rapid glycoside cleavage, were estimated by assuming that methyl replacement should generate reasonable model substrates. The rates of deamination of 9-methyladenine and 9-methylguanine were found to be similar to each other (t1/2 approximately 6000 years, 37 degrees C) and approximately 10(2)-fold slower than the rates of glycoside cleavage in 2'-deoxyadenosine and 2'-deoxyguanosine. The deamination of 2'-deoxyadenosine, 2'-deoxyguanosine, and 2'-deoxycytidine led to accelerated rates of glycoside cleavage. In the exceptional case of 2'-deoxycytidine, deamination and glycoside hydrolysis proceed at very similar rates at all temperatures. Glycoside cleavage proceeds with half-times ranging from 4 years for 2'-deoxyinosine to 40 years for 2'-deoxycytidine (37 degrees C). The rate enhancements produced by DNA glycosylases, estimated by comparison with the rates of these uncatalyzed reactions, are found to be substantially smaller than those produced by deaminases and staphylococcal nuclease.


Assuntos
DNA Glicosilases/química , DNA/metabolismo , Nucleosídeo Desaminases/química , Catálise , DNA/química , DNA Glicosilases/antagonistas & inibidores , DNA Glicosilases/metabolismo , Desaminação , Desoxirribonucleases/química , Desoxirribonucleases/metabolismo , Hidrólise , Cinética , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Nucleosídeo Desaminases/antagonistas & inibidores , Nucleosídeo Desaminases/metabolismo , Temperatura , Termodinâmica , Thermotoga maritima/enzimologia , Thermotoga maritima/fisiologia
4.
J Virol ; 81(19): 10588-96, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17652382

RESUMO

Interferons (IFNs) play a major role in the control of hepatitis B virus (HBV), whether as endogenous cytokines limiting the spread of the virus during the acute phase of the infection or as drugs for the treatment of its chronic phase. However, the mechanism by which IFNs inhibit HBV replication has so far remained elusive. Here, we show that type I and II IFN treatment of human hepatocytes induces the production of APOBEC3G (A3G) and, to a lesser extent, that of APOBEC3F (A3F) and APOBEC3B (A3B) but not that of two other cytidine deaminases also endowed with anti-HBV activity, activation-induced cytidine deaminase (AID), and APOBEC1. Most importantly, we reveal that blocking A3B, A3F, and A3G by combining RNA interference and the virion infectivity factor (Vif) protein of human immunodeficiency virus does not abrogate the inhibitory effect of IFNs on HBV. We conclude that these cytidine deaminases are not essential effectors of IFN in its action against this pathogen.


Assuntos
Citidina Desaminase/metabolismo , Vírus da Hepatite B/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Interferon Tipo I/farmacologia , Interferon gama/farmacologia , Replicação Viral/efeitos dos fármacos , Desaminase APOBEC-1 , Desaminase APOBEC-3G , Linhagem Celular Tumoral , Citidina Desaminase/antagonistas & inibidores , Citidina Desaminase/genética , Citocinas/farmacologia , Citosina Desaminase/antagonistas & inibidores , Citosina Desaminase/genética , Citosina Desaminase/metabolismo , HIV-1/efeitos dos fármacos , Vírus da Hepatite B/fisiologia , Hepatócitos/enzimologia , Humanos , Interferons , Antígenos de Histocompatibilidade Menor , Nucleosídeo Desaminases/antagonistas & inibidores , Nucleosídeo Desaminases/genética , Nucleosídeo Desaminases/metabolismo , RNA Interferente Pequeno/farmacologia , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo
5.
J Virol ; 81(15): 8201-10, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17522216

RESUMO

Human cytidine deaminases APOBEC3G (A3G) and APOBEC3F (A3F) inhibit replication of Vif-deficient human immunodeficiency virus type 1 (HIV-1). HIV-1 Vif overcomes these host restriction factors by binding to them and inducing their proteasomal degradation. The Vif-A3G and Vif-A3F interactions are attractive targets for antiviral drug development because inhibiting the interactions could allow the host defense mechanism to control HIV-1 replication. It was recently reported that the Vif amino acids D(14)RMR(17) are important for functional interaction and degradation of the previously identified Vif-resistant mutant of A3G (D128K-A3G). However, the Vif determinants important for functional interaction with A3G and A3F have not been fully characterized. To identify these determinants, we performed an extensive mutational analysis of HIV-1 Vif. Our analysis revealed two distinct Vif determinants, amino acids Y(40)RHHY(44) and D(14)RMR(17), which are essential for binding to A3G and A3F, respectively. Interestingly, mutation of the A3G-binding region increased Vif's ability to suppress A3F. Vif binding to D128K-A3G was also dependent on the Y(40)RHHY(44) region but not the D(14)RMR(17) region. Consistent with previous observations, subsequent neutralization of the D128K-A3G antiviral activity required substitution of Vif determinant D(14)RMR(17) with SEMQ, similar to the SERQ amino acids in simian immunodeficiency virus SIV(AGM) Vif, which is capable of neutralizing D128K-A3G. These studies are the first to clearly identify two distinct regions of Vif that are critical for independent interactions with A3G and A3F. Pharmacological interference with the Vif-A3G or Vif-A3F interactions could result in potent inhibition of HIV-1 replication by the APOBEC3 proteins.


Assuntos
Citosina Desaminase/metabolismo , Produtos do Gene vif/metabolismo , HIV-1/metabolismo , Nucleosídeo Desaminases/metabolismo , Proteínas Repressoras/metabolismo , Desaminase APOBEC-3G , Alanina/metabolismo , Linhagem Celular , Citidina Desaminase , Citosina Desaminase/antagonistas & inibidores , Citosina Desaminase/genética , Análise Mutacional de DNA , Produtos do Gene vif/química , Produtos do Gene vif/genética , Humanos , Mutagênese , Nucleosídeo Desaminases/antagonistas & inibidores , Nucleosídeo Desaminases/genética , Ligação Proteica , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/genética , Produtos do Gene vif do Vírus da Imunodeficiência Humana
6.
PLoS Pathog ; 3(2): e15, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17291161

RESUMO

APOBEC3G (A3G) is a potent antiretroviral deoxycytidine deaminase that, when incorporated into HIV virions, hypermutates nascent viral DNA formed during reverse transcription. HIV Vif counters the effect of A3G by depleting intracellular stores of the enzyme, thereby blocking its virion incorporation. Through pulse-chase analyses, we demonstrate that virion A3G is mainly recruited from the cellular pool of newly synthesized enzyme compared to older "mature" A3G already residing in high-molecular-mass RNA-protein complexes. Virion-incorporated A3G forms a large complex with viral genomic RNA that is clearly distinct from cellular HMM A3G complexes, as revealed by both gel filtration and biochemical fractionation. Unexpectedly, the enzymatic activity of virion-incorporated A3G is lost upon its stable association with HIV RNA. The activity of the latent A3G enzyme is ultimately restored during reverse transcription by the action of HIV RNase H. Degradation of the viral genomic RNA by RNase H not only generates the minus-strand DNA substrate targeted by A3G for hypermutation but also removes the inhibitory RNA bound to A3G, thereby enabling its function as a deoxycytidine deaminase. These findings highlight an unexpected interplay between host and virus where initiation of antiviral enzymatic activity is dependent on the action of an essential viral enzyme.


Assuntos
HIV/metabolismo , Nucleosídeo Desaminases/fisiologia , RNA Viral/metabolismo , Proteínas Repressoras/fisiologia , Ribonuclease H/metabolismo , Vírion/metabolismo , Desaminase APOBEC-3G , Citidina Desaminase , Ativação Enzimática , Produtos do Gene vif/fisiologia , HIV/genética , Humanos , Nucleosídeo Desaminases/antagonistas & inibidores , Proteínas Repressoras/antagonistas & inibidores , Transcrição Gênica , Montagem de Vírus , Produtos do Gene vif do Vírus da Imunodeficiência Humana
7.
Mol Immunol ; 44(4): 583-90, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16580072

RESUMO

HIV-1 Vif protein protects viral replication in non-permissive cells by inducing degradation of APOBEC3G via ubiquitination and proteasomal pathway, although new studies indicate a putative role in Vif's direct inhibition of APOBEC3G. APOBEC3G is member of a homologous family of proteins with cytidine deaminase activity expressed with characteristic tissue specificity, that in humans consist of APOBEC1, APOBEC2, APOBEC3A-H, APOBEC4 and the activation-induced deaminase (AID), a B lymphoid protein necessary for somatic hypermutation, gene conversion and class switch recombination. In this work we show that Vif can counteract AID's activity in E. coli in absence of specific eukaryotic co-factors necessary for AID induced somatic hypermutation, gene conversion and to stimulate class switch recombination in B-cells. We show that AID inhibition is mediated by a direct protein-protein interaction via unique amino acid D118 an homologous mutant responsible for the species-specific restriction of HIV-1 Vif protein existent for APOBEC3G. These results raise the hypothesis that Vif related proteins can act as a broad inhibitor of deaminase activity. Moreover as AID and Vif evolved in different cellular environments, these results may indicate that Vif related proteins might mimic cellular factors that interact with a structural conserved domain of cytidine deaminases during evolution.


Assuntos
Citidina Desaminase/antagonistas & inibidores , Produtos do Gene vif/genética , HIV-1 , Desaminase APOBEC-3G , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Linfócitos B/enzimologia , Linfócitos B/virologia , Citidina Desaminase/genética , Escherichia coli , Produtos do Gene vif/metabolismo , Rearranjo Gênico do Linfócito B , Humanos , Camundongos , Dados de Sequência Molecular , Mutação , Nucleosídeo Desaminases/antagonistas & inibidores , Nucleosídeo Desaminases/genética , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/genética , Produtos do Gene vif do Vírus da Imunodeficiência Humana
8.
Virology ; 349(1): 31-40, 2006 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-16460778

RESUMO

APOBEC3G and APOBEC3F exhibit antiretroviral activity primarily as a consequence of their ability to deaminate cytidines in retroviral DNA. Here, we compare the properties of APOBEC3F and APOBEC3G from human, macaque, and African green monkey (AGM). While all APOBEC proteins tested exhibited anti-HIV-1 activity, human APOBEC3F was, surprisingly, 10- to 50-fold less potent than human APOBEC3G. However, similar discrepancies in antiviral potency were not found when pairs of proteins from macaque and AGM were compared. Intrinsic differences in the ability of each APOBEC protein to induce hypermutation, rather than differences in packaging efficiency, partially accounted for variable antiretroviral activity. Each of four primate lentivirus Vif proteins reduced human and AGM APOBEC3F expression and antiviral activity, but all were only partially effective and species-specific effects were relatively minor. Overall, highly efficient and species-specific neutralization of APOBEC3G, and less efficient neutralization of APOBEC3F, appears to be a general property of Vif proteins.


Assuntos
Fármacos Anti-HIV , Citidina Desaminase/fisiologia , Desaminase APOBEC-3G , Sequência de Aminoácidos , Animais , Fármacos Anti-HIV/antagonistas & inibidores , Chlorocebus aethiops , Citidina Desaminase/antagonistas & inibidores , Citosina Desaminase/antagonistas & inibidores , Citosina Desaminase/química , Citosina Desaminase/fisiologia , Regulação da Expressão Gênica , Produtos do Gene vif/fisiologia , HIV-1/efeitos dos fármacos , HIV-1/crescimento & desenvolvimento , Humanos , Macaca mulatta , Mutação , Nucleosídeo Desaminases/antagonistas & inibidores , Nucleosídeo Desaminases/química , Nucleosídeo Desaminases/fisiologia , RNA Viral/genética , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/química , Proteínas Repressoras/fisiologia , Homologia de Sequência de Aminoácidos , Montagem de Vírus , Inativação de Vírus , Produtos do Gene vif do Vírus da Imunodeficiência Humana
9.
J Virol ; 80(6): 3112-5, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16501124

RESUMO

APOBEC3G (A3G) and related cytidine deaminases, such as APOBEC3F (A3F), are potent inhibitors of retroviruses. Formation of infectious human immunodeficiency virus type 1 (HIV-1) requires suppression of multiple cytidine deaminases by Vif. Whether HIV-1 Vif recognizes various APOBEC3 proteins through a common mechanism is unclear. The domains in Vif that mediate APOBEC3 recognitions are also poorly defined. The N-terminal region of HIV-1 Vif is unusually rich in Trp residues, which are highly conserved. In the present study, we examined the role of these Trp residues in the suppression of APOBEC3 proteins by HIV-1 Vif. We found that most of the highly conserved Trp residues were required for efficient suppression of both A3G and A3F, but some of these residues were selectively required for the suppression of A3F but not A3G. Mutant Vif molecules in which Ala was substituted for Trp79 and, to a lesser extent, for Trp11 remained competent for A3G interaction and its suppression; however, they were defective for A3F interaction and therefore could not efficiently suppress the antiviral activity of A3F. Interestingly, while the HIV-1 Vif-mediated degradation of A3G was not affected by the different C-terminal tag peptides, that of A3F was significantly influenced by its C-terminal tags. These data indicate that the mechanisms by which HIV-1 Vif recognizes its target molecules, A3G and A3F, are not identical. The fact that several highly conserved residues in Vif are required for the suppression of A3F but not that of A3G suggests a critical role for A3F in the restriction of HIV-1 in vivo.


Assuntos
Antivirais/antagonistas & inibidores , Citosina Desaminase/antagonistas & inibidores , Produtos do Gene vif/química , HIV-1/patogenicidade , Nucleosídeo Desaminases/antagonistas & inibidores , Proteínas Repressoras/antagonistas & inibidores , Triptofano/química , Desaminase APOBEC-3G , Antivirais/metabolismo , Linhagem Celular , Citidina Desaminase , Citosina Desaminase/genética , Citosina Desaminase/metabolismo , Regulação da Expressão Gênica , Produtos do Gene vif/genética , Produtos do Gene vif/metabolismo , Humanos , Mutação , Nucleosídeo Desaminases/genética , Nucleosídeo Desaminases/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transfecção , Produtos do Gene vif do Vírus da Imunodeficiência Humana
10.
Chem Res Toxicol ; 19(1): 50-7, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16411656

RESUMO

Nitric oxide (NO) is a physiologically important molecule that has been implicated in the pathophysiology of diseases associated with chronic inflammation, such as cancer. While the complicated chemistry of NO-mediated genotoxicity has been extensively study in vitro, neither the spectrum of DNA lesions nor their consequences in vivo have been rigorously defined. We have approached this problem by exposing human TK6 lymphoblastoid cells to controlled steady-state concentrations of 1.75 or 0.65 microM NO along with 186 microM O2 in a recently developed reactor that avoids the anomalous gas-phase chemistry of NO and approximates the conditions at sites of inflammation in tissues. The resulting spectrum of nucleobase deamination products was defined using a recently developed liquid chromatography/mass spectrometry (LC/MS) method, and the results were correlated with cytotoxicity and apoptosis. A series of control experiments revealed the necessity of using dC and dA deaminase inhibitors to avoid adventitious formation of 2'-deoxyuridine (dU) and 2'-deoxyinosine (dI), respectively, during DNA isolation and processing. Exposure of TK6 cells to 1.75 microM NO and 186 microM O2 for 12 h (1260 microM x min dose) resulted in 32% loss of cell viability measured immediately after exposure and 87% cytotoxicity after a 24 h recovery period. The same exposure resulted in 3.5-, 3.8-, and 4.1-fold increases in dX, dI, and dU, respectively, to reach the following levels: dX, 7 (+/- 1) per 10(6) nt; dI, 25 (+/- 2.1) per 10(6) nt; and dU, 40 (+/- 3.8) per 10(6) nt. dO was not detected above the limit of detection of 6 lesions per 10(7) nt in 50 microg of DNA. A 12 h exposure to 0.65 microM NO and 190 microM O2 (468 microM x min dose) caused 1.7-, 1.8-, and 2.0-fold increases in dX, dI, and dU, respectively, accompanied by a approximately 15% (+/- 3.6) reduction in cell viability immediately after exposure. Again, dO was not detected. These results reveal modest increases in the steady-state levels of DNA deamination products in cells exposed to relatively cytotoxic levels of NO. This could result from limited nitrosative chemistry in nuclear DNA in cells exposed to NO or high levels of formation balanced by rapid repair of nucleobase deamination lesions in DNA.


Assuntos
Dano ao DNA , Óxido Nítrico/toxicidade , Linhagem Celular Tumoral , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Coformicina/farmacologia , DNA/química , DNA/metabolismo , Desaminação , Desoxiuridina/análise , Desoxiuridina/metabolismo , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Humanos , Inosina/análogos & derivados , Inosina/análise , Inosina/metabolismo , Nucleosídeo Desaminases/antagonistas & inibidores , Nucleosídeo Desaminases/metabolismo , Oxigênio , Tetra-Hidrouridina/farmacologia
11.
J Gen Virol ; 84(Pt 6): 1437-1441, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12771412

RESUMO

We have previously observed that the expression of two thymidylate biosynthesis enzymes, dihydrofolate reductase and thymidylate synthase (TS), is upregulated in quiescent human fibroblasts infected with human cytomegalovirus (HCMV). Here, we have demonstrated that HCMV increases expression of the cellular deoxycytidylate deaminase (dCMP deaminase), which provides the substrate for TS by converting dCMP to dUMP. We observed an increase in dCMP deaminase protein levels, whereas deoxyuridine triphosphatase (dUTPase), another cellular enzyme that may provide dUMP by hydrolysing dUTP, was undetectable. The essential requirement of cellular dCMP deaminase for productive HCMV replication was further emphasized by showing that a precursor of a potent dCMP deaminase inhibitor, zebularine, suppressed virus replication and DNA synthesis. These results suggest that HCMV exploits the host's dCMP deaminase activity to replicate in quiescent cells.


Assuntos
Citomegalovirus/fisiologia , Citomegalovirus/patogenicidade , Nucleosídeo Desaminases/fisiologia , Replicação Viral/fisiologia , Células Cultivadas , Citidina/análogos & derivados , Citidina Desaminase , Citomegalovirus/efeitos dos fármacos , Citomegalovirus/crescimento & desenvolvimento , DNA Viral/biossíntese , Inibidores Enzimáticos/farmacologia , Humanos , Interfase , Nucleosídeo Desaminases/antagonistas & inibidores , Nucleosídeos de Pirimidina/farmacologia , Pirofosfatases/fisiologia , Replicação Viral/efeitos dos fármacos
12.
J Mol Biol ; 315(4): 687-97, 2002 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-11812140

RESUMO

Cytosine deaminase (CD) catalyzes the deamination of cytosine, producing uracil. This enzyme is present in prokaryotes and fungi (but not multicellular eukaryotes) and is an important member of the pyrimidine salvage pathway in those organisms. The same enzyme also catalyzes the conversion of 5-fluorocytosine to 5-fluorouracil; this activity allows the formation of a cytotoxic chemotherapeutic agent from a non-cytotoxic precursor. The enzyme is of widespread interest both for antimicrobial drug design and for gene therapy applications against tumors. The structure of Escherichia coli CD has been determined in the presence and absence of a bound mechanism-based inhibitor. The enzyme forms an (alphabeta)(8) barrel structure with structural similarity to adenosine deaminase, a relationship that is undetectable at the sequence level, and no similarity to bacterial cytidine deaminase. The enzyme is packed into a hexameric assembly stabilized by a unique domain-swapping interaction between enzyme subunits. The active site is located in the mouth of the enzyme barrel and contains a bound iron ion that coordinates a hydroxyl nucleophile. Substrate binding involves a significant conformational change that sequesters the reaction complex from solvent.


Assuntos
Escherichia coli/enzimologia , Nucleosídeo Desaminases/química , Adenosina Desaminase/química , Adenosina Desaminase/metabolismo , Sítios de Ligação , Catálise , Cristalografia por Raios X , Citosina Desaminase , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Flucitosina/metabolismo , Terapia Genética/métodos , Humanos , Modelos Moleculares , Nucleosídeo Desaminases/antagonistas & inibidores , Nucleosídeo Desaminases/metabolismo , Pró-Fármacos/metabolismo , Dobramento de Proteína , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Subunidades Proteicas , Homologia de Sequência de Aminoácidos , Solventes/metabolismo , Relação Estrutura-Atividade
13.
Curr Opin Oncol ; 8(6): 499-508, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8971470

RESUMO

Gene transfer for the treatment of cancer is a rapidly expanding field. Recent studies can be divided into four main areas: 1) transfer of suicide genes that convert inactive prodrugs into cytotoxic compounds, 2) transfer of genes encoding cytokines and stimulatory markers to enhance immunogenicity against tumors, 3) transfer of tumor-suppressor genes to block tumor cell proliferation, and 4) transfer of drug resistance genes into hematopoietic stem cells to increase their resistance to myelo-suppressive chemotherapeutic agents. This review discusses recent advances in technique and knowledge and their application to the gene therapy of cancer.


Assuntos
Terapia Genética , Neoplasias/terapia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Animais , Antivirais/uso terapêutico , Biotransformação , Citocinas/genética , Citosina Desaminase , Resistência a Múltiplos Medicamentos/genética , Resistencia a Medicamentos Antineoplásicos/genética , Inibidores Enzimáticos/uso terapêutico , Ganciclovir/uso terapêutico , Genes Supressores de Tumor , Glutationa Transferase/genética , Humanos , Interleucina-2/genética , Metiltransferases/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Nucleosídeo Desaminases/antagonistas & inibidores , O(6)-Metilguanina-DNA Metiltransferase , Pró-Fármacos/farmacocinética , Proteínas Recombinantes de Fusão/antagonistas & inibidores , Proteínas Recombinantes de Fusão/metabolismo , Simplexvirus/enzimologia , Tetra-Hidrofolato Desidrogenase/genética , Timidina Quinase/antagonistas & inibidores , Timidina Quinase/genética , Transfecção
15.
J Biol Chem ; 268(32): 24005-11, 1993 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-8226944

RESUMO

Cytosine deaminase (CDase, EC 3.5.4.1) isolated from Escherichia coli contains a catalytically essential divalent metal ion. Fe2+ was efficiently removed from the enzyme with o-phenanthroline to yield an apoenzyme with less than 5% of the catalytic activity of native enzyme. The time courses for inactivation and for removal of Fe2+ from the enzyme by o-phenanthroline were similar. Apoenzyme reconstituted with Fe2+, Mn2+, Co2+, or Zn2+ (M2+CDase) had kcat values of 185, 88, 50, and 32 s-1, respectively. The Km values of these M2+CDases for cytosine were similar (0.22-0.39 mM). Cytosine potently inhibited reconstitution of the apoenzyme with Fe2+. Fe2+CDase was rapidly inactivated by 1 mM H2O2 (t1/2 < 1 s), whereas Mn2+CDase, Co2+CDase, and Zn2+CDase were not inactivated by H2O2. CDase was also inhibited by excess divalent cations. Cu2+ and Zn2+ reversibly inhibited Fe2+CDase activity with inhibition constants of 1.8 and 5.8 microM, respectively. Cu2+ dissociated slowly from the secondary binding on CDase with a rate constant of 2 x 10(-3) s-1.


Assuntos
Metais/farmacologia , Nucleosídeo Desaminases/metabolismo , Catálise , Cátions Bivalentes , Cromatografia Líquida , Meios de Cultura , Citosina Desaminase , Eletroforese em Gel de Poliacrilamida , Escherichia coli/enzimologia , Peróxido de Hidrogênio/farmacologia , Nucleosídeo Desaminases/antagonistas & inibidores , Nucleosídeo Desaminases/isolamento & purificação
16.
Am J Physiol ; 259(3 Pt 2): H835-8, 1990 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-2396691

RESUMO

We tested the effect of the adenosine deaminase inhibitor erythro-9-(2-hydroxy-3-nonyl)adenosine (EHNA) on ischemia-reperfusion injury in isolated perfused rat heart. In the ischemia-reperfusion group (n = 10), ventricular fibrillation occurred within 3 min of reperfusion after the 40-min ischemic period. The incidence of ventricular fibrillation was 90% with a mean duration of 3.15 +/- 0.97 (SE) min. Resting tension increased significantly. By contrast, the incidence of ventricular fibrillation after reperfusion in the EHNA-treated (5 microM) group (n = 10) was 20% (P less than 0.01), and the duration was 0.30 +/- 0.21 min (P less than 0.01). Resting tension was significantly lower and around the normal level in the EHNA-treated group (P less than 0.01). Contraction amplitude and heart rate recovered to nearly normal compared with the ischemia-reperfusion group (P less than 0.01). Coronary flow was greater in the EHNA-treated group (P less than 0.01). It is concluded that EHNA protects the heart, possibly by accumulation of adenosine that benefits the hearts and by blocking the xanthine oxidase pathway for free radical generation.


Assuntos
Adenina/análogos & derivados , Inibidores de Adenosina Desaminase , Coração/fisiopatologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Nucleosídeo Desaminases/antagonistas & inibidores , Adenina/farmacologia , Animais , Circulação Coronária/efeitos dos fármacos , Coração/efeitos dos fármacos , Técnicas In Vitro , Masculino , Contração Miocárdica/efeitos dos fármacos , Traumatismo por Reperfusão Miocárdica/complicações , Perfusão , Ratos , Ratos Endogâmicos , Descanso , Fibrilação Ventricular/prevenção & controle
17.
Proc Soc Exp Biol Med ; 194(1): 36-42, 1990 May.
Artigo em Inglês | MEDLINE | ID: mdl-2139230

RESUMO

The enzyme adenosine deaminase (ADA) catalyzes the conversion of adenosine and 2'-deoxyadenosine to inosine and 2'-deoxyinosine, respectively. In the absence of ADA activity, 2'-deoxyadenosine is phosphorylated to deoxyadenosine triphosphate. This study concerned the effects of the ADA inhibitor 2'-deoxycoformycin on the murine in vitro immune response to sheep red blood cells (Mishell-Dutton cultures). In the presence of 10(-7) M 2'-deoxycoformycin or 1 mM 2'-deoxyadenosine, there was a significant increase in the plaque-forming cell response when calculated as plaques per 10(6) viable cells recovered. Cultures containing 10(-7) M 2'-deoxycoformycin retained approximately 10% of residual ADA activity of control cultures. Partial ADA deficiency was not preferentially toxic for cells capable of suppressing plaque cell generation. However, there was a decrease of recovered viable cells in all ADA-deficient cultures. There was no change in the percentage of recovered cells which were L3T4+ or Lyt 2+. A significant decrease was observed in a population of cells expressing surface immunoglobulins. The number of plaque-forming cells/10(3) recovered B cells increased significantly. We conclude that partial ADA deficiency results in selective toxicity to a population of non-antigen-specific B cells. Further studies with antigen-specific cells are necessary to determine the possible mechanism(s) by which cellular activation may prevent susceptibility to the toxic effects of ADA deficiency.


Assuntos
Inibidores de Adenosina Desaminase , Formação de Anticorpos/efeitos dos fármacos , Nucleosídeo Desaminases/antagonistas & inibidores , Pentostatina/farmacologia , Animais , Linfócitos B/efeitos dos fármacos , Desoxiadenosinas/farmacologia , Feminino , Camundongos , Linfócitos T Reguladores/efeitos dos fármacos
18.
Biochem Biophys Res Commun ; 167(2): 514-9, 1990 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-2322239

RESUMO

The adenosine deaminase inhibitors deoxycoformycin and erythro-9-(2-hydroxy-3 nonyl) adenine (EHNA) induce single-strand DNA breaks in cultured human lymphocytes. Deoxycoformycin produced a significant number of strand breaks (4-fold increase compared to controls) and EHNA induced strand breaks in a dose-dependent manner. Strand breaks stimulate repair by poly(ADP-ribosylation) which requires NAD+ as a cofactor. Niacin is a precursor of NAD+ and when preincubated with human lymphocytes prior to exposure to adenosine deaminase inhibitors, strand breakage was reduced significantly. The administration of niacin may represent an approach to decreasing the toxicity associated with these agents.


Assuntos
Adenina/análogos & derivados , Inibidores de Adenosina Desaminase , Dano ao DNA , DNA/efeitos dos fármacos , Linfócitos/citologia , Niacina/farmacologia , Nucleosídeo Desaminases/antagonistas & inibidores , Pentostatina/farmacologia , Adenina/farmacologia , Linhagem Celular , Células Cultivadas , Citometria de Fluxo , Humanos , Cinética , Linfócitos/efeitos dos fármacos
19.
Eur J Immunol ; 20(3): 611-5, 1990 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-2156710

RESUMO

An experimental model of adenosine deaminase deficiency was established on the human T cell line Jurkat by using 2'-deoxycoformycin, a strong specific inhibitor of the enzyme. When deoxyadenosine was added to the inhibited cells, the nucleotide profile was modified reproducing that found in lymphocytes from adenosine deaminase-deficient children. The metabolism of phosphoinositides, analyzed by either the release of [3H]inositol phosphates or the breakdown of 32P-prelabeled phosphatidyl inositides, was compared in normal and modified cells where dATP was accumulated. No modification in 32P labeling of phosphoinositides was detectable within the 32P-loading period. However, when the cells were stimulated by phytohemagglutinin or anti-CD3 monoclonal antibody, the phosphoinositide hydrolysis was strongly reduced in the dATP-containing lymphoblasts. This decrease was correlated with the intracellular dATP concentration.


Assuntos
Inibidores de Adenosina Desaminase , Ativação Linfocitária , Nucleosídeo Desaminases/antagonistas & inibidores , Fosfatidilinositóis/metabolismo , Linfócitos T/fisiologia , Adenosina Desaminase/deficiência , Antígenos de Diferenciação de Linfócitos T/imunologia , Complexo CD3 , Linhagem Celular , Nucleotídeos de Desoxiadenina/metabolismo , Humanos , Técnicas In Vitro , Pentostatina/farmacologia , Receptores de Antígenos de Linfócitos T/imunologia
20.
Ital J Biochem ; 39(2): 115-20, 1990.
Artigo em Inglês | MEDLINE | ID: mdl-2113897

RESUMO

Enzymatic synthesis of purine 2'-deoxyriboside was obtained by reacting purine with excess 2-deoxy-alpha-D-ribose-1-phosphate in the presence of commercial bovine nucleoside phosphorylase; the product was isolated by semipreparative reverse phase HPLC with an overall 62% yield. Purine 2'-deoxyriboside was shown to behave as a competitive inhibitor of adenosine deaminase from calf intestinal mucosa and Bacillus cereus, with apparent Ki values of 4.5 and 8.5 microM, respectively.


Assuntos
Inibidores de Adenosina Desaminase , Nucleosídeo Desaminases/antagonistas & inibidores , Nucleosídeos de Purina/biossíntese , Ribonucleosídeos/biossíntese , Animais , Bacillus cereus/enzimologia , Bovinos , Técnicas In Vitro , Mucosa Intestinal/enzimologia , Cinética , Nucleosídeos de Purina/farmacologia , Ribonucleosídeos/farmacologia , Ribosemonofosfatos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA