Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Viral Hepat ; 8(6): 400-5, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11703570

RESUMEN

We have recently shown that the replication of an HCV-poliovirus (PV) chimera that is dependent upon the hepatitis C virus (HCV) 5' untranslated region (UTR) can be inhibited by treatment with ribozymes targeting HCV RNA. To determine the antiviral effects of anti-HCV ribozyme treatment in combination with type 1 interferon (IFN), we analysed the replication of this HCV-PV chimera in HeLa cells treated with anti-HCV ribozyme and/or IFN-alpha2a, IFN-alpha2b, or consensus IFN. The anti-HCV ribozyme, or any of the IFNs alone have significant inhibitory effects on HCV-PV replication compared to control treatment (> or = 85%, P < 0.01). The maximal inhibition due to IFN treatment (94%, P < 0.01) was achieved with > or = 50 U/ml for either IFN-alpha2a or IFN-alpha2b compared to control treatment. A similar level of inhibition in viral replication could be achieved with a 5-fold lower dose of IFN if ribozyme targeting the HCV 5' UTR was given in combination. For consensus IFN, the dose could be reduced by > 12.5-fold if ribozyme targeting the HCV 5' UTR was given in combination. Conversely, the dose of ribozyme could be reduced 3-fold if given in combination with any of the IFN preparations. Moreover, treatment with low doses (1-25 U/mL) of IFN-alpha2a, IFN-alpha2b, or consensus IFN in combination with anti-HCV ribozyme resulted in > 98% inhibition of HCV-PV replication compared to control treatment (P < 0.01). These results demonstrate that IFN and ribozyme each have a beneficial antiviral effect that is augmented when given in combination.


Asunto(s)
Antivirales/farmacología , Hepacivirus/efectos de los fármacos , Interferón-alfa/uso terapéutico , ARN Catalítico/farmacología , ARN Viral/genética , Animales , Chlorocebus aethiops , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Células HeLa , Hepacivirus/genética , Hepatitis C Crónica/tratamiento farmacológico , Humanos , Interferón alfa-2 , Interferón-alfa/administración & dosificación , Poliovirus/genética , ARN Catalítico/síntesis química , Proteínas Recombinantes , Transfección , Células Vero , Replicación Viral/efectos de los fármacos
2.
Prog Drug Res ; Spec No: 111-83, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11548207

RESUMEN

A safe and effective vaccine for hepatitis B virus (HBV) has been available for nearly twenty years and currently campaigns to provide universal vaccination in developing countries are underway. Nevertheless, chronic HBV infection remains a leading cause of chronic hepatitis worldwide and there is a strong need for safe and effective antiviral therapies. Attempts to identify and develop antiviral agents to treat chronic HBV infection remains focused on nucleoside analogs such as 3TC (lamivudine), adefovir dipivoxil, (bis-POMPMEA), and others. However, advances in our understanding of the molecular biology of HBV and the development of new assays for HBV polymerase activity, such as the reconstitution of active HBV polymerase in vitro, should facilitate large screening efforts for non-nucleoside reverse transcriptase inhibitors. Recent advances have furthered our understanding of clinical resistance to lamivudine, have provided new approaches to treatment, and have offered new perspectives on the major challenges to the identification and development of antiviral agents for chronic HBV infection. Here, in an update to our previous review article that appeared in this series [59a], we focus on recent advances that have occurred in the areas of virus structure and replication, in vitro viral polymerase assays, cell culture systems, and animal models.


Asunto(s)
Antivirales/farmacología , Hepatitis B Crónica/tratamiento farmacológico , Animales , Antivirales/uso terapéutico , Modelos Animales de Enfermedad , Resistencia a Medicamentos , Quimioterapia Combinada , Humanos , Replicación Viral
3.
J Biol Chem ; 276(44): 40847-57, 2001 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-11526116

RESUMEN

To examine the role of the mitochondrial polymerase (Pol gamma) in clinically observed toxicity of nucleoside analogs used to treat AIDS, we examined the kinetics of incorporation catalyzed by Pol gamma for each Food and Drug Administration-approved analog plus 1-(2-deoxy-2-fluoro-beta-D-arabinofuranosyl)-5-iodouracil (FIAU), beta-L-(-)-2',3'-dideoxy-3'-thiacytidine (-)3TC, and (R)-9-(2-phosphonylmethoxypropyl)adenine (PMPA). We used recombinant exonuclease-deficient (E200A), reconstituted human Pol gamma holoenzyme in single turnover kinetic studies to measure K(d) (K(m)) and k(pol) (k(cat)) to estimate the specificity constant (k(cat)/K(m)) for each nucleoside analog triphosphate. The specificity constants vary more than 500,000-fold for the series ddC > ddA (ddI) > 2',3'-didehydro-2',3'-dideoxythymidine (d4T) >> (+)3TC >> (-)3TC > PMPA > azidothymidine (AZT) >> Carbovir (CBV). Abacavir (prodrug of CBV) and PMPA are two new drugs that are expected to be least toxic. Notably, the higher toxicities of d4T, ddC, and ddA arose from their 13-36-fold tighter binding relative to the normal dNTP even though their rates of incorporation were comparable with PMPA and AZT. We also examined the rate of exonuclease removal of each analog after incorporation. The rates varied from 0.06 to 0.0004 s(-1) for the series FIAU > (+)3TC approximately equal to (-)3TC > CBV > AZT > PMPA approximately equal to d4T >> ddA (ddI) >> ddC. Removal of ddC was too slow to measure (<0.00002 s(-1)). The high toxicity of dideoxy compounds, ddC and ddI (metabolized to ddA), may be a combination of high rates of incorporation and ineffective exonuclease removal. Conversely, the more effective excision of (-)3TC, CBV, and AZT may contribute to lower toxicity. FIAU is readily extended by the next correct base pair (0.13 s(-1)) faster than it is removed (0.06 s(-1)) and, therefore, is stably incorporated and highly mutagenic. We define a toxicity index for chain terminators to account for relative rates of incorporation versus removal. These results provide a method to rapidly screen new analogs for potential toxicity.


Asunto(s)
ADN Polimerasa Dirigida por ADN/metabolismo , Mitocondrias/efectos de los fármacos , Inhibidores de la Transcriptasa Inversa/toxicidad , Secuencia de Bases , ADN Polimerasa gamma , Cartilla de ADN , Humanos , Cinética , Mitocondrias/enzimología , Profármacos/metabolismo , Profármacos/toxicidad , Proteínas Recombinantes/metabolismo , Inhibidores de la Transcriptasa Inversa/metabolismo
4.
Artículo en Inglés | MEDLINE | ID: mdl-11064287

RESUMEN

The burrowing brittlestar Hemipholis elongata (Say) maintains a constant M(O2)of 3.79+/-1.47 micromol O(2) g(-1) h(-1) (for 0.2-0.3 g animals, mean+/-S.D., n=7), measured in the burrow, over a broad range of PO(2). Below the critical PO(2) of 37 mm Hg, M(O(2)) becomes dependent on the oxygen tension. M(O2) is a function of the size of H. elongata; the scaling exponent is 0. 83 and is similar to those reported for other echinoderms. The M(O2) of H. elongata is unaffected by removal from the burrow, by hypercapnia, by exposure to hydrogen sulfide, or by temperature change in the range from 20 to 32 degrees C. The relative insensitivity of H. elongata to these factors may be an adaptation to life in the highly variable estuarine and tidal creek environments where the animals are frequently found.


Asunto(s)
Equinodermos/fisiología , Oxígeno/metabolismo , Respiración , Animales , Temperatura
5.
Antivir Chem Chemother ; 10(4): 155-85, 1999 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10480736

RESUMEN

Influenza A and B viruses belong to the Orthomyxoviridae family of viruses. These viruses are responsible for severe morbidity and significant excess mortality each year. Infection with influenza viruses usually leads to respiratory involvement and can result in pneumonia and secondary bacterial infections. Vaccine approaches to the prophylaxis of influenza virus infections have been problematic owing to the ability of these viruses to undergo antigenic shift by exchanging genomic segments or by undergoing antigenic drift, consisting of point mutations in the haemagglutinin (HA) and neuraminidase (NA) genes as a result of an error-prone viral polymerase. Historically, antiviral approaches for the therapy of both influenza A and B viruses have been largely unsuccessful until the elucidation of the X-ray crystallographic structure of the viral NA, which has permitted structure-based drug design of inhibitors of this enzyme. In addition, recent advances in the elucidation of the structure and complex function of influenza HA have resulted in the discovery of a number of diverse compounds that target this viral protein. This review article will focus largely on newer antiviral agents including those that inhibit the influenza virus NA and HA. Other novel approaches that have entered clinical trials or been considered for their clinical utility will be mentioned.


Asunto(s)
Gripe Humana/tratamiento farmacológico , Animales , Antivirales/farmacología , Antivirales/uso terapéutico , Humanos , Orthomyxoviridae/efectos de los fármacos , Orthomyxoviridae/fisiología , Replicación Viral/efectos de los fármacos
6.
Virology ; 248(2): 264-74, 1998 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-9721235

RESUMEN

Entry of influenza virus into the host cell is dependent on the fusion of the viral envelope with the endosomal membrane and is mediated by a low-pH-induced change of the viral hemagglutinin (HA) to a conformation that is fusogenic. A compound related to podocarpic acid (180299) was identified that inhibits multicycle replication of influenza A/Kawasaki/86 (H1N1) virus in culture. Treatment of Madin-Darby canine kidney (MDCK) cells with 180299 at 1 h before infection resulted in the inhibition of viral protein synthesis. Addition of 20 microgram of 180299/ml at 1 h p.i. had no effect, indicating that 180299 affects an early step of the influenza viral replication cycle. Genetic analysis of reassortants between sensitive and resistant viruses demonstrated that hemagglutinin (HA) conferred the 180299-resistant (180299(r)) phenotype. Twelve independent isolates of influenza A/Kawasaki/86 were selected for resistance to 180299, and sequence analysis revealed that each of these viruses contained amino acid substitutions in the HA. These mutations are dispersed throughout the HA primary amino acid sequence and cluster in one of two regions: the interface between HA1 and HA2 and in a region near the fusion domain of HA2. When compared with the parent virus, the pH-of-inactivation of the resistant mutants was increased by 0.3 to 0.6 pH unit, suggesting that the mutant HAs undergo the conformational change at an elevated pH. Fusion of human erythrocytes to MDCK cells infected with parent influenza A/Kawasaki/86 was inhibited by 180299 (0.1-10 microgram/ml) in a concentration-dependent manner, whereas fusion of erythrocytes to MDCK cells infected with 180299(r) mutants was not affected. These results suggest that 180299 interacts with the neutral pH conformation of influenza A HA and prevents the low-pH-induced change of HA to its fusogenic conformation.


Asunto(s)
Abietanos , Antivirales/farmacología , Hemaglutininas Virales/fisiología , Fusión de Membrana/efectos de los fármacos , Orthomyxoviridae/efectos de los fármacos , Fenantrenos/farmacología , Secuencia de Aminoácidos , Animales , Células Cultivadas , Perros , Farmacorresistencia Microbiana/genética , Eritrocitos/fisiología , Hemaglutininas Virales/química , Hemaglutininas Virales/genética , Humanos , Concentración de Iones de Hidrógeno , Mutación , Orthomyxoviridae/genética , Orthomyxoviridae/fisiología , Fenantrenos/química , Conformación Proteica , Ensamble de Virus , Replicación Viral
7.
Hepatology ; 28(1): 179-91, 1998 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9657111

RESUMEN

Woodchucks were used to study the antiviral activity and toxicity of fialuridine (FIAU; 1,-2'deoxy-2'fluoro-1-beta-D-arabinofuranosyl-5-iodo-uracil). In an initial experiment, groups of six chronic woodchuck hepatitis virus (WHV) carrier woodchucks received daily doses of FIAU by intraperitoneal injection for 4 weeks. At 0.3 mg/kg/d, the antiviral effect was equivocal, but at 1.5 mg/kg/d, FIAU had significant antiviral activity. No evidence of drug toxicity was observed during the 4-week period of treatment or during posttreatment follow-up. In a second experiment, groups of nine WHV carriers or uninfected woodchucks were given 1.5 mg/kg/d of FIAU orally for 12 weeks, and the results compared with placebo-treated controls. After 4 weeks, the serum WHV-DNA concentration in the FIAU-treated carrier group was two to three logs lower than that in the placebo-treated group. After 12 weeks of FIAU treatment, serum WHV DNA was not detectable by conventional dot-blot analysis, hepatic WHV-DNA replicative intermediates (RI) had decreased 100-fold, and hepatic expression of WHV core antigen was remarkably decreased. No evidence of toxicity was observed after 4 weeks, but, after 6 to 7 weeks, food intake decreased and, after 8 weeks, the mean body weights of woodchucks treated with FIAU were significantly lower than controls. Anorexia, weight loss, muscle wasting, and lethargy became progressively severe, and all FIAU-treated woodchucks died or were euthanized 78 to 111 days after treatment began. Hepatic insufficiency (hyperbilirubinemia, decreased serum fibrinogen, elevated prothrombin time), lactic acidosis, and hepatic steatosis were characteristic findings in the final stages of FIAU toxicity in woodchucks. The syndrome of delayed toxicity in woodchucks was similar to that observed previously in humans treated with FIAU, suggesting that the woodchuck should be valuable in future investigations of the molecular mechanisms of FIAU toxicity in vivo and for preclinical toxicological evaluation of other nucleoside analogs before use in patients.


Asunto(s)
Antivirales/uso terapéutico , Arabinofuranosil Uracilo/análogos & derivados , Hepatitis B/tratamiento farmacológico , Animales , Anorexia/inducido químicamente , Antivirales/efectos adversos , Arabinofuranosil Uracilo/efectos adversos , Arabinofuranosil Uracilo/farmacocinética , Arabinofuranosil Uracilo/uso terapéutico , Portador Sano/virología , ADN Viral/análisis , Hepatitis B/sangre , Hepatitis B/patología , Antígenos del Núcleo de la Hepatitis B/análisis , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/fisiología , Hígado/metabolismo , Hígado/patología , Marmota , Músculos/efectos de los fármacos , Fases del Sueño , Factores de Tiempo , Replicación Viral/efectos de los fármacos
8.
Prog Drug Res ; 50: 259-322, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9670781

RESUMEN

Hepatitis B virus (HBV) is the leading cause of chronic hepatitis throughout the world. Notwithstanding the availability of a safe and effective vaccine, the world prevalence of HBV has not declined significantly, thus resulting in the need for a selective antiviral agent. HBV is a small, partially double-stranded DNA virus which replicates through an RNA intermediate. Most efforts to develop anti-HBV agents have been targeted to the viral DNA polymerase which possesses reverse transcriptase activity. Currently, the most promising anti-HBV agents are nucleoside analogs which interfere with viral DNA replication. Although earlier nucleoside analogs such as vidarabine (ara-A) and fialuridine (FIAU) have displayed unacceptable toxicities, newer analogs such as lamivudine (3TC), bis-POM PMEA (GS-840), lobucavir, and BMS-200,475 have demonstrated clinical utility. In particular, the use of lamivudine has generated considerable interest in the development of other L-enantiomeric nucleoside analogs for use against HBV. Here, we provide an overview of HBV structure and replication strategy and discuss the use of cell culture systems, in vitro viral polymerase systems, and animal models to identify and evaluate anti-HBV agents. We also discuss the various classes of nucleoside analogs in terms of structure, mechanism of action, status in clinical development, ability to select for resistant HBV variants, and use in combination therapies. Finally, we present a discussion of novel antiviral approaches, including antisense and gene therapy, and address the various challenges to successful anti-HBV chemotherapeutic intervention.


Asunto(s)
Antivirales/uso terapéutico , Hepatitis B Crónica/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Quimioterapia Combinada , Virus de la Hepatitis B/ultraestructura , Humanos , Replicación Viral/efectos de los fármacos
9.
Bioorg Med Chem ; 6(5): 577-85, 1998 May.
Artículo en Inglés | MEDLINE | ID: mdl-9629471

RESUMEN

The synthesis and antiviral evaluation of 21 prodrugs of 1-[2',3'-dideoxy-3'-C-(hydroxymethyl)-beta-D-erythropentofuranosyl ] cytosine 1 is reported. Cytosine N4-imine analogues were prepared by condensation of 1 with selected formamide dimethyl acetals. Amino acid substituted prodrugs were prepared from 1 or imine prodrug 2 by coupling with either N-tert-butoxycarbonyl (t-Boc)-L-valine or N-t-Boc-L- phenylalanine in the presence of dicyclohexycarbodiimide (DCC) and 4-dimethylaminopyridine (4-DMAP). Deprotection of the t-Boc protecting group was achieved with trifluoroacetic acid (TFAA) in methylene chloride. Cytosine N4-amide analogues were prepared by reaction of 1 with appropriate anhydrides in aqueous dioxane. Triacylated analogue 22 was prepared by reaction of 1 with four equivalents of benzoyl chloride in pyridine. Prodrugs were evaluated for activity against duck hepatitis B virus, herpes simplex virus types 1 and 2, human cytomegalovirus, and human immunodeficiency virus. A number of analogues were found comparable in activity to 1 with the cytosine N4-imine series more active than the amino acid substituted and cytosine N4-amide prodrugs. Slight to moderate cellular toxicity was observed with some analogues.


Asunto(s)
Antivirales/síntesis química , Didesoxinucleósidos/síntesis química , Profármacos/síntesis química , Nucleósidos de Pirimidina/síntesis química , Antivirales/farmacología , Citomegalovirus/efectos de los fármacos , Didesoxinucleósidos/farmacología , VIH/efectos de los fármacos , Virus de la Hepatitis B del Pato/efectos de los fármacos , Profármacos/farmacología , Nucleósidos de Pirimidina/farmacología , Análisis Espectral , Células Tumorales Cultivadas
10.
J Biol Chem ; 273(27): 16998-7011, 1998 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-9642264

RESUMEN

Hemoglobin (Hb) occurs in circulating red blood cells, neural tissue, and body wall muscle tissue of the nemertean worm, Cerebratulus lacteus. The neural and body wall tissue each express single major Hb components for which the amino acid sequences have been deduced from cDNA and genomic DNA. These 109-residue globins form the smallest stable Hbs known. The globin genes have three exons and two introns with splice sites in the highly conserved positions of most globin genes. Alignment of the sequences with those of other globins indicates that the A, B, and H helices are about one-half the typical length. Phylogenetic analysis indicates that shortening results in a small tendency of globins to group together regardless of their actual relationships. The neural and body wall Hbs in situ are half-saturated with O2 at 2.9 and 4.1 torr, respectively. The Hill coefficient for the neural Hb in situ, approximately 2.9, suggests that the neural Hb self-associates in the deoxy state at least to tetramers at the 2-3 mM (heme) concentration estimated in the cells. The Hb must dissociate upon oxygenation and dilution because the weight-average molecular mass of the HbO2 in vitro is only about 18 kDa at 2-3 microM heme concentration. Calculations suggest that the Hb can function as an O2 store capable of extending neuronal activity in an anoxic environment for 5-30 min.


Asunto(s)
Hemoglobinas/metabolismo , Invertebrados/metabolismo , Músculos/metabolismo , Sistema Nervioso/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , ADN , Hemoglobinas/química , Hemoglobinas/genética , Humanos , Invertebrados/genética , Datos de Secuencia Molecular , Oxígeno/metabolismo , Filogenia , Unión Proteica , Conformación Proteica , Pliegue de Proteína , Homología de Secuencia de Aminoácido
11.
Virology ; 236(1): 66-75, 1997 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-9299618

RESUMEN

A single change (E119G) in the influenza A virus N9 neuraminidase (NA) results in resistance of the enzyme to the NA inhibitor 4-Guanidino-Neu5Ac2en (4-GuDANA). This change causes a salt link between Glu119, which sits in a pocket in the bottom of the active site of the enzyme, and the 4-guanidinium moiety of the inhibitor to be lost. NA "heads" of the resistant enzyme produced only a few small crystals under conditions in which the wild-type enzyme readily formed large crystals. These small crystals were of sufficient quality to yield X-ray crystallographic data which confirmed the E119G change and demonstrated the presence of electron density representing either a strong structural-water molecule or an anionic species in place of the glutamate carboxylate. NA heads of the resistant enzyme also have greatly reduced NA activity per milligram of total protein. We have now found that the mutant NA heads consist predominantly of monomers with a few dimers and tetramers, as determined by electron microscopic analysis of the protein. The low level of enzymatic activity as well as the small number of crystals obtained were probably from the few tetramers remaining intact in the preparation. The purified wild-type and 4-GuDANA-resistant enzymes were treated with the homobifunctional NHS-ester cross linker, DTSSP. SDS-PAGE analysis of the treated enzymes clearly revealed cross-linked dimers of the wild-type enzyme. In contrast, only a small proportion of the 4-GuDANA-resistant neuraminidase was cross-linked. An examination of the known X-ray crystallographic structure of the wild-type NA reveals a salt bridge between Glu119 and Arg156 of the same monomer. Arg156 is a conserved amino acid that is situated at the interface between monomers, and a salt link between this amino acid and Glu119 may contribute to the stability of enzyme tetramers. It is suggested that the E119G alteration in the 4-GuDANA-resistant NA leads to the abrogation of this interaction and thus to the instability of the NA tetramers.


Asunto(s)
Virus de la Influenza A/enzimología , Neuraminidasa/química , Mutación Puntual , Estructura Secundaria de Proteína , Animales , Embrión de Pollo , Simulación por Computador , Reactivos de Enlaces Cruzados , Cristalización , Cristalografía por Rayos X , Dimerización , Inhibidores Enzimáticos/farmacología , Guanidinas , Humanos , Cinética , Sustancias Macromoleculares , Microscopía Electrónica , Modelos Moleculares , Neuraminidasa/metabolismo , Neuraminidasa/ultraestructura , Piranos , Ácidos Siálicos/farmacología , Especificidad por Sustrato , Zanamivir
12.
J Infect Dis ; 176 Suppl 1: S66-8, 1997 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9240698

RESUMEN

The reassortant influenza viruses, A/NWS-G70c with N9 neuraminidase (NA) and B/HK/8/73 (HG) with B/Lee/40 NA, were selected for resistance to 4-guanidino-Neu5Ac2en (4-GuDANA) by passaging the virus in stepwise increases in the concentration of 4-GuDANA. In the NA of resistant viruses, the absolutely conserved Glu 119, which lies in a pocket beneath the active site of the enzyme and interacts with the guanidinium moiety of 4-GuDANA, was changed to Gly. The mutant NA was >200-fold more resistant to 4-GuDANA than was the wild-type enzyme. During 72 h in cell culture, resistant A and B viruses displayed much less NA activity than did wild-type viruses but did undergo multicycle replication. While emergence of resistance to 4-GuDANA has not been observed in vivo, these results demonstrate that the development of resistance is possible and can be mediated by a single amino acid change in the active site of the viral NA.


Asunto(s)
Antivirales/farmacología , Inhibidores Enzimáticos/farmacología , Virus de la Influenza B/efectos de los fármacos , Neuraminidasa/antagonistas & inhibidores , Ácidos Siálicos/farmacología , Células Cultivadas , Resistencia a Medicamentos , Guanidinas , Virus de la Influenza A/efectos de los fármacos , Mutación , Neuraminidasa/genética , Piranos , Zanamivir
13.
Antiviral Res ; 34(1): 71-4, 1997 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9107387

RESUMEN

Fialuridine (FIAU) is a thymidine analog effective against hepatitis B virus. Toxicity associated with FIAU treatment included clinical signs consistent with mitochondrial dysfunction, including severe lactic acidosis. To understand further the mechanism of FIAU toxicity, we examined the effect of FIAU on DNA synthesis in mitochondria. Mitochondria isolated from livers of naive rats were treated in vitro with concentrations of FIAU or FIAU triphosphate (FIAU-TP) ranging from 0.1 to 200 microM. A 14 or 32% decrease in mitochondrial DNA synthesis compared to controls was observed when isolated mitochondria were treated with 25 microM FIAU or FIAU-TP, respectively. Since it is thought that nucleosides must be phosphorylated to inhibit DNA polymerase, studies were conducted to determine whether isolated rat mitochondria could phosphorylate FIAU. Results using lanthanum chloride precipitation and HPLC analysis showed that enzymes present in a mitochondrial lysate were capable of phosphorylating FIAU to form FIAU monophosphate.


Asunto(s)
Antivirales/metabolismo , Antivirales/farmacología , Arabinofuranosil Uracilo/análogos & derivados , Replicación del ADN/efectos de los fármacos , Mitocondrias Hepáticas/efectos de los fármacos , Animales , Arabinofuranosil Uracilo/metabolismo , Arabinofuranosil Uracilo/farmacología , Mitocondrias Hepáticas/metabolismo , Fosforilación , Ratas
14.
Nucleic Acids Res ; 24(21): 4111-6, 1996 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-8932359

RESUMEN

The thymidine analog, 1-(2-deoxy-2-fluoro-beta-D-arabino-furanosyl)-5-iodouracil (FIAU), is incorporated into DNA in cell culture and in vivo. To investigate the effect of incorporation of FIAU into DNA on the binding of transcription factors, oligonucleotide duplexes which bind specifically to activator protein 1 (AP-1) or to TFIID were synthesized and binding of these oligonucleotides to their respective proteins was studied using gel-shift analysis. When thymidine at position -3, -1, 1 or 7 (relative to the first thymidine of the core binding sequence) was replaced with FIAU, binding to AP-1 was approximately 82, 28, 86 and 51%, respectively, of the binding to the non-substituted oligonucleotide to AP-1. When thymidine at position 3 or 5 (each adjacent to the center of dyad symmetry) was replaced with FIAU, binding to AP-1 was abrogated. Oligonucleotides containing FIAU at positions -1, 3 or 5, were much less able to compete with radiolabeled wild-type oligonucleotides for binding to AP-1. In contrast, the presence of FIAU, depending on its location, resulted in the increased binding of TFIID to its consensus target DNA sequence. These results indicate that incorporation of FIAU into DNA may induce local conformational changes resulting in the altered ability of transcriptional factors to bind to their cognate DNA sequences. Additional studies demonstrated that the presence of FIAU at a position 5' to the cleavage site in the consensus sequence T*TAA (where * is the cleavage site) inhibited restriction of the oligomeric duplex by MseI.


Asunto(s)
Antivirales/farmacología , Arabinofuranosil Uracilo/análogos & derivados , ADN/efectos de los fármacos , Factor de Transcripción AP-1/metabolismo , Factores de Transcripción/metabolismo , Arabinofuranosil Uracilo/farmacología , Unión Competitiva , ADN/metabolismo , Desoxirribonucleasa BamHI/metabolismo , Desoxirribonucleasas de Localización Especificada Tipo II/metabolismo , Electroforesis en Gel de Poliacrilamida , Células HeLa , Humanos , Unión Proteica/efectos de los fármacos , Factor de Transcripción TFIID
15.
Proc Natl Acad Sci U S A ; 93(8): 3592-7, 1996 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-8622980

RESUMEN

The thymidine analog fialuridine deoxy-2-fluoro-beta-D-arabinofuranosyl)-5-iodouracil (FIAU) was toxic in trials for chronic hepatitis B infection. One mechanism postulated that defective mtDNA replication was mediated through inhibition of DNA polymerase-gamma (DNA pol-gamma), by FIAU triphosphate (FIALTP) or by triphosphates of FIAU metabolites. Inhibition kinetics and primer-extension analyses determined biochemical mechanisms of FIAU, 1-(2-deoxy-2-fluoro-beta-D-arabinofuranosyl) -5-methyluracil (FAU), 1-(2-deoxy-2-fluoro-beta-D-arabinofuranosyl)uracil triphosphate (TP) inhibition of DNA pol-gamma. dTMP incorporation by DNA pol-gamma was inhibited competitively by FIAUTP, FMAUTP, and FAUTP (K1=0.015, 0.03, and 1.0 microM, respectively). By using oliginucleotide template-primers. DNA pol-gamma incorporated each analog into DNA opposite a single adenosine efficiently without effects on DNA chain elongation. Incorporation of multiple adjacent analogs at positions of consecutive adenosines dramatically impaired chain elongation by DNA pol-gamma. Effects of FIAU, FMAU, and FAU on HepG2 cell mmtDNA abundance and ultrastructure were determined. After 14 days, mtDNA decreased by 30% with 20 microM FIAU or 20 microM FMAU and decreased less than 10% with 100 microM FAU. FIAU and FMAU disrupted mitochondria and caused accumulation of intracytoplasmic lipid droplets. Biochemical and cell biological findings suggest that FIAU and its metabolites inhibit mtDNA replication, most likely at positions of adenosine tracts, leading to decreased mtDNA and mitochondrial ultrastructural defects.


Asunto(s)
Antivirales/metabolismo , Antivirales/toxicidad , Arabinofuranosil Uracilo/análogos & derivados , ADN Polimerasa III/antagonistas & inhibidores , ADN Mitocondrial/metabolismo , Mitocondrias Hepáticas/efectos de los fármacos , Arabinofuranosil Uracilo/metabolismo , Arabinofuranosil Uracilo/toxicidad , Secuencia de Bases , Sitios de Unión , Línea Celular , Cartilla de ADN/genética , Humanos , Cinética , Microscopía Electrónica , Mitocondrias Hepáticas/ultraestructura , Datos de Secuencia Molecular
16.
Antiviral Res ; 29(2-3): 125-39, 1996 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-8739593

RESUMEN

Fialuridine (FIAU) is a thymidine nucleoside analog with activity against various herpesviruses and hepatitis B virus (HBV) in vitro and in vivo. In a clinical evaluation for its use as a treatment for chronic HBV infection, long term (HBV) in vitro and in vivo. In a clinical evaluation for its term oral administration of FIAU resulted in severe multi-organ toxicity characterized by a delayed onset and refractory lactic acidosis. These clinical manifestations led to the hypothesis that the toxicity of FIAU was mediated through mitochondrial dysfunction, possibly as a result of the inhibition of mitochondrial DNA polymerase gamma and/or incorporation of FIAU into mitochondrial DNA. In addition to describing the anti-HBV activity of FIAU, this review discusses results from in vitro experiments carried out by various laboratories in an effort to evaluate and understand more fully the mitochondrial toxicity of FIAU.


Asunto(s)
Antivirales/farmacología , Arabinofuranosil Uracilo/análogos & derivados , Virus de la Hepatitis B/efectos de los fármacos , Animales , Arabinofuranosil Uracilo/farmacología , ADN Mitocondrial/efectos de los fármacos , Complejo IV de Transporte de Electrones/metabolismo , Hepatitis B/tratamiento farmacológico , Humanos , Hígado/citología , Hígado/efectos de los fármacos
17.
Toxicol In Vitro ; 10(3): 297-303, 1996 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20650209

RESUMEN

Fialuridine (FIAU), a thymidine nucleoside analogue with anti-hepatitis B virus activity, showed clinical toxicity consistent with mitochondrial dysfunction. In vitro methods were used to understand further this toxicity. Using a sensitive and specific radioimmunoassay, FIAU was found to be present in nuclear DNA of human hepatoblastoma cells incubated for 6 days in 10 or 50 n M drug, at a level of 1 residue per 63 or 39 thymidines, respectively, and was present in mitochondrial DNA at a level of 1 residue per 2139 or 1696 thymidines, respectively. Human hepatoblastoma cells were incubated for 6 days in increasing concentrations of FIAU or, for comparative purposes, the nucleoside analogue dideoxycytidine (ddC), after which time the cells were examined by electron microscopy. At 10 mum and higher concentrations, both compounds induced morphological changes in the ultrastructure of mitochondria characterized by marked mitochondrial swelling, loss of internal cristae and dissolution of the internal matrix. These results, considered along with previously published studies, indicate that FIAU has deleterious effects in vitro on mitochondrial function and structure that occur relatively quickly but without an apparent decrease in the abundance of mitochondrial DNA.

18.
Virology ; 214(2): 642-6, 1995 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-8553569

RESUMEN

We report the selection and characterization of influenza A/NWS-G70c and B/HK/8/73 (HG) viruses which are resistant to the potent influenza neuraminidase inhibitor, 4-guanidino-Neu5Ac2en. Viruses were selected which replicated in MDCK cells in the presence of 20 micrograms/ml inhibitor. The neuraminidase of resistant viruses was > 200-fold more resistant to 4-guanidino-Neu5Ac2en than was the neuraminidase of the parent viruses. Although amounts of neuraminidase protein were similar in resistant and parent viruses, the enzyme activity of the resistant neuraminidase heads was reduced by > 95% for the substrates used. Relative to parent viruses, the resistant viruses replicated to equal or greater titers in tissue culture and in embryonated chicken eggs. Sequence analysis revealed a single nucleotide mutation in the neuraminidase gene of each virus resulting in the change of the conserved Glu 119 (which lies in a pocket beneath the active site of the enzyme) to Gly thus eliminating an electrostatic interaction with the C-4 guanidinium moiety of the inhibitor. Mutations (Asn-->Ser) at amino acids 145 and 150 were also found in the hemagglutinin gene of the B/HK/8/73 (HG) virus resistant to 4-guanidino-Neu5Ac2en. No changes were found in the hemagglutinin gene of the resistant A/NWS-G70c virus.


Asunto(s)
Antivirales/farmacología , Inhibidores Enzimáticos/farmacología , Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza B/efectos de los fármacos , Neuraminidasa/antagonistas & inhibidores , Ácidos Siálicos/farmacología , Secuencia de Aminoácidos , Animales , Línea Celular , Embrión de Pollo , Farmacorresistencia Microbiana , Guanidinas , Glicoproteínas Hemaglutininas del Virus de la Influenza , Hemaglutininas Virales/genética , Humanos , Virus de la Influenza A/enzimología , Virus de la Influenza B/enzimología , Datos de Secuencia Molecular , Mutación , Neuraminidasa/genética , Piranos , Homología de Secuencia de Aminoácido , Zanamivir
19.
Antimicrob Agents Chemother ; 39(6): 1329-35, 1995 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-7574525

RESUMEN

To identify the minimal structural elements necessary for biological activity, the rigid tricyclic nucleus of the known human immunodeficiency virus type 1 (HIV-1) reverse transcriptase (RT) inhibitor tetrahydroimidazobenzodiazepinthione was subjected to systematic bond disconnection to obtain simpler structures. A rational selection and testing of modeled analogs containing these potential pharmacophoric moieties led to the discovery of a new series of nonnucleoside inhibitors of RT. The lead compound of this new PETT series of nonnucleoside RT inhibitors, N-(2-phenylethyl)-N'-(2-thiazolyl)thiourea (LY73497), was found to inhibit HIV-1 but not HIV-2 or simian immunodeficiency virus in cell culture at micromolar concentrations. This derivative was also found to inhibit HIV-1 RT. Through an integrated effort involving synthesis and molecular modeling, compounds with nanomolar potency against HIV-1 in cell culture were developed. In these studies, LY300046-HCl was identified as a potent nonnucleoside inhibitor of HIV-1 RT possessing favorable pharmacokinetic properties.


Asunto(s)
VIH-1/efectos de los fármacos , Sustancias Intercalantes/farmacología , Inhibidores de la Transcriptasa Inversa , Tiazoles/farmacología , Triazoles/farmacología , Animales , Antivirales/farmacología , Secuencia de Bases , Benzodiazepinas/química , Encéfalo/metabolismo , Bovinos , Células Cultivadas , ADN Polimerasa Dirigida por ADN/efectos de los fármacos , ADN Polimerasa Dirigida por ADN/metabolismo , Farmacorresistencia Microbiana , Humanos , Imidazoles/química , Masculino , Datos de Secuencia Molecular , Piridinas/síntesis química , Piridinas/farmacocinética , Piridinas/farmacología , Ratas , Ratas Endogámicas F344 , Relación Estructura-Actividad , Tiazoles/síntesis química , Tiourea/análogos & derivados , Tiourea/síntesis química , Tiourea/farmacología
20.
Biochemistry ; 33(48): 14620-4, 1994 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-7981224

RESUMEN

Fialuridine [FIAU, 1-(2'-deoxy-2'-fluoro-beta-D-arabinofuranosyl)-5- iodouridine] was used in clinical trials for chronic hepatitis B virus infection and was extremely toxic. Evidence suggested targets of FIAU toxicity included mitochondria, but toxic mechanisms were unclear. Since FIAU is a thymidine analog, we reasoned that triphosphorylated FIAU (FIAUTP) could be incorporated into mitochondrial DNA by DNA pol-gamma and into genomic DNA by DNA polymerases alpha, beta, delta, and epsilon. All five purified mammalian DNA polymerases incorporated FIAUMP into the nascent DNA chain during in vitro DNA synthesis. When FIAUTP was substituted for dTTP, oligonucleotide products were generated efficiently by DNA pol-gamma and were similar to those generated in the presence of the four normal dNTPs. In contrast, oligonucleotide products generated by the four nuclear DNA polymerases in the presence of FIAUTP were significantly reduced in length relative to those generated in the presence of dTTP. In parallel kinetic assays, FIAUTP competitively inhibited the accumulation of radiolabeled dTTP into DNA by DNA pol-gamma. The Ki with DNA pol-gamma was 0.04 microM, the lowest Ki among the mammalian DNA polymerases. Competition between FIAUTP and dTTP and the relative ease of accumulation of FIAUMP in mitochondrial DNA by DNA pol-gamma in vitro together may relate to clinical FIAU toxicity.


Asunto(s)
Arabinofuranosil Uracilo/análogos & derivados , ADN Polimerasa Dirigida por ADN/metabolismo , ADN/biosíntesis , Nucleótidos de Desoxiuracil/metabolismo , Inhibidores de la Síntesis del Ácido Nucleico , Animales , Arabinofuranosil Uracilo/metabolismo , Arabinofuranosil Uracilo/farmacología , Unión Competitiva , Cinética , Mamíferos , Mitocondrias/metabolismo , Especificidad por Sustrato , Moldes Genéticos , Nucleótidos de Timina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA