Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 265
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 18(7): e1010688, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35793357

RESUMEN

Herpes simplex virus type 1 (HSV-1) is a common virus of mankind and HSV-1 infections are a significant cause of blindness. The current antiviral treatment of herpes infection relies on acyclovir and related compounds. However, acyclovir resistance emerges especially in the long term prophylactic treatment that is required for prevention of recurrent herpes keratitis. Earlier we have established antiviral siRNA swarms, targeting sequences of essential genes of HSV, as effective means of silencing the replication of HSV in vitro or in vivo. In this study, we show the antiviral efficacy of 2´-fluoro modified antiviral siRNA swarms against HSV-1 in human corneal epithelial cells (HCE). We studied HCE for innate immunity responses to HSV-1, to immunostimulatory cytotoxic double stranded RNA, and to the antiviral siRNA swarms, with or without a viral challenge. The panel of studied innate responses included interferon beta, lambda 1, interferon stimulated gene 54, human myxovirus resistance protein A, human myxovirus resistance protein B, toll-like receptor 3 and interferon kappa. Our results demonstrated that HCE cells are a suitable model to study antiviral RNAi efficacy and safety in vitro. In HCE cells, the antiviral siRNA swarms targeting the HSV UL29 gene and harboring 2´-fluoro modifications, were well tolerated, induced only modest innate immunity responses, and were highly antiviral with more than 99% inhibition of viral release. The antiviral effect of the 2'-fluoro modified swarm was more apparent than that of the unmodified antiviral siRNA swarm. Our results encourage further research in vitro and in vivo on antiviral siRNA swarm therapy of corneal HSV infection, especially with modified siRNA swarms.


Asunto(s)
Herpes Simple , Herpesvirus Humano 1 , Aciclovir/metabolismo , Aciclovir/farmacología , Antivirales/metabolismo , Antivirales/farmacología , Células Epiteliales/metabolismo , Herpes Simple/genética , Herpes Simple/terapia , Herpesvirus Humano 1/fisiología , Humanos , ARN Bicatenario/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Replicación Viral/genética
2.
Acta Pharmacol Sin ; 41(1): 129-137, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31341258

RESUMEN

Rheumatoid arthritis patients can be prescribed a combination of immunosuppressive drug leflunomide (LEF) and the antiviral drug acyclovir to reduce the high risk of infection. Acyclovir is a substrate of organic anion transporter (OAT) 1/3 and multidrug resistance-associated protein (MRP) 2. Considering the extraordinarily long half-life of LEF's active metabolite teriflunomide (TER) and the kidney injury risk of acyclovir, it is necessary to elucidate the potential impact of LEF on the disposition of acyclovir. Here we used a specific MRP inhibitor MK571 and probenecid (OAT1/3 and MRP2 inhibitor) to assess the effects of MRP2 and OAT1/3 on the pharmacokinetics and tissue distribution of acyclovir in rats. We showed that LEF and probenecid, but not MK571 significantly increased the plasma concentration of acyclovir. However, kidney and liver exposures of acyclovir were increased when coadministered with LEF, probenecid or MK571. The kidney/plasma ratio of acyclovir was increased to approximately 2-fold by LEF or probenecid, whereas it was increased to as much as 14.5-fold by MK571. Consistently, these drugs markedly decreased the urinary excretion of acyclovir. TER (0.5-100 µmol/L) dose-dependently increased the accumulation of acyclovir in MRP2-MDCK cells with an IC50 value of 4.91 µmol/L. TER (5 µmol/L) significantly inhibited the uptake of acyclovir in hOAT1/3-HEK293 cells. These results suggest that LEF/TER increased the kidney accumulation of acyclovir by inhibiting the efflux transporter MRP2, which increased its kidney/plasma ratio and renal injury risk. However, the inhibitory effects of LEF/TER on OAT1/3 reduced the tubular cells' uptake of acyclovir and increased the plasma concentration.


Asunto(s)
Aciclovir/farmacocinética , Riñón/metabolismo , Leflunamida/farmacología , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/antagonistas & inhibidores , Proteína 1 de Transporte de Anión Orgánico/antagonistas & inhibidores , Transportadores de Anión Orgánico Sodio-Independiente/antagonistas & inhibidores , Aciclovir/administración & dosificación , Aciclovir/metabolismo , Administración Intravenosa , Animales , Células Cultivadas , Crotonatos/administración & dosificación , Crotonatos/metabolismo , Crotonatos/farmacología , Perros , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Hidroxibutiratos , Leflunamida/administración & dosificación , Leflunamida/metabolismo , Células de Riñón Canino Madin Darby/efectos de los fármacos , Células de Riñón Canino Madin Darby/metabolismo , Masculino , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Nitrilos , Proteína 1 de Transporte de Anión Orgánico/metabolismo , Transportadores de Anión Orgánico Sodio-Independiente/metabolismo , Probenecid/administración & dosificación , Probenecid/metabolismo , Probenecid/farmacología , Propionatos/administración & dosificación , Propionatos/metabolismo , Propionatos/farmacología , Quinolinas/administración & dosificación , Quinolinas/metabolismo , Quinolinas/farmacología , Ratas , Ratas Sprague-Dawley , Distribución Tisular , Toluidinas/administración & dosificación , Toluidinas/metabolismo , Toluidinas/farmacología
3.
Drug Metab Dispos ; 45(2): 137-144, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27895114

RESUMEN

Despite peptide transporter 1 (PEPT1) being responsible for the bioavailability for a variety of drugs, there has been little study of its potential involvement in drug-drug interactions. Pomaglumetad methionil, a metabotropic glutamate 2/3 receptor agonist prodrug, utilizes PEPT1 to enhance absorption and bioavailability. In vitro studies were conducted to guide the decision to conduct a clinical drug interaction study and to inform the clinical study design. In vitro investigations determined the prodrug (LY2140023 monohydrate) is a substrate of PEPT1 with Km value of approximately 30 µM, whereas the active moiety (LY404039) is not a PEPT1 substrate. In addition, among the eight known PEPT1 substrates evaluated in vitro, valacyclovir was the most potent inhibitor (IC50 = 0.46 mM) of PEPT1-mediated uptake of the prodrug. Therefore, a clinical drug interaction study was conducted to evaluate the potential interaction between the prodrug and valacyclovir in healthy subjects. No effect of coadministration was observed on the pharmacokinetics of the prodrug, valacyclovir, or either of their active moieties. Although in vitro studies showed potential for the prodrug and valacyclovir interaction via PEPT1, an in vivo study showed no interaction between these two drugs. PEPT1 does not appear to easily saturate because of its high capacity and expression in the intestine. Thus, a clinical interaction at PEPT1 is unlikely even with a compound with high affinity for the transporter.


Asunto(s)
Aciclovir/análogos & derivados , Aminoácidos/metabolismo , Transportador de Péptidos 1/metabolismo , Profármacos/metabolismo , Receptores de Glutamato Metabotrópico/agonistas , Valina/análogos & derivados , Aciclovir/administración & dosificación , Aciclovir/sangre , Aciclovir/metabolismo , Aciclovir/orina , Adolescente , Adulto , Anciano , Aminoácidos/administración & dosificación , Aminoácidos/sangre , Aminoácidos/orina , Transporte Biológico , Compuestos Bicíclicos Heterocíclicos con Puentes/sangre , Compuestos Bicíclicos Heterocíclicos con Puentes/orina , Óxidos S-Cíclicos/sangre , Óxidos S-Cíclicos/orina , Interacciones Farmacológicas , Femenino , Células HeLa , Humanos , Masculino , Persona de Mediana Edad , Profármacos/administración & dosificación , Profármacos/farmacocinética , Especificidad por Sustrato , Valaciclovir , Valina/administración & dosificación , Valina/sangre , Valina/metabolismo , Valina/orina , Adulto Joven
4.
Molecules ; 22(11)2017 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-29113080

RESUMEN

Acyclovir (ACV) and penciclovir (PNV) have been commonly used during the last few decades as potent antiviral agents, especially for the treatment of herpes virus infections. In the present research their binding properties with human serum albumin (HSA) were studied using different advanced spectroscopic and in-silico methods. The interactions between ACV/PNV and HSA at the three investigated temperatures revealed a static type of binding. Extraction of the thermodynamic parameters of the ACV-HSA and PNV-HSA systems from the measured spectrofluorimetric data demonstrated spontaneous interactions with an enthalpy change (∆H°) of -1.79 ± 0.29 and -4.47 ± 0.51 kJ·mol-1 for ACV and PNV, respectively. The entropy change (∆S°) of 79.40 ± 0.95 and 69.95 ± 1.69 J·mol-1·K-1 for ACV and PNV, respectively, hence supported a potential contribution of electrostatic binding forces to the ACV-HSA and PNV-HSA systems. Putative binding of ACV/PNV to HSA, using previously reported site markers, showed that ACV/PNV were bound to HSA within subdomains IIA and IIIA (Sudlow sites I and II). Further confirmation was obtained through molecular docking studies of ACV-HSA and PNV-HSA binding, which confirmed the binding site of ACV/PNV with the most stable configurations of ACV/PNV within the HSA. These ACV/PNV conformers were shown to have free energies of -25.61 and -22.01 kJ·mol-1 for ACV within the HSA sites I and II and -22.97 and -26.53 kJ·mol-1 for PNV in HSA sites I and II, with hydrogen bonding and electrostatic forces being the main binding forces in such conformers.


Asunto(s)
Aciclovir/análogos & derivados , Aciclovir/metabolismo , Antivirales/metabolismo , Albúmina Sérica Humana/química , Albúmina Sérica Humana/metabolismo , Sitios de Unión , Guanina , Humanos , Enlace de Hidrógeno , Modelos Moleculares , Simulación del Acoplamiento Molecular , Estructura Molecular , Unión Proteica , Dominios Proteicos , Termodinámica
5.
AAPS PharmSciTech ; 18(6): 2085-2094, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28004342

RESUMEN

The dissolution and subsequent oral bioavailability of acyclovir (ACY) is limited by its poor aqueous solubility. An attempt has been made in this work to provide mechanistic insights into the solubility enhancement and dissolution of ACY by using the water-soluble carrier polyethylene glycol 6000 (PEG6000). Solid dispersions with varying ratios of the drug (ACY) and carrier (PEG6000) were prepared and evaluated by phase solubility, in vitro release studies, kinetic analysis, in situ perfusion, and in vitro permeation studies. Solid state characterization was done by powder X-ray diffraction (XRD), differential scanning calorimetry (DSC), and Fourier transform infrared (FTIR) analysis, and surface morphology was assessed by polarizing microscopic image analysis, scanning electron microscopy, atomic force microscopy, and nuclear magnetic resonance analysis. Thermodynamic parameters indicated the solubilization effect of the carrier. The aqueous solubility and dissolution of ACY was found to be higher in all samples. The findings of XRD, DSC, FTIR and NMR analysis confirmed the formation of solid solution, crystallinity reduction, and the absence of interaction between the drug and carrier. SEM and AFM analysis reports ratified the particle size reduction and change in the surface morphology in samples. The permeation coefficient and amount of ACY diffused were higher in samples in comparison to pure ACY. Stability was found to be higher in dispersions. The results suggest that the study findings provided clear mechanical insights into the solubility and dissolution enhancement of ACY in PEG6000, and such findings could lay the platform for resolving the poor aqueous solubility issues in formulation development.


Asunto(s)
Aciclovir/síntesis química , Aciclovir/metabolismo , Portadores de Fármacos/síntesis química , Portadores de Fármacos/metabolismo , Aciclovir/farmacología , Animales , Antivirales/síntesis química , Antivirales/metabolismo , Antivirales/farmacología , Disponibilidad Biológica , Rastreo Diferencial de Calorimetría/métodos , Portadores de Fármacos/farmacología , Absorción Intestinal/efectos de los fármacos , Absorción Intestinal/fisiología , Cinética , Masculino , Microscopía Electrónica de Rastreo/métodos , Polietilenglicoles/síntesis química , Polietilenglicoles/metabolismo , Polietilenglicoles/farmacología , Ratas , Ratas Sprague-Dawley , Solubilidad , Espectroscopía Infrarroja por Transformada de Fourier/métodos , Difracción de Rayos X/métodos
6.
Biochemistry ; 55(7): 1168-77, 2016 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-26836009

RESUMEN

We examined the impact of two clinically approved anti-herpes drugs, acyclovir and Forscarnet (phosphonoformate), on the exonuclease activity of the herpes simplex virus-1 DNA polymerase, UL30. Acyclovir triphosphate and Foscarnet, along with the closely related phosphonoacetic acid, did not affect exonuclease activity on single-stranded DNA. Furthermore, blocking the polymerase active site due to either binding of Foscarnet or phosphonoacetic acid to the E-DNA complex or polymerization of acyclovir onto the DNA also had a minimal effect on exonuclease activity. The inability of the exonuclease to excise acyclovir from the primer 3'-terminus results from the altered sugar structure directly impeding phosphodiester bond hydrolysis as opposed to inhibiting binding, unwinding of the DNA by the exonuclease, or transfer of the DNA from the polymerase to the exonuclease. Removing the 3'-hydroxyl or the 2'-carbon from the nucleotide at the 3'-terminus of the primer strongly inhibited exonuclease activity, although addition of a 2'-hydroxyl did not affect exonuclease activity. The biological consequences of these results are twofold. First, the ability of acyclovir and Foscarnet to block dNTP polymerization without impacting exonuclease activity raises the possibility that their effects on herpes replication may involve both direct inhibition of dNTP polymerization and exonuclease-mediated destruction of herpes DNA. Second, the ability of the exonuclease to rapidly remove a ribonucleotide at the primer 3'-terminus in combination with the polymerase not efficiently adding dNTPs onto this primer provides a novel mechanism by which the herpes replication machinery can prevent incorporation of ribonucleotides into newly synthesized DNA.


Asunto(s)
Aciclovir/farmacología , Antivirales/farmacología , Exodesoxirribonucleasas/antagonistas & inhibidores , Foscarnet/farmacología , Herpesvirus Humano 1/enzimología , Modelos Moleculares , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Proteínas Virales/antagonistas & inhibidores , Aciclovir/química , Aciclovir/metabolismo , Antivirales/química , Antivirales/metabolismo , Dominio Catalítico , ADN de Cadena Simple/química , ADN de Cadena Simple/metabolismo , ADN Polimerasa Dirigida por ADN/química , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Exodesoxirribonucleasas/química , Exodesoxirribonucleasas/genética , Exodesoxirribonucleasas/metabolismo , Foscarnet/química , Foscarnet/metabolismo , Herpesvirus Humano 1/efectos de los fármacos , Hidrólisis/efectos de los fármacos , Cinética , Estructura Molecular , Inhibidores de la Síntesis del Ácido Nucleico/química , Inhibidores de la Síntesis del Ácido Nucleico/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Ribonucleótidos/química , Ribonucleótidos/metabolismo , Especificidad por Sustrato , Proteínas Virales/química , Proteínas Virales/genética , Proteínas Virales/metabolismo
7.
Antimicrob Agents Chemother ; 59(1): 186-92, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25331707

RESUMEN

The development of deoxynucleoside triphosphate (dNTP)-based drugs requires a quantitative understanding of any inhibition, activation, or hydrolysis by off-target cellular enzymes. SAMHD1 is a regulatory dNTP-triphosphohydrolase that inhibits HIV-1 replication in human myeloid cells. We describe here an enzyme-coupled assay for quantifying the activation, inhibition, and hydrolysis of dNTPs, nucleotide analogues, and nucleotide analogue inhibitors by triphosphohydrolase enzymes. The assay facilitates mechanistic studies of triphosphohydrolase enzymes and the quantification of off-target effects of nucleotide-based antiviral and chemotherapeutic agents.


Asunto(s)
Ácido Anhídrido Hidrolasas/análisis , Bioensayo/métodos , Evaluación Preclínica de Medicamentos/métodos , Proteínas de Unión al GTP Monoméricas/análisis , Proteínas de Unión al GTP Monoméricas/metabolismo , Ácido Anhídrido Hidrolasas/genética , Ácido Anhídrido Hidrolasas/metabolismo , Aciclovir/química , Aciclovir/metabolismo , Aciclovir/farmacología , Nucleótidos de Adenina/química , Nucleótidos de Adenina/farmacología , Antivirales/química , Antivirales/metabolismo , Antivirales/farmacología , Arabinonucleósidos/química , Arabinonucleósidos/farmacología , Catálisis/efectos de los fármacos , Clofarabina , Desoxirribonucleótidos/química , Desoxirribonucleótidos/metabolismo , Relación Dosis-Respuesta a Droga , Ganciclovir/química , Ganciclovir/farmacología , VIH-1 , Hidrólisis , Proteína 1 que Contiene Dominios SAM y HD
8.
Nucleic Acids Res ; 41(16): 7793-803, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23775789

RESUMEN

Chain-terminating nucleoside analogs (CTNAs) that cause stalling or premature termination of DNA replication forks are widely used as anticancer and antiviral drugs. However, it is not well understood how cells repair the DNA damage induced by these drugs. Here, we reveal the importance of tyrosyl-DNA phosphodiesterase 1 (TDP1) in the repair of nuclear and mitochondrial DNA damage induced by CTNAs. On investigating the effects of four CTNAs-acyclovir (ACV), cytarabine (Ara-C), zidovudine (AZT) and zalcitabine (ddC)-we show that TDP1 is capable of removing the covalently linked corresponding CTNAs from DNA 3'-ends. We also show that Tdp1-/- cells are hypersensitive and accumulate more DNA damage when treated with ACV and Ara-C, implicating TDP1 in repairing CTNA-induced DNA damage. As AZT and ddC are known to cause mitochondrial dysfunction, we examined whether TDP1 repairs the mitochondrial DNA damage they induced. We find that AZT and ddC treatment leads to greater depletion of mitochondrial DNA in Tdp1-/- cells. Thus, TDP1 seems to be critical for repairing nuclear and mitochondrial DNA damage caused by CTNAs.


Asunto(s)
Antimetabolitos Antineoplásicos/toxicidad , Antivirales/toxicidad , Daño del ADN , Reparación del ADN , Hidrolasas Diéster Fosfóricas/metabolismo , Aciclovir/metabolismo , Aciclovir/toxicidad , Animales , Fármacos Anti-VIH/metabolismo , Fármacos Anti-VIH/toxicidad , Antimetabolitos Antineoplásicos/metabolismo , Antivirales/metabolismo , Línea Celular , Núcleo Celular/efectos de los fármacos , Células Cultivadas , Pollos , Citarabina/metabolismo , Citarabina/toxicidad , ADN Mitocondrial/efectos de los fármacos , ADN Mitocondrial/metabolismo , Eliminación de Gen , Ratones , Hidrolasas Diéster Fosfóricas/genética , Zalcitabina/metabolismo , Zalcitabina/toxicidad , Zidovudina/metabolismo , Zidovudina/toxicidad
9.
J Hepatol ; 61(5): 1064-72, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24862448

RESUMEN

BACKGROUND & AIMS: Hepatocellular carcinoma (HCC) is one of the most common malignancies worldwide. Although hepatectomy and transplantation have significantly improved survival, there is no effective chemotherapeutic treatment for HCC and its prognosis remains poor. Sustained activation of telomerase is essential for the growth and progression of HCC, suggesting that telomerase is a rational target for HCC therapy. Therefore, we developed a thymidine analogue pro-drug, acycloguanosyl-5'-thymidyltriphosphate (ACV-TP-T), which is specifically activated by telomerase in HCC cells and investigated its anti-tumour efficacy. METHODS: First, we verified in vitro whether ACV-TP-T was a telomerase substrate. Second, we evaluated proliferation and apoptosis in murine (Hepa1-6) and human (Hep3B, HuH7, HepG2) hepatic cancer cells treated with ACV-TP-T. Next, we tested the in vivo treatment efficacy in HBV transgenic mice that spontaneously develop hepatic tumours, and in a syngeneic orthotopic murine model where HCC cells were implanted directly in the liver. RESULTS: In vitro characterization provided direct evidence that the pro-drug was actively metabolized in liver cancer cells by telomerase to release the active form of acyclovir. Alterations in cell cycle and apoptosis were observed following in vitro treatment with ACV-TP-T. In the transgenic and orthotopic mouse models, treatment with ACV-TP-T reduced tumour growth, increased apoptosis, and reduced the proliferation of tumour cells. CONCLUSIONS: ACV-TP-T is activated by telomerase in HCC cells and releases active acyclovir that reduces proliferation and induces apoptosis in human and murine liver cancer cells. This pro-drug holds a great promise for the treatment of HCC.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma Hepatocelular/tratamiento farmacológico , Guanosina/análogos & derivados , Neoplasias Hepáticas/tratamiento farmacológico , Profármacos/uso terapéutico , Nucleótidos de Timina/uso terapéutico , Aciclovir/metabolismo , Aciclovir/uso terapéutico , Animales , Antineoplásicos/metabolismo , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Guanosina/metabolismo , Guanosina/uso terapéutico , Células Hep G2 , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/patogenicidad , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas Experimentales/tratamiento farmacológico , Neoplasias Hepáticas Experimentales/metabolismo , Neoplasias Hepáticas Experimentales/patología , Ratones , Ratones Transgénicos , Profármacos/metabolismo , Telomerasa/metabolismo , Nucleótidos de Timina/metabolismo
10.
Virol J ; 11: 34, 2014 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-24558980

RESUMEN

BACKGROUND: Herpesviruses are ubiquitous pathogens that infect and cause recurrent disease in multiple animal species. Feline herpesvirus-1 (FHV-1), a member of the alphaherpesvirus family, causes respiratory illness and conjunctivitis, and approximately 80% of domestic cats are latently infected. Oral administration of famciclovir or topical application of cidofovir has been shown in masked, placebo-controlled prospective trials to reduce clinical signs and viral shedding in experimentally inoculated cats. However, to the authors' knowledge, other drugs have not been similarly assessed or were not safe or effective. Likewise, to our knowledge, no drugs have been assessed in a placebo-controlled manner in cats with recrudescent herpetic disease. Controlled-release devices would permit long-term administration of these drugs and enhance compliance. METHODS: We therefore engineered implantable cylindrical devices made from silicone (MED-4750) impregnated with penciclovir, for long-term, steady-state delivery of this drug. RESULTS: Our data show that these devices release penciclovir with a burst of drug delivery until the tenth day of release, then at an average rate of 5.063 ± 1.704 µg per day through the next 50 days with near zero-order kinetics (in comparison to MED-4750-acyclovir devices, which show the same burst kinetics and average 2.236 ± 0.625 µg/day thereafter). Furthermore, these devices suppress primary infection of FHV-1 in a cell culture system. CONCLUSIONS: The clinical deployment of these silicone-penciclovir devices may allow long-term treatment of FHV-1 infection with a single intervention that could last the life of the host cat.


Asunto(s)
Aciclovir/análogos & derivados , Antivirales/metabolismo , Sistemas de Liberación de Medicamentos , Varicellovirus/efectos de los fármacos , Aciclovir/metabolismo , Animales , Gatos , Células Cultivadas , Guanina , Polímeros , Siliconas
11.
Biochemistry ; 52(33): 5665-74, 2013 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-23859606

RESUMEN

d-Amino acid oxidase (DAO) is a degradative enzyme that is stereospecific for d-amino acids, including d-serine and d-alanine, which are believed to be coagonists of the N-methyl-d-aspartate (NMDA) receptor. To identify a new class of DAO inhibitor(s) that can be used to elucidate the molecular details of the active site environment of DAO, manifold biologically active compounds of microbial origin and pre-existing drugs were screened for their ability to inhibit DAO activity, and several compounds were identified as candidates. One of these compounds, acyclovir (ACV), a well-known antiviral drug used for the treatment of herpesvirus infections, was characterized and evaluated as a novel DAO inhibitor in vitro. Analysis showed that ACV acts on DAO as a reversible slow-binding inhibitor, and interestingly, the time required to achieve equilibrium between DAO, ACV, and the DAO/ACV complex was highly dependent on temperature. The binding mechanism of ACV to DAO was investigated in detail by several approaches, including kinetic analysis, structural modeling of DAO complexed with ACV, and site-specific mutagenesis of an active site residue postulated to be involved in the binding of ACV. The results confirm that ACV is a novel, active site-directed inhibitor of DAO that can be a valuable tool for investigating the structure-function relationships of DAO, including the molecular details of the active site environment of DAO. In particular, it appears that ACV can serve as an active site probe to study the structural basis of temperature-induced conformational changes of DAO.


Asunto(s)
Aciclovir/metabolismo , Aciclovir/farmacología , D-Aminoácido Oxidasa/antagonistas & inhibidores , D-Aminoácido Oxidasa/metabolismo , Aciclovir/química , Algoritmos , Antivirales/química , Antivirales/metabolismo , Antivirales/farmacología , Benzoatos/química , Benzoatos/metabolismo , Benzoatos/farmacología , Dominio Catalítico/genética , D-Aminoácido Oxidasa/química , Relación Dosis-Respuesta a Droga , Humanos , Cinética , Modelos Moleculares , Estructura Molecular , Mutagénesis Sitio-Dirigida , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Temperatura
12.
Mol Pharm ; 10(2): 650-63, 2013 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-23268600

RESUMEN

The barrier epithelia of the cornea and retina control drug and nutrient access to various compartments of the human eye. While ocular transporters are likely to play a critical role in homeostasis and drug delivery, little is known about their expression, localization and function. In this study, the mRNA expression levels of 445 transporters, metabolic enzymes, transcription factors and nuclear receptors were profiled in five regions of the human eye: cornea, iris, ciliary body, choroid and retina. Through RNA expression profiling and immunohistochemistry, several transporters were identified as putative targets for drug transport in ocular tissues. Our analysis identified SLC22A7 (OAT2), a carrier for the antiviral drug acyclovir, in the corneal epithelium, in addition to ABCG2 (BCRP), an important xenobiotic efflux pump, in retinal nerve fibers and the retinal pigment epithelium. Collectively, our results provide an understanding of the transporters that serve to maintain ocular homeostasis and which may be potential targets for drug delivery to deep compartments of the eye.


Asunto(s)
Ojo/metabolismo , Perfilación de la Expresión Génica/métodos , Transportadores de Anión Orgánico ATP-Dependiente/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Aciclovir/metabolismo , Córnea/metabolismo , Humanos , Inmunohistoquímica , Técnicas In Vitro , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Transportadores de Anión Orgánico ATP-Dependiente/genética , Transportadores de Anión Orgánico Sodio-Independiente/genética , Transportadores de Anión Orgánico Sodio-Independiente/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Retina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Pharm Res ; 30(8): 2063-76, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23657675

RESUMEN

PURPOSE: Biotinylated lipid prodrugs of acyclovir (ACV) were designed to target the sodium dependent multivitamin transporter (SMVT) on the cornea to facilitate enhanced cellular absorption of ACV. METHODS: All the prodrugs were screened for in vitro cellular uptake, interaction with SMVT, docking analysis, cytotoxicity, enzymatic stability and antiviral activity. RESULTS: Uptake of biotinylated lipid prodrugs of ACV (B-R-ACV and B-12HS-ACV) was significantly higher than biotinylated prodrug (B-ACV), lipid prodrugs (R-ACV and 12HS-ACV) and ACV in corneal cells. Transepithelial transport across rabbit corneas indicated the recognition of the prodrugs by SMVT. Average Vina scores obtained from docking studies further confirmed that biotinylated lipid prodrugs possess enhanced affinity towards SMVT. All the prodrugs studied did not cause any cytotoxicity and were found to be safe and non-toxic. B-R-ACV and B-12HS-ACV were found to be relatively more stable in ocular tissue homogenates and exhibited excellent antiviral activity. CONCLUSIONS: Biotinylated lipid prodrugs demonstrated synergistic improvement in cellular uptake due to recognition of the prodrugs by SMVT on the cornea and lipid mediated transcellular diffusion. These biotinylated lipid prodrugs appear to be promising drug candidates for the treatment of herpetic keratitis (HK) and may lower ACV resistance in patients with poor clinical response.


Asunto(s)
Aciclovir/metabolismo , Aciclovir/farmacología , Antivirales/metabolismo , Antivirales/farmacología , Córnea/metabolismo , Profármacos/metabolismo , Profármacos/farmacología , Simportadores/metabolismo , Aciclovir/química , Aciclovir/farmacocinética , Animales , Antivirales/química , Antivirales/farmacocinética , Biotinilación , Línea Celular , Células Cultivadas , Humanos , Simulación del Acoplamiento Molecular , Profármacos/química , Profármacos/farmacocinética , Conejos , Virosis/tratamiento farmacológico , Virus/efectos de los fármacos
14.
J Virol ; 85(9): 4618-22, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21325417

RESUMEN

Acyclovir, a nucleoside analog, is thought to be specific for the human herpesviruses because it requires a virally encoded enzyme to phosphorylate it to acyclovir monophosphate. Recently, acyclovir triphosphate was shown to be a direct inhibitor of human immunodeficiency virus type 1 (HIV-1) reverse transcriptase. Here, we showed that acyclovir is an inhibitor of HIV-1 replication in CD4(+) T cells from cord blood that have undetectable levels of the eight human herpesviruses. Additionally, acyclovir phosphates were detected by reverse-phase-high performance liquid chromatography (RP-HPLC) and quantified in a primer extension assay from cord blood. The data support acyclovir as an inhibitor of HIV-1 replication in herpesvirus-negative cells.


Asunto(s)
Aciclovir/farmacología , Antivirales/farmacología , Linfocitos T CD4-Positivos/virología , VIH-1/efectos de los fármacos , Herpesviridae/aislamiento & purificación , Replicación Viral/efectos de los fármacos , Aciclovir/metabolismo , Adulto , Antivirales/metabolismo , Linfocitos T CD4-Positivos/química , Cromatografía Líquida de Alta Presión , Herpesviridae/enzimología , Humanos
15.
Environ Sci Technol ; 46(4): 2169-78, 2012 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-22300376

RESUMEN

The oxidation of the antiviral drug acyclovir (ACV) and its main biotransformation product carboxy-acyclovir (carboxy-ACV) by ozone was investigated. Both compounds have recently been detected in surface water, and carboxy-ACV has also been detected in drinking water. The experiments revealed a strong pH dependence of the oxidation of ACV and carboxy-ACV with reaction rate constants increasing by 4 orders of magnitude between the protonated, positively charged form (k(ox,PH(+)), ∼2.5 × 10(2) M(-1) s(-1)) and the deprotonated, negatively charged form (k(ox,P(-)), 3.4 × 10(6) M(-1) s(-1)). At pH 8 a single oxidation product was formed which was identified via LC-LTQ-Orbitrap MS and NMR as N-(4-carbamoyl-2-imino-5-oxoimidazolidin)formamido-N-methoxyacetic acid (COFA). Using Vibrio fischeri , an acute bacterial toxicity was found for COFA while carboxy-ACV revealed no toxic effects. Ozonation experiments with guanine and guanosine at pH 8 led to the formation of the respective 2-imino-5-oxoimidazolidines, confirming that guanine derivatives such as carboxy-ACV are undergoing the same reactions during ozonation. Furthermore, COFA was detected in finished drinking water of a German waterworks after ozonation and subsequent activated carbon treatment.


Asunto(s)
Aciclovir/química , Antivirales/química , Oxidantes Fotoquímicos/química , Ozono/química , Purificación del Agua , Aciclovir/metabolismo , Antivirales/metabolismo , Biotransformación , Guanina/química , Guanosina/química , Concentración de Iones de Hidrógeno , Cinética , Oxidación-Reducción , Aguas del Alcantarillado/microbiología
16.
Organogenesis ; 18(1): 2055354, 2022 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-35384798

RESUMEN

Encephalitis, the most significant of the central nervous system (CNS) diseases caused by Herpes simplex virus 1 (HSV-1), may have long-term sequelae in survivors treated with acyclovir, the cause of which is unclear. HSV-1 exhibits a tropism toward neurogenic niches in CNS enriched with neural precursor cells (NPCs), which play a pivotal role in neurogenesis. NPCs are susceptible to HSV-1. There is a paucity of information regarding the influence of HSV-1 on neurogenesis in humans. We investigated HSV-1 infection of NPCs from two individuals. Our results show (i) HSV-1 impairs, to different extents, the proliferation, self-renewing, and, to an even greater extent, migration of NPCs from these two subjects; (ii) The protective effect of the gold-standard antiherpetic drug acyclovir (ACV) varies with viral dose and is incomplete. It is also subject to differences in terms of efficacy of the NPCs derived from these two individuals. These results suggest that the effects of HSV-1 may have on aspects of NPC neurogenesis may vary among individuals, even in the presence of acyclovir, and this may contribute to the heterogeneity of cognitive sequelae across encephalitis survivors. Further analysis of NPC cell lines from a larger number of individuals is warranted.


Asunto(s)
Encefalitis , Herpes Simple , Herpesvirus Humano 1 , Células-Madre Neurales , Aciclovir/metabolismo , Aciclovir/farmacología , Aciclovir/uso terapéutico , Encefalitis/tratamiento farmacológico , Herpes Simple/tratamiento farmacológico , Herpesvirus Humano 1/metabolismo , Humanos , Neurogénesis
17.
J Virol ; 84(9): 4534-42, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20181711

RESUMEN

Ganciclovir (GCV) and acyclovir (ACV) are guanine nucleoside analogues that inhibit lytic herpesvirus replication. GCV and ACV must be monophosphorylated by virally encoded enzymes to be converted into nucleotides and incorporated into viral DNA. However, whether GCV and/or ACV phosphorylation in Epstein-Barr virus (EBV)-infected cells is mediated primarily by the EBV-encoded protein kinase (EBV-PK), the EBV-encoded thymidine kinase (EBV-TK), or both is controversial. To examine this question, we constructed EBV mutants containing stop codons in either the EBV-PK or EBV-TK open reading frame and selected for stable 293T clones latently infected with wild-type EBV or each of the mutant viruses. Cells were induced to the lytic form of viral replication with a BZLF1 expression vector in the presence and absence of various doses of GCV and ACV, and infectious viral titers were determined by a green Raji cell assay. As expected, virus production in wild-type EBV-infected 293T cells was inhibited by both GCV (50% inhibitory concentration [IC(50)] = 1.5 microM) and ACV (IC(50) = 4.1 microM). However, the EBV-PK mutant (which replicates as well as the wild-type (WT) virus in 293T cells) was resistant to both GCV (IC(50) = 19.6 microM) and ACV (IC(50) = 36.4 microM). Expression of the EBV-PK protein in trans restored GCV and ACV sensitivity in cells infected with the PK mutant virus. In contrast, in 293T cells infected with the TK mutant virus, viral replication remained sensitive to both GCV (IC(50) = 1.2 microM) and ACV (IC(50) = 2.8 microM), although susceptibility to the thymine nucleoside analogue, bromodeoxyuridine, was reduced. Thus, EBV-PK but not EBV-TK mediates ACV and GCV susceptibilities.


Asunto(s)
Aciclovir/farmacología , Antivirales/farmacología , Ganciclovir/farmacología , Herpesvirus Humano 4/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Timidina Quinasa/metabolismo , Proteínas Virales/metabolismo , Aciclovir/metabolismo , Antivirales/metabolismo , Línea Celular , Codón sin Sentido , Ganciclovir/metabolismo , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/crecimiento & desarrollo , Humanos , Concentración 50 Inhibidora , Mutagénesis Sitio-Dirigida , Fosforilación , Proteínas Serina-Treonina Quinasas/deficiencia , Timidina Quinasa/deficiencia , Proteínas Virales/genética , Replicación Viral/efectos de los fármacos
18.
Environ Sci Technol ; 45(7): 2761-9, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21388176

RESUMEN

The biotransformation of the two antiviral drugs, acyclovir (ACV) and penciclovir (PCV), was investigated in contact with activated sludge. Biodegradation kinetics were determined, and transformation products (TPs) were identified using Hybrid Linear Ion Trap- FT Mass Spectrometry (LTQ Orbitrap Velos) and 1D (1H NMR, 13C NMR) and 2D (1H,1H-COSY, 1H-(13)C-HSQC) NMR Spectroscopy. ACV and PCV rapidly dissipated in the activated sludge batch systems with half-lives of 5.3 and 3.4 h and first-order rate constants in relation to the amount of suspended solids (SS) of 4.9±0.1 L gss(-1) d(-1) and 7.6±0.3 L gss(-1) d(-1), respectively. For ACV only a single TP was found, whereas eight TPs were identified for PCV. Structural elucidation of TPs exhibited that transformation only took place at the side chain leaving the guanine moiety unaltered. The oxidation of the primary hydroxyl group in ACV resulted in the formation of carboxy-acyclovir (Carboxy-ACV). For PCV, transformation was more diverse with several enzymatic reactions taking place such as the oxidation of terminal hydroxyl groups and ß-oxidation followed by acetate cleavage. Analysis of different environmental samples revealed the presence of Carboxy-ACV in surface and drinking water with concentrations up to 3200 ng L(-1) and 40 ng L(-1), respectively.


Asunto(s)
Aciclovir/análogos & derivados , Aciclovir/metabolismo , Antivirales/metabolismo , Aguas del Alcantarillado/química , Eliminación de Residuos Líquidos/métodos , Aciclovir/análisis , Antivirales/análisis , Biotransformación , Guanina/análogos & derivados , Guanina/análisis , Guanina/metabolismo , Aguas del Alcantarillado/microbiología , Contaminantes Químicos del Agua/análisis , Contaminantes Químicos del Agua/metabolismo
19.
Can J Physiol Pharmacol ; 89(9): 675-80, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21859328

RESUMEN

The human breast cancer resistance protein (BCRP/ABCG2) is widely expressed in human tissues, including the kidney. In mice, Bcrp1 (murine BCRP ortholog) mediates the transport of acyclovir into breast milk. It is plausible that acyclovir is also a substrate for the human BCRP. The objective of the study was to determine whether acyclovir is a substrate for human BCRP. Transfected human embryonic kidney (HEK293) cells (containing the wild-type ABCG2 gene) were exposed to [8-(14)C]acyclovir (1 µmol/L) in the presence or absence of the BCRP inhibitor fumitremorgin C (FTC). Intracellular acyclovir accumulation was assessed using a liquid scintillation counter. Coexposure to FTC resulted in a significant (5-fold) increase in the intracellular accumulation of acyclovir. The results suggest that acyclovir is a substrate for human BCRP. The study is the first to provide direct evidence for the role of human BCRP in acyclovir transport and its potential significance with respect to renal tubular transport of acyclovir and the direct renal tubular insult induced by the drug.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Lesión Renal Aguda/inducido químicamente , Aciclovir/metabolismo , Aciclovir/toxicidad , Riñón/efectos de los fármacos , Riñón/metabolismo , Proteínas de Neoplasias/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/biosíntesis , Transportadoras de Casetes de Unión a ATP/genética , Lesión Renal Aguda/metabolismo , Aciclovir/farmacocinética , Transporte Biológico/efectos de los fármacos , Línea Celular Transformada , Femenino , Células HEK293 , Humanos , Túbulos Renales/metabolismo , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Transfección , Células Tumorales Cultivadas
20.
Biomed Chromatogr ; 25(11): 1189-200, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21400550

RESUMEN

Plasma estimation of valaciclovir, an antiviral drug, is challenging due to both in-vivo and ex-vivo hydrolysis to active metabolite acyclovir. A simultaneous method is described involving the solid-phase ion-exchange extraction procedure requiring 100 µL of plasma volume, a reverse-phase Lichrosphere RP Select B (125 × 6 mm, 5 µm) column and isocratic mobile phase to achieve the desired chromatographic separation. ESI-MS/MS multiple reaction monitoring in positive polarity, detected mass pairs for valaciclovir (m/z 325.5 → 152.2), acyclovir (m/z 226.3 → 152.2) and respective internal standards valganciclovir (m/z 307.1 → 220.3) and acyclovir-d4 (m/z 230.2 → 152.0). Fully fledged method validation was evaluated as per current regulatory requirements and results were deemed acceptable. The plasma samples showed extensive hydrolysis of valaciclovir when collected or processed at room temperature, without buffer stabilization prior to storage at -15°C. Our results showed that using prechilled K3 EDTA vacutainers immersed in an iced-water bath during blood sample collection, and addition of 50% orthophosphoric acid solution to plasma samples prior to storage at -50°C for at least 120 days controlled the hydrolysis of valaciclovir to acyclovir. While monitoring drug absorption into systematic circulation, the valaciclovir to acyclovir formation ratio was improved to 1:20 in healthy volunteers for the first time.


Asunto(s)
Aciclovir/análogos & derivados , Aciclovir/sangre , Aciclovir/química , Espectrometría de Masas en Tándem/métodos , Valina/análogos & derivados , Aciclovir/metabolismo , Aciclovir/farmacocinética , Cromatografía Liquida , Estabilidad de Medicamentos , Humanos , Hidrólisis , Modelos Lineales , Masculino , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Valaciclovir , Valina/sangre , Valina/química , Valina/metabolismo , Valina/farmacocinética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA