Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Naunyn Schmiedebergs Arch Pharmacol ; 393(7): 1251-1267, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32313990

RESUMO

The effects of 100 µM of 3',5'-cGMP, cAMP, cCMP, and cUMP as well as of the corresponding membrane-permeant acetoxymethyl esters on anti-CD3-antibody (OKT3)-induced IL-2 production of HuT-78 cutaneous T cell lymphoma (Sézary lymphoma) cells were analyzed. Only 3',5'-cGMP significantly reduced IL-2 production. Flow cytometric analysis of apoptotic (propidium iodide/annexin V staining) and anti-proliferative (CFSE staining) effects revealed that 3',5'-cGMP concentrations > 50 µM strongly inhibited proliferation and promoted apoptosis of HuT-78 cells (cultured in the presence of αCD3 antibody). Similar effects were observed for the positional isomer 2',3'-cGMP and for 2',-GMP, 3'-GMP, 5'-GMP, and guanosine. By contrast, guanosine and guanosine-derived nucleotides had no cytotoxic effect on peripheral blood mononuclear cells (PBMCs) or acute lymphocytic leukemia (ALL) xenograft cells. The anti-proliferative and apoptotic effects of guanosine and guanosine-derived compounds on HuT-78 cells were completely eliminated by the nucleoside transport inhibitor NBMPR (S-(4-Nitrobenzyl)-6-thioinosine). By contrast, the ecto-phosphodiesterase inhibitor DPSPX (1,3-dipropyl-8-sulfophenylxanthine) and the CD73 ecto-5'-nucleotidase inhibitor AMP-CP (adenosine 5'-(α,ß-methylene)diphosphate) were not protective. We hypothesize that HuT-78 cells metabolize guanosine-derived nucleotides to guanosine by yet unknown mechanisms. Guanosine then enters the cells by an NBMPR-sensitive nucleoside transporter and exerts cytotoxic effects. This transporter may be ENT1 because NBMPR counteracted guanosine cytotoxicity in HuT-78 cells with nanomolar efficacy (IC50 of 25-30 nM). Future studies should further clarify the mechanism of the observed effects and address the question, whether guanosine or guanosine-derived nucleotides may serve as adjuvants in the therapy of cancers that express appropriate nucleoside transporters and are sensitive to established nucleoside-derived cytostatic drugs.


Assuntos
Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Guanosina/farmacologia , Linfoma de Células T/tratamento farmacológico , Linhagem Celular Tumoral , Guanosina/administração & dosagem , Guanosina/análogos & derivados , Humanos , Concentração Inibidora 50 , Leucócitos Mononucleares/efeitos dos fármacos , Linfoma de Células T/patologia , Proteínas de Transporte de Nucleosídeos/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Mol Med Rep ; 15(6): 3999-4004, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28487988

RESUMO

Guanosine (GUO) is neuroprotective when administered acutely for the treatment of cerebral ischemia. The aim of the present study was to investigate whether delayed administration of GUO improved long­term functional recovery following stroke, as well as to explore the potential underlying mechanisms. GUO (8 mg/kg) or a vehicle was administered intraperitoneally for 7 consecutive days beginning 24 h prior to photothrombosis­induced stroke in male C57/B6J mice. Behaviour tests were performed at days 1, 3, 7, 14 and 28 post­stroke. Infarct volume was measured using Nissl staining at day 7 post­stroke. Neurogenesis and angiogenesis were evaluated by co­labelling bromodeoxyuridine (BrdU) with doublecortin (DCX), neuronal nuclei (NeuN) and von Willebrand factor, in immunohistochemical studies. Brain­derived neurotrophic factor (BDNF) and vascular endothelial growth factor (VEGF) levels in the ipsilesional brain at day 28 post­stroke were detected by western blot analysis. Delayed administration of GUO did not reduce infarct volume or affect neurological function at day 7 post­stroke; however, it did improve functional recovery from day 14 post­stroke, when compared with the vehicle group. GUO significantly increased the number of BrdU+ and BrdU+/DCX+ cells in the subventricular zone and subgranular zone at all examined time points, the number of Brdu+/NeuN+ cells in the peri­infarction region at days 14 and 28 post­stroke and microvessel density in the peri­infarction region at day 28 post­stroke compared with the vehicle group. In addition, the BDNF and VEGF levels in the ipsilesional brain were significantly elevated. Delayed administration of GUO at 24 h post­stroke enhanced neurogenesis and angiogenesis, and increased BDNF and VEGF levels, which likely contributes to long­term functional recovery following stroke.


Assuntos
Guanosina/administração & dosagem , Neovascularização Fisiológica/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Recuperação de Função Fisiológica/efeitos dos fármacos , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/fisiopatologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Isquemia Encefálica/complicações , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Modelos Animais de Doenças , Proteína Duplacortina , Expressão Gênica , Imuno-Histoquímica , Masculino , Camundongos , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Substâncias Protetoras/administração & dosagem , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/etiologia , Fatores de Tempo
3.
J Pharmacol Exp Ther ; 350(3): 719-26, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25002416

RESUMO

In cultured renal cells and isolated perfused kidneys, extracellular guanosine augments extracellular adenosine and inosine (the major renal metabolite of adenosine) levels by altering the extracellular disposition of these purines. The present study addressed whether this "guanosine-adenosine mechanism" exists in vivo. In rats (n = 15), intravenous infusions of adenosine (1 µmol/kg per minute) decreased mean arterial blood pressure (MABP) from 114 ± 4 to 83 ± 5 mm Hg, heart rate (HR) from 368 ± 11 to 323 ± 9 beats/min), and renal blood flow (RBF) from 6.2 ± 0.5 to 5.3 ± 0.6 ml/min). In rats (n = 15) pretreated with intravenous guanosine (10 µmol/kg per minute), intravenous adenosine (1 µmol/kg per minute) decreased MABP (from 109 ± 4 to 58 ± 5 mm Hg), HR (from 401 ± 10 to 264 ± 20 beats/min), and RBF (from 6.2 ± 0.7 to 1.7 ± 0.3). Two-factor analysis of variance (2F-ANOVA) revealed a significant interaction (P < 0.0001) between guanosine and adenosine for MABP, HR, and RBF. In control rats, the urinary excretion rate of endogenous inosine was 211 ± 103 ng/30 minutes (n = 9); however, in rats treated with intravenous guanosine (10 µmol/kg per minute), the excretion rate of inosine was 1995 ± 300 ng/30 minutes (n = 12; P < 0.0001 versus controls). Because adenosine inhibits inflammatory cytokine production, we also examined the effects of intravenous guanosine on endotoxemia-induced increases in tumor necrosis factor-α (TNF-α). In control rats (n = 7), lipopolysaccharide (LPS; Escherichia coli 026:B6 endotoxin; 30 mg/kg) increased plasma TNF-α from 164 ± 56 to 4082 ± 730 pg/ml, whereas in rats pretreated with intravenous guanosine (10 µmol/kg per minute; n = 6), LPS increased plasma TNF-α from 121 ± 45 to 1821 ± 413 pg/ml (2F-ANOVA interaction effect, P = 0.0022). We conclude that the guanosine-adenosine mechanism exists in vivo and that guanosine may be a useful therapeutic for reducing inflammation.


Assuntos
Adenosina/administração & dosagem , Adenosina/sangue , Guanosina/administração & dosagem , Guanosina/sangue , Animais , Interações Medicamentosas/fisiologia , Infusões Intravenosas , Masculino , Ratos , Ratos Sprague-Dawley
4.
PLoS One ; 9(2): e90693, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24587409

RESUMO

BACKGROUND AND PURPOSE: Stroke is a devastating disease. Both excitotoxicity and oxidative stress play important roles in ischemic brain injury, along with harmful impacts on ischemic cerebral tissue. As guanosine plays an important neuroprotective role in the central nervous system, the purpose of this study was to evaluate the neuroprotective effects of guanosine and putative cerebral events following the onset of permanent focal cerebral ischemia. METHODS: Permanent focal cerebral ischemia was induced in rats by thermocoagulation. Guanosine was administered immediately, 1 h, 3 h and 6 h after surgery. Behavioral performance was evaluated by cylinder testing for a period of 15 days after surgery. Brain oxidative stress parameters, including levels of ROS/RNS, lipid peroxidation, antioxidant non-enzymatic levels (GSH, vitamin C) and enzymatic parameters (SOD expression and activity and CAT activity), as well as glutamatergic parameters (EAAC1, GLAST and GLT1, glutamine synthetase) were analyzed. RESULTS: After 24 h, ischemic injury resulted in impaired function of the forelimb, caused brain infarct and increased lipid peroxidation. Treatment with guanosine restored these parameters. Oxidative stress markers were affected by ischemic insult, demonstrated by increased ROS/RNS levels, increased SOD expression with reduced SOD activity and decreased non-enzymatic (GSH and vitamin C) antioxidant defenses. Guanosine prevented increased ROS/RNS levels, decreased SOD activity, further increased SOD expression, increased CAT activity and restored vitamin C levels. Ischemia also affected glutamatergic parameters, illustrated by increased EAAC1 levels and decreased GLT1 levels; guanosine reversed the decreased GLT1 levels and did not affect the EAAC1 levels. CONCLUSION: The effects of brain ischemia were strongly attenuated by guanosine administration. The cellular mechanisms involved in redox and glutamatergic homeostasis, which were both affected by the ischemic insult, were also modulated by guanosine. These observations reveal that guanosine may represent a potential therapeutic agent in cerebral ischemia by preventing oxidative stress and excitotoxicity.


Assuntos
Lesões Encefálicas/prevenção & controle , Lesões Encefálicas/fisiopatologia , Isquemia Encefálica/fisiopatologia , Guanosina/farmacologia , Animais , Ácido Ascórbico/metabolismo , Western Blotting , Lesões Encefálicas/metabolismo , Isquemia Encefálica/etiologia , Catalase/metabolismo , Relação Dose-Resposta a Droga , Esquema de Medicação , Eletrocoagulação/efeitos adversos , Transportador 3 de Aminoácido Excitatório/metabolismo , Comportamento Exploratório/efeitos dos fármacos , Comportamento Exploratório/fisiologia , Membro Anterior/efeitos dos fármacos , Membro Anterior/fisiopatologia , Proteínas de Transporte de Glutamato da Membrana Plasmática/metabolismo , Glutamato-Amônia Ligase/metabolismo , Glutationa/metabolismo , Guanosina/administração & dosagem , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Fármacos Neuroprotetores/farmacologia , Óxido Nítrico/metabolismo , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/metabolismo
5.
Purinergic Signal ; 9(4): 655-61, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23839776

RESUMO

The aim of the present study was to evaluate if guanine-based purines may affect the gastric motor function in mouse. Thus, the influence of guanosine on the gastric emptying rate in vivo was determined and its effects on spontaneous gastric mechanical activity, detected as changes of the intraluminal pressure, were analyzed in vitro before and after different treatments. Gastric gavage of guanosine (1.75-10 mg/kg) delayed the gastric emptying. Guanosine (30 µM-1 mM) induced a concentration-dependent relaxation of isolated stomach, which was not affected by the inhibition of the purine nucleoside phosphorylase enzyme by 4'-deaza-1'-aza-2'-deoxy-1'-(9-methylene)-immucillin-H. The inhibitory response was antagonized by S-(4-nitrobenzyl)-6-thioinosine, a membrane nucleoside transporter inhibitor, but not affected by 9-chloro-2-(2-furanyl)-[1,2,4]triazolo[1,5-c]quinazolin-5-amine, a nonselective adenosine receptor antagonist, or by tetrodotoxin, a blocker of neuronal voltage-dependent Na(+) channels. Moreover, guanosine-induced effects persisted in the presence of 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, an inhibitor of soluble guanylyl cyclase or tetraethylammonium, a nonselective potassium channel blocker, but they were progressively reduced by increasing concentrations of 2'5'dideoxyadenosine, an adenylyl cyclase inhibitor. Lastly, the levels of cyclic adenosine monophosphate (cAMP), measured by ELISA, in gastric full thickness preparations were increased by guanosine. In conclusion, our data indicate that, in mouse, guanosine is able to modulate negatively the gastric motor function, reducing gastric emptying and inducing muscular relaxation. The latter is dependent by its cellular uptake and involves adenylyl cyclase activation and increase in cAMP intracellular levels, while it is independent on neural action potentials, adenosine receptors, and K(+) channel activation.


Assuntos
AMP Cíclico/metabolismo , Esvaziamento Gástrico/fisiologia , Motilidade Gastrointestinal/fisiologia , Guanosina/administração & dosagem , Músculo Liso/fisiologia , Estômago/fisiologia , Adaptação Fisiológica/efeitos dos fármacos , Adaptação Fisiológica/fisiologia , Animais , Relação Dose-Resposta a Droga , Esvaziamento Gástrico/efeitos dos fármacos , Motilidade Gastrointestinal/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso/efeitos dos fármacos , Estômago/efeitos dos fármacos
6.
Br J Pharmacol ; 159(6): 1247-63, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20132210

RESUMO

BACKGROUND AND PURPOSE: It is well known that adenine-based purines exert multiple effects on pain transmission. However, less attention has been given to the potential effects of guanine-based purines on pain transmission. The aim of this study was to investigate the effects of intraperitoneal (i.p.) and oral (p.o.) administration of guanosine on mice pain models. Additionally, investigation into the mechanisms of action of guanosine, its potential toxicity and cerebrospinal fluid (CSF) purine levels were also assessed. EXPERIMENTAL APPROACH: Mice received an i.p. or p.o. administration of vehicle (0.1 mM NaOH) or guanosine (up to 240 mg x kg(-1)) and were evaluated in several pain models. KEY RESULTS: Guanosine produced dose-dependent antinociceptive effects in the hot-plate, glutamate, capsaicin, formalin and acetic acid models, but it was ineffective in the tail-flick test. Additionally, guanosine produced a significant inhibition of biting behaviour induced by i.t. injection of glutamate, AMPA, kainate and trans-ACPD, but not against NMDA, substance P or capsaicin. The antinociceptive effects of guanosine were prevented by selective and non-selective adenosine receptor antagonists. Systemic administration of guanosine (120 mg x kg(-1)) induced an approximately sevenfold increase on CSF guanosine levels. Guanosine prevented the increase on spinal cord glutamate uptake induced by intraplantar capsaicin. CONCLUSIONS AND IMPLICATIONS: This study provides new evidence on the mechanism of action of the antinociceptive effects after systemic administration of guanosine. These effects seem to be related to the modulation of adenosine A(1) and A(2A) receptors and non-NMDA glutamate receptors.


Assuntos
Analgésicos/uso terapêutico , Guanosina/uso terapêutico , Dor/tratamento farmacológico , Administração Oral , Analgésicos/administração & dosagem , Analgésicos/líquido cefalorraquidiano , Analgésicos/farmacologia , Analgésicos/toxicidade , Animais , Comportamento Animal/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Cromatografia Líquida de Alta Pressão , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Edema/tratamento farmacológico , Guanosina/administração & dosagem , Guanosina/líquido cefalorraquidiano , Guanosina/farmacologia , Guanosina/toxicidade , Injeções Intraperitoneais , Dose Letal Mediana , Masculino , Camundongos , Atividade Motora/efeitos dos fármacos , Dor/líquido cefalorraquidiano , Dor/fisiopatologia , Limiar da Dor/efeitos dos fármacos , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo
7.
Arch Pharm Res ; 31(10): 1347-53, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18958427

RESUMO

A 1:1 mixture of acriflavine (ACF; CAS 8063-24-9) and guanosine is under evaluation in preclinical studies as a possible antitumor agent. Guanosine is known to potentiate the anti-cancer activity of ACF. We therefore investigated the pharmacokinetics of guanosine following administration of the ACF/guanosine mixture in rats. Rats were given guanosine (1 or 5 mg/kg) or ACF/guanosine (2 or 10 mg/kg) by i.v. bolus; or guanosine (3 or 15 mg/kg) or ACF/guanosine (6 or 30 mg/kg) by i.m. injection. We found that guanosine was rapidly cleared from the blood and transferred to tissues after i.m. administration of ACF/guanosine. The mean plasma half-lives (t(1/2)) at the alpha and beta phases were 0.091 and 6.86 h, or 0.09 and 7.51 h at a dose of 1 or 5 mg/kg guanosine, respectively. ACF had no effect on the plasma disappearance of guanosine following either i.v. bolus or i.m. administration of the combination mixture. Moreover, the ACF combination with guanosine did not significantly alter the values of MRT, V(dss), and CL(t) of guanosine. Guanosine exhibited linear pharmacokinetics over the dose range from 1 to 5 mg/kg for i.v. doses and 3 to 15 mg/kg for i.m. doses. The bioavailability of guanosine after i.m. administration was 84% for 3 mg/kg dose and 88% for 15 mg/kg dose. ACF had no effects on biliary and urinary excretion of guanosine after i.m. administration. The cumulative amount of guanosine in urine after i.m. administration was about 5-fold larger than that in bile, indicating that guanosine is mostly excreted into the urine. Guanosine was widely distributed in all tissues examined in this study, but was most highly concentrated in the kidney after i.m. administration, followed by slow excretion to bile or urine. ACF had no effect on the tissue distribution of guanosine following i.m. administration. These characterizations of the pharmacokinetics of guanosine after administration of the ACF/guanosine combination will be useful in providing preclinical and clinical bases for the potential application of this combination to the treatment of cancer.


Assuntos
Acriflavina , Antineoplásicos , Guanosina/farmacocinética , Animais , Área Sob a Curva , Combinação de Medicamentos , Guanosina/administração & dosagem , Indicadores e Reagentes , Injeções Intramusculares , Injeções Intravenosas , Masculino , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual
8.
Brain Res ; 1234: 50-8, 2008 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-18708036

RESUMO

It is well known that adenine-based purines exert multiple effects on pain transmission. However, less attention has been given to the potential effects of guanine-based purines (GBPs) on pain transmission. The aim of this study was to investigate the effects of intracerebroventricular (i.c.v.) guanosine and GMP on mice pain models. Mice received an i.c.v. injection of vehicle (saline or 10 muM NaOH), guanosine (5 to 400 nmol), or GMP (240 to 960 nmol). Additional groups were also pre-treated with i.c.v. injection of the A(1)/A(2A) antagonist caffeine (15 nmol), the non-selective opioid antagonist naloxone (12.5 nmol), or the 5'-nucleotidase inhibitor AOPCP (1 nmol). Measurements of CSF purine levels and cortical glutamate uptake were performed after treatments. Guanosine and GMP produced dose-dependent antinociceptive effects. Neither caffeine nor naloxone affected guanosine antinociception. Pre-treatment with AOPCP completely prevented GMP antinociception, indicating that conversion of GMP to guanosine is required for its antinociceptive effects. Intracerebroventricular administration of guanosine and GMP induced, respectively, a 180- and 1800-fold increase on CSF guanosine levels. Guanosine was able to prevent the decrease on cortical glutamate uptake induced by intraplantar capsaicin. This study provides new evidence on the mechanism of action of GBPs, with guanosine and GMP presenting antinociceptive effects in mice. This effect seems to be independent of adenosine and opioid receptors; it is, however, at least partially associated with modulation of the glutamatergic system by guanosine.


Assuntos
Analgésicos , Guanosina Monofosfato/farmacologia , Guanosina/farmacologia , Animais , Química Encefálica/efeitos dos fármacos , Capsaicina , Cromatografia Líquida de Alta Pressão , Ácido Glutâmico/metabolismo , Guanosina/administração & dosagem , Guanosina/líquido cefalorraquidiano , Guanosina Monofosfato/administração & dosagem , Guanosina Monofosfato/líquido cefalorraquidiano , Temperatura Alta , Injeções Intraventriculares , Masculino , Camundongos , Atividade Motora/efeitos dos fármacos , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Medição da Dor/efeitos dos fármacos , Equilíbrio Postural/efeitos dos fármacos , Desempenho Psicomotor/efeitos dos fármacos , Tempo de Reação/efeitos dos fármacos
9.
Arch Pharm Res ; 30(3): 372-80, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17424946

RESUMO

Preclinical studies are currently underway to examine the potential antitumor effects of a 1:1 mixture of acriflavine (ACF; CAS 8063-24-9) and guanosine. Guanosine potentiates the anticancer activity of some compounds. However, the effects of guanosine on the pharmacokinetics of ACF in mammals are unknown. Therefore, this study investigated the effects of guanosine on the pharmacokinetics of ACF after administering a 1:1 mixture of ACF and guanosine in rats. The rats were given either 10 mg/kg of the mixture or 5 mg/kg ACF via an intravenous bolus injection; or 30 mg/kg of the mixture or 15 mg/kg ACF intramuscularly. An HPLC-based method, which was validated in this laboratory, was used to analyze the levels of trypaflavine (TRF) and proflavine (PRF) in the plasma, bile, urine, and tissue homogenates. It was found that TRF and PRF were rapidly cleared from the blood and transferred to the tissues after the i.v. bolus or i.m. injection of the combination mixture. Both TRF and PRF were found to be most highly concentrated in the kidneys after the i.v. bolus or i.m. injection, followed by slow excretion to the bile or urine. Guanosine had no effect on the plasma disappearance of TRF or PRF after the i.v. bolus injection. However, guanosine led to a prolongation of the plasma levels of PRF after the i.m. administration of the combination mixture, resulting in a 2 fold increase in the bioavailability (BA) of PRF The concentrations of TRF and PRF in all the tissues examined were similar in the groups given the mixture and ACF. However, guanosine led to a prolongation of the biliary and urinary excretions of both TRF and PRF after the i.v. bolus (1.25 fold) or i.m. (1.5-2.4 folds) injection. These prolonged effects of guanosine on the plasma disappearance or urinary excretion of TRF and PRF might be one reason for the enhanced antitumor effects of ACF. However, more study will be needed to further examine this potential mechanism.


Assuntos
Acriflavina/administração & dosagem , Acriflavina/farmacocinética , Antineoplásicos/administração & dosagem , Guanosina/administração & dosagem , Guanosina/farmacologia , Animais , Bile/metabolismo , Cromatografia Líquida de Alta Pressão , Combinação de Medicamentos , Masculino , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual
10.
Xenobiotica ; 35(7): 755-73, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16323364

RESUMO

Acriflavine (ACF; CAS 8063-24-9), a mixture of trypaflavine (TRF) and proflavine (PRF) at a ratio of 2:1 is being investigated in rodents as an anticancer agent. However, its pharmacokinetics have not been investigated in mammals. Guanosine is known to potentiate the anticancer activity of some compounds. The pharmacokinetics of AG60, a 1:1 mixture of ACF and guanosine, were therefore investigated in rats. Rats were given 2 or 10 mg kg(-1) AG60 by intravenous bolus or 6 or 30 mg kg (-1) intramuscularly. An HPLC-based method was developed to analyse the levels of TRF, PRF, and their metabolites in plasma, bile, urine and tissue homogenates. The plasma concentrations of TRF and PRF decreased rapidly after intravenous administration and more slowly after intramuscular administration. Both TRF and PRF were distributed widely, most notably in the kidney, and were eliminated slowly. Three glucuronosyl conjugate metabolite peaks were tentatively identified in the bile. The intramuscular route leads to a prolongation of TRF or PRF plasma levels, and the systemic exposures for both TRF and PRF were both relatively high. These observations indicate that the intramuscular route may be the best way to administer AG60 for various clinical applications.


Assuntos
Acriflavina/farmacocinética , Anti-Infecciosos Locais/farmacocinética , Antineoplásicos/farmacocinética , Guanosina/farmacocinética , Acriflavina/administração & dosagem , Acriflavina/metabolismo , Animais , Anti-Infecciosos Locais/administração & dosagem , Anti-Infecciosos Locais/metabolismo , Antineoplásicos/administração & dosagem , Antineoplásicos/metabolismo , Guanosina/administração & dosagem , Guanosina/metabolismo , Injeções Intramusculares/métodos , Injeções Intravenosas/métodos , Masculino , Ratos , Ratos Sprague-Dawley
11.
Biochem Biophys Res Commun ; 336(2): 625-33, 2005 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-16143305

RESUMO

Mature B-2 cells expressing surface IgM and IgD proliferate upon stimulation by CD38, CD40 or lipopolysaccharide (LPS) and differentiate into IgG1-producing plasma cells in the presence of cytokines. The process of class switch recombination (CSR) from IgM to other isotypes is highly regulated by cytokines and activation-induced cytidine deaminase (AID). Blimp-1 and XBP-1 play an essential role in the terminal differentiation of switched B-2 cells to Ig-producing plasma cells. IL-5 induces AID and Blimp-1 expression in CD38- and CD40-activated B-2 cells, leading to mu to gamma1 CSR at DNA level and IgG1 production. IL-4, a well-known IgG1-inducing factor, does not induce mu to gamma1 CSR in CD38-activated B-2 cells or Blimp-1, while IL-4 induces mu to gamma1 CSR, XBP-1 expression, and IgG1 production expression in CD40-activated B-2 cells. Interestingly, the addition of 8-mercaptoguanosine (8-SGuo) with IL-4 to the culture of CD38-activated B cells can induce mu to gamma1 CSR, Blimp-1 expression, and IgG1 production. Intriguingly, 8-SGuo by itself induces AID expression in CD38-activated B cells. However, it does not induce mu to gamma1 CSR. These results imply that the mode of B-cell activation for extracellular stimulation affects the outcome of cytokine stimulation with respect to the efficiency and direction of CSR, and the requirements of the transcriptional regulator and the generation of antibody-secreting cells. Furthermore, our data suggest the requirement of additional molecules in addition to AID for CSR.


Assuntos
ADP-Ribosil Ciclase/imunologia , Antígenos CD/imunologia , Linfócitos B/imunologia , Guanosina/análogos & derivados , Switching de Imunoglobulina/efeitos dos fármacos , Switching de Imunoglobulina/imunologia , Imunoglobulina G/imunologia , Interleucina-4/administração & dosagem , Tionucleosídeos/administração & dosagem , ADP-Ribosil Ciclase 1 , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Feminino , Guanosina/administração & dosagem , Glicoproteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL
13.
Neurobiol Learn Mem ; 81(2): 137-43, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14990233

RESUMO

Extracellular guanine-based purines, mainly the nucleoside guanosine, have recently been shown to exert neuroprotective effects, which seem to be related to antagonism of the glutamatergic system. In this study, we investigated the effects of acute oral administration of guanosine on inhibitory avoidance task in rats and mice. We also studied its effects on locomotor activity, anxiety-related behaviors and mechanisms of action involving the purinergic system. Guanosine (2.0 and 7.5mg/kg, per os), administered 75min pretraining, dose-dependently impaired retention of the inhibitory avoidance task in rats and mice, an effect not prevented by the adenosine receptor antagonist caffeine. Guanosine presented no effects on locomotor activity and anxiety-related behaviors. This amnesic effect of guanosine may be compatible with inhibition of glutamatergic system and seems to be not mediated by adenosine.


Assuntos
Aprendizagem da Esquiva/efeitos dos fármacos , Guanosina/efeitos adversos , Inibição Psicológica , Aprendizagem em Labirinto/efeitos dos fármacos , Administração Oral , Animais , Comportamento Animal/efeitos dos fármacos , Guanosina/administração & dosagem , Masculino , Camundongos , Ratos , Ratos Wistar , Receptores de AMPA/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores
14.
Leuk Lymphoma ; 33(1-2): 147-53, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10194132

RESUMO

Fludarabine has shown a definite clinical activity in B-cell chronic lymphocytic leukemia (B-CLL). Recently it has been demonstrated that loxoribine, a guanine ribonucleotide derivative, is able to increase the cytotoxicity of fludarabine in B-CLL cells, in vitro. We have here extended these findings by testing the activity of loxoribine in combination with fludarabine and mafosfamide. As we have previously demonstrated, loxoribine enhances the activity of fludarabine at all concentrations, while only lower doses of mafosfamide seem to be positively affected by loxoribine. The combination of fludarabine and mafosfamide is synergistic on CLL cells, and the cytotoxic activity is increased by the addition of loxoribine. We have also evaluated the pro-apoptotic activity of each drug, both alone and in combination; these results are concordant with the cytotoxicity data, thus demonstrating that, even though loxoribine is more active in combination with fludarabine than with mafosfamide, the efficacy of the triple combination is higher than that obtained with any other agent alone or in double combination.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/toxicidade , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/toxicidade , Idoso , Idoso de 80 Anos ou mais , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Apoptose , Divisão Celular/efeitos dos fármacos , Ciclofosfamida/administração & dosagem , Ciclofosfamida/análogos & derivados , Ciclofosfamida/toxicidade , Ensaios de Seleção de Medicamentos Antitumorais , Quimioterapia Combinada , Feminino , Guanosina/administração & dosagem , Guanosina/análogos & derivados , Guanosina/toxicidade , Humanos , Leucemia Linfocítica Crônica de Células B/patologia , Masculino , Pessoa de Meia-Idade , Células Tumorais Cultivadas , Vidarabina/administração & dosagem , Vidarabina/análogos & derivados , Vidarabina/toxicidade
15.
Antiviral Res ; 35(3): 187-93, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9298758

RESUMO

9-(beta-D-1,3-Dioxolan-4-yl)guanine (DXG) exhibits potent antiviral activity against human immunodeficiency virus type 1 (HIV-1) and hepatitis B virus (HBV) in vitro. However, since DXG possesses limited aqueous solubility, a more water soluble prodrug of DXG, 9-(beta-D-1,3-dioxolan-4-yl)-2-aminopurine (APD), was synthesized. The purpose of this study was to characterize the pharmacokinetics of APD and its antiviral metabolite DXG in mice. Female NIH-Swiss mice were administered 100 mg/kg APD intravenously or orally. Serum, brain and liver were collected at selected times following prodrug administration and concentrations of APD and DXG were determined by HPLC. APD was efficiently converted to parent nucleoside DXG following both intravenous and oral administration. Biotransformation of APD to DXG likely occurs in the liver and is mediated by xanthine oxidase. Similar pharmacokinetic profiles for DXG were observed following either route of administration in serum, liver and brain. These results demonstrate that APD appears to be a promising prodrug for the delivery of DXG.


Assuntos
2-Aminopurina/análogos & derivados , Dioxolanos/metabolismo , Dioxolanos/farmacologia , Dioxolanos/farmacocinética , Guanina/análogos & derivados , Guanosina/análogos & derivados , Pró-Fármacos/farmacocinética , Purinas/farmacocinética , 2-Aminopurina/metabolismo , 2-Aminopurina/farmacologia , Administração Oral , Alopurinol/farmacologia , Animais , Biotransformação , Química Encefálica , Dioxolanos/administração & dosagem , Dioxolanos/sangue , Dioxolanos/química , Interações Medicamentosas , Feminino , Guanina/química , Guanina/metabolismo , Guanina/farmacologia , Guanosina/administração & dosagem , Guanosina/sangue , Guanosina/farmacocinética , Injeções Intravenosas , Fígado/química , Fígado/efeitos dos fármacos , Camundongos , Purinas/administração & dosagem , Purinas/sangue , Xantina Oxidase/efeitos dos fármacos , Xantina Oxidase/metabolismo
16.
Leuk Lymphoma ; 26(3-4): 343-8, 1997 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9322897

RESUMO

Purine analogues like fludarabine have been shown to be superior to conventional therapy for B-cell chronic lymphocytic leukemia (B-CLL). In order to improve the activity of fludarabine, we tested its combination with loxoribine, a guanine ribonucleotide derivative, known to enhance the sensitivity of B-CLL cells to cytotoxic drugs. B-CLL cells from 6 patients were studied; co-incubation with loxoribine 100 microM increased the activity of fludarabine by 12% to 48%, as demonstrated by XTT colorimetric assay; while 1000 microM loxoribine exerted a protective effect. Accordingly, fludarabine-induced apoptosis was enhanced by the addition of loxoribine 1000 microM (39% increase). These results indicate that the combination of loxoribine and fludarabine could be of interest in B-CLL.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Idoso , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Interações Medicamentosas , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Guanosina/administração & dosagem , Guanosina/análogos & derivados , Humanos , Leucemia Linfocítica Crônica de Células B/patologia , Masculino , Pessoa de Meia-Idade , Células Tumorais Cultivadas , Vidarabina/administração & dosagem , Vidarabina/análogos & derivados
17.
J Pharm Pharmacol ; 49(2): 216-22, 1997 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9055199

RESUMO

The anti-tumour activity of acriflavine in combination with guanosine has been evaluated in solid or ascitic tumour-implanted animal models. Guanosine is known to potentiate the anti-tumour effects of some chemotherapeutic agents. Administration of acriflavine (15 mg kg-1 day-1, i.m., 14 days) to ICR mice subcutaneously implanted with Ehrlich carcinoma resulted in approximately 30% inhibition in tumour growth. In contrast, minor tumour growth inhibition was observed in animals treated with guanosine at the same daily dose. Treatment of animals with both acriflavine and guanosine (AG60, 1:1, w/w) at 30 mg kg-1 resulted in approximately 65% inhibition in tumour growth rate. Whereas treatment with acriflavine or guanosine resulted in 70% or 30% decrease in tumour weight, respectively, treatment of tumour-implanted mice with AG60 (30 mg kg-1) resulted in a 96% decrease in tumour weight, relative to control, 14 days after tumour-cell implantation. Dose-related inhibition in tumour growth rate was also observed in animals treated with AG60, with maximum (65%) inhibition noted at a dose of 30 mg kg-1 (ED50 23 mg kg-1). Suppression of body weight increase and elevated plasma glucose levels by acriflavine or AG60 indicated that glucose utilization might be impaired. The anti-tumour effect of AG60 was also determined in CDF1 mice intraperitoneally implanted with Ehrlich ascitic tumour. Ehrlich ascitic tumour proliferation was completely suppressed by AG60 (30 mg kg-1, i.p.). Microscopic analyses of intraperitoneal touch-prints revealed that AG60 was more effective in suppressing tumour proliferation than acriflavine alone. Fluorescent microscopic examinations demonstrated that acriflavine avidly bound with Yac-1 cell plasma membrane, leading to morphological changes in the cells, such as bleb formations, swelling and ballooning. The time-related changes in tumour cell morphology by acriflavine or AG60 might represent energy depletion, followed by osmotic lysis as a result of cationic influx. Enhanced anti-tumour activity of acriflavine in combination with guanosine might be explained by the blocking of nutrient transport through selective acriflavine binding with plasma membrane and by concomitant guanosine perturbation of cellular ATP production. This study demonstrates that guanosine enhances the anti-tumour effects of acriflavine against a variety of cancer cells without serious adverse effects, providing a preclinical basis for potential application of this combination against cancer proliferation.


Assuntos
Acriflavina/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Guanosina/uso terapêutico , Acriflavina/administração & dosagem , Animais , Glicemia/efeitos dos fármacos , Carcinoma de Ehrlich/tratamento farmacológico , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Guanosina/administração & dosagem , Leucemia P388 , Masculino , Camundongos , Camundongos Endogâmicos ICR , Células Tumorais Cultivadas/efeitos dos fármacos
18.
Br J Cancer ; 76(9): 1157-62, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9365163

RESUMO

The exposure of cells to O6-benzyl-N2-acetylguanosine (BNAG) and several guanine derivatives is known to reduce the activity of O6-alkylguanine-DNA alkyltransferase (MGMT) and to enhance the sensitivity of Mer+ (methyl enzyme repair positive) tumour cells to chloroethylnitrosoureas (CENUs) in vitro and in vivo. High water solubility and the pharmacokinetic properties of BNAG make it a candidate for simultaneous administration with CENUs by the i.v. route in human clinical use. In vivo we have shown previously that BNAG significantly increases the efficiency of N'-[2-chloroethyl]-N-[2-(methylsulphonyl)ethyl]-N'-nitrosourea (cystemustine) against M4Beu melanoma cells (Mer+) through its cytostatic activity by the i.p. route, but also increases its toxicity. To investigate the toxicity of BNAG and cystemustine when administered simultaneously in mice, we compared the maximum tolerated dose and LD50 doses of cystemustine alone or in combination with 40 mg kg(-1) BNAG by the i.p. route. The toxicity of cystemustine was enhanced by a factor of almost 1.44 when combined with BNAG. To compare the therapeutic index of cystemustine alone and the cystemustine/BNAG combination, pharmacological tests were carried out in nude mice bearing Mer+ M4Beu human melanoma cells. Isotoxic doses were calculated using the 1.44 ratio. The treatments were administered three times by the i.v. route on days 1, 5 and 9 after s.c. inoculation of tumour cells. Although the toxicities of the treatments were equal, BNAG strongly enhanced tumour growth inhibition. These results demonstrate the increase of the therapeutic index of cystemustine by BNAG and justify the use of BNAG to enhance nitrosourea efficiency in vivo by i.v. co-injection.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Guanosina/análogos & derivados , Melanoma/tratamento farmacológico , Compostos de Nitrosoureia/administração & dosagem , Animais , Feminino , Guanosina/administração & dosagem , Guanosina/efeitos adversos , Humanos , Injeções Intravenosas , Camundongos , Camundongos Nus , Transplante de Neoplasias , Compostos de Nitrosoureia/efeitos adversos , Fatores de Tempo , Células Tumorais Cultivadas
19.
Int J Cancer ; 56(6): 892-9, 1994 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-7907081

RESUMO

Benzamide riboside exhibits significant cytotoxicity against a variety of human tumor cells in culture. On the basis of metabolic studies, the primary target of this drug's action appears to be IMP dehydrogenase (IMPDH). Incubation of human myelogenous leukemia K562 cells with an IC50 concentration of benzamide riboside resulted in an expansion of IMP pools (5.9-fold), with a parallel reduction in the concentration of GMP (90%), GDP (63%), GTP (55%) and dGTP (40%). On kinetic grounds, it was deduced that benzamide riboside (whose Ki versus IMPDH is 6.4 mM, while that of its 5'-monophosphate is 3.9 mM) or its 5'-monophosphate were unlikely to be responsible for inhibition of this target enzyme, IMPDH, since only micromolar concentrations of benzamide riboside were needed to exert potent inhibition of tumor-cell growth. Studies on the metabolism of this C-nucleoside have revealed the presence of a new peak eluting in the nucleoside diphosphate area on HPLC. Treatment of this peak with venom phosphodiesterase degraded it and concurrently nullified its inhibitory activity versus IMPDH; alkaline phosphatase, on the other hand, totally failed to digest the anabolite. These results suggest that the metabolite in question is the phosphodiester, benzamide adenine dinucleotide (BAD). Evidence that the inhibitor was an analog of NAD, wherein the nicotinamide moiety has been replaced by benzamide, was provided by both NMR and mass spectrometric analysis and confirmed by enzymatic synthesis. Further insight into the nature of the active principle was obtained from kinetic studies, which established that BAD competitively inhibited NAD utilization by partially purified IMPDH from K562 cells with a Ki of 0.118 microM. In concert, these studies establish that benzamide riboside exhibits potent antiproliferative activity by inhibiting IMPDH through BAD.


Assuntos
IMP Desidrogenase/antagonistas & inibidores , Nucleosídeos/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Neoplasias do Colo/tratamento farmacológico , Ensaios de Seleção de Medicamentos Antitumorais , Guanosina/administração & dosagem , Guanosina/farmacologia , Humanos , Leucemia Mieloide/tratamento farmacológico , Leucemia Mieloide/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Espectroscopia de Ressonância Magnética , Espectrometria de Massas , Nucleosídeos/química , Nucleosídeos/metabolismo , Nucleotídeos/metabolismo , Ribonucleotídeos/metabolismo , Células Tumorais Cultivadas
20.
Cell Immunol ; 147(2): 302-12, 1993 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-8453674

RESUMO

Guanine ribonucleosides which have been substituted at the N7 and/or C8 positions have been shown previously to activate natural killer (NK) cells and to act as sparing agents for interleukin-2 (IL-2) in the in vitro generation of lymphokine activated killer (LAK) cells. In this paper we examined a disubstituted guanosine, 7-allyl-8-oxoguanosine (loxoribine), for the ability to activate NK cells and to interact with IL-2 in the generation of LAK cells in vivo. Following iv administration, loxoribine enhanced murine splenic NK activity in a dose-related fashion, with optimal responses occurring at 3 mg/mouse. Enhanced lysis of YAC-1 cells was seen within 6 hr of injection and NK activity remained elevated for over 96 hr. Mature B and T cells were not required for NK activation since SCID mice responded to loxoribine within the same dose range as did the normal, immunocompetent mice. Both effector and precursor cells were eliminated by the administration of anti-asialo GM1 antibodies and NK activation was totally blocked in mice injected with anti-NK 1.1 antibodies. To test whether loxoribine would act as a sparing agent for IL-2 stimulated LAK activation, mice were injected with 2 mg loxoribine followed by twice daily administration of 10,000 units IL-2. In assays performed 48, 72, and 96 hr after injection of loxoribine, the cytolytic activity with the combination therapy exceeded the activity expected from the algebraic sum of the responses to the individual agents. Single injections of 2 mg loxoribine and 25,000 units IL-2 also stimulated NK/LAK activity, but the greatest enhancement was seen when loxoribine was administered 24 hr before the IL-2. Analysis of mRNA transcripts for the alpha chain of the IL-2 receptor indicated that gene transcription was enhanced within hours of loxoribine administration.


Assuntos
Citotoxicidade Imunológica/efeitos dos fármacos , Guanosina/análogos & derivados , Células Matadoras Naturais/efeitos dos fármacos , Ativação Linfocitária/efeitos dos fármacos , Animais , Antígenos/imunologia , Antígenos Ly , Antígenos de Superfície , Esquema de Medicação , Gangliosídeo G(M1)/imunologia , Expressão Gênica , Guanosina/administração & dosagem , Interleucina-2/administração & dosagem , Células Matadoras Ativadas por Linfocina/imunologia , Lectinas Tipo C , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Subfamília B de Receptores Semelhantes a Lectina de Células NK , Proteínas/imunologia , RNA Mensageiro/genética , Receptores de Interleucina-2/metabolismo , Baço/citologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA