Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 95
1.
J Food Sci ; 87(11): 5118-5127, 2022 Nov.
Article En | MEDLINE | ID: mdl-36250495

Hyperuricemia is a well-known cause of gout and also a risk factor for various comorbidities. Current agents like xanthine oxidase inhibitors prevent hyperuricemia, but usually induce severe side effects. Alternative strategies, such as novel dietary supplementations, are necessary for the management of hyperuricemia. Lactic acid bacteria (LAB) have been used in human diet for a long time with a good safety record. In this study, 345 LAB strains isolated from traditional fermented dairy products were tested for assimilating abilities of guanosine. Two LAB strains, Lacticaseibacillus rhamnosus 1155 (LR1155) and Limosilactobacillus fermentum 2644 (LF2644), showing great capacities of guanosine transformation and degradation were selected. Compared to LR1155, LF2644 showed a better effect with 100.00% transforming rate and 55.10% degrading rate. In an in vivo test, a hyperuricemic rat model was established and the results showed that administration of LR1155 (p < 0.01) or LF2644 (p < 0.01) prevented the rise of serum uric acid with more than 20% decrease when compared with the hyperuricemia rats. In addition, an increased fecal uric acid level was observed in LF2644 or LR1155 treated rats (LR1155-M p < 0.05, others p < 0.01). This study proved that LR1155 and LF2644 can be promising candidates of dietary supplements for prevention or improvement of hyperuricemia. PRACTICAL APPLICATION: The LAB strains tested in this study could be considered as good potential probiotic candidates for dietary supplements because of their urate-lowering effects, which provide a novel antihyperuricemic strategy with advantages of safety and sustainability.


Cultured Milk Products , Hyperuricemia , Lactobacillales , Humans , Rats , Animals , Uric Acid/metabolism , Uric Acid/therapeutic use , Hyperuricemia/drug therapy , Hyperuricemia/metabolism , Lactobacillales/metabolism , Xanthine Oxidase , Guanosine/therapeutic use
2.
Acta Neuropathol Commun ; 10(1): 150, 2022 10 23.
Article En | MEDLINE | ID: mdl-36274161

Diffuse intrinsic pontine glioma (DIPG) is an aggressive incurable brainstem tumor that targets young children. Complete resection is not possible, and chemotherapy and radiotherapy are currently only palliative. This study aimed to identify potential therapeutic agents using a computational pipeline to perform an in silico screen for novel drugs. We then tested the identified drugs against a panel of patient-derived DIPG cell lines. Using a systematic computational approach with publicly available databases of gene signature in DIPG patients and cancer cell lines treated with a library of clinically available drugs, we identified drug hits with the ability to reverse a DIPG gene signature to one that matches normal tissue background. The biological and molecular effects of drug treatment was analyzed by cell viability assay and RNA sequence. In vivo DIPG mouse model survival studies were also conducted. As a result, two of three identified drugs showed potency against the DIPG cell lines Triptolide and mycophenolate mofetil (MMF) demonstrated significant inhibition of cell viability in DIPG cell lines. Guanosine rescued reduced cell viability induced by MMF. In vivo, MMF treatment significantly inhibited tumor growth in subcutaneous xenograft mice models. In conclusion, we identified clinically available drugs with the ability to reverse DIPG gene signatures and anti-DIPG activity in vitro and in vivo. This novel approach can repurpose drugs and significantly decrease the cost and time normally required in drug discovery.


Astrocytoma , Brain Stem Neoplasms , Diffuse Intrinsic Pontine Glioma , Glioma , Humans , Mice , Animals , Diffuse Intrinsic Pontine Glioma/drug therapy , Diffuse Intrinsic Pontine Glioma/genetics , Mycophenolic Acid/therapeutic use , Glioma/drug therapy , Glioma/genetics , Glioma/metabolism , Brain Stem Neoplasms/drug therapy , Brain Stem Neoplasms/genetics , Brain Stem Neoplasms/pathology , Gene Expression , Guanosine/therapeutic use
3.
Antimicrob Agents Chemother ; 66(10): e0100822, 2022 10 18.
Article En | MEDLINE | ID: mdl-36094205

Human metapneumovirus (HMPV) is recognized as an important cause of pneumonia in infants, in the elderly, and in immunocompromised individuals worldwide. The absence of an antiviral treatment or vaccine strategy against HMPV infection creates a high burden on the global health care system. Drug repurposing has become increasingly attractive for the treatment of emerging and endemic diseases as it requires less research and development costs than traditional drug discovery. In this study, we developed an in vitro medium-throughput screening assay that allows for the identification of novel anti-HMPV drugs candidates. Out of ~2,400 compounds, we identified 11 candidates with a dose-dependent inhibitory activity against HMPV infection. Additionally, we further described the mode of action of five anti-HMPV candidates with low in vitro cytotoxicity. Two entry inhibitors, Evans Blue and aurintricarboxylic acid, and three post-entry inhibitors, mycophenolic acid, mycophenolate mofetil, and 2,3,4-trihydroxybenzaldehyde, were identified. Among them, the mycophenolic acid series displayed the highest levels of inhibition, due to the blockade of intracellular guanosine synthesis. Importantly, MPA has significant potential for drug repurposing as inhibitory levels are achieved below the approved human oral dose. Our drug-repurposing strategy proved to be useful for the rapid discovery of novel hit candidates to treat HMPV infection and provide promising novel templates for drug design.


Metapneumovirus , Paramyxoviridae Infections , Infant , Humans , Aged , Drug Repositioning , Mycophenolic Acid , Evans Blue/therapeutic use , Aurintricarboxylic Acid/therapeutic use , Paramyxoviridae Infections/drug therapy , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Guanosine/therapeutic use
4.
Antimicrob Agents Chemother ; 65(11): e0098821, 2021 10 18.
Article En | MEDLINE | ID: mdl-34424050

Every year, millions of people worldwide are infected with dengue virus (DENV), with a significant number developing severe life-threatening disease. There are currently no broadly indicated vaccines or therapeutics available for treatment of DENV infection. Here, we show that AT-281, the free base of AT-752, an orally available double prodrug of a guanosine nucleotide analog, was a potent inhibitor of DENV serotypes 2 and 3 in vitro, requiring concentrations of 0.48 and 0.77 µM, respectively, to inhibit viral replication by 50% (EC50) in Huh-7 cells. AT-281 was also a potent inhibitor of all other flaviviruses tested, with EC50 values ranging from 0.19 to 1.41 µM. Little to no cytotoxicity was observed for AT-281 at concentrations up to 170 µM. After oral administration of AT-752, substantial levels of the active triphosphate metabolite AT-9010 were formed in vivo in peripheral blood mononuclear cells of mice, rats, and monkeys. Furthermore, AT-9010 competed with GTP in RNA template-primer elongation assays with DENV2 RNA polymerase, which is essential for viral replication, with incorporation of AT-9010 resulting in termination of RNA synthesis. In AG129 mice infected with DENV D2Y98P, treatment with AT-752 significantly reduced viremia and morbidity and increased survival. The demonstrated in vitro and in vivo activity of AT-752 suggests that it is a promising compound for the treatment of dengue virus infection and is currently under evaluation in clinical studies.


Dengue Virus , Dengue , Flavivirus , Prodrugs , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Dengue/drug therapy , Guanosine/pharmacology , Guanosine/therapeutic use , Leukocytes, Mononuclear , Mice , Nucleotides/therapeutic use , Prodrugs/pharmacology , Prodrugs/therapeutic use , Rats , Virus Replication
5.
Purinergic Signal ; 17(2): 255-271, 2021 06.
Article En | MEDLINE | ID: mdl-33834349

Ischemic stroke is a major cause of morbidity and mortality worldwide and only few affected patients are able to receive treatment, especially in developing countries. Detailed pathophysiology of brain ischemia has been extensively studied in order to discover new treatments with a broad therapeutic window and that are accessible to patients worldwide. The nucleoside guanosine (Guo) has been shown to have neuroprotective effects in animal models of brain diseases, including ischemic stroke. In a rat model of focal permanent ischemia, systemic administration of Guo was effective only when administered immediately after stroke induction. In contrast, intranasal administration of Guo (In-Guo) was effective even when the first administration was 3 h after stroke induction. In order to validate the neuroprotective effect in this larger time window and to investigate In-Guo neuroprotection under global brain dysfunction induced by ischemia, we used the model of thermocoagulation of pial vessels in Wistar rats. In our study, we have found that In-Guo administered 3 h after stroke was capable of preventing ischemia-induced dysfunction, such as bilateral suppression and synchronicity of brain oscillations and ipsilateral cell death signaling, and increased permeability of the blood-brain barrier. In addition, In-Guo had a long-lasting effect on preventing ischemia-induced motor impairment. Our data reinforce In-Guo administration as a potential new treatment for brain ischemia with a more suitable therapeutic window.


Brain/physiopathology , Guanosine/administration & dosage , Guanosine/therapeutic use , Ischemic Stroke/drug therapy , Ischemic Stroke/physiopathology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/therapeutic use , Administration, Intranasal , Animals , Blood-Brain Barrier/drug effects , Cell Death/drug effects , Cerebral Veins/drug effects , Electrocoagulation , Electroencephalography/drug effects , Functional Laterality/drug effects , Ischemic Stroke/complications , Male , Movement Disorders/etiology , Movement Disorders/prevention & control , Rats , Rats, Wistar , Signal Transduction/drug effects
6.
Purinergic Signal ; 16(3): 379-387, 2020 09.
Article En | MEDLINE | ID: mdl-32725400

Parkinson's disease (PD) signs and symptoms regularly include tremor. Interestingly, the nucleoside guanosine (GUO) has already proven to be effective in reducing reserpine-induced tremulous jaw movements (TJMs) in rodent models, thus becoming a promising antiparkinsonian drug. Here, we aimed at revealing the mechanism behind GUO antiparkinsonian efficacy by assessing the role of adenosine A1 and A2A receptors (A1R and A2AR) on GUO-mediated anti-tremor effects in the reserpinized mouse model of PD. Reserpinized mice showed elevated reactive oxygen species (ROS) production and cellular membrane damage in striatal slices assessed ex vivo and GUO treatment reversed ROS production. Interestingly, while the simultaneous administration of sub-effective doses of GUO (5 mg/kg) and SCH58261 (0.01 mg/kg), an A2AR antagonist, precluded reserpine-induced TJMs, these were ineffective on reverting ROS production in ex vivo experiments. Importantly, GUO was able to reduce TJM and ROS production in reserpinized mouse lacking the A2AR, thus suggesting an A2AR-independent mechanism of GUO-mediated effects. Conversely, the administration of DPCPX (0.75 mg/kg), an A1R antagonist, completely abolished both GUO-mediated anti-tremor effects and blockade of ROS production. Overall, these results indicated that GUO anti-tremor and antioxidant effects in reserpinized mice were A1R dependent but A2AR independent, thus suggesting a differential participation of adenosine receptors in GUO-mediated effects.


Guanosine/therapeutic use , Parkinson Disease, Secondary/metabolism , Receptor, Adenosine A1/metabolism , Receptor, Adenosine A2A/metabolism , Tremor/metabolism , Adenosine A1 Receptor Antagonists/pharmacology , Adenosine A2 Receptor Antagonists , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Guanosine/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/drug therapy , Reactive Oxygen Species/metabolism , Tremor/chemically induced , Tremor/drug therapy , Xanthines/pharmacology
7.
Brain Res Bull ; 163: 31-39, 2020 10.
Article En | MEDLINE | ID: mdl-32681970

Traumatic brain injury (TBI) constitutes a heterogeneous cerebral insult induced by traumatic biomechanical forces. Mitochondria play a critical role in brain bioenergetics, and TBI induces several consequences related with oxidative stress and excitotoxicity clearly demonstrated in different experimental model involving TBI. Mitochondrial bioenergetics alterations can present several targets for therapeutics which could help reduce secondary brain lesions such as neuropsychiatric problems, including memory loss and motor impairment. Guanosine (GUO), an endogenous neuroprotective nucleoside, affords the long-term benefits of controlling brain neurodegeneration, mainly due to its capacity to activate the antioxidant defense system and maintenance of the redox system. However, little is known about the exact protective mechanism exerted by GUO on mitochondrial bioenergetics disruption induced by TBI. Thus, the aim of this study was to investigate the effects of GUO in brain cortical and hippocampal mitochondrial bioenergetics in the mild TBI model. Additionally, we aimed to assess whether mitochondrial damage induced by TBI may be related to behavioral alterations in rats. Our findings showed that 24 h post-TBI, GUO treatment promotes an adaptive response of mitochondrial respiratory chain increasing oxygen flux which it was able to protect against the uncoupling of oxidative phosphorylation (OXPHOS) induced by TBI, restored the respiratory electron transfer system (ETS) established with an uncoupler. Guanosine treatment also increased respiratory control ratio (RCR), an indicator of the state of mitochondrial coupling, which is related to the mitochondrial functionality. In addition, mitochondrial bioenergetics failure was closely related with locomotor, exploratory and memory impairments. The present study suggests GUO treatment post mild TBI could increase GDP endogenous levels and consequently increasing ATP levels promotes an increase of RCR increasing OXPHOS and in substantial improve mitochondrial respiration in different brain regions, which, in turn, could promote an improvement in behavioral parameters associated to the mild TBI. These findings may contribute to the development of future therapies with a target on failure energetic metabolism induced by TBI.


Brain Concussion/drug therapy , Energy Metabolism/drug effects , Guanosine/therapeutic use , Locomotion/drug effects , Memory, Long-Term/drug effects , Mitochondria/drug effects , Animals , Brain Concussion/metabolism , Brain Concussion/pathology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Energy Metabolism/physiology , Guanosine/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Locomotion/physiology , Male , Memory, Long-Term/physiology , Mitochondria/metabolism , Mitochondria/pathology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Rats , Rats, Wistar
8.
Sanid. mil ; 75(3): 135-142, jul.-sept. 2019. ilus, graf, tab
Article Es | IBECS | ID: ibc-187449

Antecedentes: España por su situación geopolítica no está exenta de amenazas y desafíos asociados a enfermedades infecciosas víricas como las «fiebres hemorrágicas víricas». Implementar acciones para disminuir estos riesgos frente a agentes víricos con el desarrollo de un tratamiento de urgencia por vía intravenosa resulta de gran interés. Objetivos: Desarrollar dos formulaciones de Ribavirina 100 mg/ml con distintos excipientes y condiciones de fabricación para buscar la formulación más estable. Material y métodos: Se ha utilizado ribavirina, distintos tampones y agua para inyectables. Se ha evaluado alteraciones físicas, riqueza y pH siguiendo especificaciones de la monografía de la Farmacopea Europea. Resultados: Se exponen resultados de pH y riqueza de ambas formulaciones con y sin esterilización terminal. Conclusiones: Los resultados obtenidos cumplen especificaciones para ambas formulaciones, demostrando una estabilidad química tras su conservación en condiciones de estudios a largo plazo durante 12 meses y acelerados durante 6 meses. El desarrollo de la ribavirina 100 mg/ml (10% w/v) inyectable satisface una necesidad terapéutica relacionada con el tratamiento de primera elección en enfermedades víricas altamente contagiosas que carecen de tratamientos eficaces


Background: Spain, due to its geopolitical situation, is not exempt from the threats and challenges associated with viral infectious diseases such as the «viral hemorrhagic fevers». Implementing actions to reduce these risks with the development of an emergency treatment against these viral agents for intravenous is of great interest. Objective: Develop two formulations of Ribavirin 100 mg/ml with different excipients and manufacturing conditions to look for the most stable formulation. Material and methods: Ribavirin, various buffers and water for injections have been used. Physical alterations, richness and pH have been evaluated following specifications of European Pharmacopoeia monograp. Results: Results of pH and richness of both formulations are exposed with and without terminal sterilization. Conclusions: The results obtained meet specifications for both formulations, demonstrating chemical stability after preservation under long-term study conditions for 12 months and accelerated for 6 months. The development of injectable 100 mg/ml ribavirin (10% w/v) satisfies a therapeutic need related to the treatment of first choice in highly contagious viral diseases that lack treatments


Humans , Ribavirin/therapeutic use , Emergency Treatment/trends , Communicable Diseases/drug therapy , Guanosine/therapeutic use , Injections/methods , Antiviral Agents/therapeutic use , Ribavirin/pharmacology , Homeopathic Dosage/pharmacology
9.
Neurosci Lett ; 704: 141-144, 2019 06 21.
Article En | MEDLINE | ID: mdl-30974229

Traumatic brain injury (TBI) represents one of the leading causes of death worldwide. Its pathophysiology involves several neurochemical events including mitochondrial dysfunction. Since mitochondrial respiration plays a key role in cell survival, pharmacological interventions targeting mitochondrial function have been highlighted as a powerful tool against the neurodegenerative process triggered by TBI. Guanosine (GUO), a neuroprotective molecule in different neurological disorders involving neurotoxicity, has shown protective properties after TBI, however its mechanism of action is not well understood in the central nervous system (CNS). Therefore, the aim of this study is to evaluate the possible target receptor involved in the protective GUO effects on TBI-induced mitochondrial dysfunction in the cerebral cortex of rats. Results show that a single dose of GUO (7.5 mg/kg) injected 40 min after a fluid percussion injury (FPI) protects against loss of mitochondrial membrane potential and increase of reactive oxygen species 8 h post-TBI. These effects were specifically blocked by a pretreatment (10 min after TBI) with an A1 adenosine receptor antagonist (DPCPX 1 mg/kg). In contrast, pretreatment with an A2A adenosine receptor antagonist (SCH 58261 0.05 mg/kg) did not alter GUO effects. These findings suggest that acute GUO neuroprotection following TBI involves the modulation of the adenosinergic system, especially A1 adenosine receptor.


Brain Injuries, Traumatic/drug therapy , Guanosine/pharmacology , Mitochondria/drug effects , Neuroprotective Agents/pharmacology , Receptor, Adenosine A1/metabolism , Receptors, Adenosine A2/metabolism , Animals , Brain Injuries, Traumatic/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Guanosine/therapeutic use , Male , Membrane Potential, Mitochondrial , Mitochondria/physiology , Neuroprotective Agents/therapeutic use , Rats, Wistar , Reactive Oxygen Species/metabolism
10.
Mol Neurobiol ; 56(5): 3145-3158, 2019 May.
Article En | MEDLINE | ID: mdl-30105669

Traumatic brain injury (TBI) is a leading cause of disability worldwide, triggering chronic neurodegeneration underlying cognitive and mood disorder still without therapeutic prospects. Based on our previous observations that guanosine (GUO) attenuates short-term neurochemical alterations caused by TBI, this study investigated the effects of chronical GUO treatment in behavioral, molecular, and morphological disturbances 21 days after trauma. Rats subject to TBI displayed mood (anxiety-like) and memory dysfunction. This was accompanied by a decreased expression of both synaptic (synaptophysin) and plasticity proteins (BDNF and CREB), a loss of cresyl violet-stained neurons, and increased astrogliosis and microgliosis in the hippocampus. Notably, chronic GUO treatment (7.5 mg/kg i.p. daily starting 1 h after TBI) prevented all these TBI-induced long-term behavioral, neurochemical, and morphological modifications. This neuroprotective effect of GUO was abrogated in the presence of the adenosine A1 receptor antagonist DPCPX (1 mg/kg) but unaltered by the adenosine A2A receptor antagonist SCH58261 (0.05 mg/kg). These findings show that a chronic GUO treatment prevents the long-term mood and memory dysfunction triggered by TBI, which involves adenosinergic receptors.


Behavior, Animal/drug effects , Brain Injuries, Traumatic/drug therapy , Guanosine/therapeutic use , Receptors, Purinergic P1/metabolism , Animals , Anxiety/drug therapy , Anxiety/etiology , Biomarkers/metabolism , Brain Injuries, Traumatic/complications , Gliosis/complications , Gliosis/pathology , Guanosine/pharmacology , Hippocampus/drug effects , Hippocampus/pathology , Male , Memory Disorders/complications , Microglia/drug effects , Microglia/pathology , Models, Biological , Motor Activity/drug effects , Neuronal Plasticity/genetics , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Rats, Wistar
11.
Vet Res Commun ; 42(4): 309-313, 2018 Dec.
Article En | MEDLINE | ID: mdl-30094586

Peste des petits ruminants virus (PPRV) causes highly contagious diseases in domestic and particular wild small ruminants, leading to substantial economic loss. The development of effective and cheap antiviral medications shall help to circumvent this emerging burden. In this study, we found that ribavirin, a competitive inosine-5'-monophosphate dehydrogenase (IMPDH) inhibitor, significantly inhibits the replication of PPRV. As IMPDH is a key enzyme in purine nucleotide synthesis, supplementation of exogenous guanosine attenuate the anti-PPRV effect of ribavirin. Interestingly, an uncompetitive IMPDH inhibitor, mycophenolic acid (MPA), exerted more potent antiviral effect again PPRV. Similarly, this effect was largely restored upon supplementation of guanosine. Thus, we have demonstrated that the IMPDH inhibitors ribavirin and MPA combat PPRV infection through purine nucleotide depletion. Because both regimens have been widely used in the clinic for treating viral infection or organ rejection in transplantation patients for decades, respectively, repurposing these existing safe and cheap medications may provide a new avenue for combating PPRV infection.


Antiviral Agents/therapeutic use , Mycophenolic Acid/therapeutic use , Peste-des-Petits-Ruminants/drug therapy , Peste-des-petits-ruminants virus/drug effects , Ribavirin/therapeutic use , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/pharmacology , Chlorocebus aethiops , Drug Synergism , Drug Therapy, Combination , Guanosine/administration & dosage , Guanosine/pharmacology , Guanosine/therapeutic use , Mycophenolic Acid/administration & dosage , Mycophenolic Acid/pharmacology , Ribavirin/administration & dosage , Ribavirin/pharmacology , Vero Cells/virology , Virus Replication/drug effects
12.
Article En | MEDLINE | ID: mdl-29530841

Norovirus infections are a significant health and economic burden globally, accounting for hundreds of millions of cases of acute gastroenteritis every year. In the absence of an approved norovirus vaccine, there is an urgent need to develop antivirals to treat chronic infections and provide prophylactic therapy to limit viral spread during epidemics and pandemics. Toll-like receptor (TLR) agonists have been explored widely for their antiviral potential, and several are progressing through clinical trials for the treatment of human immunodeficiency virus (HIV) and hepatitis B virus (HBV) and as adjuvants for norovirus viruslike particle (VLP) vaccines. However, norovirus therapies in development are largely direct-acting antivirals (DAAs) with fewer compounds that target the host. Our aim was to assess the antiviral potential of TLR7 agonist immunomodulators on norovirus infection using the murine norovirus (MNV) and human Norwalk replicon models. TLR7 agonists R-848, Gardiquimod, GS-9620, R-837, and loxoribine were screened using a plaque reduction assay, and each displayed inhibition of MNV replication (50% effective concentrations [EC50s], 23.5 nM, 134.4 nM, 0.59 µM, 1.5 µM, and 79.4 µM, respectively). RNA sequencing of TLR7-stimulated cells revealed a predominant upregulation of innate immune response genes and interferon (IFN)-stimulated genes (ISGs) that are known to drive an antiviral state. Furthermore, the combination of R-848 and the nucleoside analogue (NA) 2'C-methylcytidine elicited a synergistic antiviral effect against MNV, demonstrating that combinational therapy of host modulators and DAAs might be used to reduce drug cytotoxicity. In summary, we have identified that TLR7 agonists display potent inhibition of norovirus replication and are a therapeutic option to combat norovirus infections.


Antiviral Agents/therapeutic use , Caliciviridae Infections/drug therapy , Toll-Like Receptor 7/metabolism , Aminoquinolines/therapeutic use , Animals , Cell Line , Guanosine/analogs & derivatives , Guanosine/therapeutic use , Humans , Imidazoles/therapeutic use , Imiquimod/therapeutic use , Mice , Pteridines/therapeutic use , RAW 264.7 Cells , Toll-Like Receptor 7/agonists , Virus Replication/drug effects
13.
J Pharmacol Exp Ther ; 363(3): 358-366, 2017 12.
Article En | MEDLINE | ID: mdl-28928119

8-Aminoguanosine induces diuresis, natriuresis, glucosuria, and antikaliuresis. These effects could be mediated via 8-aminoguanosine's metabolism to 8-aminoguanine. In this study, we tested this hypothesis in anesthetized rats. First, we demonstrated that at 55- to 85-minutes post-i.v. administration, 8-aminoguanosine and 8-aminoguanine (33.5 µmol/kg) significantly increased urine volume [ml/30 min: 8-aminoguanosine from 0.3 ± 0.1 to 0.9 ± 0.1 (mean ± S.E.M.; n = 7); 8-aminoguanine from 0.3 ± 0.1 to 1.5 ± 0.2 (n = 8)], sodium excretion (µmol/30 min: 8-aminoguanosine from 12 ± 5 to 109 ± 21; 8-aminoguanine from 18 ± 8 to 216 ± 31), and glucose excretion (µg/30 min: 8-aminoguanosine from 18 ± 3 to 159 ± 41; 8-aminoguanine from 17 ± 3 to 298 ± 65). Both compounds significantly decreased potassium excretion (µmol/30 min: 8-aminoguanosine from 62 ± 7 to 39 ± 9; 8-aminoguanine from 61 ± 10 to 34 ± 6). Next, we administered 8-aminoguanosine and 8-aminoguanine i.v. (33.5 µmol/kg) and measured renal interstitial (microdialysis probes) 8-aminoguanosine and 8-aminoguanine. The i.v. administration of 8-aminoguanosine and 8-aminoguanine similarly increased renal medullary interstitial levels of 8-aminoguanine [nanograms per milliliter; 8-aminoguanosine from 4 ± 1 to 1025 ± 393 (n = 6), and 8-aminoguanine from 2 ± 1 to 1069 ± 407 (n = 6)]. Finally, we determine the diuretic, natriuretic, glucosuric, and antikaliuretic effects of intrarenal artery infusions of 8-aminoguanosine and 8-aminoguanine (0.1, 0.3, and 1 µmol/kg/min). 8-Aminoguanine increased urine volume and sodium and glucose excretion by the ipsilateral kidney, yet had only mild effects at the highest dose in the contralateral kidney. Intrarenal infusions of 8-aminoguanosine did not induce diuresis, natriuresis, or glucosuria in either the ipsilateral or contralateral kidney, yet decreased potassium excretion in the ipsilateral kidney. Together these data confirm that the diuretic, natriuretic, and glucosuric effects of 8-aminoguanosine are not direct, but require metabolism to 8-aminoguanine. However, 8-aminoguanosine has direct antikaliuretic effects.


Diuretics/pharmacology , Glycosuria/urine , Guanine/analogs & derivatives , Guanosine/analogs & derivatives , Hyperkalemia/drug therapy , Natriuretic Agents/pharmacology , Animals , Diuretics/metabolism , Guanine/metabolism , Guanine/pharmacology , Guanosine/metabolism , Guanosine/pharmacology , Guanosine/therapeutic use , Hyperkalemia/metabolism , Kidney Medulla/drug effects , Kidney Medulla/metabolism , Male , Natriuretic Agents/metabolism , Rats, Sprague-Dawley , Urodynamics/drug effects
14.
Mol Neurobiol ; 54(5): 3137-3148, 2017 07.
Article En | MEDLINE | ID: mdl-27052954

The nucleoside guanosine (GUO) increases glutamate uptake by astrocytes and acts as antioxidant, thereby providing neuroprotection against glutamatergic excitotoxicity, as we have recently demonstrated in an animal model of chronic hepatic encephalopathy. Here, we investigated the neuroprotective effect of GUO in an acute ammonia intoxication model. Adult male Wistar rats received an intraperitoneal (i.p.) injection of vehicle or GUO 60 mg/kg, followed 20 min later by an i.p. injection of vehicle or 550 mg/kg of ammonium acetate. Afterwards, animals were observed for 45 min, being evaluated as normal, coma (i.e., absence of corneal reflex), or death status. In a second cohort of rats, video-electroencephalogram (EEG) recordings were performed. In a third cohort of rats, the following were measured: (i) plasma levels of glucose, transaminases, and urea; (ii) cerebrospinal fluid (CSF) levels of ammonia, glutamine, glutamate, and alanine; (iii) glutamate uptake in brain slices; and (iv) brain redox status and glutamine synthetase activity in cerebral cortex. GUO drastically reduced the lethality rate and the duration of coma. Animals treated with GUO had improved EEG traces, decreased CSF levels of glutamate and alanine, lowered oxidative stress in the cerebral cortex, and increased glutamate uptake by astrocytes in brain slices compared with animals that received vehicle prior to ammonium acetate administration. This study provides new evidence on mechanisms of guanine-derived purines in their potential modulation of glutamatergic system, contributing to GUO neuroprotective effects in a rodent model of by acute ammonia intoxication.


Ammonia/toxicity , Guanosine/pharmacology , Neuroprotective Agents/pharmacology , Animals , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Brain/metabolism , Coma/blood , Coma/cerebrospinal fluid , Coma/chemically induced , Coma/drug therapy , Disease Models, Animal , Electroencephalography , Guanosine/therapeutic use , Male , Neuroprotective Agents/therapeutic use , Oxidation-Reduction , Oxidative Stress/drug effects , Rats, Wistar
15.
Mol Neurobiol ; 54(1): 423-436, 2017 01.
Article En | MEDLINE | ID: mdl-26742520

Accumulating evidences indicate that endogenous modulators of excitatory synapses in the mammalian brain are potential targets for treating neuropsychiatric disorders. Indeed, glutamatergic and adenosinergic neurotransmissions were recently highlighted as potential targets for developing innovative anxiolytic drugs. Accordingly, it has been shown that guanine-based purines are able to modulate both adenosinergic and glutamatergic systems in mammalian central nervous system. Here, we aimed to investigate the potential anxiolytic-like effects of guanosine and its effects on the adenosinergic and glutamatergic systems. Acute/systemic guanosine administration (7.5 mg/kg) induced robust anxiolytic-like effects in three classical anxiety-related paradigms (elevated plus maze, light/dark box, and round open field tasks). These guanosine effects were correlated with an enhancement of adenosine and a decrement of glutamate levels in the cerebrospinal fluid. Additionally, pre-administration of caffeine (10 mg/kg), an unspecific adenosine receptors' antagonist, completely abolished the behavioral and partially prevented the neuromodulatory effects exerted by guanosine. Although the hippocampal glutamate uptake was not modulated by guanosine (both ex vivo and in vitro protocols), the synaptosomal K+-stimulated glutamate release in vitro was decreased by guanosine (100 µM) and by the specific adenosine A1 receptor agonist, 2-chloro-N 6-cyclopentyladenosine (CCPA, 100 nM). Moreover, the specific adenosine A1 receptor antagonist 8-cyclopentyl-1,3-dipropylxanthine (DPCPX, 100 nM) fully reversed the inhibitory guanosine effect in the glutamate release. The pharmacological modulation of A2a receptors has shown no effect in any of the evaluated parameters. In summary, the guanosine anxiolytic-like effects seem closely related to the modulation of adenosinergic (A1 receptors) and glutamatergic systems.


Adenosine A1 Receptor Antagonists/pharmacology , Adenosine/metabolism , Anti-Anxiety Agents/therapeutic use , Glutamic Acid/metabolism , Guanosine/therapeutic use , Receptor, Adenosine A1/metabolism , Animals , Anti-Anxiety Agents/pharmacology , Anxiety/drug therapy , Anxiety/metabolism , Guanosine/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Male , Neurotransmitter Agents/metabolism , Rats , Rats, Wistar
16.
Mol Neurobiol ; 54(10): 7585-7596, 2017 12.
Article En | MEDLINE | ID: mdl-27830534

Traumatic brain injury (TBI) is one of the most common types of brain injuries that cause death or persistent neurological disturbances in survivors. Most of the promising experimental drugs were not effective in clinical trials; therefore, the development of TBI drugs represents a huge unmet need. Guanosine, an endogenous neuroprotective nucleoside, has not been evaluated in TBI to the best of our knowledge. Therefore, the present study evaluated the effect of guanosine on TBI-induced neurological damage. Our findings showed that a single dose of guanosine (7.5 mg/kg, intraperitoneally (i.p.) injected 40 min after fluid percussion injury (FPI) in rats protected against locomotor and exploratory impairments 8 h after injury. The treatment also protected against neurochemical damage to the ipsilateral cortex, glutamate uptake, Na+/K+-ATPase, glutamine synthetase activity, and alterations in mitochondrial function. The inflammatory response and brain edema were also reduced by this nucleoside. In addition, guanosine protected against neuronal death and caspase 3 activation. Therefore, this study suggests that guanosine plays a neuroprotective role in TBI and can be exploited as a new pharmacological strategy.


Brain Injuries, Traumatic/prevention & control , Guanosine/therapeutic use , Inflammation Mediators/antagonists & inhibitors , Mitochondria/drug effects , Neurons/drug effects , Neuroprotective Agents/therapeutic use , Animals , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Cell Count/methods , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Guanosine/pharmacology , Inflammation/metabolism , Inflammation/pathology , Inflammation/prevention & control , Inflammation Mediators/metabolism , Male , Mitochondria/metabolism , Mitochondria/pathology , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/pharmacology , Rats , Rats, Wistar
17.
J Pharmacol Exp Ther ; 359(3): 420-435, 2016 Dec.
Article En | MEDLINE | ID: mdl-27679494

In vivo, guanine moieties in DNA, RNA, guanine nucleotides, or guanosine or guanine per se can undergo nitration (for example, by peroxynitrite) or hydroxylation (for example, by superoxide anion) on position 8 of the purine ring. Subsequent catabolism of these modified biomolecules leads to the production of a diverse group of 8-nitro, 8-amino, and 8-hydroxy guanosine and guanine compounds. Indeed, studies suggest the in vivo existence of 8-nitroguanosine, 8-nitroguanine, 8-aminoguanosine, 8-aminoguanine, 8-hydroxyguanosine, 8-hydroxy-2'-deoxyguanosine, and 8-hydroxyguanine. Since a multitude of these compounds exist in vivo, and since the renal effects of 8-substituted guanosine and guanine compounds are entirely unknown, we examined the effects of guanosine, guanine, 8-nitroguanosine, 8-nitroguanine, 8-hydroxyguanosine, 8-hydroxyguanine, 8-hydroxy-2'-deoxyguanosine, 8-aminoguanosine, and 8-aminoguanine (33.5 µmol/kg/min; intravenous infusion for 115 minutes) on excretion of sodium, potassium, and glucose in rats. Guanosine, 8-nitroguanosine, and 8-hydroxy-2'-deoxyguanosine had minimal natriuretic activity. Guanine, 8-nitroguanine, 8-hydroxyguanosine, and 8-hydroxyguanine had moderate natriuretic activity (increased sodium excretion by 9.4-, 7.8-, 7.1-, and 8.6-fold, respectively). In comparison with all other compounds, 8-aminoguanosine and 8-aminoguanine were highly efficacious and increased sodium excretion by 26.6- and 17.2-fold, respectively, exceeding that of a matched dose of amiloride (13.6-fold increase). 8-Aminoguanosine and 8-aminoguanine also increased glucose excretion by 12.1- and 12.2-fold, respectively, and decreased potassium excretion by 69.1 and 71.0%, respectively. Long-term radiotelemetry studies demonstrated that oral 8-aminoguanosine and 8-aminoguanine (5 mg/kg/day) suppressed deoxycorticosterone/salt-induced hypertension. These experiments demonstrate that some naturally occurring 8-substitued guanosine and guanine compounds, particularly 8-aminoguanosine and 8-aminoguanine, are potent and efficacious potassium-sparing diuretics/natriuretics that may represent a novel class of antihypertensive diuretics.


Antihypertensive Agents/pharmacology , Diuretics/pharmacology , Glycosuria/drug therapy , Guanine/analogs & derivatives , Guanosine/analogs & derivatives , Natriuresis/drug effects , Animals , Antihypertensive Agents/therapeutic use , Diuretics/therapeutic use , Guanine/pharmacology , Guanine/therapeutic use , Guanosine/pharmacology , Guanosine/therapeutic use , Male , Rats , Rats, Sprague-Dawley
18.
Purinergic Signal ; 12(1): 149-59, 2016 Mar.
Article En | MEDLINE | ID: mdl-26695181

In addition to its intracellular roles, the nucleoside guanosine (GUO) also has extracellular effects that identify it as a putative neuromodulator signaling molecule in the central nervous system. Indeed, GUO can modulate glutamatergic neurotransmission, and it can promote neuroprotective effects in animal models involving glutamate neurotoxicity, which is the case in brain ischemia. In the present study, we aimed to investigate a new in vivo GUO administration route (intranasal, IN) to determine putative improvement of GUO neuroprotective effects against an experimental model of permanent focal cerebral ischemia. Initially, we demonstrated that IN [(3)H] GUO administration reached the brain in a dose-dependent and saturable pattern in as few as 5 min, presenting a higher cerebrospinal GUO level compared with systemic administration. IN GUO treatment started immediately or even 3 h after ischemia onset prevented behavior impairment. The behavior recovery was not correlated to decreased brain infarct volume, but it was correlated to reduced mitochondrial dysfunction in the penumbra area. Therefore, we showed that the IN route is an efficient way to promptly deliver GUO to the CNS and that IN GUO treatment prevented behavioral and brain impairment caused by ischemia in a therapeutically wide time window.


Brain Ischemia/drug therapy , Guanosine/administration & dosage , Guanosine/therapeutic use , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/therapeutic use , Stroke/drug therapy , Administration, Intranasal , Animals , Behavior, Animal , Brain Ischemia/psychology , Cerebral Infarction/pathology , Cerebral Infarction/prevention & control , Dose-Response Relationship, Drug , Exploratory Behavior/drug effects , Guanosine/cerebrospinal fluid , Guanosine/pharmacokinetics , Male , Mitochondria/drug effects , Neuroprotective Agents/cerebrospinal fluid , Neuroprotective Agents/pharmacokinetics , Rats , Rats, Wistar , Stroke/psychology
19.
Pharmacol Biochem Behav ; 127: 7-14, 2014 Dec.
Article En | MEDLINE | ID: mdl-25316306

Guanosine is a guanine-based purine that modulates glutamate uptake and exerts neurotrophic and neuroprotective effects. In a previous study, our group demonstrated that this endogenous nucleoside displays antidepressant-like properties in a predictive animal model. Based on the role of oxidative stress in modulating depressive disorders as well as on the association between the neuroprotective and antioxidant properties of guanosine, here we investigated if its antidepressant-like effect is accompanied by a modulation of hippocampal oxidant/antioxidant parameters. Adult Swiss mice were submitted to an acute restraint stress protocol, which is known to cause behavioral changes that are associated with neuronal oxidative damage. Animals submitted to ARS exhibited an increased immobility time in the forced swimming test (FST) and the administration of guanosine (5mg/kg, p.o.) or fluoxetine (10mg/kg, p.o., positive control) before the exposure to stressor prevented this alteration. Moreover, the significantly increased levels of hippocampal malondialdehyde (MDA; an indicator of lipid peroxidation), induced by ARS were not observed in stressed mice treated with guanosine. Although no changes were found in the hippocampal levels of reduced glutathione (GSH), the group submitted to ARS procedure presented enhanced glutathione peroxidase (GPx), glutathione reductase (GR), superoxide dismutase (SOD) activities and reduced catalase (CAT) activity in the hippocampus. Guanosine was able to prevent the alterations in GPx, GR, CAT activities, and in SOD/CAT activity ratio, but potentiated the increase in SOD activity elicited by ARS. Altogether, the present findings indicate that the observed antidepressant-like effects of guanosine might be related, at least in part, to its capability of modulating antioxidant defenses and mitigating hippocampal oxidative damage induced by ARS.


Guanosine/therapeutic use , Hippocampus/metabolism , Oxidative Stress/physiology , Stress, Psychological/drug therapy , Stress, Psychological/metabolism , Swimming/psychology , Acute Disease , Animals , Female , Guanosine/pharmacology , Hippocampus/drug effects , Mice , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Restraint, Physical , Stress, Psychological/psychology
20.
J Hepatol ; 61(5): 1064-72, 2014 Nov.
Article En | MEDLINE | ID: mdl-24862448

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.


Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Guanosine/analogs & derivatives , Liver Neoplasms/drug therapy , Prodrugs/therapeutic use , Thymine Nucleotides/therapeutic use , Acyclovir/metabolism , Acyclovir/therapeutic use , Animals , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Guanosine/metabolism , Guanosine/therapeutic use , Hep G2 Cells , Hepatitis B virus/genetics , Hepatitis B virus/pathogenicity , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Prodrugs/metabolism , Telomerase/metabolism , Thymine Nucleotides/metabolism
...