Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 74
1.
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
2.
Exp Neurol ; 334: 113459, 2020 12.
Article En | MEDLINE | ID: mdl-32891670

Ketamine has been reported to exert a prophylactic effect against stress-induced depressive-like behavior by modulating the guanosine-based purinergic system. However, the molecular pathways underlying its prophylactic effect and whether guanosine also elicits a similar effect remain to be determined. Here, we investigated the prophylactic effect of ketamine and guanosine against corticosterone (CORT - 20 mg/kg, p.o.)-induced depressive-like behavior in mice. Furthermore, we characterized if the prophylactic response may be associated with mTORC1-driven signaling in the hippocampus and prefrontal cortex. A single administration of ketamine (5 mg/kg, i.p.), but not guanosine (1 or 5 mg/kg, p.o.), given 1 week before the pharmacological stress prevented CORT-induced depressive-like behavior in the tail suspension test (TST) and splash test (SPT). Fluoxetine treatment for 3 weeks did not prevent CORT-induced behavioral effects. A single administration of subthreshold doses of ketamine (1 mg/kg, i.p.) plus guanosine (5 mg/kg, p.o.) partially prevented the CORT-induced depressive-like behavior in the SPT. Additionally, CORT reduced Akt (Ser473) and GSK-3ß (Ser9) phosphorylation and PSD-95, GluA1, and synapsin immunocontent in the hippocampus, but not in the prefrontal cortex. No alterations on mTORC1/p70S6K immunocontent were found in both regions in any experimental group. CORT-induced reductions on PSD-95, GluA1, and synapsin immunocontent were prevented only by ketamine treatment. Collectively, these findings suggest that ketamine, but not guanosine, exerts a prophylactic effect against depressive-like behavior, an effect associated with the stimulation of long-lasting pro-synaptogenic signaling in the hippocampus.


Corticosterone/toxicity , Depression/prevention & control , Guanosine/administration & dosage , Ketamine/administration & dosage , Pre-Exposure Prophylaxis/methods , Synapses/physiology , Animals , Antidepressive Agents/administration & dosage , Depression/chemically induced , Depression/psychology , Hindlimb Suspension/adverse effects , Hindlimb Suspension/psychology , Hippocampus/drug effects , Hippocampus/physiology , Male , Mice , Signal Transduction/drug effects , Signal Transduction/physiology , Synapses/drug effects
3.
Mol Neurobiol ; 57(11): 4790-4809, 2020 Nov.
Article En | MEDLINE | ID: mdl-32789760

Amyloid-ß oligomers (AßOs) toxicity causes mitochondrial dysfunction, leading to synaptic failure in Alzheimer's disease (AD). Considering presynaptic high energy demand and tight Ca2+ regulation, impairment of mitochondrial function can lead to deteriorated neural activity and cell death. In this study, an AD mouse model induced by ICV (intracerebroventricular) injection of AßOs was used to investigate the toxicity of AßOs on presynaptic function. As a therapeutic approach, GUO (guanosine) was given by oral route to evaluate the neuroprotective effects on this AD model. Following 24 h and 48 h from the model induction, behavioral tasks and biochemical analyses were performed, respectively. AßOs impaired object recognition (OR) short-term memory and reduced glutamate uptake and oxidation in the hippocampus. Moreover, AßOs decreased spare respiratory capacity, reduced ATP levels, impaired Ca2+ handling, and caused mitochondrial swelling in hippocampal synaptosomes. Guanosine crossed the BBB, recovered OR short-term memory, reestablished glutamate uptake, recovered mitochondrial Ca2+ homeostasis, and partially prevented mitochondrial swelling. Therefore, this endogenous purine presented a neuroprotective effect on presynaptic mitochondria and should be considered for further studies in AD models.


Amyloid beta-Peptides/toxicity , Calcium/metabolism , Guanosine/pharmacology , Homeostasis , Mitochondria/metabolism , Neuroprotection/drug effects , Presynaptic Terminals/metabolism , Amyloid beta-Peptides/administration & dosage , Animals , Gene Expression Regulation/drug effects , Glutamic Acid/metabolism , Guanosine/administration & dosage , Hippocampus/drug effects , Hippocampus/metabolism , Homeostasis/drug effects , Male , Memory/drug effects , Mice , Mitochondria/drug effects , Mitochondria/ultrastructure , Oxidative Stress/drug effects , Presynaptic Terminals/drug effects , Synaptosomes/metabolism , Synaptosomes/ultrastructure
4.
Neurochem Int ; 139: 104797, 2020 10.
Article En | MEDLINE | ID: mdl-32652267

Augmentative treatment is considered the best second-option when a first-choice drug has partial limitations, particularly by allowing antidepressant dose reduction. Considering that ketamine has significant knock-on effects, this study investigated the effects of a single coadministration with subthreshold doses of ketamine plus guanosine in a corticosterone (CORT)-induced animal model of depression and the role of anti-inflammatory and antioxidant pathways. CORT administration (20 mg/kg, p.o. for 21 days) increased the immobility time in the tail suspension test (TST) and the grooming latency in the splash test (SPT), as well as reduced the total time of grooming in the SPT. These behavioral alterations were accompanied by impaired hippocampal slices viability, elevated immunocontent of nuclear factor-kappa B (NF-κB) and indoleamine-2,3-dioxygenase 1 (IDO-1), and reduced immunocontent of glucocorticoids receptor (GR), glutamate transporter (GLT-1), nuclear factor-erythroid 2-related factor 2 (Nrf2), and heme oxygenase-1 (HO-1) in the hippocampus. CORT also decreased the thioredoxin reductase activity in the hippocampus, while reduced the glutathione reductase activity and non-protein thiols levels in both hippocampus and prefrontal cortex. In addition, elevated content of malondialdehyde and protein carbonyl was also observed in the hippocampus and prefrontal cortex of CORT-treated mice. Of note, a single administration of ketamine (0.1 mg/kg, i.p.) plus guanosine (0.01 mg/kg, p.o.) attenuated the depressive-like behavior and hippocampal slices impairments induced by CORT. The behavioral response obtained by the combined administration of these drugs was paralleled by the reestablishment of the CORT-induced molecular alterations on hippocampal GR, NF-κB, IDO-1, and GLT-1 immunocontent. Moreover, CORT-induced alterations on the antioxidant enzyme activity and oxidative stress markers were partially restored by ketamine plus guanosine treatment. Taken together, these findings suggest that guanosine might potentiate the effects of ketamine on inflammatory and oxidative markers that are elevated in depression.


Antidepressive Agents/administration & dosage , Guanosine/administration & dosage , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Ketamine/administration & dosage , NF-kappa B/antagonists & inhibitors , Receptors, Glucocorticoid/antagonists & inhibitors , Animals , Corticosterone/toxicity , Depression/chemically induced , Depression/drug therapy , Depression/metabolism , Disease Models, Animal , Drug Therapy, Combination , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Male , Mice , NF-kappa B/metabolism , Receptors, Glucocorticoid/metabolism
5.
Naunyn Schmiedebergs Arch Pharmacol ; 393(7): 1251-1267, 2020 07.
Article En | MEDLINE | ID: mdl-32313990

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.


Apoptosis/drug effects , Cell Proliferation/drug effects , Guanosine/pharmacology , Lymphoma, T-Cell/drug therapy , Cell Line, Tumor , Guanosine/administration & dosage , Guanosine/analogs & derivatives , Humans , Inhibitory Concentration 50 , Leukocytes, Mononuclear/drug effects , Lymphoma, T-Cell/pathology , Nucleoside Transport Proteins/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Xenograft Model Antitumor Assays
6.
J Psychiatr Res ; 115: 103-112, 2019 08.
Article En | MEDLINE | ID: mdl-31128500

The ketamine's potential for the treatment of refractory depression and anxiety has been considered one the most important discoveries in the last years, however, repeated use of ketamine is limited due to its side/adverse effects. Therefore, the search for effective augmentation strategies that may reduce ketamine doses is welcome. Therefore, this study sought to augment the effect of ketamine by guanosine in the novelty-suppressed feeding (NSF) test, a behavioral paradigm able to detect depression/anxiety-related behavior. Acute administration of guanosine (0.05 mg/kg, p.o.), similar to ketamine (1 mg/kg, i.p.), produced a rapid behavioral response in mice submitted to NSF test. Moreover, the coadministration of sub-effective doses of guanosine (0.01 mg/kg, p.o.) and ketamine (0.1 mg/kg, i.p.) was effective in mice submitted to NSF test. Subsequently, the intracellular mechanism underpinning the augmentation effect of ketamine by guanosine was investigated. Our results suggest that augmentation response of ketamine by guanosine in the NSF test probably involves the activation of mTOR signaling, since the treatment with rapamycin (0.2 nmol/site, i.c.v., a selective mTOR inhibitor) completely abolished this effect. This augmentation strategy also increased mTOR phosphorylation (Ser2448) in the hippocampus, reinforcing the role of mTOR in this augmentation response. However, no changes in the p70S6K, PSD-95, GluA1, and synapsin immunocontents were found in the hippocampus of ketamine plus guanosine-treated mice. Overall, results provide evidence that guanosine is able to augment the effect of ketamine in the NSF test via mTOR activation, a finding that might have therapeutic implications for the management of depression/anxiety.


Behavior, Animal/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Exploratory Behavior/drug effects , Guanosine/pharmacology , Hippocampus/drug effects , Ketamine/pharmacology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/drug effects , Animals , Drug Synergism , Excitatory Amino Acid Antagonists/administration & dosage , Guanosine/administration & dosage , Ketamine/administration & dosage , Mice
7.
Arch Razi Inst ; 74(4): 357-364, 2019 12.
Article En | MEDLINE | ID: mdl-31939252

Un-methylated cytosine-phosphate-guanosine oligodeoxynucleotides (CpG-ODN) has been considered as a powerful vaccine adjuvant and recognition of CpG-ODN by chicken leukocytes promotes their ability to fight against infections. In our study, efficacy of different routes of CpG-ODN application as an adjuvant on immune responses (antibody titer together with leukogram) following vaccination against Newcastle disease (ND) has been evaluated in broiler chickens (Ross-308). The results indicated that routes of CpG-ODN administration influence immune responses and comparison effectiveness of CpG-OND delivery routes showed that group vaccinated by eye-drop application had the highest antibody titer than that of the group injected intramuscularly (im) and the difference was significant (p = 0.04) on day 35 of age. Antibody titer of the group treated with Clone 30 plus CpG-ODN via eye-drop route was higher than that of the group vaccinated with clone 30 alone on days 28 and 35 of age and the difference was significant (p = 0.04). Co-administration of both vaccine and CpG improved outcome of leukogram of the chickens on days 21 to 42 of age and among the treated groups, WBC of the group received both vaccine and CpG by eye-drop route significantly (p < 0.05) differed from that of the group vaccinated with clone 30 alone on days 28 and 35 but not on day 42 of age. Average final body weight of the control group did not significantly differ from those of the treated groups at end of the experiment. In conclusion, co-administration of ND vaccine plus CpG-ODN via eye-drop route improves immune responses.


Adjuvants, Immunologic/pharmacology , Chickens , Immunity, Humoral/drug effects , Newcastle Disease/prevention & control , Oligodeoxyribonucleotides/immunology , Poultry Diseases/prevention & control , Viral Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Cytosine/administration & dosage , Cytosine/immunology , Guanosine/administration & dosage , Guanosine/immunology , Newcastle disease virus/drug effects , Oligodeoxyribonucleotides/administration & dosage , Phosphates/administration & dosage , Phosphates/immunology , Viral Vaccines/administration & dosage
8.
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
9.
Neurochem Res ; 43(7): 1476-1489, 2018 Jul.
Article En | MEDLINE | ID: mdl-29855847

Guanosine (GUO) has neuroprotective effects in experimental models of brain diseases involving glutamatergic excitotoxicity in male animals; however, its effects in female animals are poorly understood. Thus, we investigated the influence of gender and GUO treatment in adult male and female Wistar rats submitted to focal permanent cerebral ischemia in the motor cortex brain. Female rats were subdivided into non-estrogenic and estrogenic phase groups by estrous cycle verification. Immediately after surgeries, the ischemic animals were treated with GUO or a saline solution. Open field and elevated plus maze tasks were conducted with ischemic and naïve animals. Cylinder task, immunohistochemistry and infarct volume analyses were conducted only with ischemic animals. Female GUO groups achieved a full recovery of the forelimb symmetry at 28-35 days after the insult, while male GUO groups only partially recovered at 42 days, in the final evaluation. The ischemic insult affected long-term memory habituation to novelty only in female groups. Anxiety-like behavior, astrocyte morphology and infarct volume were not affected. Regardless the estrous cycle, the ischemic injury affected differently female and male animals. Thus, this study points that GUO is a potential neuroprotective compound in experimental stroke and that more studies, considering the estrous cycle, with both genders are recommended in future investigation concerning brain diseases.


Brain Ischemia/prevention & control , Cerebral Cortex/drug effects , Guanosine/administration & dosage , Neuroprotective Agents/administration & dosage , Sex Characteristics , Animals , Brain Ischemia/pathology , Cerebral Cortex/pathology , Female , Male , Maze Learning/drug effects , Maze Learning/physiology , Rats , Rats, Wistar , Recovery of Function/drug effects , Recovery of Function/physiology
10.
J Bacteriol ; 200(14)2018 07 15.
Article En | MEDLINE | ID: mdl-29712876

In Staphylococcus aureus, the global transcriptional regulator CodY modulates the expression of hundreds of genes in response to the availability of GTP and the branched-chain amino acids isoleucine, leucine, and valine (ILV). CodY DNA-binding activity is high when GTP and ILV are abundant. When GTP and ILV are limited, CodY's affinity for DNA drops, altering expression of CodY-regulated targets. In this work, we investigated the impact of guanine nucleotides (GNs) on S. aureus physiology and CodY activity by constructing a guaA null mutant (ΔguaA strain). De novo biosynthesis of guanine monophosphate is abolished due to the guaA mutation; thus, the mutant cells require exogenous guanosine for growth. We also found that CodY activity was reduced when we knocked out guaA, activating the Agr two-component system and increasing secreted protease activity. Notably, in a rich, complex medium, we detected an increase in alternative sigma factor B activity in the ΔguaA mutant, which results in a 5-fold increase in production of the antioxidant pigment staphyloxanthin. Under biologically relevant flow conditions, ΔguaA cells failed to form robust biofilms when limited for guanine or guanosine. Transcriptome sequencing (RNA-Seq) analysis of the S. aureus transcriptome during growth in guanosine-limited chemostats revealed substantial CodY-dependent and -independent alterations of gene expression profiles. Importantly, these changes increase production of proteases and δ-toxin, suggesting that S. aureus exhibits a more invasive lifestyle when limited for guanosine. Further, gene products upregulated under GN limitation, including those necessary for lipoic acid biosynthesis and sugar transport, may prove to be useful drug targets for treating Gram-positive infections.IMPORTANCEStaphylococcus aureus infections impose a serious economic burden on health care facilities and patients because of the emergence of strains resistant to last-line antibiotics. Understanding the physiological processes governing fitness and virulence of S. aureus in response to environmental cues is critical for developing efficient diagnostics and treatments. De novo purine biosynthesis is essential for both fitness and virulence in S. aureus since inhibiting production cripples S. aureus's ability to cause infection. Here, we corroborate these findings and show that blocking guanine nucleotide synthesis severely affects S. aureus fitness by altering metabolic and virulence gene expression. Characterizing pathways and gene products upregulated in response to guanine limitation can aid in the development of novel adjuvant strategies to combat S. aureus infections.


Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial/drug effects , Guanine/administration & dosage , Repressor Proteins/metabolism , Staphylococcus aureus/metabolism , Bacterial Proteins/genetics , Biofilms , Genotype , Guanine/metabolism , Guanine/pharmacology , Guanosine/administration & dosage , Guanosine/metabolism , RNA, Bacterial , Repressor Proteins/genetics , Sequence Analysis, RNA , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Transcriptome , Virulence Factors
11.
Mol Med Rep ; 15(6): 3999-4004, 2017 Jun.
Article En | MEDLINE | ID: mdl-28487988

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.


Guanosine/administration & dosage , Neovascularization, Physiologic/drug effects , Neurogenesis/drug effects , Recovery of Function/drug effects , Stroke/pathology , Stroke/physiopathology , Animals , Brain/metabolism , Brain/pathology , Brain Ischemia/complications , Brain Ischemia/metabolism , Brain Ischemia/pathology , Disease Models, Animal , Doublecortin Protein , Gene Expression , Immunohistochemistry , Male , Mice , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Protective Agents/administration & dosage , Stroke/drug therapy , Stroke/etiology , Time Factors
12.
J Neurotrauma ; 34(7): 1318-1328, 2017 04 01.
Article En | MEDLINE | ID: mdl-27931151

Traumatic brain injury (TBI) is a highly complex multi-factorial disorder. Experimental trauma involves primary and secondary injury cascades that underlie delayed neuronal dysfunction and death. Mitochondrial dysfunction and glutamatergic excitotoxicity are the hallmark mechanisms of damage. Accordingly, a successful pharmacological intervention requires a multi-faceted approach. Guanosine (GUO) is known for its neuromodulator effects in various models of brain pathology, specifically those that involve the glutamatergic system. The aim of the study was to investigate the GUO effects against mitochondrial damage in hippocampus and cortex of rats subjected to TBI, as well as the relationship of this effect with the glutamatergic system. Adult male Wistar rats were subjected to a unilateral moderate fluid percussion brain injury (FPI) and treated 15 min later with GUO (7.5 mg/kg) or vehicle (saline 0.9%). Analyses were performed in hippocampus and cortex 3 h post-trauma and revealed significant mitochondrial dysfunction, characterized by a disrupted membrane potential, unbalanced redox system, decreased mitochondrial viability, and complex I inhibition. Further, disruption of Ca2+ homeostasis and increased mitochondrial swelling was also noted. Our results showed that mitochondrial dysfunction contributed to decreased glutamate uptake and levels of glial glutamate transporters (glutamate transporter 1 and glutamate aspartate transporter), which leads to excitotoxicity. GUO treatment ameliorated mitochondrial damage and glutamatergic dyshomeostasis. Thus, GUO might provide a new efficacious strategy for the treatment acute physiological alterations secondary to TBI.


Amino Acid Transport System X-AG/metabolism , Brain Injuries, Traumatic , Glutamic Acid/metabolism , Guanosine/pharmacology , Mitochondrial Diseases , Animals , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Disease Models, Animal , Guanosine/administration & dosage , Male , Mitochondrial Diseases/drug therapy , Mitochondrial Diseases/etiology , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/physiopathology , Oxidation-Reduction/drug effects , Rats , Rats, Wistar
13.
Eur J Neurosci ; 43(8): 1006-15, 2016 Apr.
Article En | MEDLINE | ID: mdl-26779605

Guanosine is a purine nucleoside that occurs naturally in the central nervous system, exerting trophic effects. Given its neuroprotective properties, the potential of guanosine as an antidepressant has been recently examined. Within this context, the present study sought to investigate the effects of chronic treatment with guanosine on the tail suspension test (TST), open field test and adult hippocampal neurogenesis. Swiss mice were administered guanosine for 21 days (5 mg/kg/day, p.o.) and subsequently submitted to the TST and open-field test. Following behavioural testing, animals were killed and the brains were processed for immunohistochemical analyses of hippocampal cell proliferation and neuronal differentiation. Animals treated with guanosine showed a reduction in immobility time in the TST without alterations in locomotor activity, confirming the antidepressant-like effect of this compound. Quantitative microscopic analysis did not reveal significant alterations in the numbers of Ki-67- and proliferating cell nuclear antigen (PCNA)-positive cells in the hippocampal dentate gyrus (DG) of guanosine-treated mice. However, guanosine treatment resulted in a significant increase in the number of immature neurons, as assessed by immunohistochemistry for the neurogenic differentiation protein. Interestingly, this effect was localized to the ventral hippocampal DG, a functionally distinct region of this structure known to regulate emotional and motivational behaviours. Taken together, our results suggest that the antidepressant-like effect of chronic guanosine treatment is associated with an increase in neuronal differentiation, reinforcing the notion that this nucleoside may be an endogenous mood modulator.


Guanosine/pharmacology , Hippocampus/drug effects , Neurogenesis , Neurons/drug effects , Neuroprotective Agents/pharmacology , Animals , Female , Guanosine/administration & dosage , Hippocampus/cytology , Hippocampus/physiology , Locomotion , Mice , Neurons/cytology , Neurons/physiology , Neuroprotective Agents/administration & dosage
14.
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
15.
Eur J Drug Metab Pharmacokinet ; 41(5): 567-74, 2016 Oct.
Article En | MEDLINE | ID: mdl-25898809

IDX184 is a phosphoramidate prodrug of 2'-methylguanosine-5'-monophosphate, developed to treat patients infected with hepatitis C virus. A mass balance study of radiolabeled IDX184 and pharmacokinetic studies of IDX184 in portal vein-cannulated monkeys revealed relatively low IDX184 absorption but higher exposure of IDX184 in the portal vein than in the systemic circulation, indicating >90 % of the absorbed dose was subject to hepatic extraction. Systemic exposures to the main metabolite, 2'-methylguanosine (2'-MeG), were used as a surrogate for liver levels of the pharmacologically active entity 2'-MeG triphosphate, and accordingly, systemic levels of 2'-MeG in the monkey were used to optimize formulations for further clinical development of IDX184. Capsule formulations of IDX184 delivered acceptable levels of 2'-MeG in humans; however, the encapsulation process introduced low levels of the genotoxic impurity ethylene sulfide (ES), which necessitated formulation optimization. Animal pharmacokinetic data guided the development of a tablet with trace levels of ES and pharmacokinetic performance equal to that of the clinical capsule in the monkey. Under fed conditions in humans, the new tablet formulation showed similar exposure to the capsule used in prior clinical trials.


Guanosine Monophosphate/analogs & derivatives , Guanosine/analogs & derivatives , Liver/drug effects , Prodrugs/administration & dosage , Prodrugs/pharmacokinetics , Animals , Capsules/administration & dosage , Capsules/pharmacokinetics , Chemistry, Pharmaceutical/methods , Guanosine/administration & dosage , Guanosine/pharmacokinetics , Guanosine Monophosphate/administration & dosage , Guanosine Monophosphate/pharmacokinetics , Haplorhini , Humans , Male , Tablets/administration & dosage , Tablets/pharmacokinetics
16.
PLoS One ; 9(9): e105577, 2014.
Article En | MEDLINE | ID: mdl-25184445

Hyperuricemia is well known as the cause of gout. In recent years, it has also been recognized as a risk factor for arteriosclerosis, cerebrovascular and cardiovascular diseases, and nephropathy in diabetic patients. Foods high in purine compounds are more potent in exacerbating hyperuricemia. Therefore, the development of probiotics that efficiently degrade purine compounds is a promising potential therapy for the prevention of hyperuricemia. In this study, fifty-five lactic acid bacteria isolated from Chinese sauerkraut were evaluated for the ability to degrade inosine and guanosine, the two key intermediates in purine metabolism. After a preliminary screening based on HPLC, three candidate strains with the highest nucleoside degrading rates were selected for further characterization. The tested biological characteristics of candidate strains included acid tolerance, bile tolerance, anti-pathogenic bacteria activity, cell adhesion ability, resistance to antibiotics and the ability to produce hydrogen peroxide. Among the selected strains, DM9218 showed the best probiotic potential compared with other strains despite its poor bile resistance. Analysis of 16S rRNA sequences showed that DM9218 has the highest similarity (99%) to Lactobacillus plantarum WCFS1. The acclimated strain DM9218-A showed better resistance to 0.3% bile salt, and its survival in gastrointestinal tract of rats was proven by PCR-DGGE. Furthermore, the effects of DM9218-A in a hyperuricemia rat model were evaluated. The level of serum uric acid in hyperuricemic rat can be efficiently reduced by the intragastric administration of DM9218-A (P<0.05). The preventive treatment of DM9218-A caused a greater reduction in serum uric acid concentration in hyperuricemic rats than the later treatment (P<0.05). Our results suggest that DM9218-A may be a promising candidate as an adjunctive treatment in patients with hyperuricemia during the onset period of disease. DM9218-A also has potential as a probiotic in the prevention of hyperuricemia in the normal population.


Diet/adverse effects , Gastrointestinal Tract/microbiology , Hyperuricemia/diet therapy , Lactobacillaceae/isolation & purification , Lactobacillus plantarum/isolation & purification , Probiotics/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Bacterial Adhesion/drug effects , Bile Acids and Salts/pharmacology , Brassica/microbiology , Fermentation , Gastrointestinal Tract/metabolism , Guanosine/administration & dosage , Guanosine/adverse effects , Humans , Hydrogen Peroxide/pharmacology , Hyperuricemia/chemically induced , Hyperuricemia/metabolism , Inosine/administration & dosage , Inosine/adverse effects , Lactic Acid/metabolism , Lactobacillaceae/drug effects , Lactobacillaceae/genetics , Lactobacillaceae/metabolism , Lactobacillus plantarum/drug effects , Lactobacillus plantarum/genetics , Lactobacillus plantarum/metabolism , Male , Microbial Sensitivity Tests , Probiotics/metabolism , RNA, Ribosomal, 16S/genetics , Rats , Rats, Wistar , Uric Acid/blood
17.
J Pharmacol Exp Ther ; 350(3): 719-26, 2014 Sep.
Article En | MEDLINE | ID: mdl-25002416

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.


Adenosine/administration & dosage , Adenosine/blood , Guanosine/administration & dosage , Guanosine/blood , Animals , Drug Interactions/physiology , Infusions, Intravenous , Male , Rats , Rats, Sprague-Dawley
18.
PLoS One ; 9(2): e90693, 2014.
Article En | MEDLINE | ID: mdl-24587409

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.


Brain Injuries/prevention & control , Brain Injuries/physiopathology , Brain Ischemia/physiopathology , Guanosine/pharmacology , Animals , Ascorbic Acid/metabolism , Blotting, Western , Brain Injuries/metabolism , Brain Ischemia/etiology , Catalase/metabolism , Dose-Response Relationship, Drug , Drug Administration Schedule , Electrocoagulation/adverse effects , Excitatory Amino Acid Transporter 3/metabolism , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Forelimb/drug effects , Forelimb/physiopathology , Glutamate Plasma Membrane Transport Proteins/metabolism , Glutamate-Ammonia Ligase/metabolism , Glutathione/metabolism , Guanosine/administration & dosage , Lipid Peroxidation/drug effects , Male , Neuroprotective Agents/pharmacology , Nitric Oxide/metabolism , Rats, Wistar , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
19.
Purinergic Signal ; 9(4): 655-61, 2013 Dec.
Article En | MEDLINE | ID: mdl-23839776

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.


Cyclic AMP/metabolism , Gastric Emptying/physiology , Gastrointestinal Motility/physiology , Guanosine/administration & dosage , Muscle, Smooth/physiology , Stomach/physiology , Adaptation, Physiological/drug effects , Adaptation, Physiological/physiology , Animals , Dose-Response Relationship, Drug , Gastric Emptying/drug effects , Gastrointestinal Motility/drug effects , Male , Mice , Mice, Inbred C57BL , Muscle, Smooth/drug effects , Stomach/drug effects
20.
J Neurosci Res ; 91(2): 262-72, 2013 Feb.
Article En | MEDLINE | ID: mdl-23151946

After ischemic stroke, early thrombolytic therapy to reestablish tissue perfusion improves outcome but triggers a cascade of deleterious cellular and molecular events. Using a collaborative approach, our groups examined the effects of guanosine (Guo) in response to ischemic reperfusion injury in vitro and in vivo. In a transient middle cerebral artery occlusion (MCAO) in rats, Guo significantly reduced infarct volume in a dose-dependent manner when given systemically either immediately before or 30 min, but not 60 min, after the onset of the 5.5-hr reperfusion period. In a separate experiment, Guo significantly reduced infarct volume after 24 hr of reperfusion when administered 5 min before reperfusion. Western blot analysis did not reveal any significant changes either in endoplasmic reticulum (ER) stress proteins (GRP 78 and 94) or HSP 70 or in levels of m-calpain. In vitro oxygen and glucose deprivation (OGD) significantly increased production of both reactive oxygen species (ROS) and interleukin-8 (IL-8) in the primary astrocytes. Guo did not alter ROS or IL-8 production when given to the astrocytes before OGD. However, Guo when added to the cells prior to or 30 min after reperfusion significantly reduced IL-8 release but not ROS formation. Our study revealed a dose- and time-dependent protective effect of Guo on reperfusion injury in vitro and vivo. The mechanisms by which Guo exerts its effect are independent of unfolded proteins in ER or the level of intracellular calcium or ROS formation. However, the effect may be induced, at least partially, by inhibiting IL-8, a marker of reperfusion-triggered proinflammatory events.


Brain Infarction/prevention & control , Guanosine/administration & dosage , Infarction, Middle Cerebral Artery/drug therapy , Neuroprotective Agents/administration & dosage , Reperfusion Injury/prevention & control , Analysis of Variance , Animals , Animals, Newborn , Astrocytes/drug effects , Astrocytes/metabolism , Brain Infarction/etiology , Cells, Cultured , Gene Expression Regulation/drug effects , Glucose/deficiency , Heat-Shock Proteins/metabolism , Hypoxia , Interleukin-8/metabolism , Male , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Reperfusion/adverse effects , Reperfusion Injury/complications , Time Factors
...