Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
Add more filters










Publication year range
1.
J Biol Regul Homeost Agents ; 27(3): 673-80, 2013.
Article in English | MEDLINE | ID: mdl-24152836

ABSTRACT

In previous studies, we have found that extracellular guanosine can stimulate endogenous progenitor/stem cell proliferation in the spinal cord following chronic injury and in the subventricular zone of the brains of rats afflicted with Parkinson's Disease. In this study, using neural stem cells isolated from one-day old rats, we found that guanosine could stimulate neural stem cell proliferation, and that the proliferation was not due to the guanosine metabolism mechanism since guanine, which is interconverted by an ecto-purine nucleoside phosphorylase from guanosine, has no stimulating effect on the proliferation of neural stem cells. We determined that second messenger cAMP was involved in the pathway as results showed that 100 microM guanosine stimulated cAMP accumulation. Using western blot analysis, we found that 100 microM guanosine can activate the phosphorylation of CREB without changing the total amount of CREB. In conclusion, guanosine can stimulate neural stem cell proliferation, and the cAMP-CREB pathway is involved in this biological effect.


Subject(s)
Cell Proliferation/drug effects , Cyclic AMP Response Element-Binding Protein/physiology , Cyclic AMP/physiology , Guanosine/pharmacology , Neural Stem Cells/drug effects , Signal Transduction , Animals , Female , Guanine/pharmacology , Male , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Phosphorylation , Rats , Rats, Wistar
2.
Panminerva Med ; 54(1 Suppl 4): 43-51, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23241934

ABSTRACT

Parkinson's disease (PD) is a pathological condition characterized by a progressive neurodegeneration of dopaminergic neurons with the consequent reduction of dopamine content in the substantia nigra. The neurotoxin 6-hydroxydopamine (6-OHDA) is widely used to mimic the neuropathology of PD in both in vivo and in vitro experimental models. We found that, as expected, in dopaminergic human SH-SY5Y neuroblastoma cells the toxin reduced cell viability causing programmed cell death as assessed by an increase in DNA fragmentation. We also examined, in these cells, the activation/inactivation of several pro and anti apoptotic signaling pathways by 6-OHDA including p-38 kinase (p-38), c-Jun N-terminal kinase (JNK), protein kinase B (also known as Akt), glycogen synthase kinase-3ß (GSK3ß), and Bcl-2 protein. Guanine-based purines, exert neuroprotective effects and we previously reported that guanosine activates cell survival pathways including PI3K/Akt/PKB signaling in different kinds of cells including glia and neuroblastoma cells. In the present study we found that guanosine (300 µM) protected SH-SY5Y neuroblastoma cells when they were exposed to 6-OHDA, promoting their survival. Guanosine reduced the 6-OHDA mediated activation of p-38 and JNK. Moreover the nucleoside potentiated the early increase in the phosphorylation of the anti-apoptotic kinase Akt and the increase in the expression of the anti-apoptotic Bcl-2 protein induced by 6-OHDA. In summary our results show that guanosine results to be neuroprotective in a recognized in vitro model of PD thus suggesting that it could represent a new potential pharmacological tool to be studied in the therapeutic approach to PD.


Subject(s)
Guanosine/therapeutic use , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , Substantia Nigra/drug effects , Apoptosis , Cell Line, Tumor , Cell Proliferation , Cell Survival , Culture Media/pharmacology , DNA Fragmentation , Dose-Response Relationship, Drug , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Oxidopamine/adverse effects , Phosphorylation , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
3.
J Biol Regul Homeost Agents ; 26(1): 51-65, 2012.
Article in English | MEDLINE | ID: mdl-22475097

ABSTRACT

Guanosine has long been known as an endogenous purine nucleoside deeply involved in the modulation of several intracellular processes, especially G-protein activity. More recently, it has been reported to act as an extracellular signaling molecule released from neurons and, more markedly, from astrocytes either in basal conditions or after different kinds of stimulation including hypoxia. Moreover, in vivo studies have shown that guanosine plays an important role as both a neuroprotective and neurotrophic agent in the central nervous system. Specific high-affinity binding sites for this nucleoside have been found on membrane preparations from rat brain. The present study was undertaken to investigate the distribution and metabolic profiles of guanosine after administering the nucleoside to gain a better understanding of the biological effects of this potential drug candidate. Rats were given an intraperitonal (i.p.) injection of 2, 4, 8 or 16 mg/kg of guanosine combined with 0.05% of [3H]guanosine. Plasma samples were collected 7.5, 15, 30, 60 and 90 min after the guanosine-mixture administration and analyzed by either a liquid scintillation counter or by HPLC connected to a UV and to an on-line radiochemical detector to measure the levels of guanosine and its metabolic products guanine, xanthine and uric acid. The levels of guanosine, guanine and xanthine were also measured in brain, lung, heart, kidney and liver tissue homogenates at the defined time points after the injection of 8 mg/kg of the guanosine-mixture. We found that the levels of radioactivity in plasma increased linearly in a dose- and time-dependent manner. Guanosine was widely distributed in all tissues examined in the present study, at almost twice its usual levels. In addition, guanine levels dramatically increased in all the organs. Interestingly, enzymatic analysis of the plasma samples showed the presence of a soluble purine nucleoside phosphorylase, a key enzyme in the purine salvage pathway and nucleoside catabolism. Since guanosine has been shown to be neuroprotective and astrocytes have been reported to play critical roles in mediating neuronal survival and functions in different neurodegenerative disorders, we also performed uptake and release.


Subject(s)
Guanosine/pharmacokinetics , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Brain/cytology , Brain/metabolism , Cells, Cultured , Guanine/metabolism , Guanosine/administration & dosage , Guanosine/blood , Injections, Intraperitoneal , Kidney/metabolism , Liver/metabolism , Lung/metabolism , Male , Myocardium/metabolism , Purine-Nucleoside Phosphorylase/blood , Purines/metabolism , Rats , Rats, Sprague-Dawley , Xanthine/metabolism
4.
J Biol Regul Homeost Agents ; 24(1): 19-25, 2010.
Article in English | MEDLINE | ID: mdl-20385068

ABSTRACT

Cancers contain a 'side population' (SP), a subset of cells that is greatly enriched in stem cells and which contains malignant progenitors. SP cells are characterised by high efflux capability for Hoechst 33342 dye and for anti-cancer therapeutic agents through transporters; ABCG2 (ATP-binding cassette transporter G2) is currently most closely associated with the SP phenotype. Guanosine is an important intercellular signalling molecule; it stimulates stem cell proliferation in vivo and affects cholesterol efflux in vitro through activation of ABCG transporter (ABCG1), raising the possibility that it might also affect ABCG2 and hence the SP. We examined the effects of guanosine on the SP of A549 lung cancer cells. Fluorescence-activated cell sorting (FACS) revealed that exposure to 10 microM guanosine significantly decreased the proportion of SP cells after 48 hours but not after 6 hours. In contrast, Western blot analysis showed that 10 microM guanosine significantly decreased ABCG2 expression after 6 hours, but not after 48 hours. These data demonstrate that guanosine affects both the proportion of SP cells and ABCG2 transporters, but the lack of correlation between ABCG2 expression and the SP phenotype indicates that transporters other than ABCG2 are involved in maintaining the SP phenotype in A549 lung cancer cells.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Guanosine/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Neoplasm Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Benzimidazoles , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Fluorescent Dyes , Gene Expression/drug effects , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Neoplasm Proteins/genetics , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Phenotype
5.
Int J Immunopathol Pharmacol ; 20(2): 301-16, 2007.
Article in English | MEDLINE | ID: mdl-17624242

ABSTRACT

Under pathological conditions brain cells release ATP at concentrations reported to activate P2X(7) ionotropic receptor subtypes expressed in both neuronal and glial cells. In the present study we report that the most potent P2X(7) receptor agonist BzATP stimulates the expression of the metabotropic ATP receptor P2Y(2) in cultured rat brain astrocytes. In other cell types several kinds of stimulation, including stress or injury, induce P2Y(2) expression that, in turn, is involved in different cell reactions. Similarly, it has recently been found that in astrocytes and astrocytoma cells P2Y(2) sites can trigger neuroprotective pathways through the activation of several mechanisms, including the induction of genes for antiapoptotic factors, neurotrophins, growth factors and neuropeptides. Here we present evidence that P2Y(2) mRNA expression in cultured astrocytes peaks 6 h after BzATP exposure and returns to basal levels after 24 h. This effect was mimicked by high ATP concentrations (1 mM) and was abolished by P2X(7)-antagonists oATP and BBG. The BzATP-evoked P2Y(2) receptor up-regulation in cultured astrocytes was coupled to an increased UTP-mediated intracellular calcium response. This effect was inhibited by oATP and BBG and by P2Y(2)siRNA, thus supporting evidence of increased P2Y(2) activity. To further investigate the mechanisms by which P2X(7) receptors mediated the P2Y(2) mRNA up-regulation, the cells were pre-treated with the chelating agent EGTA, or with inhibitors of mitogen-activated kinase (MAPK) (PD98059) or protein kinase C, (GF109203X). Each inhibitor significantly reduced the extent to which BzATP induced P2Y(2) mRNA. Both BzATP and ATP (1 mM) increased ERK1/2 activation. P2X(7)-induced ERK1/2 phosphorylation was unaffected by pre-treatment of astrocytes with EGTA whereas it was inhibited by GF109203X. Phorbol-12-myristate-13-acetate (PMA), an activator of PKCs, rapidly increased ERK1/2 activation. We conclude that activation of P2X(7) receptors in astrocytes enhances P2Y(2) mRNA expression by a mechanism involving both calcium influx and PKC/MAPK signalling pathways.


Subject(s)
Astrocytes/metabolism , Brain/metabolism , Gene Expression Regulation/physiology , RNA, Messenger/biosynthesis , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2/physiology , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/physiology , Animals , Brain/cytology , Brain/embryology , Cells, Cultured , Rats , Receptors, Purinergic P2/biosynthesis , Receptors, Purinergic P2X7 , Receptors, Purinergic P2Y2
6.
Spinal Cord ; 45(10): 671-7, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17228353

ABSTRACT

STUDY DESIGN: Retrospective, descriptive study. OBJECTIVE: To determine if patients with metastatic spinal cord compression (MSCC) make significant functional gains through rehabilitation. To study survival and predictors of survival in MSCC. To explore predictive factors for high or low functional gains in MSCC. SETTING: Inpatient neuro-oncology rehabilitation ward, Henderson General Hospital, Hamilton, Canada. METHODS: Clinical records were examined for 63 inpatients with MSCC. Demographics, treatment of MSCC, length of rehabilitation, admission, and discharge Functional Independence Measure (FIM) scores, Tokuhashi score and survival data were collected. Statistical analyses included nonparametric comparisons, Kaplan-Meier analyses, Cox regression, and exploratory logistic regression. RESULTS: FIM score improved from 83 to 102 (P<0.0001). Estimated median survival from time of rehabilitation was 10.0 months. Kaplan-Meier analysis showed longer survival in patients with high Tokuhashi scores (9-15) compared to low scores (0-8) (P<0.005); and high FIM change (>13) compared to low FIM change (< or =13) (P<0.02). Cox regression revealed that high FIM gain and high Tokuhashi score were prognostic factors. Logistic regression showed Tokuhashi score (odds ratio (OR)=1.30, 95% confidence interval (CI)=1.04-1.62) and length of rehabilitation (OR=1.04, 95% confidence interval (CI)=1.01-1.07) were associated with high FIM gain. CONCLUSIONS: Rehabilitation improves functional outcomes in MSCC. Patients who had a high Tokuhashi score and achieved high functional gains after rehabilitation had longer survival. Tokuhashi score and length of rehabilitation were associated with high FIM gain. The Tokuhashi score can help identify patients with good prognosis and potential for improvement during rehabilitation.


Subject(s)
Recovery of Function , Spinal Cord Compression/etiology , Spinal Cord Compression/rehabilitation , Spinal Neoplasms/mortality , Spinal Neoplasms/secondary , Activities of Daily Living , Disability Evaluation , Female , Humans , Kaplan-Meier Estimate , Length of Stay , Male , Middle Aged , Prognosis , Retrospective Studies
7.
Int J Immunopathol Pharmacol ; 19(2): 293-308, 2006.
Article in English | MEDLINE | ID: mdl-16831297

ABSTRACT

Among P2 metabotropic ATP receptors, P2Y2 subtype seems to be peculiar as its upregulation triggers important biological events in different cells types. In non-stimulated cells including astrocytes, P2Y2 receptors are usually expressed at levels lower than P2Y1 sites, however the promoter region of the P2Y2 receptors has not yet been studied and little is known about the mechanisms underlying the regulation of the expression of this ATP receptor. We showed that not only UTP and ATP are the most potent and naturally occurring agonist for P2Y2 sites, but also guanosine induced an up-regulation of astrocyte P2Y2 receptor mRNA evaluated by Northern blot analysis. We also focused our attention on this nucleoside since in our previous studies it was reported to be released by cultured astrocytes and to exert different neuroprotective effects. UTP and guanosine-evoked P2Y2 receptor up-regulation in rat brain cultured astrocytes was linked to an increased P2Y2-mediated intracellular calcium response, thus suggesting an increased P2Y2 activity. Actinomycin D, a RNA polymerase inhibitor, abrogated both UTP and guanosine-mediated P2Y2 up-regulation, thus indicating that de novo transcription was required. The effect of UTP and guanosine was also evaluated in astrocytes pretreated with different inhibitors of signal transduction pathways including ERK, PKC and PKA reported to be involved in the regulation of other cell surface receptor mRNAs. The results show that ERK1-2/MAPK pathway play a key role in the P2Y2 receptor up-regulation mediated by either UTP or guanosine. Moreover, our data suggest that PKA is also involved in guanosine-induced transcriptional activation of P2Y2 mRNA and that increased intracellular calcium levels and PKC activation may also mediate P2Y2 receptor up-regulation triggered by UTP. The extracellular release of ATP under physiological and pathological conditions has been widely studied. On the contrary, little is known about the release of pyrimidines and in particular of UTP. Here we show that astrocytes are able to release UTP, either at rest or during and following hypoxia/hypoglycemia obtained by submitting the cells to glucose-oxygen deprivation (OGD). Interestingly, also P2Y2 receptor mRNA increased by about two-fold the control values when the cultures were submitted to OGD. It has been recently reported that P2Y2 receptors can play a protective role in astrocytes, thus either guanosine administration or increased extracellular concentrations of guanosine and UTP reached locally following CNS injury may increase P2Y2-mediated biological events aimed at promoting a protective astrocyte response.


Subject(s)
Astrocytes/metabolism , Brain Chemistry/drug effects , Brain/cytology , Guanosine/pharmacology , Receptors, Purinergic P2/biosynthesis , Up-Regulation/drug effects , Uridine Triphosphate/pharmacology , Adenosine Triphosphate/pharmacology , Animals , Astrocytes/drug effects , Blotting, Northern , Calcium/metabolism , Cell Hypoxia/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Chromatography, High Pressure Liquid , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/physiology , Extracellular Space/drug effects , Extracellular Space/metabolism , Glucose/deficiency , Pyrimidines/metabolism , RNA/analysis , RNA/biosynthesis , Rats , Receptors, Purinergic P2Y2 , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Stroke/metabolism
8.
Int J Immunopathol Pharmacol ; 18(3): 417-30, 2005.
Article in English | MEDLINE | ID: mdl-16164825

ABSTRACT

Astrocytes have been recognized as important elements in controlling inflammatory as well as immune processes in the central nervous system (CNS). Recently, glial cells have been shown to produce cysteinyl leukotrienes (CysLTs) which are known lipid mediators of inflammation and whose extracellular concentrations rise under different pathological conditions in the brain. In the same conditions also extracellular concentrations of ATP dramatically increase reaching levels able to activate P2X7 ionotropic receptors for which an emerging role in neuroinflammation and neurodegeneration has been claimed. RTPCR analysis showed that primary cultures of rat brain astrocytes express P2X7 receptors. Application of the selective P2X7 agonist benzoyl benzoly ATP (BzATP) markedly increased [Ca2+]i which was mediated by a calcium influx from the extracellular milieu. The P2X7 antagonist, oATP, suppressed the BzATP-induced calcium increase. Consistent with the evidence that increased calcium levels activate the leukotriene biosynthetic pathway, challenge of astrocytes with either the calcium ionophore A23187 or BzATP significantly increased CysLT production and the cell pre-treatment with EGTA abolished these effects. Again the P2X7 antagonist prevented the BzATP-mediated CysLT efflux, whereas the astrocyte pretreatment with MK-571, a CysLT1 receptor antagonist, was ineffective. The astrocyte pre-treatment with a cocktail of inhibitors of ATP binding cassette (ABC) proteins reduced the BzATP-mediated CysLT production confirming that ABC transporters are involved in the release of CysLTs. The astrocyte P2X7- evoked rise of CysLT efflux was abolished in the presence of MK-886, an inhibitor of 5-lipoxygenase activating protein (FLAP) whose expression, along with that of 5-lipoxygenase (5-LO) was reported by Northern Blot analysis. The stimulation of P2X7 induced an up-regulation of FLAPmRNA that was reduced by the antagonist oATP. These data suggest that in rat brain cultured astrocytes P2X7ATP receptors may participate in the control of CysLT release thus further supporting a role for extracellular ATP as an integral component of the inflammatory brain response.


Subject(s)
Astrocytes/metabolism , Brain/cytology , Cysteine/biosynthesis , Cysteine/metabolism , Leukotrienes/biosynthesis , Leukotrienes/metabolism , Receptors, Purinergic P2/metabolism , 5-Lipoxygenase-Activating Proteins , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Affinity Labels/pharmacology , Animals , Astrocytes/cytology , Astrocytes/drug effects , Calcimycin/pharmacology , Carrier Proteins/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Chelating Agents/pharmacology , Cysteine/chemistry , Dose-Response Relationship, Drug , Egtazic Acid/pharmacology , Indoles/pharmacology , Ionophores/pharmacology , Leukotriene Antagonists/pharmacology , Leukotrienes/chemistry , Lipoxygenase Inhibitors/pharmacology , Membrane Proteins/metabolism , Propionates/pharmacology , Purinergic P2 Receptor Antagonists , Quinolines/pharmacology , RNA, Messenger/metabolism , Rats , Up-Regulation
9.
Neuroscience ; 128(4): 697-712, 2004.
Article in English | MEDLINE | ID: mdl-15464278

ABSTRACT

Guanosine 5' triphosphate (GTP), acting synergistically with the nerve growth factor (NGF), enhances the proportion of neurite-bearing cells in cultures of PC12 rat pheochromocytoma cells. We studied the transduction mechanisms activated by GTP in PC12 cells and found that addition of GTP (100 microM) increased intracellular calcium concentration ([Ca(2+)](i)) in cells that were between 60 and 70% confluent. Addition of GTP also enhanced activation of NGF-induced extracellular regulated kinases (ERKs) and induced Ca(2+) mobilization. This mobilization, due to the activation of voltage-sensitive and ryanodine-sensitive calcium channels, as well as pertussis toxin-sensitive purinoceptors, modulates Ca(2+)-activated K(+) channels not involved in activation of ERKs. The results presented here indicate that GTP-triggered [Ca(2+)](i) increase may be a key event in GTP signal transduction, which can modulate activity of ERKs. The physiological importance of the GTP effect lies in its capacity to interact with the NGF-activated pathway to enhance neurite outgrowth from PC12 cells.


Subject(s)
Cell Differentiation/physiology , Extracellular Space/drug effects , Gallic Acid/analogs & derivatives , Guanosine Triphosphate/physiology , Nerve Growth Factor/physiology , PC12 Cells/cytology , Pyridoxal Phosphate/analogs & derivatives , Signal Transduction/physiology , Suramin/analogs & derivatives , Animals , Barbiturates/metabolism , Blotting, Western/methods , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Cell Count/methods , Chelating Agents/pharmacology , Clotrimazole/pharmacology , Diagnostic Imaging/methods , Dose-Response Relationship, Drug , Drug Synergism , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Fluorescent Antibody Technique/methods , Fluorescent Dyes/metabolism , Gallic Acid/pharmacology , Growth Inhibitors/pharmacology , Ionomycin/pharmacology , Ionophores/pharmacology , Isoxazoles/metabolism , Membrane Potentials/drug effects , Microscopy, Confocal/methods , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neurites/drug effects , Neurites/physiology , Nifedipine/pharmacology , Pertussis Toxin/pharmacology , Pyridoxal Phosphate/pharmacology , Rats , Suramin/pharmacology , Time Factors , Triazines/pharmacology
10.
Int J Immunopathol Pharmacol ; 17(3): 353-66, 2004.
Article in English | MEDLINE | ID: mdl-15461869

ABSTRACT

Extracellular non-adenine based purines are neuroprotective. Preliminary studies indicate that administration of the synthetic purine 4-[[3-(1,6 dihydro-6-oxo-9-purine-9-yl)-1-oxypropyl] amino] benzoic acid (AIT-082, leteprinim potassium) to rats immediately after acute spinal cord injury (SCI), improves functional outcome. The effects of potential new agents are often compared to methylprednisolone (MPSS). We evaluated the effects of AIT-082 and MPSS, separately and in combination, on the functional and morphological outcome of acute SCI in adult rats. After standardized T11-12 spinal cord compression rats were given intraperitoneally one of the following: vehicle (saline); MPSS (30 mg/kg or 60 mg/kg body weight, first dose 15 min after crush); AIT-082 (60 mg/kg body weight daily, first dose 15 min after crush); or AIT-082 plus MPSS. After 1, 3, or 21 days, the rats were perfused for histological analysis. AIT-082 administrations significantly reduced locomotor impairment from 121 days post-operatively. At 1 and 3 days post injury, AIT-082-treatment reduced tissue swelling, tissue loss and astrogliosis at the injured cords but did not alter the extent of hemorrhage and the number of macrophages and/or microglia. MPSS reduced hemorrhage and the number of macrophages and/or microglia, but did not alter astrogliosis. At 21 days, either AIT-082 or MPSS administration improved function and morphology similarly (less tissue loss and astrogliosis). In contrast, administration of AIT-082 and MPSS together abolished the beneficial effects observed when either drug was given individually. These results suggest that MPSS and AIT-082 may exert their beneficial effects through different and potentially antagonistic pathways.


Subject(s)
Aminobenzoates/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Hypoxanthines/therapeutic use , Methylprednisolone/therapeutic use , Neuroprotective Agents/therapeutic use , Spinal Cord Injuries/drug therapy , Aminobenzoates/administration & dosage , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Drug Interactions , Female , Gliosis/pathology , Hindlimb/physiology , Hypoxanthines/administration & dosage , Immunohistochemistry , Locomotion/physiology , Methylprednisolone/administration & dosage , Nerve Crush , Neuroprotective Agents/administration & dosage , Rats , Rats, Wistar , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Treatment Outcome
11.
J Neurosci Res ; 65(2): 184-9, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11438987

ABSTRACT

Purinergic receptors play an important role in the regulation of free cytosolic calcium concentration ([Ca(2+)](i)) in astrocytes. In the present study, 10 microM adenosine caused an increase in [Ca(2+)](i) in 85% of the cultures studied, i.e., primary cultures of mouse astrocytes, differentiated by culturing in the presence of dibutyryl cyclic AMP. Antagonist sensitivity and rapid desensitization suggested that it did so by acting on A3 receptors. Another biologically important purine, guanosine, also caused an increase in astrocytic [Ca(2+)](i) (at concentrations of 0.1-100 microM). Although this response did not show the same rapid desensitization as the response to adenosine, it may also have been exerted on an A3 receptor. It supports this idea that inosine also caused an increase in [Ca(2+)](i), because inosine is known to activate A3 receptors in mast cells and structurally is even more closely related to guanosine than is adenosine.


Subject(s)
Adenosine/pharmacology , Astrocytes/drug effects , Brain/drug effects , Calcium/metabolism , Cytosol/drug effects , Guanosine/pharmacology , Purinergic P1 Receptor Agonists , Adenosine/metabolism , Animals , Astrocytes/cytology , Astrocytes/metabolism , Brain/cytology , Brain/metabolism , Cells, Cultured/cytology , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Cytosol/metabolism , Dose-Response Relationship, Drug , Extracellular Space/drug effects , Extracellular Space/metabolism , Guanosine/metabolism , Mice , Phosphodiesterase Inhibitors , Purinergic P1 Receptor Antagonists , Quinazolines/pharmacology , Receptor, Adenosine A3 , Receptors, Purinergic P1/metabolism , Theophylline/pharmacology , Triazoles/pharmacology , Xanthines/pharmacology
12.
Exp Neurol ; 169(2): 392-9, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11358452

ABSTRACT

4-[[3-(1,6-dihydro-6-oxo-9-purin-9-yl)-1-oxopropyl]amino]benzoic acid (AIT-082) is an hypoxanthine derivative that stimulates in vitro neurite outgrowth and the production of adenosine and neurotrophins from astrocytes. These effects may predict an in vivo neuroprotective activity of the drug. Thus, we evaluated whether AIT-082 protected against a long-term excitotoxicity of hippocampal neurons following status epilepticus induced in rats by i.p. injection of kainate (12 mg/kg). The epileptogenic effect of kainate was evaluated by monitoring behavioral signs and by electroencephalographic (EEG) recording (80% of the animals showed status epilepticus with a latency of 96.8 +/- 7.4 min starting from the injection). In surviving rats (40% of the injected animals) the neurotoxic effect was evaluated by measuring glutamic acid decarboxylase (GAD) activity, as an index of loss of hippocampal GABAergic neurons, by evaluating the body weight after 7 days and by histological examination of hippocampi. The GAD activity was reduced by 44 +/- 8%, and neuronal loss (about 70%) was found in the CA3c, the CA1 area, and in the dentate gyrus. A single dose of diazepam (20 mg/kg; i.p., 20 min before the kainate injection) almost completely inhibited both seizures and neurotoxicity, ensuring survival of animals. AIT-082 (60 mg/kg/day; i.p., for 7 days, starting from 20 min before the kainate injection) did not modify the seizures caused by kainate but, like diazepam, it decreased kainate-induced mortality, the reduction of GAD activity, and the loss of hippocampal neurons. These data confirm that AIT-082 is of potential interest for the experimental therapy of neurodegenerative disorders.


Subject(s)
Aminobenzoates , Electroencephalography/drug effects , Hippocampus/physiology , Hypoxanthines , Kainic Acid/toxicity , Neurons/physiology , Neuroprotective Agents/pharmacology , Purines/pharmacology , Seizures/prevention & control , Status Epilepticus/prevention & control , Status Epilepticus/physiopathology , Analysis of Variance , Animals , Cell Survival/drug effects , Dentate Gyrus/drug effects , Dentate Gyrus/pathology , Dentate Gyrus/physiology , Diazepam/pharmacology , Face , Glutamate Decarboxylase/metabolism , Hippocampus/drug effects , Hippocampus/pathology , Male , Motor Activity/drug effects , Motor Activity/physiology , Mouth , Neurons/drug effects , Neurons/pathology , Rats , Rats, Sprague-Dawley , Seizures/chemically induced , Seizures/physiopathology , Status Epilepticus/chemically induced , Time Factors , gamma-Aminobutyric Acid/metabolism
13.
Int J Dev Neurosci ; 19(4): 395-414, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11378300

ABSTRACT

Astrocytes are involved in multiple brain functions in physiological conditions, participating in neuronal development, synaptic activity and homeostatic control of the extracellular environment. They also actively participate in the processes triggered by brain injuries, aimed at limiting and repairing brain damages. Purines may play a significant role in the pathophysiology of numerous acute and chronic disorders of the central nervous system (CNS). Astrocytes are the main source of cerebral purines. They release either adenine-based purines, e.g. adenosine and adenosine triphosphate, or guanine-based purines, e.g. guanosine and guanosine triphosphate, in physiological conditions and release even more of these purines in pathological conditions. Astrocytes express several receptor subtypes of P1 and P2 types for adenine-based purines. Receptors for guanine-based purines are being characterised. Specific ecto-enzymes such as nucleotidases, adenosine deaminase and, likely, purine nucleoside phosphorylase, metabolise both adenine- and guanine-based purines after release from astrocytes. This regulates the effects of nucleotides and nucleosides by reducing their interaction with specific membrane binding sites. Adenine-based nucleotides stimulate astrocyte proliferation by a P2-mediated increase in intracellular [Ca2+] and isoprenylated proteins. Adenosine also, via A2 receptors, may stimulate astrocyte proliferation, but mostly, via A1 and/or A3 receptors, inhibits astrocyte proliferation, thus controlling the excessive reactive astrogliosis triggered by P2 receptors. The activation of A1 receptors also stimulates astrocytes to produce trophic factors, such as nerve growth factor, S100beta protein and transforming growth factor beta, which contribute to protect neurons against injuries. Guanosine stimulates the output of adenine-based purines from astrocytes and in addition it directly triggers these cells to proliferate and to produce large amount of neuroprotective factors. These data indicate that adenine- and guanine-based purines released in large amounts from injured or dying cells of CNS may act as signals to initiate brain repair mechanisms widely involving astrocytes.


Subject(s)
Adenine/physiology , Astrocytes/physiology , Brain Diseases/metabolism , Brain Injuries/metabolism , Brain/metabolism , Guanine/physiology , Nerve Tissue Proteins/physiology , Receptors, Purinergic P1/physiology , Receptors, Purinergic P2/physiology , Adenosine Triphosphate/physiology , Animals , Astrocytes/drug effects , Brain/pathology , Brain Diseases/pathology , Brain Injuries/pathology , Cell Division , Chickens , Energy Metabolism , Extracellular Space/metabolism , Guanosine Triphosphate/physiology , Humans , Ion Transport , Mice , Nerve Growth Factors/physiology , Neuroprotective Agents/pharmacology , Nucleosides/physiology , Nucleotides/physiology , Rats , Signal Transduction , Transforming Growth Factor beta/physiology
14.
Neuroscience ; 96(4): 817-24, 2000.
Article in English | MEDLINE | ID: mdl-10727799

ABSTRACT

Extracellular guanosine 5' triphosphate (GTP) enhances nerve growth factor-dependent neurite outgrowth from rat pheochromocytoma (PC12) cells; cultures of PC12 cells exposed to GTP and nerve growth factor together contain significantly more neurite-bearing cells than do those exposed to either nerve growth factor or GTP alone [Gysbers J. W. and Rathbone M. P. (1996) Int. J. devl Neurosci. 14, 19-34]. PC12 cells contain specific cell surface binding sites for extracellular GTP, which do not bind ATP or uridine 5' triphosphate. Exposure of PC12 cells to extracellular GTP (300microM) produced a robust and sustained increase in intracellular Ca(2+) ([Ca(2+)](i)), different from the transient response to the addition of ATP. The GTP-induced [Ca(2+)](i) increase was blocked by the L-type calcium channel inhibitor, nifedipine. The L-type Ca(2+) channel inhibitors, nifedipine or verapamil, also inhibited the enhancement of neurite outgrowth by GTP, but did not affect neurite outgrowth stimulated by nerve growth factor alone. Pre-treatment of PC12 cells with ryanodine (0.5-50microM) depleted calcium from internal stores and prevented the further release of calcium by GTP. Similarly, pre-treatment of PC12 cells with thapsigargin (an inhibitor of internal store Ca(2+)/ATPase) or dantrolene (which blocks Ca(2+) release from some of these stores) also reduced the enhancement of neurite outgrowth by GTP. Therefore, Ca(2+)-induced Ca(2+) release from specific stores, present in PC12 cells, is involved in the enhancement of nerve growth factor-induced neurite outgrowth by GTP, possibly acting at specific binding sites on the cell surface. GTP is proving to be an important extracellular trophic modulator in the central nervous system. These studies show that the neuritogenic actions of GTP involve moderate but sustained increases in intracellular Ca(2+) which are likely due to activation of L-type Ca(2+) channels and Ca(2+)-induced Ca(2+) release from intracellular stores. These effects of extracellular GTP are likely mediated at the cell surface and may be related to specific GTP binding sites which are distinct from G-proteins and from hitherto described purine nucleotide (P2) receptors. These data indicate a mechanism whereby the neuritogenic effects of GTP are mediated and emphasize the importance of considering GTP as a neurotrophic mediator.


Subject(s)
Calcium/metabolism , Extracellular Space/metabolism , Guanosine Triphosphate/metabolism , Guanosine Triphosphate/pharmacology , Intracellular Fluid/metabolism , Nerve Growth Factor/metabolism , Nerve Growth Factor/pharmacology , Neurites/drug effects , Neurites/metabolism , Animals , Binding Sites/drug effects , Binding Sites/physiology , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/drug effects , Calcium Channels, L-Type/metabolism , Cell Culture Techniques , Enzyme Inhibitors/pharmacology , Nifedipine/pharmacology , PC12 Cells , Protein Kinase C/drug effects , Protein Kinase C/metabolism , Radioligand Assay , Rats , Receptors, Cell Surface/drug effects , Receptors, Cell Surface/metabolism , Thapsigargin/pharmacology
15.
Glia ; 29(3): 202-11, 2000 Feb 01.
Article in English | MEDLINE | ID: mdl-10642747

ABSTRACT

Extracellular adenosine (Ado) and ATP stimulate astrocyte proliferation through activation of P(1) and P(2) purinoceptors. Extracellular GTP and guanosine (Guo), however, that do not bind strongly to these receptors, are more effective mitogens than ATP and Ado. Exogenous Guo, like GTP and 5'-guanosine-betagamma-imidotriphosphate (GMP-PNP), dose-dependently stimulated proliferation of rat cultured astrocytes; potency order GMP-PNP > GTP > or = Guo. The mitogenic effect of Guo was independent of the extracellular breakdown of GTP to Guo, because GMP-PNP, a GTP analogue resistant to hydrolysis, was the most mitogenic. In addition to a direct effect on astrocytes, Guo exerts its proliferative activity involving Ado. Exogenous Guo, indeed, enhanced the extracellular levels of endogenous Ado assayed by HPLC in the medium of cultured astrocytes. Culture pretreatment with Ado deaminase (ADA), that converts Ado into inosine, reduced but did not abolish Guo-induced astrocyte proliferation whereas erythro-9-(2-hydroxy-3-nonyl)adenine (EHNA), that inhibits ADA activity, amplified Guo effect. Moreover, the mitogenic activity of Guo was partly inhibited by 8-cyclopentyl-1,3-dipropylxanthine and alloxazine, antagonists of Ado A(1) and A(2B) receptors, respectively. Also microglia seem to be a target for the action of Guo. Indeed, the mitogenic effect of Guo on astrocytes was: i) increased proportionally to the number of microglial cells present in the astrocyte cultures; ii) amplified when purified cultures of astrocytes were supplemented with conditioned medium deriving from Guo-pretreated microglial cultures. These data indicate that the mitogenic effects exerted by exogenous Guo on rat astrocytes are mediated via complex mechanisms involving extracellular Ado and microglia-derived soluble factors.


Subject(s)
Adenosine/physiology , Astrocytes/cytology , Microglia/physiology , Animals , Cell Division/physiology , Cells, Cultured , Extracellular Space/metabolism , Fetus , Mitogens/physiology , Purines/chemistry , Purines/metabolism , Rats
16.
Glia ; 25(1): 93-8, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9888301

ABSTRACT

Brain ischemia stimulates release from astrocytes of adenine-based purines, particularly adenosine, which is neuroprotective. Guanosine, which has trophic properties that may aid recovery following neurological damage, is present in high local concentrations for several days after focal cerebral ischemia. We investigated whether guanine-based purines, like their adenine-based counterparts, were released from astrocytes and whether their release increased following hypoxia/hypoglycemia. HPLC analysis of culture medium of rat astrocytes showed spontaneous release of endogenous guanine-based purines at a higher rate than their adenine-based counterparts. The concentration of guanosine (approximately 120 nM) and adenosine (approximately 43 nM) in the culture medium remained constant, whereas concentrations of adenine and guanine nucleotides, particularly GMP, and their metabolites increased with time. Exposure of the cultures to hypoxia/hypoglycemia for 30 min increased the extracellular concentration of adenine-based purines by 2.5-fold and of guanine-based purines by 3.5-fold. Following hypoxia/hypoglycemia extracellular adenine nucleotide levels increased further. Adenosine concentration increased, but not proportionally to nucleotide levels. Accumulation of adenosine metabolites indicated it was rapidly metabolized. Conversely, the concentrations of extracellular guanine-based nucleotides remained elevated and the concentration of guanosine continued to increase. These data indicate that astrocytes are a major source of guanine-based purines, the release of which is markedly increased following hypoxia/hypoglycemia, permitting them to exert neurotrophic effects.


Subject(s)
Astrocytes/metabolism , Cell Hypoxia/physiology , Guanine/metabolism , Hypoglycemia/metabolism , Purines/metabolism , Adenosine/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Differentiation , Cells, Cultured , Chromatography, High Pressure Liquid , Rats , Spectrophotometry, Ultraviolet
17.
Prog Neurobiol ; 59(6): 663-90, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10845757

ABSTRACT

In addition to their well known roles within cells, purine nucleotides such as adenosine 5' triphosphate (ATP) and guanosine 5' triphosphate (GTP), nucleosides such as adenosine and guanosine and bases, such as adenine and guanine and their metabolic products xanthine and hypoxanthine are released into the extracellular space where they act as intercellular signaling molecules. In the nervous system they mediate both immediate effects, such as neurotransmission, and trophic effects which induce changes in cell metabolism, structure and function and therefore have a longer time course. Some trophic effects of purines are mediated via purinergic cell surface receptors, whereas others require uptake of purines by the target cells. Purine nucleosides and nucleotides, especially guanosine, ATP and GTP stimulate incorporation of [3H]thymidine into DNA of astrocytes and microglia and concomitant mitosis in vitro. High concentrations of adenosine also induce apoptosis, through both activation of cell-surface A3 receptors and through a mechanism requiring uptake into the cells. Extracellular purines also stimulate the synthesis and release of protein trophic factors by astrocytes, including bFGF (basic fibroblast growth factor), nerve growth factor (NGF), neurotrophin-3, ciliary neurotrophic factor and S-100beta protein. In vivo infusion into brain of adenosine analogs stimulates reactive gliosis. Purine nucleosides and nucleotides also stimulate the differentiation and process outgrowth from various neurons including primary cultures of hippocampal neurons and pheochromocytoma cells. A tonic release of ATP from neurons, its hydrolysis by ecto-nucleotidases and subsequent re-uptake by axons appears crucial for normal axonal growth. Guanosine and GTP, through apparently different mechanisms, are also potent stimulators of axonal growth in vitro. In vivo the extracellular concentration of purines depends on a balance between the release of purines from cells and their re-uptake and extracellular metabolism. Purine nucleosides and nucleotides are released from neurons by exocytosis and from both neurons and glia by non-exocytotic mechanisms. Nucleosides are principally released through the equilibratory nucleoside transmembrane transporters whereas nucleotides may be transported through the ATP binding cassette family of proteins, including the multidrug resistance protein. The extracellular purine nucleotides are rapidly metabolized by ectonucleotidases. Adenosine is deaminated by adenosine deaminase (ADA) and guanosine is converted to guanine and deaminated by guanase. Nucleosides are also removed from the extracellular space into neurons and glia by transporter systems. Large quantities of purines, particularly guanosine and, to a lesser extent adenosine, are released extracellularly following ischemia or trauma. Thus purines are likely to exert trophic effects in vivo following trauma. The extracellular purine nucleotide GTP enhances the tonic release of adenine nucleotides, whereas the nucleoside guanosine stimulates tonic release of adenosine and its metabolic products. The trophic effects of guanosine and GTP may depend on this process. Guanosine is likely to be an important trophic effector in vivo because high concentrations remain extracellularly for up to a week after focal brain injury. Purine derivatives are now in clinical trials in humans as memory-enhancing agents in Alzheimer's disease. Two of these, propentofylline and AIT-082, are trophic effectors in animals, increasing production of neurotrophic factors in brain and spinal cord. Likely more clinical uses for purine derivatives will be found; purines interact at the level of signal-transduction pathways with other transmitters, for example, glutamate. They can beneficially modify the actions of these other transmitters.


Subject(s)
Brain/metabolism , Neuroglia/metabolism , Neurons/metabolism , Purines/metabolism , Animals , Humans
18.
Expert Opin Investig Drugs ; 8(8): 1255-62, 1999 Aug.
Article in English | MEDLINE | ID: mdl-15992149

ABSTRACT

The synthetic purine 4-[[3-(1,6 dihydro-6-oxo-9-purin-9-yl)-1-oxypropyl] amino] benzoic acid (AIT-082, Neotrofin, leteprinim potassium) possesses several biological properties of note: it stimulates outgrowth of neurites from PC12 cells and neurones, stimulates synthesis and/or release of neurotrophic factors from astrocytes, enhances nerve fibre regeneration in vivo and enhances of memory in animals and humans. AIT-082 also protects against glutamate neurotoxicity in vitro and in vivo, which has led to successful tests of AIT-082 in animal models of acute central nervous system injury. In such cases, AIT-082 probably functions by both acutely reducing glutamate excitotoxicity and, over a longer period, by enhancing neuronal sprouting and functional recovery.

19.
Alzheimer Dis Assoc Disord ; 12 Suppl 2: S36-45, 1998.
Article in English | MEDLINE | ID: mdl-9769028

ABSTRACT

This article reviews the effects of extracellular purine bases, nucleosides, and nucleotides as intracellular signaling molecules with trophic effects on cells after insults to the brain and spinal cord. Astrocytes are the principal source of extracellular purines in brain after injury, ischemia, or trauma. In vitro and in vivo extracellular purines have both immediate and long-term trophic effects, including stimulation of astrocyte and neuronal differentiation, mitosis, morphogenesis, apoptosis, and stimulation of growth and trophic factor synthesis. The effects of the nucleoside adenosine and the nucleotide adenosine triphosphate (ATP) are mediated principally via specific receptors on the cell surface coupled to a series of signaling cascades. Unlike adenosine and ATP, guanosine and guanosine triphosphate (GTP) do not act at classical purine receptors. However, they exert similar effects on astrocytes, apparently by causing the astrocytes to release large amounts of adenosine and ATP over prolonged periods. The release of adenosine and ATP may be related to the effects of guanosine on the purine nucleoside transporters in the cell membrane, whereas the release of ATP may be due to the effects of GTP on the ATP-binding cassette (ABC) proteins. Physiologically, the effects of guanosine are important because this nucleoside, unlike adenosine, remains elevated for prolonged periods after brain injury.


Subject(s)
Adenosine Triphosphate/metabolism , Adenosine/metabolism , Astrocytes/physiology , Purines/metabolism , Receptors, Purinergic/physiology , Signal Transduction/physiology , Animals , Brain/cytology , Cell Division/physiology , Cells, Cultured , Humans , Rats
20.
Brain Res Bull ; 45(3): 297-9, 1998.
Article in English | MEDLINE | ID: mdl-9510422

ABSTRACT

The significance of guanine nucleotides and nucleosides in neurodegenerative disorders is suggested by recent reports that these molecules enhance neurite branching and astrocyte proliferation. The objective of this study was to investigate the influence of increased dopamine metabolism, produced by 5-day treatment of rabbits with reserpine (2 mg/kg) or levodopa (LD) (50 mg/kg), on striatal concentrations of guanosine, guanine, and their metabolites. Reserpine treatment decreased striatal guanosine by 41% and increased guanine by 50%, while LD decreased guanosine by 48% (all p < 0.01 vs. vehicle-treated controls). LD also increased guanine by 22% (not statistically significant). Xanthine and uric acid concentrations were unchanged. Because of the neurotrophic properties of guanosine and guanine, changes in striatal concentrations of these purines secondary to increased dopamine (DA) turnover may have relevance for survival of remaining dopaminergic neurons in Parkinson's disease (PD).


Subject(s)
Corpus Striatum/metabolism , Dopamine Agents/pharmacology , Dopamine/metabolism , Guanine/metabolism , Guanosine/metabolism , Reserpine/pharmacology , Animals , Corpus Striatum/cytology , Levodopa/pharmacology , Male , Neurons/drug effects , Rabbits
SELECTION OF CITATIONS
SEARCH DETAIL
...