Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
2.
J Biol Regul Homeost Agents ; 32(6): 1349-1353, 2018.
Article in English | MEDLINE | ID: mdl-30574738

ABSTRACT

Exracellular matrix (ECM) consists of a plethora of proteins and polysaccharides, which aggregate into an organized network connected to the surface of the producing cells. It is structurally and functionally present in all components of tissues and organs and represents the substrate on which cells adhere, migrate, proliferate and differentiate, influencing their survival, shape and function. In response to acute (trauma) or chronic (degenerative) insults, brain ECM modifies its composition and function, actively contributing to "scar forming" gliosis or tissue degeneration/remodelling. Moreover, morphological changes in dendritic spines associated with extracellular matrix remodeling play key roles in rewiring synaptic circuitry pertinent to memory formation. In the present report, we collected the main acquisitions on the functional interplay between ECM alterations and the adenine-/guaninebased purine system with particular regard on how purine compounds and their respective receptors may affect and be affected by changes of the cerebral ECM.


Subject(s)
Brain/physiology , Central Nervous System/physiology , Extracellular Matrix/physiology , Receptors, Purinergic/physiology , Humans
3.
Int J Immunopathol Pharmacol ; 29(2): 168-79, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26684641

ABSTRACT

The development of pharmacokinetics led this science to achieve a relevant role in the investigation of new chemical entities for therapeutic application, and has allowed a series of new useful realizations of out of patent drugs like prolonged release and delayed release formulations, therapeutic delivery system (TDS) for drugs to be active in systemic circulation avoiding the first pass effect, orodispersible and effervescent formulations, intramuscular and subcutaneous depot formulations acting over a long period, oral inhalatory systems, and drug association at fixed dose. The above applications had pharmacokinetics as protagonist and have required the support from bioanalytical methods to assay drug concentrations, even in pg·mL(-1) of plasma, that really have paralleled the synergic development of pharmacokinetics.The complexity of the above realizations required specific guidelines from the regulatory authorities, mainly the US FDA and EU EMA, which have normalized and, in most cases, simplified the above applications admitting some waivers of in vivo bioequivalence.However, this review highlights some critical points, not yet focused on by operating guidelines, which need to be clarified by regulatory authorities. One of the most relevant issues is about the planning and conducting bioavailability and bioequivalence trials with endogenous substances, that possess own homeostatic equilibria with fluctuations, in some cases with specific rhythms, like melatonin and female sex hormones. The baseline subtraction required by guidelines to define the net contribute to the exogenous absorbed drug in most cases is a non-solvable problem.


Subject(s)
Pharmaceutical Preparations/chemistry , Pharmaceutical Preparations/metabolism , Animals , Chemistry, Pharmaceutical/methods , Humans , Pharmaceutical Research/methods , Therapeutic Equivalency , United States , United States Food and Drug Administration
4.
Panminerva Med ; 54(1 Suppl 4): 53-8, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23241935

ABSTRACT

Guanosine has been reported to exert neuroprotective effects. We recently reported that, following intraperitoneal (i.p.) injection to rats, it resulted to be widely distributed. Its metabolic product guanine also rapidly increased in all the tissues, including brain, after i.p. injection of guanosine and consistently we found a significant enzymatic activity of a soluble purine nucleoside phosphorylase in the plasma of the treated animals. In this study the effect of per os administration of guanosine or guanine to rats submitted to passive avoidance task has been evaluated. Guanosine (4 and 8 mg/kg) administered pretraining impaired retention in the passive avoidance task and was unable to prevent the amnesic effect caused by 100 mg/kg N-omega-nitro-l-arginine methyl ester (L-NAME), an inhibitor of the nitric oxide synthase (NOS) known to reduce the capability of treated animals to acquire or retain informations in several learning tasks. On the contrary, guanine (4 and 8 mg/kg), which per se did not modify the latency to step-trough in the passive avoidance task, when administered pretraining 15 min before L-NAME prevented, in a dose dependent manner, the amnesic effect of the NOS inhibitor. Moreover the nucleobase was able to rescue the memory trace also when administered after training. Neither guanosine nor guanine had effects on locomotor activity. These results indicate that guanine can exert important biological activities which may be different from those mediated by its precursor guanosine, thus this evenience should be taken into account when the biological effects of guanosine are evaluated.


Subject(s)
Guanine/therapeutic use , Guanosine/therapeutic use , Learning/drug effects , Memory/drug effects , NG-Nitroarginine Methyl Ester/therapeutic use , Purines/chemistry , Administration, Oral , Animals , Avoidance Learning/drug effects , Brain/drug effects , Locomotion , Male , NG-Nitroarginine Methyl Ester/pharmacology , Neuroprotective Agents/chemistry , Neuroprotective Agents/metabolism , Neuroprotective Agents/therapeutic use , Rats , Rats, Wistar
5.
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
6.
Panminerva Med ; 2012 Nov 09.
Article in English | MEDLINE | ID: mdl-23138719

ABSTRACT

Guanosine has been reported to exert neuroprotective effects. We recently reported that, following intraperitoneal (i.p.) injection to rats, it resulted to be widely distributed. Its metabolic product guanine also rapidly increased in all the tissues, including brain, after i.p. injection of guanosine and consistently we found a significant enzymatic activity of a soluble purine nucleoside phosphorylase in the plasma of the treated animals. In this study the effect of per os administration of guanosine or guanine to rats submitted to passive avoidance task has been evaluated. Guanosine (4 and 8 mg/kg) administered pretraining impaired retention in the passive avoidance task and was unable to prevent the amnesic effect caused by 100 mg/kg N-omega-nitro-l-arginine methyl ester (L-NAME), an inhibitor of the nitric oxide synthase (NOS) known to reduce the capability of treated animals to acquire or retain informations in several learning tasks. On the contrary, guanine (4 and 8 mg/kg), which per se did not modify the latency to step-trough in the passive avoidance task, when administered pretraining 15 min before L-NAME prevented, in a dose dependent manner, the amnesic effect of the NOS inihibitor. Moreover the nucleobase was able to rescue the memory trace also when administered after training. Neither guanosine nor guanine had effects on locomotor activity. These results indicate that guanine can exert important biological activities which may be different from those mediated by its precursor guanosine, thus this evenience should be taken into account when the biological effects of guanosine are evaluated.

7.
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
8.
J Biol Regul Homeost Agents ; 24(3): 297-306, 2010.
Article in English | MEDLINE | ID: mdl-20846477

ABSTRACT

Amyloid-beta (Abeta) peptide aggregation forms such as soluble oligomers (O) have a causal role in neuronal dysfunction and death associated with Alzheimer?s Disease (AD). The main efforts for the development of neuroprotective drugs are therefore focused on preventing Abeta production, aggregation or downstream neurotoxic events. We therefore investigated the effect of guanosine (GUO), a guanine based purine, that exerts neurotrophic and neuroprotective effects. The GUO showed the ability to reduce neuronal death in terms of apoptosis, but not necrosis, elicited by Abeta1-42O in human neuroblastoma SH-SY5Y cells. The neuroprotective effect was recorded only when the GUO was added simultaneously to treatment of the SH-SY5Y cells with Abeta1-42O. By contrast, the GUO treatment of SH-SY5Y cells before and after the appearance of beta1-42O toxicity had no neuroprotective effects. The employment of specific inhibitors showed the involvement of neuronal survival pathways, such as PI3K?Akt and MAPK-ERK for the GUO anti-apoptotic effects observed. In parallel, the SH-SY5Y cells treated with GUO, in experimental conditions similar to those adopted to evaluate neuronal death, showed a marked decrease of the early reactive oxygen species formation induced by Abeta1-42O and pro-oxidant H2O2. In the same neuronal model, GUO was also shown to inhibit the extra- and intra-cellular Abeta1-42 release as well as the beta-secretase activity evoked by H2O2 pro-oxidant action. Based on these findings, GUO and other guanine based purines appear to be a promising class of compounds with neuroprotective properties that may play an important role in the therapy of AD.


Subject(s)
Amyloid beta-Peptides/toxicity , Guanosine/pharmacology , Neuroblastoma/metabolism , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Peptide Fragments/toxicity , Antioxidants/pharmacology , Cell Line, Tumor , Humans , MAP Kinase Signaling System , Neuroblastoma/pathology , Proto-Oncogene Proteins c-akt/metabolism
9.
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
10.
Minerva Ginecol ; 59(2): 175-81, 2007 Apr.
Article in Italian | MEDLINE | ID: mdl-17505459

ABSTRACT

In the last decade, ''fertiloscopy'', a new mini-invasive diagnostic technique, is becoming more and more popular: it is a good alternative to the diagnostic laparoscopy, a standard procedure but surely not harmless, very often capable to discover pathologies in asymptomatic patients. Fertiloscopy allows the visualization of the posterior pelvis (posterior face of the uterus, ovaries, tubes and intestinal ansae with the rectum), with a technique of introducing an optical device in the pouch of Douglas, through the posterior vaginal fornix, under previous general or local anesthesia. When fertiloscopy is performed under local anesthesia, it can comfortably be carried out in out-patient departments and it is generally well tolerated by patients, who follow the whole procedure on the monitor. Moreover, it is possible to perform small interventions, such as adhesiolysis, ovarian drilling, coagulation of endometriosis spots and to perform chromosalpingoscopy and salpingoscopy, important investigations in the diagnostic iter of unexplained female infertility. With fertiloscopy, the patient, therefore, can avoid a real surgical intervention, such as diagnostic laparoscopy, and also uncomfortable examinations, such as hysterosalpingography.


Subject(s)
Endoscopy/methods , Infertility, Female/diagnosis , Infertility, Female/surgery , Endoscopes , Equipment Design , Fallopian Tubes , Female , Humans , Intestines , Ovary , Uterus
11.
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
12.
Neurobiol Aging ; 27(1): 98-104, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16298245

ABSTRACT

Quantitative proton magnetic resonance spectroscopy (MRS) was used to determine region-specific metabolic changes in young and aged animals subjected to a long-term hypoxic-ischemic injury. Focal ischemia, which was studied as an experimental stroke model, was induced in 3- and 24-month-old rats by unilateral common carotid artery occlusion associated with 24 h of hypoxia. Eight metabolites were quantified from extracts in three different brain regions (hippocampus, frontoparietal and occipital cortices) from both the ipsilateral and contralateral sides. Our findings showed significant differences in lactate and myo-inositol concentration values in the hippocampus of the aged rats as compared to the same area of the young adult group under normoxic conditions. After hypoxia-ischemia (HI), the most relevant changes in metabolite concentrations were found in the hippocampal region of both young and aged groups as compared to their age-matched controls. Of the three brain areas under investigation, the hippocampus proved to be particularly susceptible to the prolonged hypoxia-ischemia perturbation. The effects were more evident in the aged animals.


Subject(s)
Aging/metabolism , Brain/metabolism , Hypoxia-Ischemia, Brain/metabolism , Magnetic Resonance Spectroscopy/methods , Neurotransmitter Agents/metabolism , Age Factors , Animals , Brain Chemistry , Chronic Disease , Female , Neurotransmitter Agents/analysis , Protons , Rats , Rats, Wistar , Time Factors , Tissue Distribution
13.
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
14.
Int J Immunopathol Pharmacol ; 18(2): 255-68, 2005.
Article in English | MEDLINE | ID: mdl-15888248

ABSTRACT

Inflammation is widely recognized as contributing to the pathology of acute and chronic neurodegenerative conditions. Microglial cells are pathologic sensors in the brain and activated microglia have been viewed as detrimental. Leukotriene, including cysteinyl leukotrienes (CysLTs) are suggested to be involved in brain inflammation and neurological diseases and ATP, by its receptors is a candidate for microglia activation. A23187 (10 microM) stimulated microglia to co-release CysLTs and [3H] adenine based purines ([3H] ABPs), mainly ATP. The biosynthetic production of CysLTs was abolished by 10 microM MK-886, an inhibitor of 5-lipoxygenase-activating protein activity. RT-PCR analysis showed that microglia expressed both CysLT1 / CysLT2 receptors, P2Y1ATP receptors and several members of the ATP binding cassette (ABC) transporters including MRP1, MRP4 and Pgp. The increase in [Ca2+]i elicited by LTD4 (0.1 microM) and 2MeSATP (100 microM), agonists for CysLT- and P2Y1-receptors, was abolished by the respective antagonists, BAYu9773 (0.5 microM) and suramin (50 microM). The stimulation of both receptor subtypes, induced a concomitant increase in the release of both [3H] ABPs and CysLTs that was blocked by the antagonists and significantly reduced by a cocktail of ABC transporter inhibitors, BAPTA/AM (intracellular Ca2+ chelator) and staurosporine (0.1 microM, PKC blocker). P2Y antagonist was unable to antagonise the effects of LTD4 and BAYu9773 did not reduce the effects of 2MeSATP. These data suggest that: i) the efflux of purines and cysteinyl-leukotrienes is specifically and independently controlled by the two receptor types, ii) calcium, PKC and the ABC transporter system can reasonably be considered common mechanisms underlying the release of ABPs and CysLTs from microglia. The blockade of P2Y1 or CysLT1/CysLT2 receptors by specific antagonists that abolished the raise in [Ca2+]i and drastically reduced the concomitant efflux of both compounds, as well as the effects of BAPTA and staurosporine support this hypothesis. In conclusion, the data of the present study suggest a cross talk between the purine and leukotriene systems in a possible autocrine/paracrine control of the microglia-mediated initiation and progression of an inflammatory response.


Subject(s)
Cysteine/biosynthesis , Leukotrienes/biosynthesis , Membrane Proteins/metabolism , Microglia/metabolism , Purines/biosynthesis , Receptors, Leukotriene/metabolism , Receptors, Purinergic P2/metabolism , ATP-Binding Cassette Transporters/metabolism , Animals , Brain/cytology , Calcium/metabolism , Cells, Cultured , Membrane Proteins/antagonists & inhibitors , Microglia/drug effects , Purinergic P2 Receptor Antagonists , Rats , Receptor Cross-Talk , Receptors, Purinergic P2Y1
15.
J Cereb Blood Flow Metab ; 21(9): 1013-33, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11524608

ABSTRACT

Metabotropic glutamate (mGlu) receptors have been considered as potential targets for neuroprotective drugs, but the lack of specific drugs has limited the development of neuroprotective strategies in experimental models of acute or chronic central nervous system (CNS) disorders. The advent of potent and centrally available subtype-selective ligands has overcome this limitation, leading to an extensive investigation of the role of mGlu receptor subtypes in neurodegeneration during the last 2 years. Examples of these drugs are the noncompetitive mGlu1 receptor antagonists, CPCCOEt and BAY-36-7620; the noncompetitive mGlu5 receptor antagonists, 2-methyl-6-(phenylethynyl)pyridine, SIB-1893, and SIB-1757; and the potent mGlu2/3 receptor agonists, LY354740 and LY379268. Pharmacologic blockade of mGlu1 or mGlu5 receptors or pharmacologic activation of mGlu2/3 or mGlu4/7/8 receptors produces neuroprotection in a variety of in vitro or in vivo models. MGlu1 receptor antagonists are promising drugs for the treatment of brain ischemia or for the prophylaxis of neuronal damage induced by synaptic hyperactivity. MGlu5 receptor antagonists may limit neuronal damage induced by a hyperactivity of N-methyl-d-aspartate (NMDA) receptors, because mGlu5 and NMDA receptors are physically and functionally connected in neuronal membranes. A series of observations suggest a potential application of mGlu5 receptor antagonists in chronic neurodegenerative disorders, such as amyotrophic lateral sclerosis and Alzheimer disease. MGlu2/3 receptor agonists inhibit glutamate release, but also promote the synthesis and release of neurotrophic factors in astrocytes. These drugs may therefore have a broad application as neuroprotective agents in a variety of CNS disorders. Finally, mGlu4/7/8 receptor agonists potently inhibit glutamate release and have a potential application in seizure disorders. The advantage of all these drugs with respect to NMDA or AMPA receptor agonists derives from the evidence that mGlu receptors do not "mediate," but rather "modulate" excitatory synaptic transmission. Therefore, it can be expected that mGlu receptor ligands are devoid of the undesirable effects resulting from the inhibition of excitatory synaptic transmission, such as sedation or an impairment of learning and memory.


Subject(s)
Brain Diseases/drug therapy , Neuroprotective Agents/pharmacology , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Animals , Humans
16.
J Neurochem ; 78(3): 435-45, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11483646

ABSTRACT

The mGlu2/3 receptor agonists 4-carboxy-3-hydroxyphenylglycine (4C3HPG) and LY379268 attenuated NMDA toxicity in primary cultures containing both neurons and astrocytes. Neuroprotection was abrogated by PD98059 and LY294002, which inhibit the mitogen activated protein kinase (MAPK) and the phosphatidylinositol-3-kinase (PI-3-K) pathways, respectively. Cultured astrocytes lost the ability to produce transforming growth factor-beta1 (TGF-beta1) in response to mGlu2/3 receptor agonists when co-incubated with PD98059 or LY294002. As a result, the glial medium was no longer protective against NMDA toxicity. Activation of the MAPK and PI-3-K pathways in cultured astrocytes treated with 4C3HPG or LY379268 was directly demonstrated by an increase in the phosphorylated forms of ERK-1/2 and Akt. Similarly to that observed in the culture, intracerebral or systemic injections of mGlu2/3 receptor agonists enhanced TGF-beta1 formation in the rat or mouse caudate nucleus, and this effect was reduced by PD98059. PD98059 also reduced the ability of LY379268 to protect striatal neurons against NMDA toxicity. These results suggest that activation of glial mGlu2/3 receptors induces neuroprotection through the activation of the MAPK and PI-3-K pathways leading to the induction of TGF-beta.


Subject(s)
Amino Acids/pharmacology , Astrocytes/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Glycine/pharmacology , MAP Kinase Signaling System/physiology , Neurons/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Metabotropic Glutamate/metabolism , Animals , Astrocytes/metabolism , Blotting, Northern , Cells, Cultured , Chromones/pharmacology , Corpus Striatum/cytology , Corpus Striatum/metabolism , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Flavonoids/pharmacology , Glycine/analogs & derivatives , Immunoblotting , Immunohistochemistry , Male , Mice , Morpholines/pharmacology , N-Methylaspartate/toxicity , Neurons/metabolism , Neuroprotective Agents/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1
17.
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
18.
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
19.
Brain Res ; 892(1): 78-85, 2001 Feb 16.
Article in English | MEDLINE | ID: mdl-11172751

ABSTRACT

Presynaptic inhibition is one of the major control mechanisms in the CNS. Previously we reported that A1 adenosine receptors are highly concentrated in the brain, including optic tectum, of trout and that they inhibited the release of glutamate. The optic tectum is heavily innervated by cholinergic nerve terminals. We have investigated whether A1 receptors inhibit the presynaptic release of acetylcholine and whether the inhibition is triggered by calcium. The release of [3H]ACh evoked by 30 mM KCl was Ca2+ dependent and it was dose-dependently inhibited by the A1 adenosine receptor agonist 2-chloro-N(6)-cyclopentyladenosine (CCPA) ranging between 10 nM to 100 microM. The maximum of inhibition was reached at 10 microM. The A1 receptor antagonist 8-cyclopentyltheopylline (CPT, 10 microM), reversed almost completely the inhibition induced by CCPA 10 microM. In Fura-2/AM loaded synaptosomes, K(+) depolarization raised [Ca2+](i) by about 64%. CCPA (10 microM) reduced the K(+)-evoked Ca2+ influx increase by about 48% and this effect was completely antagonised by CPT 10 microM. Synaptosome pretreatment with different Ca2+ channel blockers differently affected K(+)-evoked Ca2+ influx. This was not significantly modified by nifedipine (1 microM, L-type blocker) nor by omega-agatoxin IVA (0.3 microM, P/Q-type blocker), whereas about 50% reduction was shown by 0.5 microMomega-conotoxin GVIA (N-type blocker). Neurochemical parameters associated with cholinergic transmission and the density of A(1) adenosine receptors were measured in the trout optic tectum 12 days after unilateral eye ablation. A significant drop of both acetylcholinesterase (AChE) activity (24%) and choline acetyltransferase (CAT) activity (32%) was observed in deafferentated optic tectum, whereas the high affinity choline uptake did not parallel the decrease in enzyme activity. Eye ablation caused a marked decrease (43%) of A1 receptor density without changing the affinity. The K(+)-evoked release of [3H]ACh from synaptosomes of deafferentated was not modify as well as the efficacy of 10 microMCCPA in decreasing [3H]ACh release was not apparently modified.


Subject(s)
Acetylcholine/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Calcium Channel Blockers/pharmacology , Calcium/physiology , Receptors, Purinergic P1/physiology , Superior Colliculi/physiology , Synaptosomes/physiology , Theophylline/analogs & derivatives , Theophylline/pharmacology , Acetylcholinesterase/metabolism , Afferent Pathways/physiology , Animals , Choline O-Acetyltransferase/metabolism , Kinetics , Membrane Potentials/drug effects , Nifedipine/pharmacology , Potassium Chloride/pharmacology , Purinergic P1 Receptor Agonists , Synaptosomes/drug effects , Tritium , Trout , omega-Agatoxin IVA/pharmacology , omega-Conotoxin GVIA/pharmacology
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...