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










Publication year range
1.
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
2.
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
3.
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
4.
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
5.
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.

6.
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
7.
J Biol Regul Homeost Agents ; 25(1): 57-69, 2011.
Article in English | MEDLINE | ID: mdl-21382274

ABSTRACT

Mesenchymal stem cells (MSC), isolated from dental tissues, are largely studied for future application in regenerative dentistry. In this study, we used MSC obtained from human dental pulp (DPSC) of normal impacted third molars that, when cultured in lineage-specific inducing media, differentiate into osteoblasts and adipocytes (evaluated by Alizarin Red S and Red Oil O stainings, respectively), thus showing a multipotency. We confirmed that DPSC, grown under undifferentiating conditions, are negative for hematopoietic (CD45, CD31, CD34, CD144) and positive for mesenchymal (CD29, CD90, CD105, CD166, CD146, STRO-1) markers, that underwent down-regulation when cells were grown in osteogenic medium for 3 weeks. In this condition, they also exhibit an increase in the expression of osteogenic markers (RUNX-2, alkaline phosphatase) and extracellular calcium deposition, whereas the expression of receptors (VEGFR-1 and -2) for vascular endothelial growth factors (VEGF) and related VEGF binding proteins was similar to that found in undifferentiated DPSC. Exposure of DPSC growing under undifferentiating or osteogenic conditions to VEGF-A165 peptide (10-40 ng/ml) for 8 days dose- and time-dependently increased the number of proliferating cells without inducing differentiation towards endothelial lineage, as evaluated by the lack of expression of specific markers (CD31, CD34, CD144). Additionally, exposure of DPSC cultured in osteogenic medium to VEGF-A165 for a similar period enhanced cell differentiation towards osteoblasts as evaluated after 14 and 21 days by Alizarin Red S staining and alkaline phosphatase activity quantification. These findings may have clinical implications possibly facilitating tissue repair and remodeling.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Dental Pulp/metabolism , Mesenchymal Stem Cells/metabolism , Osteogenesis/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Adolescent , Antigens, Differentiation/metabolism , Cells, Cultured , Dental Pulp/cytology , Female , Humans , Male , Mesenchymal Stem Cells/cytology
8.
Int J Immunopathol Pharmacol ; 21(2): 309-18, 2008.
Article in English | MEDLINE | ID: mdl-18547474

ABSTRACT

The purpose of this study is to characterise the expression of matrix extracellular phosphoglycoprotein (MEPE) in cultured mesenchymal cells isolated from human dental papilla (PaMCs) of impacted third molars either before or during differentiation of these cells into osteo/odontoblasts. PaMCs, like mesenchymal cells deriving from human dental pulp (DPMCs), resulted positive for a number of mesenchymal markers including CD146 and STRO-1. During the first week in culture they showed a faster proliferation rate than DPMCs, coupled to an earlier down-regulation of MEPE. Also when the cells were further cultured in osteogenic medium (containing beta-glycerophosphate, ascorbic acid and dexamethasone) for 40 days, MEPE down-regulation coupled to an increased expression of osteogenic markers, such as osteocalcin and alkaline phosphatase, occurred earlier in PaMCs than in DPMCs. Thus, our data, indicating that also in PaMCs MEPE expression is higher when cells proliferate, whereas it is downregulated as cells differentiated, are in favour of a role of MEPE as an early regulator of odontogenic differentiation. We also confirm the superior proliferative potential of PaMCs in comparison with DPMCs, coupled to a more rapid induction of osteogenic differentiation. Therefore, these cells represent an optimal source to be conveniently used for dental tissue engineering and tooth regeneration.


Subject(s)
Dental Papilla/physiology , Extracellular Matrix Proteins/biosynthesis , Glycoproteins/biosynthesis , Mesenchymal Stem Cells/physiology , Osteoblasts/physiology , Osteocytes/physiology , Phosphoproteins/biosynthesis , Adolescent , Adult , Anthraquinones , Antigens, CD/metabolism , Blotting, Northern , Blotting, Western , Calcification, Physiologic/physiology , Cell Adhesion/physiology , Cell Differentiation/physiology , Cell Proliferation , Child , Flow Cytometry , Humans , Male , Mesenchymal Stem Cells/metabolism , Osteoblasts/metabolism , Osteocytes/metabolism , RNA/biosynthesis , RNA/isolation & purification
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.
Int J Immunopathol Pharmacol ; 20(4): 819-26, 2007.
Article in English | MEDLINE | ID: mdl-18179737

ABSTRACT

In this study we investigated the in vitro behaviour, morphostructure and extracellular matrix synthesis of human dental follicular stem cells (hDFSCs) isolated from human dental bud, which resulted to be positive for mesenchymal markers (CD29, CD90, CD146 and CD166) by FACS analysis. Cells were analysed by light and electronic microscopy to evaluate their biological response either at week 1, that is before differentiation, or at weeks 3-6, when they had been cultured in osteogenic medium onto a highly porous natural scaffold material (Bio-Oss). Microscopy analysis of primary culture cells showed they had a mesenchymal stem cell-like morphostructure, spindle shaped, similar to the culture of mesenchymal stem cells derived from adult bone marrow. Also, after osteogenic differentiation, these analyses indicate typical osteoblast morphostructure and reveale a tri-dimensional organization of the cells and deposition of extracellular matrix (ECM) in close contact with biomaterial. This approach would allow to personalize the scaffold for bone tissue engineering in order to accelerate the process of osteogenesis.


Subject(s)
Biocompatible Materials , Durapatite , Stem Cells/ultrastructure , Tissue Scaffolds , Tooth/cytology , Cell Differentiation , Cells, Cultured , Extracellular Matrix/ultrastructure , Fibroblasts/physiology , Fibroblasts/ultrastructure , Flow Cytometry , Humans , Microscopy, Electron, Scanning , Phenotype , Porosity , Stem Cells/immunology , Stem Cells/physiology , Tissue Engineering , Tooth/physiology , Tooth/ultrastructure
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.
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
13.
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
14.
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
15.
J Neurochem ; 88(5): 1179-85, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15009673

ABSTRACT

An overactivation of alpha-amino-3-hydroxy-5-methylisoxazole-4-propionate (AMPA)/kainate receptors has been implicated in the pathophysiology of oligodendrocyte damage in demyelinating disorders of the CNS. We decided to examine the effect of testosterone on excitotoxic death of oligodendrocytes because a gender difference exists in the incidence and disease course of multiple sclerosis. Short-term pure cultures of oligodendrocytes (4 days in vitro) were exposed to a brief pulse with kainate or AMPA + cyclothiazide for the induction of excitotoxicity. Exposure to testosterone enantate was slightly toxic per se and amplified both AMPA and kainate toxicity. Testosterone treatment induced all gene targets of p53, and amplified the induction of these genes induced by kainate. The effect of testosterone was mediated by the activation of androgen receptors and was resistant to the aromatase inhibitors, dl-aminoglutethimide and 4-hydroxyandrost-4-ene-3,17-dione. Testosterone treatment also potentiated the stimulation of 45Ca2+ influx induced by AMPA + cyclothiazide or kainate without changing the expression of the glutamate receptor (GluR) 1, -2/3, and -4 subunits of AMPA receptors or the GluR6/7 subunits of kainate receptors. We conclude that testosterone amplifies excitotoxic damage of oligodendrocytes acting at an early step of the death cascade triggered by AMPA/kainate receptors.


Subject(s)
Neurotoxins/toxicity , Oligodendroglia/drug effects , Testosterone/pharmacology , Animals , Benzothiadiazines/toxicity , Calcium/metabolism , Calcium/pharmacokinetics , Cells, Cultured , Drug Synergism , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Gene Expression/drug effects , Kainic Acid/toxicity , L-Lactate Dehydrogenase/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism , Quinoxalines/pharmacology , Rats , Receptors, AMPA/antagonists & inhibitors , Receptors, AMPA/metabolism , Receptors, Androgen/drug effects , Receptors, Androgen/metabolism , Receptors, Kainic Acid/antagonists & inhibitors , Receptors, Kainic Acid/metabolism , Time Factors , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/toxicity
16.
Minerva Cardioangiol ; 51(4): 405-9, 2003 Aug.
Article in Italian | MEDLINE | ID: mdl-12900722

ABSTRACT

Venous aneurysms are uncommon. Venous dilatation and large varices could be treated with injections of foam and sclerosing agents followed by local compression which obliterate or thrombose the aneurysmal space. However, the use of foam-sclerotherapy to obliterate venous aneurysms has never been reported before. The obstruction or sclerosis of the aneurysmal space should be obtained without altering the femoral flow. The patient is a 65-year old lady without history of trauma involving the femoral region. A compressible lump is present at the third, internal part of the left femoral fold. The lump is painless and easily obliterated by compression. Color duplex shows a large cavity (4 x 4 x 6 cm) and venous flow is visible. Flow produces a venous jet, visible with color and ejecting from the common femoral vein. A first injection is made under ultrasound guide using a foaming agent: 10 ml of foam including 4 ml of 3% sclerosing solution are injected. During the injection the femoral vein is compressed with the probe to avoid passage of the sclerosing foam into the femoral vein. After 2 weeks, ultrasound indicate only a partial occlusion of the aneurysm. The procedure is repeated and after 2 more weeks the ultrasound scan shows a complete occlusion of the aneurysm. The femoral vein and the long saphenous vein are patent. After 12 weeks the situation is unchanged and the results appear to be permanent. This new method, never described before is an important, minimally invasive method in case of venous aneurysms. Results from larger studies should be available to define indications and modalities of treatment.


Subject(s)
Aneurysm/therapy , Femoral Vein , Sclerosing Solutions/therapeutic use , Sclerotherapy , Surface-Active Agents/therapeutic use , Aged , Aneurysm/diagnostic imaging , Emulsions , Female , Femoral Vein/diagnostic imaging , Humans , Sclerosing Solutions/administration & dosage , Surface-Active Agents/administration & dosage , Ultrasonography, Doppler, Color , Ultrasonography, Interventional
17.
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
18.
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
19.
Glia ; 27(3): 275-81, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10457374

ABSTRACT

Pharmacological activation of A(1) adenosine receptor with 2-chloro-N6-cyclopentyladenosine (CCPA) or mGlu3 metabotropic glutamate receptors with (2S,2'R,3'R)-2-(2', 3'-dicarboxycyclopropyl)glycine (DCG-IV) or aminopyrrolidine-2R, 4R-dicarboxylate (2R,4R-APDC) enhanced the release of nerve growth factor (NGF) or S-100beta protein from rat cultured astrocytes. Stimulation of release by CCPA and DCG-IV or 2R,4R-APDC was inhibited by the A(1) adenosine receptor antagonist 8-cyclopentyl-1, 3-dipropylxanthine and by the mGlu2/3 receptor antagonist (2S,1'S, 2'S,3'R)-2-(2'-carboxy-3'-phenylcyclopropyl)glycine (PCCG-4), respectively. Time-course studies revealed a profound difference between the release of S-100beta protein and the release of NGF in response to extracellular signals. Stimulation of S-100beta protein exhibited rapid kinetics, peaking after 1 h of drug treatment, whereas the enhancement of NGF release was much slower, requiring at least 6 h of A(1) adenosine or mGlu3 receptor activation. In addition, stimulation of NGF but not S-100beta release was substantially reduced in cultures treated with the protein synthesis inhibitor cycloheximide. In addition, a 6-8 h treatment of cultured astrocytes with A(1) or mGlu3 receptor agonists increased the levels of both NGF mRNA and NGF-like immunoreactive proteins, including NGF prohormone. We conclude that activation of A(1) adenosine or mGlu3 receptors produces pleiotropic effects in astrocytes, stimulating the synthesis and/or the release of protein factors. Astrocytes may therefore become targets for drugs that stimulate the local production of neurotrophic factors in the CNS, and this may provide the basis for a novel therapeutic strategy in chronic neurodegenerative disorders.


Subject(s)
Astrocytes/physiology , Cyclopropanes/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Glycine/analogs & derivatives , Nerve Growth Factors/genetics , Neuroglia/physiology , Proline/analogs & derivatives , Receptors, Metabotropic Glutamate/physiology , Receptors, Purinergic P1/physiology , S100 Proteins/genetics , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Astrocytes/cytology , Astrocytes/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/physiology , Gene Expression Regulation/drug effects , Glycine/pharmacology , Kinetics , Nerve Growth Factors/metabolism , Neuroglia/cytology , Proline/pharmacology , Rats , Receptors, Metabotropic Glutamate/antagonists & inhibitors , S100 Calcium Binding Protein beta Subunit , S100 Proteins/metabolism , Transcription, Genetic
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...