Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Ig Sanita Pubbl ; 77(2): 447-458, 2021.
Article in Italian | MEDLINE | ID: mdl-34314406

ABSTRACT

The rapid spread of Covid-19 epidemic led to a change in the organizational strategies of all Italian healthcare facilities. From January 31, 2020 (starting date of the state of national health emergency) Asst Valle Olona has prepared a reorganization of the supply units passing from a traditional division system to a structure that foresees the presence of traditional wards and a set of areas dedicated exclusively to the of Covid 19 treatments.The study aims to represent the method used for the redistribution of human capital in the new areas for identified, mapped, evaluated and reordered skills. The method may guarantee assistance to Covid-19 patients with the greatest number of human resources available and adequately trained.


Subject(s)
COVID-19 , Pandemics , Humans , Italy/epidemiology , SARS-CoV-2
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.
Clin Ter ; 168(5): e317-e319, 2017.
Article in English | MEDLINE | ID: mdl-29044354

ABSTRACT

INTRODUCTION: Chronic diseases are the leading cause of death and disability in almost all over the world; in Europe causing over 9 million deaths per year according to WHO estimates. A promising health organization model for chronic disease management is represented by the Chronic Care Model (CCM). In the 12th district of the ASL Roma 2 since 4 years was implemented a CCM for the management of patients affected by diabetes and/or at high cardiovascular risk. OBJECTIVE: Aim of this study is to evaluate the effectiveness of the Chronic Care Model (CCM) for the management of chronic disease in terms of mortality reduction, avoidable hospitalizations reduction and improvement of clinical parameters. MATERIALS AND METHODS: A retrospective cohort study will involve patients of 12th district of the ASL Roma 2 affected by diabetes and at high cardiovascular risk assisted through the CCM. Their health outcomes will be compared with those of patients in the same clinical conditions, residents in the same district but not assisted with CCM. The sample will be composed by adults (> 18 years) with a diagnosis of diabetes mellitus type 2 (DM2) or metabolic syndrome and / or arterial hypertension (IT) and two or more risk factors. Outcomes will be mortality from all causes and from causes related to DM and IT, preventable hospitalizations as defined in the Prevention Quality Indicators (PQI) by the Agency for Healthcare Research and Quality, and 10 clinical parameters. The data sources will be the records of causes of death (RENCAM), the hospital discharge records (SDO) and information systems for primary healthcare. CONCLUSION: Data from the experience of CCM in Tuscany seem promising especially in the evaluation of patient satisfaction and clinical outcomes particularly on cardiovascular and neurological complications and long-term mortality.


Subject(s)
Chronic Disease/therapy , Long-Term Care , Models, Organizational , Adult , Cardiovascular Diseases/therapy , Chronic Disease/mortality , Cohort Studies , Diabetes Mellitus, Type 2/therapy , Europe , Hospitalization , Humans , Hypertension/therapy , Metabolic Syndrome/therapy , Patient Satisfaction , Primary Health Care , Retrospective Studies , Risk Factors , United States
4.
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
5.
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
6.
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
7.
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.

8.
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
9.
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
10.
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
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.
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
15.
Phys Rev E Stat Nonlin Soft Matter Phys ; 63(3 Pt 2): 036304, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11308765

ABSTRACT

We suggest the existence of a characteristic inner scale xi for helicity dissipation in a regime of hydrodynamic fully developed turbulence and estimate it on dimensional grounds. This scale is always larger than the Kolmogorov scale eta and their ratio eta/xi vanishes in the high Reynolds number limit, so the flow will always be helicity free in the small scales. These ideas are illustrated in a shell model of turbulence.

16.
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
17.
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
18.
Cardiologia ; 43(4): 387-93, 1998 Apr.
Article in Italian | MEDLINE | ID: mdl-9659796

ABSTRACT

Recent studies suggest that A1 adenosine receptor antagonists may prevent reperfusion injury in the lung and heart. The pathophysiology of this protective effect is unclear; a possible inhibition of superoxide anion release from neutrophils, or leukocyte activation and platelet aggregation are reported. We tested the hypothesis of a blood-independent cardioprotection following A1 adenosine receptor antagonism with 1,3 dipropyl,8-cyclopentylxanthine (DPCPX). Isolated working rat hearts were submitted to 10 and 20 min global ischemia in order to assess functional alterations, necrosis enzyme and purine release in coronary effluent, arrhythmias, heart weight, ultrastructural morphometry and microvascular permeability by FITC-albumin diffusion technique. DPCPX (100 nM) was administered to the perfusion buffer before ischemia. In untreated hearts we detected a significant impairment of function, associated with a significant enzyme and purine release, myocardial edema and ultrastructural damage. In DPCPX-treated hearts functional and histological damage was significantly reduced compared to controls. Moreover, a significant reduction in postischemic endothelial permeability (FITC-albumin diffusion, p < 0.02) and ultrastructural damage was observed. Our data suggest that A1 adenosine receptor antagonism with DPCPX significantly reduces ischemia-reperfusion damage in isolated, crystalloid perfused rat heart by a direct reduction of endothelium damage, fluid diffusion within the interstitium and improvement of coronary microcirculation.


Subject(s)
Adenosine/antagonists & inhibitors , Coronary Circulation/drug effects , Microcirculation/drug effects , Receptors, Purinergic P1/drug effects , Animals , Disease Models, Animal , Myocardial Ischemia , Myocardial Reperfusion/adverse effects , Rats , Rats, Wistar
20.
J Pept Res ; 49(4): 308-23, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9176814

ABSTRACT

The Mycobacterium tuberculosis chaperonin 10 protein and fragments corresponding to sequences 59-99, 51-99 and 26-99 were synthesised by the solid-phase methodology using a double coupling protocol and without the aid of capping agents. After the final acid cleavage using the low TFMSA-high HF protocol the polypeptides were purified by either the ion exchange chromatography/RP-HPLC combination or the isoelectric separation carried out in solution and followed by semi-preparative RP-HPLC. Comparison of the results obtained through the two approaches indicated that in general the isoelectricfocusing/HPLC combination was superior both in terms of recovery of final material and its purity. The advantages found were as follows: (i) Unlike ion exchange chromatography, no tailoring of the separation conditions is required, (ii) Several consecutive focusings can be carried out in progressively narrower pH gradients. This increases the separation resolution without the need of changing other separation parameters, (iii) Very little manipulation is needed, and each focusing requires 3-5h. (iv) Full compatibility with non-ionic denaturants such as 8 M urea. This increases solubility so that using the ROTOFOR instrument described here 50-100 mg crude polypeptide can be processed daily. Thus the isoelectric focusing technique carried out in solution is a valid and inexpensive alternative to ion exchange chromatography.


Subject(s)
Chaperonin 10/chemical synthesis , Peptide Fragments/chemical synthesis , Amino Acid Sequence , Chaperonin 10/chemistry , Chaperonin 10/isolation & purification , Chromatography, High Pressure Liquid/methods , Chromatography, Ion Exchange/methods , Indicators and Reagents , Isoelectric Focusing/methods , Molecular Sequence Data , Mycobacterium tuberculosis , Peptide Fragments/chemistry , Peptide Fragments/isolation & purification , Solutions , Spectrometry, Mass, Secondary Ion
SELECTION OF CITATIONS
SEARCH DETAIL
...