Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Neurochem ; 109(2): 562-72, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19222701

ABSTRACT

Transgenic mice that express human equilibrative nucleoside transporter subtype 1 (hENT1) under the control of a neuron-specific enolase promoter have been generated. Southern blot and PCR revealed the presence of the transgene in five founder mice. Mice from each founder line were examined by reverse transcriptase (RT)-PCR and found to express hENT1 in RNA isolated from whole brain, cerebral cortex, striatum, hippocampus, and cerebellum but not liver, kidney, heart, lung or skeletal muscle. Cortical synaptosomes prepared from transgenic mice had significantly increased [(3)H]adenosine uptake and [(3)H]nitrobenzylthioinosine binding, relative to samples from wild-type mice. In behavioral tests, transgenic mice had altered responses to caffeine and ethanol, two drugs that inhibit and enhance, respectively, adenosine receptor activity. Caffeine-induced locomotor stimulation was attenuated whereas the hypnotic effect of ethanol was enhanced in transgenic mice. Caffeine was more potent in inhibiting ethanol-induced motor incoordination in wild-type than in transgenic mice. No differences in expression of mouse genes for adenosine receptors, nucleoside transporters, or purine metabolizing enzymes were detected by RT-PCR analyses. These data indicate that expression of hENT1 in neurons does not trigger adaptive changes in expression of adenosine-related genes. Instead, hENT1 expression affects dynamic changes in endogenous adenosine levels, as revealed by altered behavioral responses to drugs that affect adenosine receptor signalling.


Subject(s)
Equilibrative Nucleoside Transporter 1/biosynthesis , Equilibrative Nucleoside Transporter 1/genetics , Gene Expression Regulation/physiology , Neurons/metabolism , Adenosine/metabolism , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Caffeine/administration & dosage , Caffeine/antagonists & inhibitors , Equilibrative Nucleoside Transporter 1/physiology , Ethanol/administration & dosage , Gene Expression Regulation/drug effects , Humans , Mice , Mice, Transgenic , Neurons/drug effects , Neurons/physiology , PC12 Cells , Rats , Receptors, Purinergic P1/biosynthesis , Receptors, Purinergic P1/genetics , Up-Regulation/genetics
2.
Can J Physiol Pharmacol ; 87(10): 850-8, 2009 Oct.
Article in English | MEDLINE | ID: mdl-20052011

ABSTRACT

The nucleoside adenosine (ADO) is a neuromodulator in brain. ADO and its metabolite inosine (INO) have been shown to increase cell viability in stroke models. During ischemia, extracellular levels of both ADO and INO are increased. In this study, we treated rat cortical neurons with N-methyl-D-aspartate (NMDA) to initiate excitotoxicity and then investigated the mechanisms of ADO and INO release. NMDA induced a significant increase in ADO and INO production. The effect of NMDA receptor antagonists on NMDA-evoked ADO and INO release was examined. MK-801 (1 micromol/L), a potent antagonist that lacks receptor subunit selectivity, completely blocked evoked release of both ADO and INO. Memantine (10 micromol/L), a lower affinity antagonist that also lacks subunit selectivity, blocked INO, but not ADO, release. Ifenprodil (10 micromol/L), an inhibitor selective for NMDA receptors containing the NR2B subunit, completely blocked evoked ADO and INO release. NVP-AAM077 (NVP, 0.4 micromol/L), an inhibitor selective for NMDA receptors containing the NR2A subunit, did not significantly block evoked release of either ADO or INO. Removal of extracellular Ca2+ abolished NMDA-evoked release of both ADO and INO. BAPTA (25 micromol/L), which chelates intracellular Ca2+, had no significant effect on either ADO or INO release unless extracellular Ca2+ was also removed. Inhibitors of Ca2+/calmodulin-dependent protein kinase II (CaMKII) prevented NMDA-evoked ADO and INO release and decreased nucleoside transporter function. These data indicate that NMDA-evoked ADO and INO release is dependent on subunit composition of NMDA receptors. As well, NMDA-evoked ADO and INO release requires nucleoside transporters and extracellular Ca2+ and is enhanced by activation of CaMKII.


Subject(s)
Adenosine/metabolism , Calcium/physiology , Inosine/metabolism , N-Methylaspartate/pharmacology , Neurons/metabolism , Animals , Calcium Signaling/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2/physiology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Chelating Agents/pharmacology , Dizocilpine Maleate/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Neurons/drug effects , Piperidines/pharmacology , Purines/pharmacology , Quinoxalines/pharmacology , Rats , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/physiology
3.
J Neurosci Res ; 86(15): 3447-55, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18627033

ABSTRACT

Adenosine (ADO) is an important neuromodulator in brain. During pathophysiological events such as stroke or brain trauma, ADO levels can increase up to 100-fold. We tested the hypothesis that astrocytes are important for the removal of ADO produced by neurons and for the metabolism of ADO to inosine (INO) and hypoxanthine (HX). We used four different cell culture preparations: cortical neurons, cortical astrocytes, cocultures of neurons and astrocytes, and neurons transiently cocultured with astrocytes on transwell filters. These cultures were treated with N-methyl-D-aspartate (NMDA), because NMDA receptor activation is a common factor among many causes of neurotoxicity. NMDA significantly increased extracellular ADO, INO, and HX levels from cultured cortical neurons by 3-, 3.5-, and 2-fold, respectively. In cocultures, NMDA significantly increased INO, by 4.5-fold, and HX, by 3-fold, but did not increase ADO levels. There was no NMDA-evoked purine production from astrocytes. Inhibition of purine nucleoside phosphorylase (PNP) significantly decreased HX production from both neurons and cocultures to less than 30% of control levels. The transient addition of astrocytes to neurons during NMDA treatment significantly increased HX and decreased ADO levels compared with neurons alone. In addition, increasing the number of astrocytes was directly correlated with an increased capacity of ADO metabolism to INO and HX. In conclusion, NMDA evoked the production of ADO, INO, and HX from neurons. In the presence of astrocytes, there was significantly less ADO and more HX produced. Thus, ADO produced by neurons is subject to metabolism by astrocytes, a process that may limit its neuromodulatory actions.


Subject(s)
Adenosine/metabolism , Astrocytes/metabolism , Brain/metabolism , Hypoxanthine/metabolism , Neurons/metabolism , Animals , Astrocytes/drug effects , Cells, Cultured , Coculture Techniques , Excitatory Amino Acid Agonists/pharmacology , N-Methylaspartate/pharmacology , Neurons/drug effects , Purine-Nucleoside Phosphorylase/metabolism , Rats , Rats, Sprague-Dawley
4.
J Neurosci Res ; 86(12): 2641-9, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18478552

ABSTRACT

Adenosine (ADO) is produced by cultured neurons and astrocytes, albeit by different pathways, during in vitro stroke models (Parkinson and Xiong [2004] J. Neurochem. 88:1305-1312). Expression of ecto-5' nucleotidase (e-N), the enzyme responsible for extracellular dephosphorylation of AMP to ADO, is more abundant in astrocytes than neurons. Therefore, we tested the hypothesis that N-methyl-D-aspartate (NMDA) evokes ADO release per se from neurons, whereas dephosphorylation of extracellular adenine nucleotides contributes to NMDA-evoked ADO production in the presence of astrocytes. We used four different cell preparations-cortical rat neurons, cortical rat astrocytes, cocultures of neurons and astrocytes, and transient cocultures of neurons with astrocytes on transwell filters-to show that astrocytes contribute to NMDA-evoked increases in extracellular ADO. NMDA significantly increased ADO and inosine (INO) production from cultured cortical neurons but only increased extracellular INO production from cocultures. In neurons, the equilibrative nucleoside transport (ENT) inhibitor dipyridamole (DPR) prevented NMDA-evoked ADO and INO production, whereas the e-N inhibitor alpha,beta-methylene ADP (AOPCP) had no effect. Conversely, from both cocultures and transient cocultures DPR significantly decreased NMDA-evoked INO but not ADO generation. AOPCP inhibited NMDA-evoked production of both ADO and INO from transient cocultures. In the absence of astrocytes, NMDA evoked release of intracellular ADO and INO from cultured cortical neurons through ENT. However, in the presence of astrocytes, extracellular conversion of adenine nucleotides to ADO contributed significantly to NMDA-evoked production of this purine.


Subject(s)
Astrocytes/metabolism , Cerebral Cortex/metabolism , Neurons/metabolism , Purines/metabolism , Animals , Astrocytes/cytology , Astrocytes/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Coculture Techniques , N-Methylaspartate/pharmacology , Neurons/cytology , Neurons/drug effects , Purines/agonists , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology
5.
J Neurosci Res ; 84(4): 801-8, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16862552

ABSTRACT

In brain, levels of adenosine increase up to 100-fold during cerebral ischemia. Based on in vitro studies, both astrocytes and neurons contribute to this adenosine release. Neurons release adenosine per se whereas astrocytes release adenine nucleotides that are metabolized to adenosine extracellularly. In contrast, inosine is released from both cell types via a nucleoside transporter. C6 glioma cells, which are derived from astrocytes, release inosine but not adenosine. The present study investigated the relative expression of purine metabolizing enzymes and transporters in neurons, astrocytes and C6 glioma cells by real-time PCR analysis. In agreement with the extracellular formation of adenosine and intracellular formation of inosine by astrocytes, the present study showed high expression of ecto 5'-nucleotidase and AMP deaminase type 3 in astrocytes. The lack of adenosine release from C6 glioma cells was consistent with the absence of expression of the AMP-preferring cytosolic 5'-nucleotidase in these cells. The predominance of nitrobenzylthioinosine (NBMPR) insensitive equilibrative nucleoside transport (ENT2) in all three cell types was consistent with the greater activity of this isoform in comparison to NBMPR-sensitive ENT1 in these rat cells. Thus, cell type differences in adenosine formation and release are primarily a function of differences in expression of purinergic enzymes and transporters.


Subject(s)
Adenosine/metabolism , Astrocytes/metabolism , Enzymes/metabolism , Gene Expression/physiology , Glioma/metabolism , Inosine/metabolism , Neurons/metabolism , Nucleotide Transport Proteins/metabolism , Prosencephalon/cytology , Animals , Cells, Cultured , Cloning, Molecular/methods , Enzymes/genetics , Mice , Nucleotide Transport Proteins/genetics , RNA, Messenger/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction/methods , Tritium/metabolism
6.
Brain Res ; 1082(1): 23-31, 2006 Apr 12.
Article in English | MEDLINE | ID: mdl-16516170

ABSTRACT

During cerebral ischemia, dysregulated glutamate release activates N-methyl-d-aspartate (NMDA) receptors which promotes excitotoxicity and intracellular acidosis. Ischemia also induces cellular adenosine (ADO) release, which activates ADO receptors and reduces neuronal injury. The aim of this research was to determine if decreasing intracellular pH (pH(i)) enhances ADO release from neurons. Rat forebrain neurons were incubated with NMDA, acetate, propionate, 5-(N)-ethyl-N-isopropyl amiloride (EIPA) or low pH buffer. pH(i) was determined with the fluorescent dye 2',7'-bis(2-carboxyethyl)-5(6)-carboxyfluorescein acetoxymethyl ester (BCECF-AM) and cellular release of ADO was assayed. NMDA decreased pH(i) and increased ADO release from neurons. Acetate and propionate decreased pH(i) and evoked ADO release from neurons. EIPA, an inhibitor of sodium hydrogen exchanger 1 (NHE1), enhanced the acidosis in neurons but did not enhance ADO release. Decreasing extracellular pH (pH(e)) to 6.8 or 6.45 significantly decreased pH(i) in neurons, but was not consistently associated with increased ADO release. The main finding of this study was that acidosis per se did not enhance ADO release from neurons.


Subject(s)
Acidosis/physiopathology , Adenosine/metabolism , Neurons/metabolism , Prosencephalon/cytology , Acetates/pharmacology , Amiloride/analogs & derivatives , Amiloride/pharmacology , Analysis of Variance , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Drug Interactions , Embryo, Mammalian , Excitatory Amino Acid Agonists/pharmacology , Fluoresceins/pharmacokinetics , Hydrogen-Ion Concentration , Methylamines/pharmacology , Models, Biological , N-Methylaspartate/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Purines/pharmacokinetics , Rats , Time Factors , Tritium/pharmacokinetics
7.
Cancer Biol Ther ; 5(1): 10-9, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16410718

ABSTRACT

Classical chemotherapy, that specifically targets rapidly proliferating cells, has been in existence for over eighty years and has proven to be fully successful in only a limited number of cancers. Thus, this review focuses on a novel, emerging approach for cancer therapy that uses alternative, and more unique features of cancer cells. This new approach facilitates the selective targeting of cancer, while sparing normal, non-transformed cells. Examples of molecules that kill cancer cells selectively are: apoptin, E4orf4, viral protein R (VpR), and Brevinin-2R. Below we focus on apoptin, a product of the third open reading frame (VP3) of the chicken anemia virus. Besides discussing apoptin's mechanism of action, we also provide concise insight into the biology of a chicken anemia virus infection. Since apoptin's cancer-selective toxicity depends on its nuclear localization, we broadly discuss mechanism(s) involved in its nuclear retention (both nuclear import and export). We also discuss recent findings on apoptin's molecular mechanism of action, with a focus on the role of Nur77 in apoptin's nucleo-cytoplasmic signaling. Finally, we compare the current findings on apoptin to the mechanism of cancer selective toxicity of E4orf4. In the 'summary' -section, besides highlighting important issues related to cancer-selective therapy, we also discuss concurrent approaches towards therapy personalization, particularly those related to the in vivo-, and real time cancer-therapy efficacy monitoring, using "lab-on-the-chip" and other emerging technologies.


Subject(s)
Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Capsid Proteins/metabolism , Capsid Proteins/pharmacology , Neoplasms/drug therapy , Active Transport, Cell Nucleus , Amino Acid Sequence , Animals , Antineoplastic Agents/therapeutic use , Capsid Proteins/therapeutic use , Chicken anemia virus/genetics , Chicken anemia virus/physiology , DNA-Binding Proteins/physiology , Humans , Molecular Sequence Data , Nuclear Localization Signals , Nuclear Receptor Subfamily 4, Group A, Member 1 , Receptors, Cytoplasmic and Nuclear/physiology , Receptors, Steroid/physiology , Transcription Factors/physiology , Viral Proteins/therapeutic use
8.
Article in English | MEDLINE | ID: mdl-16303224

ABSTRACT

There is growing evidence from both uncontrolled and controlled clinical studies that lamotrigine (LTG) significantly augments clozapine (CLZ) in the treatment of refractory schizophrenia (RS) [Dursun, S.M., McIntosh, D., Milliken, H., 1999. Clozapine plus lamotrigine in treatment-resistant schizophrenia. Arch. Gen. Psychiatry 56, 950; Dursun, S.M., Deakin, J.F.W., 2001. Augmenting antipsychotic treatment with lamotrigine or topiramate in patients with treatment-resistant schizophrenia: a naturalistic case-series outcome study. J. Psychopharmacol. 15, 297-301; Tiihonen, J., Hallikainen, T., Ryynanen, O.P., Repo-Tiihonen, E., Kotilinen, I., Eronen, M., Toivonen, P., Wahlbeck, K., Putkonen, A., 2003. Lamotrigine in treatment-resistant schizophrenia; a randomized placebo-controlled cross over trial. Biol. Psychiatry 54, 1241-1248; Kremer, I., Vass, A., Gorelik, I., Bar, G., Blanaru, M., Javitt, D.C., Heresco-Levy, U., 2004. Placebo-controlled trial of lamotrigine added to conventional and atypical antipsychotics in schizophrenia. Biol. Psychiatry. 56, 441-446]. However, the precise mechanism of action of this synergistic augmentation between clozapine and lamotrigine remains unclear. Therefore, the goal of this research is to explore the mechanism of action of this synergistic interaction between CLZ and LTG, utilizing a pharmacological animal model of schizophrenia by using phencyclidine (PCP). The effects of CLZ plus LTG were assessed by measuring PCP-induced hyper-locomotion and stereotyped behaviours in rats. Adult male rats (250-300 g) were pre-treated via intra-peritoneal (i.p.) injection with vehicle or drug 30 min before a PCP (5 mg/kg) or saline challenge. The behaviours were recorded and analysed for a 90-min period using the Etho Vision-computer based system. PCP produced hyper-locomotion, which was maximal at 30 min. LTG (10 mg/kg) significantly increased hyperlocomotion induced with PCP. However, a combination treatment of CLZ (5 mg/kg) plus LTG (10 mg/kg) significantly blocked the potentiation of PCP-induced hyper-locomotion observed with LTG (10 mg/kg) alone. Furthermore, the PCP-induced locomotion in the combination CLZ plus LTG-treated rats was significantly decreased when compared to vehicle. Therefore, LTG at doses that do not induce ataxia enhanced PCP-induced hyper-locomotion in rats, whereas the combination of LTG and CLZ significantly decreased PCP-induced hyper-locomotion consistent with clinical data.


Subject(s)
Antidepressive Agents/therapeutic use , Antipsychotic Agents/therapeutic use , Clozapine/therapeutic use , Hyperkinesis/drug therapy , Phencyclidine , Triazines/therapeutic use , Analysis of Variance , Animals , Behavior, Animal , Disease Models, Animal , Drug Interactions , Hyperkinesis/chemically induced , Hyperkinesis/physiopathology , Lamotrigine , Male , Motor Activity/drug effects , Rats , Rats, Sprague-Dawley , Time Factors
9.
Arch Immunol Ther Exp (Warsz) ; 53(4): 308-20, 2005.
Article in English | MEDLINE | ID: mdl-16088315

ABSTRACT

In this review we focus on peptide- and peptidomimetic-based approaches that target autoimmune diseases and some pathologies of the central nervous system. Special attention is given to asthma, allergic rhinitis, osteoarthritis, and Alzheimer's disease, but other related pathologies are also reviewed, although to a lesser degree. Among others, drugs like Diacerhein and its active form Rhein, Pralnacasan, Anakinra (Kineret), Omalizumab, an antibody "BION-1", directed against the common beta-chain of cytokine receptors, are described below as well as attempts to target beta-amyloid peptide aggregation. Parts of the review are also dedicated to targeting of pathologic conditions in the brain and in other tissues with peptides as well as methods to deliver larger molecules through the "blood--brain barrier" by exploring receptor-mediated transport, or elsewhere in the body by using peptides as carriers through cellular membranes. In addition to highlighting current developments in the field, we also propose, for future drug targets, the components of the inflammasome protein complex, which is believed to initiate the activation of caspase- 1 dependent signaling events, as well as other pathways that signal inflammation. Thus we discuss the possibility of targeting inflammasome components for negative or positive modulation of an inflammatory response.


Subject(s)
Arthritis/therapy , Asthma/therapy , Autoimmune Diseases/therapy , Central Nervous System Diseases/therapy , Peptides/therapeutic use , Amyloid beta-Peptides/chemistry , Animals , Anti-Asthmatic Agents/pharmacology , Cell Membrane/metabolism , Humans , Inflammation , Models, Biological , Peptides/chemistry
10.
Neurol Res ; 27(2): 153-60, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15829178

ABSTRACT

OBJECTIVES: Adenosine is an endogenous nucleoside that signals through G-protein coupled receptors. Extracellular adenosine is required for receptor activation and two pathways have been identified for formation and cellular release of adenosine. The CLASSICAL pathway relies on intracellular formation of adenosine from adenine nucleotides and cellular efflux of adenosine via equilibrative nucleoside transporters (ENTs). The ALTERNATE pathway involves cellular release of adenine nucleotides, hydrolysis via ecto-5'-nucleotidases and extracellular formation of adenosine. METHODS: A rat model of cerebral ischemia and primary cultures of rat forebrain astrocytes and neurons were used. RESULTS: Using a rat model of cerebral ischemia, the ENT1 inhibitor nitrobenzylmercaptopurine ribonucleoside (NBMPR) significantly increased post-ischemic forebrain adenosine levels and significantly decreased hippocampal neuron injury relative to saline-treatment. NBMPR-induced increases in adenosine receptor activation were not detected, suggesting that altering the intracellular:extracellular distribution of adenosine can affect ischemic outcome. Using primary cultures of rat forebrain astrocytes and neurons, adenosine release was evoked by ischemic-like conditions. Dipyridamole, an inhibitor of ENTs, was more effective at inhibiting adenosine release from neurons than from astrocytes. In contrast, alpha , beta-methylene ADP, an inhibitor of ecto-5'-nucleotidase, was effective at inhibiting adenosine release from astrocytes, but not from neurons. Thus, during ischemic-like conditions, neurons released adenosine via the CLASSICAL pathway, while astrocytes released adenosine via the ALTERNATE pathway. DISCUSSION: These cell type differences in pathways for adenosine formation during ischemia may allow transport inhibitors to block simultaneously adenosine release from neurons and adenosine uptake into astrocytes. In principle, this could improve neuronal ATP levels without decreasing adenosine receptor activation.


Subject(s)
Adenosine/metabolism , Astrocytes/metabolism , Brain Ischemia/metabolism , Neurons/metabolism , Prosencephalon/cytology , Thioinosine/analogs & derivatives , Affinity Labels/pharmacology , Animals , Cells, Cultured , Deoxyglucose/pharmacology , Dipyridamole/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Glucose/deficiency , Hypoxia/metabolism , Inosine/pharmacology , Models, Biological , Oligomycins/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Purines/metabolism , Rats , Thioinosine/pharmacology , Tritium/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...