Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
J Neurochem ; 157(4): 1118-1137, 2021 05.
Article in English | MEDLINE | ID: mdl-32998179

ABSTRACT

Chronic exposure to ethanol is associated with enhanced leakiness in the brain microvessel endothelial cells that form the blood-brain barrier (BBB). As previous studies suggested Wnt/ß-catenin signaling could improve the BBB phenotype of brain endothelial cells, we examined the extent to which Wnt signaling is altered following ethanol exposure, using both a cell culture model of the BBB and mice exposed to ethanol, and the ability of Wnt activation to reverse the permeability effects of ethanol. The human brain endothelial cells, hCMEC/D3, were exposed to ethanol (17-200 mM) for various periods of time (0-96 hr) and Wnt signaling, as well as expression of downstream genes influencing BBB integrity in the cell monolayers were monitored. Determination of Wnt signaling in both brain homogenates and brain microvessels from mice exposed to ethanol was also performed. The effects of ethanol on the permeability of the hCMEC/D3 monolayers were examined using both small molecular weight (sodium fluorescein) and large molecular weight (IRdye 800CW PEG) fluorescent markers. Exposure of hCMEC/D3 to ethanol (50 mM) caused a down-regulation of Wnt/ß-catenin signaling, a reduction of tight junction protein expression and up-regulation of plasmalemma vesicle associated protein (PLVAP). A similar reduction in Wnt/ß-catenin activity in both cortical brain homogenates and isolated cortical cerebral microvessels were observed in mice. Other areas such as cerebellum and striatum displayed as much as 3-6 fold increases in Dkk-1, an endogenous Wnt inhibitor. Ethanol exposure caused significant changes in both sodium fluorescein and IRdye 800CW PEG permeability (2-fold compared to control). The ethanol-induced increases in permeability were attenuated by treatment with known Wnt activators (i.e. LiCl or Wnt3a). Additional screens of CNS active agents with possible Wnt activity indicated fluoxetine could also prevent the permeability effects of ethanol. These studies suggest that ethanol-induced changes in brain microvessel permeability can be reversed through activation of Wnt signaling.


Subject(s)
Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Ethanol/toxicity , Wnt Signaling Pathway/physiology , Animals , Capillary Permeability/drug effects , Capillary Permeability/physiology , Humans , Male , Mice, Inbred C57BL
2.
PLoS One ; 14(9): e0222234, 2019.
Article in English | MEDLINE | ID: mdl-31509576

ABSTRACT

Prostatic acid phosphatase (PAP), which is secreted by prostate, increases in some diseases such as prostate cancer. PAP is also present in the central nervous system. In this study we reveal that α-synuclein (Snca) gene is co-deleted/mutated in PAP null mouse. It is indicated that mice deficient in transmembrane PAP display neurological alterations. By using immunohistochemistry, cerebellar cortical neurons and zone and stripes pattern were studied in Pap-/- ;Snca-/- mouse cerebellum. We show that the Pap-/- ;Snca-/- cerebellar cortex development appears to be normal. Compartmentation genes expression such as zebrin II, HSP25, and P75NTR show the zone and stripe phenotype characteristic of the normal cerebellum. These data indicate that although aggregation of PAP and SNCA causes severe neurodegenerative diseases, PAP -/- with absence of the Snca does not appear to interrupt the cerebellar architecture development and zone and stripe pattern formation. These findings question the physiological and pathological role of SNCA and PAP during cerebellar development or suggest existence of the possible compensatory mechanisms in the absence of these genes.


Subject(s)
Acid Phosphatase/metabolism , Cerebellar Cortex/metabolism , alpha-Synuclein/metabolism , Acid Phosphatase/genetics , Acid Phosphatase/physiology , Animals , Cerebellum/metabolism , Gene Expression/genetics , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/genetics , Motor Activity/physiology , Nerve Tissue Proteins/metabolism , Transcriptome/genetics , alpha-Synuclein/genetics , alpha-Synuclein/physiology
4.
J Pharmacol Exp Ther ; 358(1): 31-8, 2016 07.
Article in English | MEDLINE | ID: mdl-27189965

ABSTRACT

Ethanol has many pharmacological effects, including increases in endogenous adenosine levels and adenosine receptor activity in brain. Ethanol consumption is associated with both positive and negative health outcomes, but tolerance to the behavioral effects of ethanol can lead to increased consumption, which increases the risk of negative health outcomes. The present study was performed to test whether a 7-day treatment with ethanol is linked to reduced adenosine signaling and whether this is a consequence of reduced ecto-5'-nucleotidase activity. Wild-type (CD73(+/+)) and ecto-5'-nucleotidase-deficient (CD73(-/-)) mice were treated with ethanol (2 g/kg) or saline for 7 days. In CD73(+/+) mice, repeated ethanol treatment reduced the hypothermic and ataxic effects of acute ethanol, indicating the development of tolerance to the acute effects of ethanol. In CD73(+/+) mice, this 7-day ethanol treatment led to increased hippocampal synaptic activity and reduced adenosine A1 receptor activity under both basal and low Mg(2+) conditions. These effects of ethanol tolerance were associated with an 18% decrease in activity of ecto-5'-nucleotidase activity in hippocampal cell membranes. In contrast, ethanol treatment was not associated with changes in synaptic activity or adenosine signaling in hippocampus from CD73(-/-) mice. These data indicate that ethanol treatment is associated with a reduction in adenosine signaling through adenosine A1 receptors in hippocampus, mediated, at least in part, via reduced ecto-5'-nucleotidase activity.


Subject(s)
5'-Nucleotidase/metabolism , Adenosine/metabolism , Drug Tolerance , Ethanol/pharmacology , Hippocampus/drug effects , Signal Transduction/drug effects , 5'-Nucleotidase/genetics , Animals , Ethanol/blood , Hippocampus/enzymology , Hippocampus/metabolism , Male , Mice, Knockout , Receptor, Adenosine A1/genetics , Receptor, Adenosine A1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Synaptic Transmission/drug effects
5.
J Neuroimaging ; 26(4): 403-5, 2016 07.
Article in English | MEDLINE | ID: mdl-27082948

ABSTRACT

2-(18) F-fluorodeoxy-D-glucose (FDG) is a glucose analog that is taken up by cells and phosphorylated. The amount of FDG accumulated by cells is a measure of the rate of glycolysis, which reflects cellular activity. As the levels and actions of the neuromodulator adenosine are dynamically regulated by neuronal activity, this study was designed to test whether endogenous adenosine affects tissue accumulation of FDG as assessed by positron emission tomography (PET) or by postmortem analysis of tissue radioactivity. Rats were given an intraperitoneal injection of the adenosine A1 receptor antagonist 8-cyclopentyl-1,3-dipropyl-xanthine (DPCPX, 3 mg/kg), the adenosine kinase inhibitor ABT-702 (3 mg/kg), or vehicle 10 minutes prior to an intravenous injection of FDG (15.4 ± 0.7 MBq per rat). Rats were then subjected to a 15 minute static PET scan. Reconstructed images were normalized to FDG PET template for rats and standard uptake values (SUVs) were calculated. To examine the regional effect of active treatment compared to vehicle, statistical parametric mapping analysis was performed. Whole-brain FDG uptake was not affected by drug treatment. Significant regional hypometabolism was detected, particularly in cerebellum, of DPCPX- and ABT-702 treated rats, relative to vehicle-treated rats. Thus, endogenous adenosine can affect FDG accumulation although this effect is modest in quiescent rats.


Subject(s)
Adenosine/physiology , Brain/diagnostic imaging , Brain/physiology , Fluorodeoxyglucose F18 , Positron-Emission Tomography , Animals , Brain Mapping , Glucose , Glycolysis/physiology , Humans , Image Processing, Computer-Assisted , Male , Morpholines/pharmacology , Positron-Emission Tomography/methods , Pyrimidines/pharmacology , Rats , Receptor, Adenosine A1/drug effects , Xanthines/pharmacology
6.
Purinergic Signal ; 10(4): 603-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25129451

ABSTRACT

ATP is a gliotransmitter released from astrocytes. Extracellularly, ATP is metabolized by a series of enzymes, including ecto-5'-nucleotidase (eN; also known as CD73) which is encoded by the gene 5NTE and functions to form adenosine (ADO) from adenosine monophosphate (AMP). Under ischemic conditions, ADO levels in brain increase up to 100-fold. We used astrocytes cultured from 5NTE (+/+) or 5NTE (-/-) mice to evaluate the role of eN expressed by astrocytes in the production of ADO and inosine (INO) in response to glucose deprivation (GD) or oxygen-glucose deprivation (OGD). We also used co-cultures of these astrocytes with wild-type neurons to evaluate the role of eN expressed by astrocytes in the production of ADO and INO in response to GD, OGD, or N-methyl-D-aspartate (NMDA) treatment. As expected, astrocytes from 5NTE (+/+) mice produced adenosine from AMP; the eN inhibitor α,ß-methylene ADP (AOPCP) decreased ADO formation. In contrast, little ADO was formed by astrocytes from 5NTE (-/-) mice and AOPCP had no significant effect. GD and OGD treatment of 5NTE (+/+) astrocytes and 5NTE (+/+) astrocyte-neuron co-cultures produced extracellular ADO levels that were inhibited by AOPCP. In contrast, these conditions did not evoke ADO production in cultures containing 5NTE (-/-) astrocytes. NMDA treatment produced similar increases in ADO in both 5NTE (+/+) and 5NTE (-/-) astrocyte-neuron co-cultures; dipyridamole (DPR) but not AOPCP inhibited ADO production. These results indicate that eN is prominent in the formation of ADO from astrocytes but in astrocyte-neuron co-cultures, other enzymes or pathways contribute to rising ADO levels in ischemia-like conditions.


Subject(s)
5'-Nucleotidase/metabolism , Adenosine/biosynthesis , Astrocytes/metabolism , Inosine/biosynthesis , Neurons/metabolism , Animals , Cell Hypoxia/physiology , Cells, Cultured , Coculture Techniques , Ischemia/metabolism , Mice , Mice, Knockout
7.
Int J Nanomedicine ; 8: 961-70, 2013.
Article in English | MEDLINE | ID: mdl-23494517

ABSTRACT

BACKGROUND: Aminosilane-coated iron oxide nanoparticles (AmS-IONPs) have been widely used in constructing complex and multifunctional drug delivery systems. However, the biocompatibility and uptake characteristics of AmS-IONPs in central nervous system (CNS)-relevant cells are unknown. The purpose of this study was to determine the effect of surface charge and magnetic field on toxicity and uptake of AmS-IONPs in CNS-relevant cell types. METHODS: The toxicity and uptake profile of positively charged AmS-IONPs and negatively charged COOH-AmS-IONPs of similar size were examined using a mouse brain microvessel endothelial cell line (bEnd.3) and primary cultured mouse astrocytes and neurons. Cell accumulation of IONPs was examined using the ferrozine assay, and cytotoxicity was assessed by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. RESULTS: No toxicity was observed in bEnd.3 cells at concentrations up to 200 µg/mL for either AmS-IONPs or COOH-AmS-IONPs. AmS-IONPs at concentrations above 200 µg/mL reduced neuron viability by 50% in the presence or absence of a magnetic field, while only 20% reductions in viability were observed with COOH-AmS-IONPs. Similar concentrations of AmS-IONPs in astrocyte cultures reduced viability to 75% but only in the presence of a magnetic field, while exposure to COOH-AmS-IONPs reduced viability to 65% and 35% in the absence and presence of a magnetic field, respectively. Cellular accumulation of AmS-IONPs was greater in all cell types examined compared to COOH-AmS-IONPs. Rank order of cellular uptake for AmS-IONPs was astrocytes > bEnd.3 > neurons. Accumulation of COOH-AmS-IONPs was minimal and similar in magnitude in different cell types. Magnetic field exposure enhanced cellular accumulation of both AmS- and COOH-AmS-IONPs. CONCLUSION: Both IONP compositions were nontoxic at concentrations below 100 µg/mL in all cell types examined. At doses above 100 µg/mL, neurons were more sensitive to AmS-IONPs, whereas astrocytes were more vulnerable toward COOH-AmS-IONPs. Toxicity appears to be dependent on the surface coating as opposed to the amount of iron-oxide present in the cell.


Subject(s)
Drug Carriers/pharmacokinetics , Magnetite Nanoparticles/administration & dosage , Magnetite Nanoparticles/chemistry , Silanes/pharmacokinetics , Analysis of Variance , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Cell Line , Cell Survival/drug effects , Drug Carriers/chemistry , Drug Carriers/pharmacology , Mice , Neurons/drug effects , Neurons/metabolism , Silanes/chemistry , Silanes/pharmacology
8.
Acta Pharmacol Sin ; 34(1): 60-6, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23064722

ABSTRACT

Adenosine is a neuromodulator with its level increasing up to 100-fold during ischemic events, and attenuates the excitotoxic neuronal injury. Adenosine is produced both intracellularly and extracellularly, and nucleoside transport proteins transfer adenosine across plasma membranes. Adenosine levels and receptor-mediated effects of adenosine are regulated by intracellular ATP consumption, cellular release of ATP, metabolism of extracellular ATP (and other adenine nucleotides), adenosine influx, adenosine efflux and adenosine metabolism. Recent studies have used genetically modified mice to investigate the relative contributions of intra- and extracellular pathways for adenosine formation. The importance of cortical or hippocampal neurons as a source or a sink of adenosine under basal and hypoxic/ischemic conditions was addressed through the use of transgenic mice expressing human equilibrative nucleoside transporter 1 (hENT1) under the control of a promoter for neuron-specific enolase. From these studies, we conclude that ATP consumption within neurons is the primary source of adenosine in neuronal cultures, but not in hippocampal slices or in vivo mice exposed to ischemic conditions.


Subject(s)
Adenosine/metabolism , Brain Ischemia/metabolism , Brain Ischemia/pathology , Brain/metabolism , Brain/pathology , Adenosine Triphosphate/metabolism , Animals , Brain/blood supply , Humans , Neurons/metabolism , Neurons/pathology , Nucleoside Transport Proteins/metabolism
9.
Int J Toxicol ; 31(5): 467-76, 2012.
Article in English | MEDLINE | ID: mdl-23064701

ABSTRACT

Insect repellent N,N-diethyl-m-toluamide (DEET) and sunscreen oxybenzone have shown a synergistic percutaneous enhancement when applied concurrently. Both compounds are extensively metabolized in vivo into a series of potentially toxic metabolites: 2 metabolites of DEET, N,N-diethyl-m-hydroxymethylbenzamide (DHMB) and N-ethyl-m-toluamide (ET), and 3 metabolites of oxybenzone, 2,4-dihydroxybenzophenone (DHB), 2,2-dihydroxy-4-methoxybenzophenone (DMB), and 2,3,4-trihydroxybenzophenone (THB). In this study, the metabolites were extensively distributed following intravenous and topical skin administration of DEET and oxybenzone in rats. Combined application enhanced the disposition of all DEET metabolites in the liver but did not consistently affect the distribution of oxybenzone metabolites. The DHMB appeared to be the major metabolite for DEET, while THB and its precursor DHB were the main metabolites for oxybenzone. Repeated once-daily topical application for 30 days led to higher concentrations of DEET metabolites in the liver. Hepatoma cell studies revealed a decrease in cellular proliferation from all metabolites as single and combined treatments, most notably at 72 hours. Increased accumulation of DHMB and ET in the liver together with an ability to reduce cellular proliferation at achievable plasma concentrations indicated that simultaneous exposure to DEET and oxybenzone might have the potential to precipitate adverse effects in a rat animal model.


Subject(s)
Benzophenones/pharmacokinetics , DEET/pharmacokinetics , Insect Repellents/pharmacokinetics , Sunscreening Agents/pharmacokinetics , Administration, Cutaneous , Administration, Intravenous , Animals , Benzophenones/administration & dosage , Benzophenones/blood , Benzophenones/urine , Cell Line, Tumor , Cell Proliferation/drug effects , DEET/administration & dosage , DEET/blood , DEET/urine , Drug Synergism , Insect Repellents/administration & dosage , Insect Repellents/blood , Insect Repellents/urine , Rats , Rats, Sprague-Dawley , Skin Absorption , Sunscreening Agents/administration & dosage , Tissue Distribution
10.
PLoS One ; 7(6): e39772, 2012.
Article in English | MEDLINE | ID: mdl-22761898

ABSTRACT

Adenosine, through activation of its A(1) receptors, has neuroprotective effects during hypoxia and ischemia. Recently, using transgenic mice with neuronal expression of human equilibrative nucleoside transporter 1 (hENT1), we reported that nucleoside transporter-mediated release of adenosine from neurons was not a key mechanism facilitating the actions of adenosine at A(1) receptors during hypoxia/ischemia. The present study was performed to test the importance of CD73 (ecto-5'-nucleotidase) for basal and hypoxic/ischemic adenosine production. Hippocampal slice electrophysiology was performed with CD73(+/+) and CD73(-/-) mice. Adenosine and ATP had similar inhibitory effects in both genotypes, with IC(50) values of approximately 25 µM. In contrast, ATP was a less potent inhibitor (IC(50) = 100 µM) in slices from mice expressing hENT1 in neurons. The inhibitory effects of ATP in CD73(+/+) and CD73(-/-) slices were blocked by the adenosine A(1) receptor antagonist 8-cyclopentyl-1,3-dipropylxanthine (DPCPX) and were enhanced by the nucleoside transport inhibitor S-(4-nitrobenzyl)-6-thioinosine (NBTI), consistent with effects that are mediated by adenosine after metabolism of ATP. AMP showed a similar inhibitory effect to ATP and adenosine, indicating that the response to ATP was not mediated by P2 receptors. In comparing CD73(-/-) and CD73(+/+) slices, hypoxia and oxygen-glucose deprivation produced similar depression of synaptic transmission in both genotypes. An inhibitor of tissue non-specific alkaline phosphatase (TNAP) was found to attenuate the inhibitory effects of AMP and ATP, increase basal synaptic activity and reduce responses to oxygen-glucose deprivation selectively in slices from CD73(-/-) mice. These results do not support an important role for CD73 in the formation of adenosine in the CA1 area of the hippocampus during basal, hypoxic or ischemic conditions, but instead point to TNAP as a potential source of extracellular adenosine when CD73 is absent.


Subject(s)
5'-Nucleotidase/physiology , Adenosine Triphosphate/pharmacology , Glucose/metabolism , Hippocampus/drug effects , Hypoxia/physiopathology , Oxygen/metabolism , Synapses/drug effects , Adenosine/metabolism , Animals , Base Sequence , DNA Primers , Hippocampus/physiology , In Vitro Techniques , Inhibitory Concentration 50 , Mice , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Synapses/physiology
11.
Exp Transl Stroke Med ; 4(1): 4, 2012 Mar 12.
Article in English | MEDLINE | ID: mdl-22409811

ABSTRACT

BACKGROUND: Activation of adenosine A1 receptors has neuroprotective effects in animal stroke models. Adenosine levels are regulated by nucleoside transporters. In vitro studies showed that neuron-specific expression of human equilibrative nucleoside transporter 1 (hENT1) decreases extracellular adenosine levels and adenosine A1 receptor activity. In this study, we tested the effect of hENT1 expression on cortical infarct size following intracerebral injection of the vasoconstrictor endothelin-1 (ET-1) or saline. METHODS: Mice underwent stereotaxic intracortical injection of ET-1 (1 µl; 400 pmol) or saline (1 µl). Some mice received the adenosine receptor antagonist caffeine (25 mg/kg, intraperitoneal) 30 minutes prior to ET-1. Perfusion and T2-weighted magnetic resonance imaging (MRI) were used to measure cerebral blood flow (CBF) and subsequent infarct size, respectively. RESULTS: ET-1 reduced CBF at the injection site to 7.3 ± 1.3% (n = 12) in hENT1 transgenic (Tg) and 12.5 ± 2.0% (n = 13) in wild type (Wt) mice. At 48 hours following ET-1 injection, CBF was partially restored to 35.8 ± 4.5% in Tg and to 45.2 ± 6.3% in Wt mice; infarct sizes were significantly greater in Tg (9 ± 1.1 mm3) than Wt (5.4 ± 0.8 mm3) mice. Saline-treated Tg and Wt mice had modest decreases in CBF and infarcts were less than 1 mm3. For mice treated with caffeine, CBF values and infarct sizes were not significantly different between Tg and Wt mice. CONCLUSIONS: ET-1 produced greater ischemic injury in hENT1 Tg than in Wt mice. This genotype difference was not observed in mice that had received caffeine. These data indicate that hENT1 Tg mice have reduced ischemia-evoked increases in adenosine receptor activity compared to Wt mice.

12.
Alcohol Clin Exp Res ; 36(7): 1117-25, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22309182

ABSTRACT

Recent studies have demonstrated that the function of glia is not restricted to the support of neuronal function. Especially, astrocytes are essential for neuronal activity in the brain. Astrocytes actively participate in synapse formation and brain information processing by releasing or uptaking gliotransmitters such as glutamate, d-serine, adenosine 5'-triphosphate (ATP), and adenosine. In the central nervous system, adenosine plays an important role in regulating neuronal activity as well as in controlling other neurotransmitter systems such as GABA, glutamate, and dopamine. Ethanol (EtOH) increases extracellular adenosine levels, which regulates the ataxic and hypnotic/sedative (somnogenic) effects of EtOH. Adenosine signaling is also involved in the homeostasis of major inhibitory/excitatory neurotransmission (i.e., GABA or glutamate) through neuron-glial interactions, which regulates the effect of EtOH and sleep. Adenosine transporters or astrocytic SNARE-mediated transmitter release regulates extracellular or synaptic adenosine levels. Adenosine then exerts its function through several adenosine receptors and regulates glutamate levels in the brain. This review presents novel findings on how neuron-glial interactions, particularly adenosinergic signaling and glutamate uptake activity involving glutamate transporter 1 (GLT1), are implicated in alcoholism and sleep disorders.


Subject(s)
Adenosine/metabolism , Alcoholism/metabolism , Cell Communication/physiology , Glutamic Acid/metabolism , Neuroglia/metabolism , Neurons/metabolism , Signal Transduction/physiology , Sleep Wake Disorders/metabolism , Alcoholism/pathology , Animals , Excitatory Amino Acid Transporter 2 , Glutamate Plasma Membrane Transport Proteins/metabolism , Humans , Neuroglia/physiology , Neurons/physiology , SNARE Proteins/metabolism , Sleep Wake Disorders/pathology
13.
Biopharm Drug Dispos ; 32(7): 369-79, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21812003

ABSTRACT

The insect repellent N,N-diethyl-m-toluamide (DEET) and sunscreen oxybenzone (OBZ) have been shown to produce synergistic permeation enhancement when applied concurrently in vitro and in vivo. The disposition of both compounds following intravenous administration (2 mg/kg of DEET or OBZ) and topical skin application (100 mg/kg of DEET and 40 mg/kg of OBZ) was determined in male Sprague-Dawley rats. Pharmacokinetic analysis was also conducted using compartmental and non-compartmental methods. A two-compartment model was deemed the best fit for intravenous administration. The DEET and oxybenzone permeated across the skin to accumulate in blood, liver and kidney following topical skin application. Combined use of DEET and oxybenzone accelerated the disappearance of both compounds from the application site, increased their distribution in the liver and significantly decreased the apparent elimination half-lives of both compounds (p < 0.05). Hepatoma cell studies revealed toxicity from exposure to all treatment concentrations, most notably at 72 h. Although DEET and oxybenzone were capable of mutually enhancing their percutaneous permeation and systemic distribution from topical skin application, there was no evidence of increased hepatotoxic deficits from concurrent application.


Subject(s)
Benzophenones/administration & dosage , Benzophenones/pharmacokinetics , DEET/administration & dosage , DEET/pharmacokinetics , Insect Repellents/pharmacokinetics , Sunscreening Agents/pharmacokinetics , Administration, Topical , Animals , Area Under Curve , Benzophenones/blood , Cell Line, Tumor , DEET/blood , Half-Life , Injections, Intravenous , Insect Repellents/administration & dosage , Insect Repellents/blood , Male , Rats , Rats, Sprague-Dawley , Sunscreening Agents/administration & dosage
14.
Behav Brain Res ; 224(1): 44-9, 2011 Oct 10.
Article in English | MEDLINE | ID: mdl-21645551

ABSTRACT

Adenosine concentrations are regulated by purinergic enzymes and nucleoside transporters. Transgenic mice with neuronal expression of human equilibrative nucleoside transporter 1 (hENT1) have been generated (Parkinson et al., 2009 [7]). The present study tested the hypothesis that mice homozygous and heterozygous for the transgene exhibit differences in hENT1 mRNA and protein expression, and in behavioral responses to caffeine and ethanol, two drugs with adenosine-dependent actions. Real time polymerase chain reaction (PCR) was used to identify mice heterozygous and homozygous for the transgene. Gene expression, determined by real time PCR of cDNA reverse transcribed from cerebral cortex RNA, was 3.8-fold greater in homozygous mice. Protein abundance, determined by radioligand binding assays using 0.14nM [(3)H]S-(4-nitrobenzyl)-6-thioinosine ([(3)H]NBTI), was up to 84% greater in cortex synaptosome membranes from homozygous than from heterozygous mice. In western blots with an antibody specific for hENT1, a protein of approximately 40kDa was strongly labelled in cortex samples from homozygous mice, weakly labelled in samples from heterozygous mice and absent from samples from wild type mice. In behavioral assays, transgenic mice showed a greater response to ethanol and a reduced response to caffeine than wild type littermates; however, no significant differences between heterozygous and homozygous mice were detected. These data indicate that the difference in ENT1 function between wild type and heterozygous mice was greater than that between heterozygous and homozygous mice. Therefore, either heterozygous or homozygous hENT1 transgenic mice can be used in studies of ENT1 regulation of adenosine levels and adenosine dependent behaviors.


Subject(s)
Behavior, Animal/physiology , Equilibrative Nucleoside Transporter 1/metabolism , Gene Expression Regulation/genetics , Adenosine Kinase/genetics , Adenosine Kinase/metabolism , Analysis of Variance , Animals , Behavior, Animal/drug effects , Caffeine/pharmacology , Cerebral Cortex/metabolism , Equilibrative Nucleoside Transporter 1/genetics , Ethanol/pharmacology , Humans , Mice , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/genetics , Protein Binding/drug effects , Protein Binding/genetics , Thioinosine/analogs & derivatives , Thioinosine/pharmacokinetics , Tritium/pharmacokinetics
15.
Curr Top Med Chem ; 11(8): 948-72, 2011.
Article in English | MEDLINE | ID: mdl-21401500

ABSTRACT

Pyrimidine and purine nucleosides and their derivatives have critical functions and pharmacological applications in the brain. Nucleosides and nucleobases are precursors of nucleotides, which serve as the energy-rich currency of intermediary metabolism and as precursors of nucleic acids. Nucleosides (e.g., adenosine) and nucleotides are key signaling molecules that modulate brain function through interaction with cell surface receptors. Brain pathologies involving nucleosides and their metabolites range from epilepsy to neurodegenerative disorders and psychiatric conditions to cerebrovascular ischemia. Nucleoside analogs are used clinically in the treatment of brain cancer and viral infections. Nucleosides are hydrophilic molecules, and transportability across cell membranes via specialized nucleoside transporter (NT) proteins is a critical determinant of their metabolism and, for nucleoside drugs, their pharmacologic actions. In mammals, there are two types of nucleoside transport process: bidirectional equilibrative processes driven by chemical gradients, and unidirectional concentrative processes driven by sodium (and proton) electrochemical gradients. In mammals, these processes, both of which are present in brain, are mediated by members of two structurally unrelated membrane protein families (ENT and CNT, respectively). In this Chapter, we review current knowledge of cellular, physiological, pathophysiological and therapeutic aspects of ENT and CNT distribution and function in the mammalian brain, including studies with NT inhibitors and new research involving NT knockout and transgenic mice. We also describe recent progress in functional and molecular studies of ENT and CNT proteins, and summarize emerging evidence of other transporter families with demonstrated or potential roles in the transport of nucleosides and their derivatives in the brain.


Subject(s)
Anion Transport Proteins/metabolism , Brain/metabolism , Nucleoside Transport Proteins/metabolism , Nucleosides , Protein Isoforms/metabolism , Proton Pumps/metabolism , Animals , Anion Transport Proteins/genetics , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/therapeutic use , Antiviral Agents/chemical synthesis , Antiviral Agents/therapeutic use , Biological Transport/physiology , Blood-Brain Barrier/metabolism , Brain/physiopathology , Cell Membrane/metabolism , Epilepsy/drug therapy , Epilepsy/metabolism , Epilepsy/physiopathology , Humans , Mice , Mice, Knockout , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology , Nucleoside Transport Proteins/classification , Nucleoside Transport Proteins/genetics , Nucleosides/metabolism , Nucleosides/pharmacology , Protein Isoforms/genetics , Proton Pumps/genetics , Psychotic Disorders/drug therapy , Psychotic Disorders/metabolism , Psychotic Disorders/physiopathology , Rats , Signal Transduction , Structure-Activity Relationship , Xenopus
16.
J Neurochem ; 118(1): 4-11, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21395582

ABSTRACT

Activation of adenosine A(1) receptors inhibits excitatory synaptic transmission. Equilibrative nucleoside transporters (ENTs) regulate extracellular adenosine levels; however, the role of neuronal ENTs in adenosine influx and efflux during cerebral ischemia has not been determined. We used mice with neuronal expression of human ENT type 1 and wild type (Wt) littermates to compare responses to in vitro hypoxic or ischemic conditions. Extracellular recordings in the CA1 region of hippocampal slices from transgenic (Tg) mice revealed increased basal synaptic transmission, relative to Wt slices, and an absence of 8-cyclopentyl-1,3-dipropyl-xanthine mediated augmentation of excitatory neurotransmission. Adenosine (10-100 µM) had a reduced potency for inhibiting synaptic transmission in slices from Tg mice; inhibitory concentration 50% values were approximately 25 and 50 µM in Wt and Tg slices, respectively. Potency of the A(1) receptor agonist N(6) -cyclopentyladenosine (1 nM-1 µM) was unchanged. Transient hypoxia or oxygen-glucose deprivation produced greater inhibition of excitatory neurotransmission in slices from Wt than Tg, mice. The ENT1 inhibitor S-(4-nitrobenzyl)-6-thioinosine abolished these differences. Taken together, our data provide evidence that neuronal ENTs reduce hypoxia- and ischemia-induced increases in extracellular adenosine levels and suggest that inhibition of neuronal adenosine transporters may be a target for the treatment of cerebral ischemia.


Subject(s)
Adenosine/metabolism , Equilibrative Nucleoside Transporter 1/metabolism , Hippocampus/pathology , Hypoxia/pathology , Neurons/drug effects , Action Potentials/drug effects , Action Potentials/physiology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine A1 Receptor Antagonists/pharmacology , Animals , Dose-Response Relationship, Drug , Equilibrative Nucleoside Transporter 1/genetics , Female , Glucose/deficiency , Glutathione/analogs & derivatives , Glutathione/pharmacology , Humans , In Vitro Techniques , Mice , Mice, Transgenic , Neurons/physiology , Patch-Clamp Techniques , Protein Binding/drug effects , Protein Binding/physiology , Purinergic P1 Receptor Agonists/pharmacology , Statistics, Nonparametric , Synaptic Transmission/drug effects , Synaptic Transmission/genetics , Thioinosine/analogs & derivatives , Thioinosine/pharmacokinetics , Tritium/pharmacokinetics , Xanthines/pharmacology
17.
Biochem Biophys Res Commun ; 406(1): 146-51, 2011 Mar 04.
Article in English | MEDLINE | ID: mdl-21303656

ABSTRACT

Humans are capable of sensing five basic tastes which are sweet, sour, salt, umami and bitter. Of these, bitter taste perception provides protection against ingestion of potentially toxic substances. Bitter taste is sensed by bitter taste receptors (T2Rs) that belong to the G-protein coupled receptors (GPCRs) superfamily. Humans have 25 T2Rs that are expressed in the oral cavity, gastrointestinal (GI) neuroendocrine cells and airway cells. Electrophysiological studies of the brain neurons show that the neurons are able to respond to different tastants. However, the presence of bitter taste receptors in brain cells has not been elucidated. In this report using RT-PCR, and immunohistochemistry analysis we show that T2Rs are expressed in multiple regions of the rat brain. RT-PCR analysis revealed the presence of T2R4, T2R107 and T2R38 transcripts in the brain stem, cerebellum, cortex and nucleus accumbens. The bitter receptor T2R4 was selected for further analysis at the transcript level by quantitative real time PCR and at the protein level by immunohistochemistry. To elucidate if the T2R4 expressed in these cells is functional, assays involving G-protein mediated calcium signaling were carried out. The functional assays showed an increase in intracellular calcium levels after the application of exogenous ligands for T2R4, denatonium benzoate and quinine to these cultured cells, suggesting that endogenous T2R4 expressed in these cells is functional. We discuss our results in terms of the physiological relevance of bitter receptor expression in the brain.


Subject(s)
Brain/metabolism , Receptors, G-Protein-Coupled/biosynthesis , Taste Buds/metabolism , Taste , Animals , Brain/cytology , Female , Male , Neuroglia/metabolism , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/genetics , Taste Buds/cytology , Transcription, Genetic
18.
Int J Toxicol ; 29(6): 594-603, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20959613

ABSTRACT

Insect repellent N,N-diethyl-m-toluamide (DEET) and sunscreen oxybenzone are capable of enhancing skin permeation of each other when applied simultaneously. We carried out a cellular study in rat astrocytes and neurons to assess cell toxicity of DEET and oxybenzone and a 30-day study in Sprague-Dawley rats to characterize skin permeation and tissue disposition of the compounds. Cellular toxicity occurred at 1 µg/mL for neurons and 7-day treatment for astrocytes and neurons. DEET and oxybenzone permeated across the skin to accumulate in blood, liver, and brain after repeated topical applications. DEET disappeared from the application site faster than oxybenzone. Combined application enhanced the disposition of DEET in liver. No overt sign of behavioral toxicity was observed from several behavioral testing protocols. It was concluded that despite measurable disposition of the study compounds in vivo, there was no evidence of neurotoxicological deficits from repeated topical applications of DEET, oxybenzone, or both.


Subject(s)
Benzophenones/pharmacokinetics , DEET/pharmacokinetics , Insect Repellents/pharmacokinetics , Skin/drug effects , Sunscreening Agents/pharmacokinetics , Administration, Topical , Animals , Astrocytes/drug effects , Behavior, Animal/drug effects , Benzophenones/administration & dosage , Benzophenones/blood , Benzophenones/toxicity , Cell Survival/drug effects , Cells, Cultured , DEET/administration & dosage , DEET/blood , DEET/toxicity , Drug Synergism , Female , Fetus/cytology , Half-Life , Insect Repellents/administration & dosage , Insect Repellents/blood , Insect Repellents/toxicity , Male , Neurons/drug effects , Permeability/drug effects , Random Allocation , Rats , Rats, Sprague-Dawley , Skin/metabolism , Sunscreening Agents/administration & dosage , Sunscreening Agents/toxicity , Tissue Distribution
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...