Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
Add more filters











Publication year range
1.
Biomolecules ; 11(5)2021 04 24.
Article in English | MEDLINE | ID: mdl-33923147

ABSTRACT

The blood-brain barrier (BBB) plays an important protective role in the central nervous system and maintains its homeostasis. It regulates transport into brain tissue and protects neurons against the toxic effects of substances circulating in the blood. However, in the case of neurological diseases or primary brain tumors, i.e., gliomas, the higher permeability of the blood-derived substances in the brain tissue is necessary. Currently applied methods of treatment for the primary brain neoplasms include surgical removal of the tumor, radiation therapy, and chemotherapy. Despite the abovementioned treatment methods, the prognosis of primary brain tumors remains bad. Moreover, chemotherapy options seem to be limited due to low drug penetration into the cancerous tissue. Modulation of the blood-brain barrier permeability may contribute to an increase in the concentration of the drug in the CNS and thus increase the effectiveness of therapy. Interestingly, endothelial cells in cerebral vessels are characterized by the presence of adenosine 2A receptors (A2AR). It has been shown that substances affecting these receptors regulate the permeability of the BBB. The mechanism of increasing the BBB permeability by A2AR agonists is the actin-cytoskeletal reorganization and acting on the tight junctions. In this case, the A2AR seems to be a promising therapy target. This article aims to assess the possibility of increasing the BBB permeability through A2AR agonists to increase the effectiveness of chemotherapy and to improve the results of cancer therapy.


Subject(s)
Blood-Brain Barrier/metabolism , Neoplasms/metabolism , Receptors, Adenosine A2/metabolism , Animals , Biological Transport , Blood-Brain Barrier/drug effects , Brain/drug effects , Brain/metabolism , Endothelial Cells/metabolism , Humans , Neoplasms/therapy , Neurons/metabolism , Permeability , Receptors, Adenosine A2/physiology , Signal Transduction/drug effects , Tight Junctions/drug effects , Tight Junctions/metabolism
2.
Neuroscience ; 422: 32-43, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31678341

ABSTRACT

A stroke-like event follows seizures which may be responsible for the postictal state and a contributing factor to the development of seizure-induced brain abnormalities and behavioral dysfunction associated with epilepsy. Caffeine is the world's most popular drug with ∼85% of people in the USA consuming it daily. Thus, persons with epilepsy are likely to have caffeine in their body and brain during seizures. This preclinical study investigated the effects of acute caffeine on local hippocampal tissue oxygenation pre and post seizure. We continuously measured local oxygen levels in the CA1 region of the hippocampus and utilized the electrical kindling model in rats. Rats were acutely administered either caffeine, or one of its metabolites, or agonists and antagonists at adenosine sub-receptor types or ryanodine receptors prior to the elicitation of seizures. Acute caffeine administration caused a significant drop in pre-seizure hippocampal pO2. Following a seizure, caffeine, as well as two of its metabolites paraxanthine, and theophylline, increased the time below the severe hypoxic threshold (10 mmHg). Likewise, the specific A2A receptor antagonist, SCH-58261, mimicked caffeine by causing a significant drop in pre-seizure pO2 and the area and time below the severe hypoxic threshold. Moreover, the A2A receptor agonist, CGS-21680 was able to prevent the effect of both caffeine and SCH-58261 adding further evidence that caffeine is likely acting through the A2A receptor. Clinical tracking and investigations are needed to determine the effect of caffeine on postictal symptomology and blood flow in persons with epilepsy.


Subject(s)
Caffeine/adverse effects , Hypoxia/physiopathology , Receptors, Adenosine A2/physiology , Seizures/physiopathology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , CA1 Region, Hippocampal/metabolism , Caffeine/analogs & derivatives , Caffeine/antagonists & inhibitors , Dose-Response Relationship, Drug , Hypoxia/complications , Kindling, Neurologic/drug effects , Male , Oxygen/metabolism , Phenethylamines/pharmacology , Pyrimidines/antagonists & inhibitors , Pyrimidines/pharmacology , Rats , Receptors, Adenosine A2/drug effects , Seizures/complications , Triazoles/antagonists & inhibitors , Triazoles/pharmacology
3.
J Neurochem ; 142(5): 620-623, 2017 09.
Article in English | MEDLINE | ID: mdl-28736837

ABSTRACT

Alcohol causes adenosine buildup, which inhibits wake-active neurons via adenosine A1 receptors thus disinhibiting sleep active neurons and also stimulates sleep-active neurons via A2A receptors, causing sleep. This editorial highlights the study entitled, "Lesions of the basal forebrain cholinergic neurons attenuates sleepiness and adenosine after alcohol consumption" by Sharma and colleagues. They report that the wake-promoting basal forebrain (BF) cholinergic neurons play a crucial role in mediating acute alcohol-induced sleep via adenosinergic signaling.


Subject(s)
Adenosine/metabolism , Alcohol Drinking/metabolism , Basal Forebrain/physiology , Cholinergic Neurons/physiology , Homeostasis/physiology , Sleep/physiology , Alcohol Drinking/adverse effects , Alcohol Drinking/trends , Animals , Basal Forebrain/drug effects , Cholinergic Neurons/drug effects , Homeostasis/drug effects , Humans , Receptor, Adenosine A1/physiology , Receptors, Adenosine A2/physiology , Sleep/drug effects , Wakefulness/drug effects , Wakefulness/physiology
4.
Addict Biol ; 21(2): 407-21, 2016 Mar.
Article in English | MEDLINE | ID: mdl-25612195

ABSTRACT

Addiction to methamphetamine (METH) is a global health problem for which there are no approved pharmacotherapies. The adenosine 2A (A2 A ) receptor presents a potential therapeutic target for METH abuse due to its modulatory effects on striatal dopamine and glutamate transmission. Notably, A2 A receptor signalling has been implicated in the rewarding effects of alcohol, cocaine and opiates; yet, the role of this receptor in METH consumption and seeking is essentially unknown. Therefore, the current study used A2 A knockout (KO) mice to assess the role of A2 A in behaviours relevant to METH addiction. METH conditioned place preference was absent in A2 A KO mice compared with wild-type (WT) littermates. Repeated METH treatment produced locomotor sensitization in both genotypes; however, sensitization was attenuated in A2 A KO mice in a dose-related manner. METH intravenous self-administration was intact in A2 A KO mice over a range of doses and schedules of reinforcement. However, the motivation to self-administer was reduced in A2 A KO mice. Regression analysis further supported the observation that the motivation to self-administer METH was reduced in A2 A KO mice even when self-administration was similar to WT mice. Sucrose self-administration was also reduced in A2 A KO mice but only at higher schedules of reinforcement. Collectively, these data suggest that A2 A signalling is critically required to integrate rewarding and motivational properties of both METH and natural rewards.


Subject(s)
Central Nervous System Stimulants/pharmacology , Methamphetamine/pharmacology , Receptors, Adenosine A2/physiology , Reward , Analysis of Variance , Animals , Conditioning, Operant , Dose-Response Relationship, Drug , Infusions, Intravenous , Locomotion/drug effects , Male , Mice, Knockout , Motor Activity/drug effects , Reinforcement, Psychology , Self Administration , Sucrose/pharmacology , Sweetening Agents/pharmacology
5.
J Cardiovasc Pharmacol ; 66(1): 25-34, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25706370

ABSTRACT

This study was undertaken to determine and confer the cardioprotective effects of the adenosine A2 receptor (A2AR) and its impact on myocardial autophagy in the setting of reperfusion. We established a rat ischemia model by subjecting rats to 30 minutes of ischemia (I) and 120 minutes of reperfusion (R). The A2AR agonists CGS21680 (A2aAR specific) and BAY60-6583 (A2bAR specific) were administered separately and in combination 5 minutes before reperfusion (postconditioning). No visible improvements in the rats' hemodynamic changes in response to either CGS or BAY were observed compared with untreated control groups (I/R). BAY significantly reduced infarct sizes, whereas CGS did not. Electron microscopy, enzyme-linked immunosorbent assay and TUNEL apoptosis staining results demonstrated that CGS and BAY play cardioprotective roles by maintaining mitochondria structural stability, decreasing serum cardiac troponin I (cTnI) concentrations and decreasing the number of apoptotic cells. CGS21680 and BAY60-6583 slightly increased the expression (vs. I/R group) of Bcl-2 and significantly attenuated the expression of Beclin-1, LC3B, and LAMP-2, as analyzed by Western blot, compared with the I/R alone group. Notably, BAY60-6583 exerts a predominant effect on mitochondria structural stabilization, apoptotic inhibition, and attenuation of LC3B/LAMP-2 expression. No synergistic effects were observed for the 2 agonists. Our data suggest that A2AR-mediated cardioprotection is associated with Beclin-1-induced autophagy downregulation in the setting of reperfusion. A2bAR activation exerts stronger cardioprotective effects against I/R injury compared with A2aAR.


Subject(s)
Autophagy/physiology , Down-Regulation/physiology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Receptors, Adenosine A2/physiology , Adenosine A2 Receptor Agonists/pharmacology , Animals , Autophagy/drug effects , Down-Regulation/drug effects , Male , Rats , Rats, Sprague-Dawley
6.
J. physiol. biochem ; 71(1): 133-140, mar. 2015.
Article in English | IBECS | ID: ibc-133910

ABSTRACT

Under physiological conditions, insulin secretion from pancreatic beta-cells is tightly regulated by different factors, including nutrients, nervous system, and other hormones. Pancreatic beta-cells are also influenced by paracrine and autocrine interactions. The results of rodent studies indicate that adenosine is present within pancreatic islets and is implicated in the regulation of insulin secretion; however, effects depend on adenosine and glucose concentrations. Moreover, species differences in adenosine action were found. In rat islets, low adenosine was demonstrated to decrease glucose-induced insulin secretion and this effect is mediated via adenosine A1 receptor. In the presence of high adenosine concentrations, other mechanisms are activated and glucose-induced insulin secretion is increased. It is also well established that suppression of adenosine action increases insulin-secretory response of beta-cells to glucose. In mouse islets, low adenosine concentrations do not significantly affect insulin secretion. However, in the presence of higher adenosine concentrations, potentiation of glucose-induced insulin secretion was demonstrated. It is also known that upon stimulation of insulin secretion, both rat and mouse islets release ATP. In rat islets, ATP undergoes extracellular conversion to adenosine. However, mouse islets are unable to convert extracellularly ATP to adenosine and adenosine arises from intracellular ATP degradation


Subject(s)
Animals , Adenosine/pharmacokinetics , Insulin-Secreting Cells/physiology , Insulin , Receptors, Adenosine A2/physiology , Islets of Langerhans/physiology , Models, Animal
7.
Nat Commun ; 5: 5115, 2014 Oct 08.
Article in English | MEDLINE | ID: mdl-25296113

ABSTRACT

Opsin, the rhodopsin apoprotein, was recently shown to be an ATP-independent flippase (or scramblase) that equilibrates phospholipids across photoreceptor disc membranes in mammalian retina, a process required for disc homoeostasis. Here we show that scrambling is a constitutive activity of rhodopsin, distinct from its light-sensing function. Upon reconstitution into vesicles, discrete conformational states of the protein (rhodopsin, a metarhodopsin II-mimic, and two forms of opsin) facilitated rapid (>10,000 phospholipids per protein per second) scrambling of phospholipid probes. Our results indicate that the large conformational changes involved in converting rhodopsin to metarhodopsin II are not required for scrambling, and that the lipid translocation pathway either lies near the protein surface or involves membrane packing defects in the vicinity of the protein. In addition, we demonstrate that ß2-adrenergic and adenosine A2A receptors scramble lipids, suggesting that rhodopsin-like G protein-coupled receptors may play an unexpected moonlighting role in re-modelling cell membranes.


Subject(s)
Phospholipid Transfer Proteins/physiology , Receptors, Adrenergic, beta-2/physiology , Rhodopsin/physiology , Animals , Cattle , Opsins/physiology , Protein Conformation , Receptors, Adenosine A2/physiology
8.
J Neurosci ; 33(7): 3135-50, 2013 Feb 13.
Article in English | MEDLINE | ID: mdl-23407968

ABSTRACT

Gap junctions in retinal photoreceptors suppress voltage noise and facilitate input of rod signals into the cone pathway during mesopic vision. These synapses are highly plastic and regulated by light and circadian clocks. Recent studies have revealed an important role for connexin36 (Cx36) phosphorylation by protein kinase A (PKA) in regulating cell-cell coupling. Dopamine is a light-adaptive signal in the retina, causing uncoupling of photoreceptors via D4 receptors (D4R), which inhibit adenylyl cyclase (AC) and reduce PKA activity. We hypothesized that adenosine, with its extracellular levels increasing in darkness, may serve as a dark signal to coregulate photoreceptor coupling through modulation of gap junction phosphorylation. Both D4R and A2a receptor (A2aR) mRNAs were present in photoreceptors, inner nuclear layer neurons, and ganglion cells in C57BL/6 mouse retina, and showed cyclic expression with partially overlapping rhythms. Pharmacologically activating A2aR or inhibiting D4R in light-adapted daytime retina increased photoreceptor coupling. Cx36 among photoreceptor terminals, representing predominantly rod-cone gap junctions but possibly including some rod-rod and cone-cone gap junctions, was phosphorylated in a PKA-dependent manner by the same treatments. Conversely, inhibiting A2aR or activating D4R in daytime dark-adapted retina decreased Cx36 phosphorylation with similar PKA dependence. A2a-deficient mouse retina showed defective regulation of photoreceptor gap junction phosphorylation, fairly regular dopamine release, and moderately downregulated expression of D4R and AC type 1 mRNA. We conclude that adenosine and dopamine coregulate photoreceptor coupling through opposite action on the PKA pathway and Cx36 phosphorylation. In addition, loss of the A2aR hampered D4R gene expression and function.


Subject(s)
Gap Junctions/physiology , Receptors, Dopamine/physiology , Receptors, Purinergic P1/physiology , Retinal Cone Photoreceptor Cells/physiology , Retinal Rod Photoreceptor Cells/physiology , Adenylyl Cyclases/metabolism , Animals , Chromatography, High Pressure Liquid , Connexins/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Dark Adaptation/physiology , Gap Junctions/metabolism , Gene Expression/physiology , Image Processing, Computer-Assisted , Immunohistochemistry , In Situ Hybridization , In Vitro Techniques , Mice , Mice, Inbred C57BL , Phosphorylation , Real-Time Polymerase Chain Reaction , Receptors, Adenosine A2/genetics , Receptors, Adenosine A2/physiology , Receptors, Dopamine/genetics , Receptors, Dopamine D4/biosynthesis , Receptors, Dopamine D4/genetics , Receptors, Purinergic P1/genetics , Gap Junction delta-2 Protein
9.
Addict Biol ; 18(5): 812-25, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23301633

ABSTRACT

There is emerging evidence that the adenosinergic system might be involved in drug addiction and alcohol dependence. We have already demonstrated the involvement of A2A receptors (A2AR) in ethanol-related behaviours in mice. Here, we investigated whether the A2AR agonist CGS 21680 can reduce ethanol operant self-administration in both non-dependent and ethanol-dependent Wistar rats. To rule out a potential involvement of the A1R in the effects of CGS 21680, we also tested its effectiveness to reduce ethanol operant self-administration in both heterozygous and homozygous A1R knockout mice. Our results demonstrated that CGS 21680 (0.065, 0.095 and 0.125 mg/kg, i.p.) had a bimodal effect on 10% ethanol operant self-administration in non-dependent rats. The intermediate dose was also effective in reducing 2% sucrose self-administration. Interestingly, the intermediate dose reduced 10% ethanol self-administration in dependent animals more effectively (75% decrease) when compared with non-dependent animals (57% decrease). These results suggest that the A2AR are involved in CGS 21680 effects since the reduction of ethanol self-administration was not dependent upon the presence of A1R in mice. In conclusion, our findings demonstrated the effectiveness of the A2AR agonist CGS 21680 in a preclinical model of alcohol addiction and suggested that the adenosinergic pathway is a promising target to treat alcohol addiction.


Subject(s)
Adenosine A2 Receptor Agonists/pharmacology , Adenosine/analogs & derivatives , Alcoholism/drug therapy , Drug-Seeking Behavior/drug effects , Ethanol/administration & dosage , Phenethylamines/pharmacology , Adenosine/administration & dosage , Adenosine/pharmacology , Adenosine A2 Receptor Agonists/administration & dosage , Alcoholism/metabolism , Analysis of Variance , Animals , Conditioning, Operant/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Food Preferences/drug effects , Male , Mice , Mice, Knockout , Motivation/drug effects , Phenethylamines/administration & dosage , Rats , Rats, Wistar , Receptor, Adenosine A1/genetics , Receptor, Adenosine A1/physiology , Receptors, Adenosine A2/physiology , Reinforcement Schedule , Reward , Self Administration , Sucrose/administration & dosage
10.
Biol Pharm Bull ; 35(7): 1091-5, 2012.
Article in English | MEDLINE | ID: mdl-22791157

ABSTRACT

The Goto-Kakizaki (GK) rat is a non-obese and spontaneous model of mild Type 2 diabetes mellitus. In the present study, we compared the regulatory mechanisms of endogenous norepinephrine (NE) release from sympathetic nerves of caudal arteries of 12-week-old GK rats and age-matched normal Wistar rats. Electrical stimulation (ES) evoked significant NE release from caudal arteries of Wistar and GK rats. The amounts of NE released by ES were almost equal in Wistar and GK rats, although the NE content in caudal artery of GK rats was significantly lower than that of Wistar rats. We examined the effects of an α2-adrenoceptor agonist, clonidine (CLO), and an α2-adrenoceptor antagonist, yohimbine (YOH), on the release of endogenous NE evoked by ES. CLO significantly reduced NE release from caudal arteries of Wistar but not GK rats. On the other hand, YOH significantly increased NE release from both rats. Furthermore, we examined the effects of an A1-adenosine receptor agonist, 2-chloroadenosine (2CA), and an A1-adenosine receptor antagonist, 8-sulfophenyltheophylline (8SPT), on the release of endogenous NE evoked by ES. 2CA significantly reduced NE release from caudal arteries of Wistar but not GK rats. On the other hand, 8SPT did not affect NE release from both rats. These results suggest that the dysfunction of negative feedback regulation of NE release via presynaptic receptors on sympathetic nerves in GK rats may be involved in the autonomic nervous system dysfunction associated with diabetic autonomic neuropathy.


Subject(s)
Adrenergic Neurons/physiology , Arteries/innervation , Diabetes Mellitus, Type 2/physiopathology , Norepinephrine/physiology , Sympathetic Nervous System/physiopathology , Adrenergic alpha-2 Receptor Agonists/pharmacology , Adrenergic alpha-2 Receptor Antagonists/pharmacology , Animals , Arteries/physiopathology , Clonidine/pharmacology , Electric Stimulation , In Vitro Techniques , Rats , Rats, Wistar , Receptor, Adenosine A1/physiology , Receptors, Adenosine A2/physiology , Yohimbine/pharmacology
11.
Sleep ; 35(6): 861-9, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22654205

ABSTRACT

STUDY OBJECTIVE: Sleep responses to chronic sleep restriction (CSR) might be very different from those observed after short-term total sleep deprivation. For example, after sleep restriction continues for several consecutive days, animals no longer express compensatory increases in daily sleep time and sleep intensity. However, it is unknown if these allostatic, or adaptive, sleep responses to CSR are paralleled by behavioral and neurochemical measures of sleepiness. DESIGN: This study was designed to investigate CSR-induced changes in (1) sleep time and intensity as a measure of electrophysiological sleepiness, (2) sleep latency as a measure of behavioral sleepiness, and (3) brain adenosine A1 (A1R) and A2a receptor (A2aR) mRNA levels as a putative neurochemical correlate of sleepiness. SUBJECTS: Male Sprague-Dawley rats INTERVENTIONS: A 5-day sleep restriction (SR) protocol consisting of 18-h sleep deprivation and 6-h sleep opportunity each day. MEASUREMENT AND RESULTS: Unlike the first SR day, rats did not sleep longer or deeper on days 2 through 5, even though they exhibited significant elevations of behavioral sleepiness throughout all 5 SR days. For all SR days and recovery day 1, A1R mRNA in the basal forebrain was maintained at elevated levels, whereas A2aR mRNA in the frontal cortex was maintained at reduced levels. CONCLUSION: CSR LEADS TO A DECOUPLING OF SLEEPINESS FROM SLEEP TIME AND SLEEP INTENSITY, SUGGESTING THAT THERE ARE AT LEAST TWO DIFFERENT SLEEP REGULATORY SYSTEMS: one mediating sleepiness (homeostatic) and the other mediating sleep time/intensity (allostatic). The time course of changes observed in adenosine receptor mRNA levels suggests that the basal forebrain and cortical adenosine system might mediate sleepiness rather than sleep time or intensity.


Subject(s)
Prosencephalon/chemistry , Receptors, Purinergic P1/analysis , Sleep Deprivation/physiopathology , Sleep/physiology , Wakefulness/physiology , Animals , Electroencephalography , Male , Methyltransferases , Nuclear Proteins , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Receptor, Adenosine A1/analysis , Receptor, Adenosine A1/physiology , Receptors, Adenosine A2/analysis , Receptors, Adenosine A2/physiology , Receptors, Purinergic P1/physiology
12.
Epilepsy Res ; 100(1-2): 157-67, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22401823

ABSTRACT

Although adenosine is widely assumed to be an endogenous anticonvulsant its role in epileptogenesis is still contradictory. Using slices from the dorsal (DH) and the vental (VH) rat hippocampus and extracellular recordings from the CA3 field we aimed to determine the effects of endogenous adenosine on the expression and long-term maintenance of epileptiform activity induced by blockade of adenosine receptors types A(1) (A(1)R) and A(2) (A(2)R) under conditions of low magnesium. We found that the A(1)Rs blockade induced persistent epileptiform discharges (PED) more frequently in VH (by 52%) than in DH (by 31%). The induction of PED upon an additional blockade of A(2)Rs increased in VH (by 48%) but decreased in DH (by 74%). Remarkably, the increment in VH was prevented by a blockade of NMDARs. A blockade of A(2)Rs increased the NMDAR-mediated component of evoked synaptic potential in both VH and DH (by ~100%) but suppressed the non-NMDAR-mediated component in DH but not VH. A blockade of A(1)Rs induced PED equally in DH (76%) and VH (80%) via a NMDAR-independent mechanism. A blockade of A(2)Rs under blockade of A(1)Rs and NMDARs reduced the PED to 17% in DH and to 38% in VH. These findings show that A(2)Rs play a different role in the long-term maintenance of epileptiform activity between DH and VH and suggest that endogenous activation of A(2)Rs facilitates NMDAR-independent induction of PED in both hippocampal poles, but suppresses NMDAR-dependent induction of PED in VH.


Subject(s)
Adenosine/physiology , Epilepsy/metabolism , Hippocampus/physiology , Receptors, Adenosine A2/metabolism , Receptors, N-Methyl-D-Aspartate/physiology , Adenosine A2 Receptor Antagonists/pharmacology , Adenosine A2 Receptor Antagonists/therapeutic use , Animals , Epilepsy/physiopathology , Epilepsy/prevention & control , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Hippocampus/drug effects , Male , Rats , Rats, Wistar , Receptors, Adenosine A2/physiology , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Synaptic Potentials/physiology
13.
Acta Physiol (Oxf) ; 205(3): 403-10, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22356216

ABSTRACT

AIM: The precise mechanisms underlying reflex cutaneous vasodilatation during hyperthermia remain unresolved. The purpose of this study was to investigate a potential contribution of adenosine A1/A2 receptor activation to reflex cutaneous vasodilatation. METHODS: Eight subjects were equipped with four microdialysis fibres on the left forearm, and each fibre was randomly assigned one of four treatments: (1) lactated Ringer's (control); (2) 4 mm of the non-selective A1/A2 adenosine receptor antagonist theophylline; (3) 10 mm L-NAME to inhibit nitric oxide (NO) synthase; and (4) combined 4 mm theophylline and 10 mm L-NAME. Laser-Doppler flowmetry (LDF) was used as an index of skin blood flow, and blood pressure was measured beat-by-beat via photoplethysmography and verified via brachial auscultation. Whole-body heat stress to raise oral temperature 0.8 °C above baseline was induced via water-perused suits. Cutaneous vascular conductance (CVC) was calculated as LDF/mean arterial pressure and normalized to maximal (%CVC max) via infusion of 28 mm nitroprusside and local heating to 43 °C. RESULTS: There was no difference between control (65 ± 5%CVC max) and theophylline (63 ± 5%CVC max) sites. L-NAME (44 ± 4%CVC max) and theophylline + L-NAME (32 ± 3%CVC max) sites were significantly attenuated compared to both control and theophylline only sites (P<0.05), and combined theophylline + L-NAME sites were significantly reduced compared to L-NAME only sites (P<0.05). CONCLUSION: These data suggest A1/A2 adenosine receptor activation does not directly contribute to cutaneous active vasodilatation; however, a role for A1/A2 adenosine receptor activation is unmasked when NO synthase is inhibited.


Subject(s)
Heat-Shock Response/physiology , Receptor, Adenosine A1/physiology , Receptors, Adenosine A2/physiology , Skin/blood supply , Vasodilation/physiology , Adult , Enzyme Inhibitors/pharmacology , Female , Humans , Male , Microdialysis , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/drug effects , Nitroprusside/pharmacology , Purinergic P1 Receptor Antagonists/pharmacology , Receptor, Adenosine A1/drug effects , Receptors, Adenosine A2/drug effects , Theophylline/pharmacology , Vasodilation/drug effects , Vasodilator Agents/pharmacology
14.
J Neurosci ; 31(37): 13272-80, 2011 Sep 14.
Article in English | MEDLINE | ID: mdl-21917810

ABSTRACT

The blood-brain barrier (BBB) is comprised of specialized endothelial cells that form the capillary microvasculature of the CNS and is essential for brain function. It also poses the greatest impediment in the treatment of many CNS diseases because it commonly blocks entry of therapeutic compounds. Here we report that adenosine receptor (AR) signaling modulates BBB permeability in vivo. A(1) and A(2A) AR activation facilitated the entry of intravenously administered macromolecules, including large dextrans and antibodies to ß-amyloid, into murine brains. Additionally, treatment with an FDA-approved selective A(2A) agonist, Lexiscan, also increased BBB permeability in murine models. These changes in BBB permeability are dose-dependent and temporally discrete. Transgenic mice lacking A(1) or A(2A) ARs showed diminished dextran entry into the brain after AR agonism. Following treatment with a broad-spectrum AR agonist, intravenously administered anti-ß-amyloid antibody was observed to enter the CNS and bind ß-amyloid plaques in a transgenic mouse model of Alzheimer's disease (AD). Selective AR activation resulted in cellular changes in vitro including decreased transendothelial electrical resistance, increased actinomyosin stress fiber formation, and alterations in tight junction molecules. These results suggest that AR signaling can be used to modulate BBB permeability in vivo to facilitate the entry of potentially therapeutic compounds into the CNS. AR signaling at brain endothelial cells represents a novel endogenous mechanism of modulating BBB permeability. We anticipate these results will aid in drug design, drug delivery and treatment options for neurological diseases such as AD, Parkinson's disease, multiple sclerosis and cancers of the CNS.


Subject(s)
Blood-Brain Barrier/metabolism , Receptor, Adenosine A1/physiology , Receptors, Adenosine A2/physiology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/immunology , Animals , Antibodies/metabolism , Blood-Brain Barrier/drug effects , Cells, Cultured , Dextrans/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelial Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Permeability , Purinergic P1 Receptor Agonists/pharmacology , Purines/pharmacology , Pyrazoles/pharmacology , Receptor, Adenosine A1/genetics , Receptors, Adenosine A2/genetics , Tight Junctions/metabolism
15.
Basic Clin Pharmacol Toxicol ; 109(3): 203-7, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21496211

ABSTRACT

Adenosinergic systems have been implicated in anxiety-like states, as caffeine can induce a state of anxiety in human beings. Caffeine is an antagonist at A(1) and A(2) adenosine receptors but it remains unclear whether anxiety is mediated by one or both of these. As the adenosinergic system is rather conserved, we opted to pursue these questions using zebrafish, a widely used model organism in genetics and developmental biology. Zebrafish adenosine 1. 2A.1 and 2A.2 receptors conserve histidine residues in TM6 and TM7 that are responsible for affinity in bovine A1 receptor. We investigated the effects of caffeine, PACPX (an A(1) receptor antagonist) and 1,3-dimethyl-1-propargylxanthine (DMPX) (an A(2) receptor antagonist) on anxiety-like behaviour and locomotor activity of zebrafish in the scototaxis test as well as evaluated the effects of these drugs on pigment aggregation. Caffeine increased anxiety at the dose of 100 mg/kg, while locomotion at the dose of 10 mg/kg was increased. Both doses of 10 and 100 mg/kg induced pigment aggregation. PACPX, on the other hand, increased anxiety at a dose of 6 mg/kg and induced pigment aggregation at the doses of 0.6 and 6 mg/kg, but did not produce a locomotor effect. DMPX, in turn, increased locomotion at the dose of 6 mg/kg but did not produce any effect on pigment aggregation or anxiety-like behaviour. These results indicate that blockade of A(1)-R, but not A(2)-R, induces anxiety and autonomic arousal, while the blockade of A(2)-R induces hyperlocomotion. Thus, as in rodents, caffeine's anxiogenic and arousing effects are probably mediated by A(1) receptors in zebrafish and its locomotor activating effect is probably mediated by A(2) receptors.


Subject(s)
Adenosine A1 Receptor Antagonists/pharmacology , Anxiety/chemically induced , Arousal/drug effects , Caffeine/pharmacology , Receptor, Adenosine A1/physiology , Zebrafish/metabolism , Adenosine A2 Receptor Antagonists/pharmacology , Animals , Anxiety/metabolism , Behavior, Animal/drug effects , Cattle , Darkness , Dose-Response Relationship, Drug , Melanophores/metabolism , Mice , Molecular Sequence Data , Motor Activity/drug effects , Pigments, Biological/metabolism , Receptor, Adenosine A1/metabolism , Receptors, Adenosine A2/metabolism , Receptors, Adenosine A2/physiology , Sequence Alignment
16.
Can J Physiol Pharmacol ; 89(3): 187-96, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21423292

ABSTRACT

It has been hypothesized that an interaction among adenosine A(1) receptors, protein kinase C (PKC) activation, and ATP-sensitive potassium channels (K(ATP)) mediates ischemic preconditioning in experiments on different animal species. The purpose of this study was to determine if activation of K(ATP) is functionally coupled to A(1) receptors and (or) PKC activation during metabolic inhibition (MI) in guinea pig ventricular myocytes. Perforated-patch using nystatin and conventional whole-cell recording methods were used to observe the effects of adenosine and adenosine-receptor antagonists on the activation of K(ATP) currents during MI induced by application of 2,4-dinitrophenol (DNP) and 2-deoxyglucose (2DG) without glucose, in the presence or absence of a PKC activator, phorbol 12-myristate 13-acetate (PMA). Adenosine accelerated the time course activation of K(ATP) currents during MI under the intact intracellular condition or dialyzed condition with l mmol/L ATP in the pipette solution. The accelerated effect of adenosine activation of K(ATP) under MI was not reversed by a nonselective Al adenosine receptor antagonist, 8-(p-sulfophenyl)theophylline (SPT), or a specific Al adenosine receptor antagonist, 8-cyclopentyl-1,3-dipropylxanthine (DPCPX). However, the adenosine A(2) receptor antagonist alloxazine reversed the time course activation of the K(ATP) current under MI. An adenylate cyclase activator, forskolin, did not further abbreviate the time course activation of K(ATP) with or without adenosine. Application of a PKC blocker, chelerythrine, reversed the time course activation of K(ATP) by adenosine under MI. In addition, pretreatment with a PKC activator, PMA, had similar effects to adenosine, while adenosine did not further shorten the time required for activation of K(ATP) currents during MI with PMA pretreatment. There is no direct evidence of activation of K(ATP) currents by adenosine A(1) receptor during metabolic inhibition under our experimental condition. However, adenosine A(2) receptor activation is involved in the K(ATP) channel activation in the guinea pig ventricular myocytes, of which effect is not mediated through the increase in intracellular cAMP. Adenosine seems to interact with PKC activation to open K(ATP) during MI, but a possible link between the adenosine A(2) receptor and PKC activation in this process needs further elucidation.


Subject(s)
Heart Ventricles/cytology , Heart Ventricles/metabolism , KATP Channels/antagonists & inhibitors , KATP Channels/metabolism , Myocytes, Cardiac/metabolism , Receptors, Adenosine A2/physiology , Animals , Guinea Pigs , Heart Ventricles/drug effects , Myocytes, Cardiac/drug effects , Purinergic P1 Receptor Agonists/pharmacology , Purinergic P1 Receptor Antagonists/pharmacology
17.
J Allergy Clin Immunol ; 127(2): 454-461.e1-9, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21281872

ABSTRACT

BACKGROUND: Water-soluble components from pollen modulate dendritic cell (DC) functions, such as IL-12 secretion and 3'-5'-cyclic adenosine monophosphate (cAMP) signaling and migration, possibly contributing to the establishment of a T(H)2-dominated immune response against pollen. Because these effects could not solely be attributed to the previously identified pollen-associated lipid mediators, the pollen metabolome was analyzed for candidate immunomodulatory substances. OBJECTIVE: We sought to perform an analysis of the effect of pollen-associated adenosine on DC function and T(H) cell differentiation. METHODS: Fractions of aqueous pollen extracts (APEs) were generated by means of ultrafiltration and were subjected simultaneously to biological tests and metabolome analysis (ultra-high-resolution mass spectrometry) and ultraperformance liquid chromatography. Effects of pollen-derived adenosine on monocyte-derived DC cAMP signaling, cytokine response, and capacity to differentiate T(H) cells were studied. RESULTS: The less than 3-kd fraction of APEs comprised thousands of substances, including adenosine in micromolar concentrations. Pollen-derived adenosine mediated A2 receptor-dependent induction of cAMP and inhibition of IL-12p70 in DCs. APEs digested with adenosine deaminase failed to mediate IL-12 inhibition. DCs of nonatopic donors exposed to APEs showed an adenosine-dependent reduced capacity to differentiate T(H)1 cells and an enhanced capacity to induce regulatory T cells and IL-10. DCs of atopic donors failed to induce IL-10 but instead induced IL-5 and IL-13. CONCLUSION: This study identifies adenosine out of thousands of metabolites as a potent immunoregulatory substance in pollen. It acts on the level of the DC, with differential effects in atopic and nonatopic donors.


Subject(s)
Adenosine/physiology , Dendritic Cells/physiology , Metabolome , Rhinitis, Allergic, Seasonal/etiology , T-Lymphocytes, Helper-Inducer/immunology , Adult , Cyclic AMP/biosynthesis , Humans , Interferon-gamma/biosynthesis , Interleukin-10/biosynthesis , Interleukin-12/biosynthesis , Receptors, Adenosine A2/physiology , Rhinitis, Allergic, Seasonal/immunology , T-Lymphocytes, Regulatory/physiology
18.
Handb Exp Pharmacol ; (200): 331-48, 2011.
Article in English | MEDLINE | ID: mdl-20859802

ABSTRACT

Caffeine is widely used to promote wakefulness and counteract fatigue induced by restriction of sleep, but also to counteract the effects of caffeine abstinence. Adenosine is a physiological molecule, which in the central nervous system acts predominantly as an inhibitory neuromodulator. Adenosine is also a sleep-promoting molecule. Caffeine binds to adenosine receptors, and the antagonism of the adenosinergic system is believed to be the mechanism through which caffeine counteracts sleep in humans as well as in other species. The sensitivity for caffeine varies markedly among individuals. Recently, genetic variations in genes related to adenosine metabolism have provided at least a partial explanation for this variability. The main effects of caffeine on sleep are decreased sleep latency, shortened total sleep time, decrease in power in the delta range, and sleep fragmentation. Caffeine may also decrease the accumulation of sleep propensity during waking, thus inducing long-term harmful effects on sleep quality.


Subject(s)
Caffeine/pharmacology , Sleep/drug effects , Adenosine/physiology , Animals , Humans , Receptor, Adenosine A1/physiology , Receptors, Adenosine A2/physiology , Sleep/physiology
19.
J Neuroimmunol ; 224(1-2): 85-92, 2010 Jul 27.
Article in English | MEDLINE | ID: mdl-20570369

ABSTRACT

We characterized the role of adenosine receptor (AR) subtypes in the modulation of glutamatergic neurotransmission by the chemokine fractalkine (CX3CL1) in mouse hippocampal CA1 neurons. CX(3)CL1 causes a reversible depression of excitatory postsynaptic current (EPSC), which is abolished by the A(3)R antagonist MRS1523, but not by A(1)R (DPCPX) or A(2A)R (SCH58261) antagonists. Consistently, CX3CL1-induced EPSC depression is absent in slices from A(3)R(-/-) but not A(1)R(-/-) or A(2A)R(-/-) mice. Further, A(3)R stimulation causes similar EPSC depression. In cultured neurons, CX3CL1-induced depression of AMPA current shows A(1)R-A(3)R pharmacology. We conclude that glutamatergic depression induced by released adenosine requires the stimulation of different ARs.


Subject(s)
CA1 Region, Hippocampal/immunology , CA1 Region, Hippocampal/metabolism , Chemokine CX3CL1/physiology , Excitatory Postsynaptic Potentials/immunology , Neural Inhibition/immunology , Receptors, Purinergic P1/physiology , Synaptic Transmission/immunology , Adenosine A1 Receptor Antagonists , Adenosine A2 Receptor Antagonists , Adenosine A3 Receptor Antagonists , Animals , CA1 Region, Hippocampal/ultrastructure , Cells, Cultured , Excitatory Postsynaptic Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Inhibition/genetics , Organ Culture Techniques , Patch-Clamp Techniques , Presynaptic Terminals/immunology , Presynaptic Terminals/metabolism , Receptor, Adenosine A1/deficiency , Receptor, Adenosine A1/physiology , Receptor, Adenosine A3/deficiency , Receptor, Adenosine A3/physiology , Receptors, Adenosine A2/deficiency , Receptors, Adenosine A2/physiology , Receptors, Purinergic P1/deficiency , Receptors, Purinergic P1/genetics , Synaptic Transmission/genetics
20.
Am J Physiol Lung Cell Mol Physiol ; 298(6): L755-67, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20228181

ABSTRACT

We have previously demonstrated that adenosine plus homocysteine enhanced endothelial basal barrier function and protected against agonist-induced barrier dysfunction in vitro through attenuation of RhoA activation by inhibition of isoprenylcysteine-O-carboxyl methyltransferase. In the current study, we tested the effect of elevated adenosine on pulmonary endothelial barrier function in vitro and in vivo. We noted that adenosine alone dose dependently enhanced endothelial barrier function. While adenosine receptor A(1) or A(3) antagonists were ineffective, an adenosine transporter inhibitor, NBTI, or a combination of DPMX and MRS1754, antagonists for adenosine receptors A(2A) and A(2B), respectively, partially attenuated the barrier-enhancing effect of adenosine. Similarly, inhibition of both A(2A) and A(2B) receptors with siRNA also blunted the effect of adenosine on barrier function. Interestingly, inhibition of both transporters and A(2A)/A(2B) receptors completely abolished adenosine-induced endothelial barrier enhancement. The adenosine receptor A(2A) and A(2B) agonist, NECA, also significantly enhanced endothelial barrier function. These data suggest that both adenosine transporters and A(2A) and A(2B) receptors are necessary for exerting maximal effect of adenosine on barrier enhancement. We also found that adenosine enhanced Rac1 GTPase activity and overexpression of dominant negative Rac1 attenuated adenosine-induced increases in focal adhesion complexes. We further demonstrated that elevation of cellular adenosine by inhibition of adenosine deaminase with Pentostatin significantly enhanced endothelial basal barrier function, an effect that was also associated with enhanced Rac1 GTPase activity and with increased focal adhesion complexes and adherens junctions. Finally, using a non-inflammatory acute lung injury (ALI) model induced by alpha-naphthylthiourea, we found that administration of Pentostatin, which elevated lung adenosine level by 10-fold, not only attenuated the development of edema before ALI but also partially reversed edema after ALI. The data suggest that adenosine deaminase inhibition may be useful in treatment of pulmonary edema in settings of ALI.


Subject(s)
Receptors, Adenosine A2/physiology , Acute Lung Injury/chemically induced , Acute Lung Injury/complications , Adenosine/pharmacology , Adenosine Deaminase Inhibitors , Adherens Junctions/drug effects , Animals , Cattle , Endothelium/metabolism , Endothelium, Vascular/cytology , Focal Adhesions/metabolism , Lung/metabolism , Male , Nucleoside Transport Proteins/physiology , Pentostatin/pharmacology , Pentostatin/therapeutic use , Pulmonary Edema/prevention & control , Rats , Rats, Sprague-Dawley , Receptor, Adenosine A2A/physiology , Receptor, Adenosine A2B/physiology , Thiourea/analogs & derivatives , rac1 GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL