Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 826
Filter
1.
Pharmacol Rep ; 71(4): 676-681, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31200233

ABSTRACT

BACKGROUND: Adenosine, an endogenous nucleoside, modulates the release of monoamines, e.g., noradrenaline, serotonin, and dopamine in the brain. Both nonselective and selective stimulation of adenosine receptors produce symptoms of depression in some animal models. Therefore, the main objective of our study was to assess the influence of a selective adenosine A1 receptor antagonist (DPCPX) and a selective adenosine A2A receptor antagonist (DMPX) on the activity of agomelatine and tianeptine. METHODS: The forced swim test (FST) and tail suspension test (TST) were performed to assess the effects of DPCPX and DMPX on the antidepressant-like activity of agomelatine and tianeptine. Drug serum and brain levels were analyzed using HPLC. RESULTS: Co-administration of agomelatine (20 mg/kg) or tianeptine (15 mg/kg) with DMPX (3 mg/kg), but not with DPCPX (1 mg/kg), significantly reduced the immobility time both in the FST and TST in mice. These effects were not associated with an enhancement in animals' spontaneous locomotor activity. The observed changes in the mouse behavior after concomitant injection of DMPX and the tested antidepressant agents were associated with elevated brain concentration of agomelatine and tianeptine. CONCLUSION: Our study shows a synergistic action of the selective A2A receptor antagonist and the studied antidepressant drugs, and a lack of such interaction in the case of the selective A1 receptor antagonist. The interaction between DMPX and agomelatine/tianeptine at least partly occurs in the pharmacokinetic phase. A combination of a selective A2A receptor antagonist and an antidepressant may be a new strategy for treating depression.


Subject(s)
Acetamides/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Antidepressive Agents/pharmacology , Depression/drug therapy , Theobromine/analogs & derivatives , Thiazepines/pharmacology , Acetamides/pharmacokinetics , Adenosine A1 Receptor Antagonists/pharmacokinetics , Adenosine A1 Receptor Antagonists/pharmacology , Adenosine A2 Receptor Antagonists/pharmacokinetics , Animals , Antidepressive Agents/pharmacokinetics , Brain/drug effects , Brain/metabolism , Depression/blood , Depression/metabolism , Drug Synergism , Male , Mice , Motor Activity/drug effects , Swimming , Theobromine/pharmacokinetics , Theobromine/pharmacology , Thiazepines/pharmacokinetics , Xanthines/pharmacokinetics , Xanthines/pharmacology
2.
PLoS One ; 14(6): e0218619, 2019.
Article in English | MEDLINE | ID: mdl-31237895

ABSTRACT

(-)-Incarvillateine (INCA) is a natural product that has garnered attention due to its purported analgesic effects and historical use as a pain reliever in China. α-Truxillic acid monoesters (TAMEs) constitute a class of inhibitors targeting fatty acid binding protein 5 (FABP5), whose inhibition produces analgesia in animal models. The structural similarity between INCA and TAMEs motivated us to assess whether INCA exerts its antinociceptive effects via FABP inhibition. We found that, in contrast to TAMEs, INCA did not exhibit meaningful binding affinities toward four human FABP isoforms (FABP3, FABP4, FABP5 and FABP7) in vitro. INCA-TAME, a putative monoester metabolite of INCA that closely resembles TAMEs also lacked affinity for FABPs. Administration of INCA to mice produced potent antinociceptive effects while INCA-TAME was without effect. Surprisingly, INCA also potently suppressed locomotor activity at the same dose that produces antinociception. The motor suppressive effects of INCA were reversed by the adenosine A2 receptor antagonist 3,7-dimethyl-1-propargylxanthine. Collectively, our results indicate that INCA and INCA-TAME do not inhibit FABPs and that INCA exerts potent antinociceptive and motor suppressive effects at equivalent doses. Therefore, the observed antinociceptive effects of INCA should be interpreted with caution.


Subject(s)
Alkaloids/pharmacology , Analgesics/pharmacology , Locomotion/drug effects , Monoterpenes/pharmacology , Nociception/drug effects , Receptors, Adenosine A2/metabolism , Adenosine A2 Receptor Agonists/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Animals , Fatty Acid-Binding Proteins/metabolism , Humans , Male , Mice , Protein Binding , Theobromine/analogs & derivatives , Theobromine/pharmacology
3.
Eur J Med Chem ; 174: 76-86, 2019 Jul 15.
Article in English | MEDLINE | ID: mdl-31029946

ABSTRACT

Reactive oxygen species (ROS) play crucial roles in maintaining redox balance and regulating physiological processes, ROS levels in cancer cells are relatively higher than those in normal cells. Therefore, elevating cellular ROS levels may be a viable strategy for selective killing of cancer cells. In this work, we synthesized a series of new theobromine derivatives and evaluated their cytotoxicity against gastric cancer cells MGC-803, SGC-7901 and HGC-27. Particularly, MQS-14 potently inhibited cell growth of MGC-803, SGC-7901 and HGC-27 cells at low micromolar levels. Mechanistic studies showed that compound MQS-14 decreased cell viability of MGC-803 cells and inhibited cell division revealed by the CFDA and EdU staining assays. MQS-14 increased cellular ROS levels and activated the MAPK pathway accompanied by the decreased p-ERK and increased p-JNK expression. MQS-14 also induced DNA damage and apoptosis in MGC-803 cells. To conclude, MQS-14 induced cell death of MGC-803 cells partly through elevating cellular ROS levels.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Reactive Oxygen Species/metabolism , Theobromine/analogs & derivatives , Theobromine/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Division/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Damage/drug effects , Humans , MAP Kinase Signaling System/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Molecular Structure , Structure-Activity Relationship , Theobromine/chemical synthesis
4.
Neurotox Res ; 35(2): 344-352, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30267268

ABSTRACT

Unsatisfactory therapeutic effects of currently used antidepressants force to search for new pharmacological treatment strategies. Recent research points to the relationship between depressive disorders and the adenosinergic system. Therefore, the main goal of our studies was to evaluate the effects of DMPX (3 mg/kg, i.p.), which possesses selectivity for adenosine A2A receptors versus A1 receptors, on the activity of imipramine (15 mg/kg, i.p.), escitalopram (2.5 mg/kg, i.p.), and reboxetine (2 mg/kg, i.p.) given in subtherapeutic doses. The studies carried out using the forced swim and tail suspension tests in mice showed that DMPX at a dose of 6 and 12 mg/kg exerts antidepressant-like effect and does not affect the locomotor activity. Co-administration of DMPX at a dose of 3 mg/kg with the studied antidepressant drugs caused the reduction of immobility time in both behavioral tests. The observed effect was not associated with an increase in the locomotor activity. To evaluate whether the observed effects were due to a pharmacokinetic/pharmacodynamic interaction, the levels of the antidepressants in blood and brain were measured using high-performance liquid chromatography. It can be assumed that the interaction between DMPX and imipramine was exclusively pharmacodynamic in nature, whereas an increased antidepressant activity of escitalopram and reboxetine was at least partly related to its pharmacokinetic interaction with DMPX.


Subject(s)
Adenosine A2 Receptor Antagonists/administration & dosage , Antidepressive Agents/administration & dosage , Hindlimb Suspension/psychology , Receptor, Adenosine A2A/metabolism , Swimming/psychology , Theobromine/analogs & derivatives , Animals , Depression/drug therapy , Depression/metabolism , Depression/psychology , Dose-Response Relationship, Drug , Drug Synergism , Hindlimb Suspension/methods , Locomotion/drug effects , Locomotion/physiology , Male , Mice , Swimming/physiology , Theobromine/administration & dosage
5.
Sci Rep ; 5: 16107, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26527075

ABSTRACT

Incarvillea sinensis is a Bignoniaceae plant used to treat rheumatism and relieve pain in traditional Chinese medicine. As a major component of I. sinensis, incarvillateine has shown analgesic activity in mice formalin tests. Using a series of animal models, this study further evaluated the effects of incarvillateine against acute, inflammatory, and neuropathic pain. Incarvillateine (10 or 20 mg/kg, i.p.) dose-dependently attenuated acetic acid-induced writhing, but did not affect thermal threshold in the hot plate test. In a Complete Freund's Adjuvant model, incarvillateine inhibited both thermal hyperalgesia and paw edema, and increased interleukin-1ß levels. Additionally, incarvillateine attenuated mechanical allodynia induced by spared nerve injury or paclitaxel, whereas normal mechanical sensation was not affected. Incarvillateine did not affect locomotor activity and time on the rotarod at analgesic doses, and no tolerance was observed after 7 consecutive daily doses. Moreover, incarvillateine-induced antinociception was attenuated by theophylline, 1,3-dipropyl-8-cyclopentylxanthine, and 3,7-dimethyl-1-propargylxanthine, but not naloxone, indicating that the effects of incarvillateine on chronic pain were related to the adenosine system, but not opioid system. These results indicate that incarvillateine is a novel analgesic compound that is effective against inflammatory and neuropathic pain, and that its effects are associated with activation of the adenosine system.


Subject(s)
Adenosine/metabolism , Alkaloids/pharmacology , Analgesics/pharmacology , Bignoniaceae/chemistry , Monoterpenes/pharmacology , Alkaloids/chemistry , Alkaloids/therapeutic use , Analgesics/chemistry , Analgesics/therapeutic use , Animals , Antineoplastic Agents, Phytogenic/toxicity , Bignoniaceae/metabolism , Disease Models, Animal , Edema/chemically induced , Edema/prevention & control , Freund's Adjuvant/chemistry , Hyperalgesia/etiology , Hyperalgesia/prevention & control , Interleukin-1beta/metabolism , Medicine, Chinese Traditional , Mice , Monoterpenes/chemistry , Monoterpenes/therapeutic use , Motor Activity/drug effects , Paclitaxel/toxicity , Pain Measurement/drug effects , Theobromine/analogs & derivatives , Theobromine/pharmacology , Theophylline/pharmacology , Xanthines/pharmacology
6.
Biomed Res Int ; 2015: 248976, 2015.
Article in English | MEDLINE | ID: mdl-26347229

ABSTRACT

Neuronal apoptosis inhibitory protein (NAIP) and survivin might play an important role in testicular function. We investigated the effect of PDRN, an agonist of adenosine A2A receptor, on testicular NAIP and survivin expression in an experimental model of varicocele. After the creation of experimental varicocele (28 days), adolescent male Sprague-Dawley rats were randomized to one of the following treatments lasting 21 days: vehicle, PDRN (8 mg/kg i.p., daily), PDRN + 3,7-dimethyl-propargylxanthine (DMPX, a specific adenosine A2A-receptor antagonist, 0.1 mg/kg i.p., daily), varicocelectomy, and varicocelectomy + PDRN (8 mg/kg i.p., daily). Sham-operated animals were used as controls. Animals were then euthanized and testis expression of NAIP and survivin was evaluated through qRT-PCR, western blot, and immunohistochemical analysis. Spermatogenetic activity was also assessed. NAIP and survivin expressions were significantly reduced following varicocele induction when compared to sham animals whereas PDRN-treated rats showed an increase in NAIP and survivin levels. Immunohistochemistry revealed an enhanced expression of NAIP and survivin with a characteristic pattern of cellular localization following PDRN treatment. Moreover, administration of PDRN significantly restored spermatogenic function in varicocele rats. PDRN may represent a rational therapeutic option for accelerating recovery from depressed testicular function through a strategic modulation of apoptosis in experimental varicocele.


Subject(s)
Fertility/drug effects , Microtubule-Associated Proteins/biosynthesis , Neuronal Apoptosis-Inhibitory Protein/biosynthesis , Oligodeoxyribonucleotides/pharmacology , Testis/metabolism , Varicocele/metabolism , Adenosine A2 Receptor Antagonists/pharmacology , Animals , Disease Models, Animal , Gene Expression Regulation/drug effects , Male , Rats , Receptors, Adenosine A2/metabolism , Spermatogenesis/drug effects , Survivin , Testis/pathology , Theobromine/analogs & derivatives , Theobromine/pharmacology , Varicocele/pathology
7.
Respir Physiol Neurobiol ; 200: 80-9, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24914466

ABSTRACT

Methylxanthines like caffeine and theophylline have long been used to treat apnea of prematurity. Despite their success in stimulating neonatal breathing, their mechanism of action remains poorly understood. Methylxanthines can act as both non-specific adenosine receptor antagonists and inhibitors of cAMP-dependent phosphodiesterases, sarcoplasmic/endoplasmic reticulum calcium ATPases or receptor-coupled anion channels, depending on the dose used. Though there is evidence for methylxanthine action at the level of the carotid body, the consensus is that methylxanthines stimulate the respiratory centers of the brainstem. Here we used the in situ neonatal rat working heart-brainstem preparation and the ex vivo neonatal rat carotid body preparation to test the hypothesis that methylxanthines act at the level of the carotid body. We conclude that although the neonatal carotid body has active adenosine receptors, the effects of methylxanthine therapy are likely mediated centrally, predominantly via inhibition of cAMP-dependent phosphodiesterase-4.


Subject(s)
Brain Stem/drug effects , Carotid Body/drug effects , Respiration/drug effects , Theophylline/pharmacology , Xanthines/pharmacology , Adenosine A1 Receptor Antagonists/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Animals , Animals, Newborn , Apnea/drug therapy , Apnea/physiopathology , Brain Stem/physiology , Carotid Body/physiology , Central Nervous System Stimulants , Enkephalin, Ala(2)-MePhe(4)-Gly(5)- , Phrenic Nerve/drug effects , Phrenic Nerve/physiology , Quinazolines/pharmacology , Rats, Sprague-Dawley , Theobromine/analogs & derivatives , Theobromine/pharmacology , Tissue Culture Techniques , Triazoles/pharmacology
8.
Alcohol Clin Exp Res ; 38(3): 629-40, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24117724

ABSTRACT

BACKGROUND: The cAMP-dependent protein kinase (PKA) signaling transduction pathway has been shown to play an important role in the modulation of several ethanol (EtOH)-induced behavioral actions. In vivo, short-term exposure to EtOH up-regulates the cAMP-signaling cascade. Interestingly, different Ca(2+) -dependent cAMP-PKA cascade mediators play a critical role in the neurobehavioral response to EtOH, being of special relevance to the Ca(2+) -dependent adenylyl cyclases 1 and 8. We hypothesize an intracellular PKA activation elicited by EtOH administration, which may be regulated by a Ca(2+) -dependent mechanism as an early cellular response. Thus, the present work aims to explore the role of Ca(2+) (internal and external) on the EtOH-activated PKA cascade. METHODS: Swiss male mice received an intraperitoneal injection of EtOH (0 or 4 g/kg), and brains were dissected following a temporal pattern (7, 15, 30, 45, 90, or 120 minutes). Either the enzymatic PKA activity or its fingerprint was analyzed on different brain areas (cortex, hypothalamus, hippocampus, and striatum). To explore the role of Ca(2+) on the EtOH-activated PKA cascade, mice were pretreated with diltiazem (0 or 20 mg/kg), dantrolene (0 or 5 mg/kg), or 3,7-Dimethyl-1-(2-propynyl)xanthine (0 or 1 mg/kg) 30 minutes before EtOH (4 g/kg) administration. After 45 minutes of EtOH administration, brains were removed and dissected to measure the PKA activity or its fingerprint. RESULTS: Results from these experiments showed an EtOH-dependent activation of PKA in different brain areas. Manipulations involving a disruption of intracellular Ca(2+) release from the endoplasmic reticulum resulted in a decreased EtOH-induced activation of PKA. On the contrary, extracellular-to-cytoplasm Ca(2+) manipulations did not prevent the PKA activation by EtOH. CONCLUSIONS: Altogether, these results show the critical role of stored Ca(2+) as an intracellular mediator of different neurobiological actions of EtOH and provide further evidence of a possible new target for EtOH within the central nervous system.


Subject(s)
Brain/drug effects , Calcium/metabolism , Central Nervous System Depressants/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Ethanol/pharmacology , Adenosine A2 Receptor Antagonists , Animals , Brain/enzymology , Calcium Channel Blockers , Endoplasmic Reticulum/metabolism , Enzyme Activation/drug effects , Male , Mice , Theobromine/analogs & derivatives
9.
J Clin Periodontol ; 40(1): 26-32, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23033941

ABSTRACT

AIM: Adenosine receptors modulate inflammation in periodontal tissues. No data are available regarding the effects of adenosine A(2A) receptor stimulation in experimental periodontitis (EPD). The aim of this study was to investigate the effects of polynucleotides (also known as polydeoxyribonucleotide, PDRN), a ligand of A(2A) receptor, in EPD in rats. MATERIALS AND METHODS: EPD was induced ligating the cervix of the lower left first molar. Sham-EPD had no ligature. After 7 days, EPD animals were randomized to a daily treatment with vehicle gel or 0.75% PDRN gel or PDRN gel with a specific A(2A) antagonist (DMPX). Treatments lasted 7 days. Animals were then euthanized and the periodontium and surrounding gingival tissue were excised for histological evaluation and bio-molecular analysis of inflammatory (p-JNK, p-ERK, TNF-α, IL-6, HMGB-1) and apoptotic proteins (BAX and Bcl-2). RESULTS: Vehicle-treated EPD rats showed severe inflammatory infiltrate in both gingival and periodontal ligament, as well as an enhanced expression of p-JNK, p-ERK, TNF-α, IL-6, HMGB-1 and BAX and a reduction in Bcl-2. PDRN gel restored the histological features, blunted inflammatory and apoptotic proteins expression and preserved Bcl-2 expression. DMPX abrogated PDRN positive effects. CONCLUSION: Our data suggest that adenosine receptor stimulation by PDRN might represent a new therapeutic strategy for periodontitis.


Subject(s)
Inflammation Mediators/pharmacology , Periodontitis/metabolism , Polydeoxyribonucleotides/pharmacology , Receptors, Purinergic P1/drug effects , Theobromine/analogs & derivatives , Adenosine A2 Receptor Antagonists/pharmacology , Alveolar Bone Loss/metabolism , Animals , Extracellular Signal-Regulated MAP Kinases/metabolism , HMGB1 Protein/metabolism , Interleukin-6/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Proto-Oncogene Proteins c-bcl-2/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley , Theobromine/pharmacology , Tumor Necrosis Factor-alpha/metabolism , bcl-2-Associated X Protein/metabolism
10.
Adv Exp Med Biol ; 765: 225-230, 2013.
Article in English | MEDLINE | ID: mdl-22879037

ABSTRACT

Alterations in glomerular filtration rate (GFR) are one of the earliest indications of altered kidney function in diabetes. Adenosine regulates GFR through tubuloglomerular feedback mechanism acting on adenosine A1 receptor. In addition, adenosine can directly regulate vascular tone by acting on A1 and A2 receptors expressed in afferent and efferent arterioles. Opposite to A1 receptors, A2 receptors mediate vasorelaxation. This study investigates the involvement of adenosine A2 receptors in regulation of renal blood flow (RBF) and GFR in control and diabetic kidneys. GFR was measured by inulin clearance and RBF by a transonic flow probe placed around the renal artery. Measurements were performed in isoflurane-anesthetized normoglycemic and alloxan-diabetic C57BL/6 mice during baseline and after acute administration of 3,7-dimethyl-1-propargylxanthine (DMPX), a selective A2 receptor antagonist. GFR and RBF were lower in diabetic mice compared to control (258 ± 61 vs. 443 ± 33 µl min(-1) and 1,083 ± 51 vs. 1,405 ± 78 µl min(-1)). In control animals, DMPX decreased RBF by -6%, whereas GFR increased +44%. DMPX had no effects on GFR and RBF in diabetic mice. Sodium excretion increased in diabetic mice after A2 receptor blockade (+78%). In conclusion, adenosine acting on A2 receptors mediates an efferent arteriolar dilatation which reduces filtration fraction (FF) and maintains GFR within normal range in normoglycemic mice. However, this regulation is absent in diabetic mice, which may contribute to reduced oxygen availability in the diabetic kidney.


Subject(s)
Arterioles/pathology , Diabetes Mellitus, Experimental/physiopathology , Glomerular Filtration Rate , Hemodynamics/physiology , Kidney/physiopathology , Receptors, Adenosine A2/metabolism , Renal Circulation/physiology , Adenosine/metabolism , Adenosine A2 Receptor Antagonists/pharmacology , Animals , Arterioles/metabolism , Diabetes Mellitus, Experimental/drug therapy , Kidney/blood supply , Male , Mice , Mice, Inbred C57BL , Receptors, Adenosine A2/chemistry , Renal Circulation/drug effects , Theobromine/analogs & derivatives , Theobromine/pharmacology
11.
J Neurosci ; 32(40): 13945-55, 2012 Oct 03.
Article in English | MEDLINE | ID: mdl-23035103

ABSTRACT

After hypoxia, a critical adverse outcome is the inability to create new memories. How anterograde amnesia develops or resolves remains elusive, but a link to brain-based IL-1 is suggested due to the vital role of IL-1 in both learning and brain injury. We examined memory formation in mice exposed to acute hypoxia. After reoxygenation, memory recall recovered faster than memory formation, impacting novel object recognition and cued fear conditioning but not spatially cued Y-maze performance. The ability of mice to form new memories after hypoxia/reoxygenation was accelerated in IL-1 receptor 1 knockout (IL-1R1 KO) mice, in mice receiving IL-1 receptor antagonist (IL-1RA), and in mice given the caspase 1 inhibitor Ac-YVAD-CMK. Mechanistically, hypoxia/reoxygenation more than doubled caspase 1 activity in the brain, which was localized to the amygdala compared to the hippocampus. This reoxygenation-dependent activation of caspase 1 was prevented by broad-spectrum adenosine receptor (AR) antagonism with caffeine and by targeted A1/A2A AR antagonism with 8-cyclopentyl-1,3-dipropylxanthine plus 3,7-dimethyl-1-propargylxanthine. Additionally, perfusion of adenosine activated caspase 1 in the brain, while caffeine blocked this action by adenosine. Finally, resolution of anterograde amnesia was improved by both caffeine and by targeted A1/A2A AR antagonism. These findings indicate that amygdala-based anterograde amnesia after hypoxia/reoxygenation is sustained by IL-1ß generated through adenosine-dependent activation of caspase 1 after reoxygenation.


Subject(s)
Adenosine/physiology , Amnesia, Anterograde/enzymology , Amygdala/physiology , Caspase 1/physiology , Hypoxia, Brain/complications , Adenosine/pharmacology , Amino Acid Chloromethyl Ketones/pharmacology , Amnesia, Anterograde/etiology , Amnesia, Anterograde/physiopathology , Amygdala/drug effects , Amygdala/enzymology , Animals , Caffeine/pharmacology , Caspase 1/drug effects , Caspase Inhibitors/pharmacology , Conditioning, Classical/drug effects , Conditioning, Classical/physiology , Cues , Enzyme Activation , Fear/drug effects , Fear/physiology , Hypoxia, Brain/physiopathology , Interleukin 1 Receptor Antagonist Protein/pharmacology , MAP Kinase Signaling System , Male , Maze Learning/drug effects , Maze Learning/physiology , Mental Recall , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxygen/metabolism , Oxygen/pharmacology , Receptors, Interleukin-1 Type I/deficiency , Receptors, Purinergic P1/physiology , Recognition, Psychology/drug effects , Recognition, Psychology/physiology , Theobromine/analogs & derivatives , Theobromine/pharmacology , Xanthines/pharmacology
12.
Neuroscience ; 201: 46-56, 2012 Jan 10.
Article in English | MEDLINE | ID: mdl-22119641

ABSTRACT

Hypocretin neurons in the lateral hypothalamus, a new wakefulness-promoting center, have been recently regarded as an important target involved in endogenous adenosine-regulating sleep homeostasis. The GABAergic synaptic transmissions are the main inhibitory afferents to hypocretin neurons, which play an important role in the regulation of excitability of these neurons. The inhibitory effect of adenosine, a homeostatic sleep-promoting factor, on the excitatory glutamatergic synaptic transmissions in hypocretin neurons has been well documented, whether adenosine also modulates these inhibitory GABAergic synaptic transmissions in these neurons has not been investigated. In this study, the effect of adenosine on inhibitory postsynaptic currents (IPSCs) in hypocretin neurons was examined by using perforated patch-clamp recordings in the acute hypothalamic slices. The findings demonstrated that adenosine suppressed the amplitude of evoked IPSCs in a dose-dependent manner, which was completely abolished by 8-cyclopentyltheophylline (CPT), a selective antagonist of adenosine A1 receptor but not adenosine A2 receptor antagonist 3,7-dimethyl-1-(2-propynyl) xanthine. A presynaptic origin was suggested as following: adenosine increased paired-pulse ratio as well as reduced GABAergic miniature IPSC frequency without affecting the miniature IPSC amplitude. Further findings demonstrated that when the frequency of electrical stimulation was raised to 10 Hz, but not 1 Hz, a time-dependent depression of evoked IPSC amplitude was detected in hypocretin neurons, which could be partially blocked by CPT. However, under a higher frequency at 100 Hz stimulation, CPT had no action on the depressed GABAergic synaptic transmission induced by such tetanic stimulation in these hypocretin neurons. These results suggest that endogenous adenosine generated under certain stronger activities of synaptic transmissions exerts an inhibitory effect on GABAergic synaptic transmission in hypocretin neurons by activation of presynaptic adenosine A1 receptors, which may finely regulate the excitability of these neurons as well as eventually modulate the sleep-wakefulness.


Subject(s)
Adenosine/pharmacology , GABAergic Neurons/cytology , Hypothalamus/cytology , Intracellular Signaling Peptides and Proteins/metabolism , Neural Inhibition/drug effects , Neuropeptides/metabolism , Presynaptic Terminals/drug effects , Synaptic Transmission/drug effects , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Animals, Newborn , Drug Interactions , Electric Stimulation , Excitatory Amino Acid Antagonists/pharmacology , GABA Agents/pharmacology , GABAergic Neurons/drug effects , Green Fluorescent Proteins/genetics , In Vitro Techniques , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Transgenic , Neuropeptides/genetics , Orexins , Patch-Clamp Techniques , Purinergic Antagonists/pharmacology , Theobromine/analogs & derivatives , Theobromine/pharmacology , Theophylline/analogs & derivatives , Theophylline/pharmacology , Valine/analogs & derivatives , Valine/pharmacology
13.
Nitric Oxide ; 26(1): 1-8, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22119149

ABSTRACT

This study examined the cytoprotective mechanisms of a combination of ischemic preconditioning (IPC) and allopurinol against liver injury caused by ischemia/reperfusion (I/R). Allopurinol (50mg/kg) was intraperitoneally administered 18 and 1h before sustained ischemia. A rat liver was preconditioned by 10 min of ischemia, followed by 10 min of reperfusion, and then subjected to 90 min of ischemia, followed by 5h of reperfusion. Rats were pretreated with adenosine deaminase (ADA), 3,7-dimethyl-1-[2-propargyl]-xanthine (DMPX), and N-nitro-l-arginine methyl ester (l-NAME) before IPC. Hepatic nitrite and nitrate and eNOS protein expression levels were increased by the combination of IPC and allopurinol. This increase was attenuated by ADA, DMPX, and l-NAME. I/R induced an increase in alanine aminotransferase activity, whereas it decreased the hepatic glutathione level. A combination of IPC and allopurinol attenuated these changes, which were abolished by ADA, DMPX, and l-NAME. The increase in the liver wet weight-to-dry weight ratio after I/R was attenuated by the combination of IPC and allopurinol. In contrast, hepatic bile flow was decreased after I/R, which was attenuated by the combination of IPC and allopurinol. These changes were restored by l-NAME. I/R induced a decrease in the level of mitochondrial dehydrogenase, whereas it increased mitochondrial swelling. A combination of IPC and allopurinol attenuated these changes, which were restored by ADA, DMPX, and l-NAME. Our findings suggest that a combination of IPC and allopurinol reduces post-ischemic hepatic injury by enhancing NO generation.


Subject(s)
Allopurinol/pharmacology , Ischemic Preconditioning , Liver/blood supply , Liver/metabolism , Nitric Oxide/metabolism , Adenosine Deaminase/metabolism , Adenosine Deaminase/pharmacology , Alanine Transaminase/blood , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , Cyclic GMP/metabolism , Glutathione/metabolism , Ischemia/metabolism , Liver/drug effects , Male , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Mitochondrial Swelling/drug effects , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III/metabolism , Organ Size/drug effects , Rats , Rats, Sprague-Dawley , Theobromine/analogs & derivatives , Theobromine/pharmacology
14.
Neurosci Lett ; 490(1): 63-7, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21167909

ABSTRACT

Adenosine is an inhibitory modulator of neuronal transmission, including GABAergic transmission in the hypothalamus. It is known that the local GABAergic inputs tonically inhibit the hypothalamic paraventricular neurons projecting to the rostral ventrolateral medulla (RVLM; PVN-RVLM neurons) which regulate sympathetic outflow. In this study, we examined the effects of adenosine on GABAergic synaptic transmission in the PVN-RVLM neurons using whole cell patch-clamp combined with the retrograde labeling technique. Adenosine (100 µM) reversibly decreased the frequency of miniature IPSCs (from 3.41 ± 0.75 to 2.19 ± 0.49 Hz) in a concentration-dependent manner (IC50 = 1.0 µM) without affecting the amplitude and the decay time constant of miniature IPSCs. Adenosine increased the paired-pulse ratio of evoked IPSCs from 1.19 ± 0.05 to 2.28 ± 0.09 (P<0.001). The effects of adenosine was mimicked by a selective A1 receptor agonist (CHA, 10 µM), and blocked by a selective A1 receptor antagonist (DPCPX, 2 µM), but not by a selective A2 receptor antagonist (DMPX, 10 µM). In conclusion, the results showed that adenosine inhibits synaptic GABA release via presynaptic A1 receptors in the PVN-RVLM neurons, indicating a potential of adenosine A1 receptors in regulating sympathetic tone in normal and disease states.


Subject(s)
Adenosine/pharmacology , Inhibitory Postsynaptic Potentials/drug effects , Medulla Oblongata/physiology , Neurons/drug effects , Paraventricular Hypothalamic Nucleus/cytology , Receptor, Adenosine A1/metabolism , gamma-Aminobutyric Acid/metabolism , Adenosine/analogs & derivatives , Adenosine A1 Receptor Agonists/pharmacology , Adenosine A1 Receptor Antagonists/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Afferent Pathways/physiology , Animals , Biophysics , Electric Stimulation , Male , Neural Inhibition/drug effects , Neurons/physiology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Theobromine/analogs & derivatives , Theobromine/pharmacology , Xanthines/pharmacology , gamma-Aminobutyric Acid/pharmacology
15.
Hippocampus ; 21(3): 265-80, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20054814

ABSTRACT

Stellate neurons in layer II entorhinal cortex (EC) provide the main output from the EC to the hippocampus. It is believed that adenosine plays a crucial role in neuronal excitability and synaptic transmission in the CNS, however, the function of adenosine in the EC is still elusive. Here, the data reported showed that adenosine hyperpolarized stellate neurons in a concentration-dependent manner, accompanied by a decrease in firing frequency. This effect corresponded to the inhibition of the hyperpolarization-activated, cation nonselective (HCN) channels. Surprisingly, the adenosine-induced inhibition was blocked by 3 µM 8-cyclopentyl-1,3-dipropylxanthine (DPCPX), a selective A(1) receptor antagonists, but not by 10 µM 3,7-dimethyl-1-propargylxanthine (DMPX), a selective A(2) receptor antagonists, indicating that activation of adenosine A(1) receptors were responsible for the direct inhibition. In addition, adenosine reduced the frequency but not the amplitude of miniature EPSCs and IPSCs, suggesting that the global depression of glutamatergic and GABAergic transmission is mediated by a decrease in glutamate and GABA release, respectively. Again the presynaptic site of action was mediated by adenosine A(1) receptors. Furthermore, inhibition of spontaneous glutamate and GABA release by adenosine A(1) receptor activation was mediated by voltage-dependent Ca(2+) channels and extracellular Ca(2+) . Therefore, these findings revealed direct and indirect mechanisms by which activation of adenosine A(1) receptors on the cell bodies of stellate neurons and on the presynaptic terminals could regulate the excitability of these neurons.


Subject(s)
Adenosine , Entorhinal Cortex/metabolism , Neural Inhibition/physiology , Receptor, Adenosine A1/metabolism , Synaptic Transmission/physiology , Adenosine/metabolism , Adenosine/pharmacology , Adenosine A1 Receptor Antagonists/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Animals , Calcium/metabolism , Calcium Channels/metabolism , Entorhinal Cortex/cytology , Entorhinal Cortex/drug effects , Excitatory Postsynaptic Potentials/physiology , Glutamic Acid/metabolism , Neural Inhibition/drug effects , Neurons/drug effects , Neurons/metabolism , Organ Culture Techniques , Patch-Clamp Techniques , Presynaptic Terminals/drug effects , Rats , Rats, Sprague-Dawley , Receptor, Adenosine A1/drug effects , Receptors, Adenosine A2/drug effects , Receptors, Adenosine A2/metabolism , Synaptic Transmission/drug effects , Theobromine/analogs & derivatives , Theobromine/pharmacology , Xanthines/pharmacology , gamma-Aminobutyric Acid/metabolism
16.
Neuroreport ; 20(11): 1013-8, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19491711

ABSTRACT

Multiple studies indicate that adenosine released in the basal forebrain during prolonged wakefulness could affect recovery sleep. It is still unclear which of adenosine receptors provide its sleep-modulating effects in the basal forebrain. We infused adenosine A1 and A2A receptors antagonists into the rat basal forebrain during sleep deprivation and compared characteristics of recovery non-rapid eye movement (non-REM) sleep (its amount and non-REM sleep delta power) after sleep deprivation, and after sleep deprivation combined with perfusion of antagonists. A1 receptor antagonist significantly reduced recovery sleep amount and delta power, whereas A2A receptor antagonist had no effect on recovery sleep. We conclude that adenosine can promote recovery non-REM sleep when acting through A1 receptors in the basal forebrain.


Subject(s)
Homeostasis/physiology , Prosencephalon/physiology , Receptor, Adenosine A1/metabolism , Receptor, Adenosine A2A/metabolism , Sleep/physiology , Adenosine A1 Receptor Antagonists , Adenosine A2 Receptor Antagonists , Analysis of Variance , Animals , Catheterization , Electrodes, Implanted , Electroencephalography , Homeostasis/drug effects , Male , Prosencephalon/drug effects , Rats , Rats, Wistar , Sleep/drug effects , Sleep Deprivation/drug therapy , Theobromine/analogs & derivatives , Theobromine/pharmacology , Theophylline/analogs & derivatives , Theophylline/pharmacology
17.
Curr Drug Saf ; 4(2): 97-102, 2009 May.
Article in English | MEDLINE | ID: mdl-19442100

ABSTRACT

The effects exerted by P1 and P2 as well as by A1 and A2 agonists and antagonists purinoceptor on the acute opiate withdrawal induced by morphine were investigated in vitro. Following a 4 min in vitro exposure to morphine, the guinea-pig isolated ileum exhibited a strong contracture after the addition of naloxone. The P1 purinoceptor agonist, adenosine, was able dose-dependently to reduce morphine withdrawal whereas alpha,beta-methylene ATP (APCPP), a P2 purinoceptor agonist, increased morphine withdrawal. Caffeine, a P1 purinoceptor antagonist, was able significantly and in a concentration dependent manner to increase morphine withdrawal whereas quinidine, a P2 receptor antagonist, reduced it. Also, the adenosine A1 receptor agonist, N6-Cyclopentyladenosine (CPA) was able to reduce dose-dependently naloxone-precipitated withdrawal whereas the selective adenosine A(2A) receptor agonist CGS 21680 increased the naloxone-precipitated withdrawal phenomenon. Dipyridamole, a blocker of adenosine reuptake, induced a significant reduction of morphine dependence. Caffeine, an adenosine receptor antagonist, significantly increased the naloxone-precipitated withdrawal effect in a concentration dependent manner. The same effect was observed with 8-phenyltheophylline (8PT), an A1 adenosine receptor antagonist, whereas 3,7-dimethyl-1-propargylxanthine (DMPX), an A2 adenosine receptor antagonist, reduced the naloxone-precipitated withdrawal phenomenon. The results of our experiments indicate that P1 and P2 as well as A1 and A2 purinoceptor agonists and antagonists are able to influence opiate withdrawal in vitro, suggesting an important functional interaction between the purinergic system and opioid withdrawal.


Subject(s)
Morphine/pharmacology , Narcotics/pharmacology , Receptor, Adenosine A1/physiology , Receptors, Adenosine A2/physiology , Receptors, Purinergic P2/physiology , Substance Withdrawal Syndrome , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine A1 Receptor Agonists , Adenosine A1 Receptor Antagonists , Adenosine A2 Receptor Agonists , Adenosine A2 Receptor Antagonists , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Analgesics, Opioid/pharmacology , Animals , Caffeine/pharmacology , Dipyridamole/pharmacology , Dose-Response Relationship, Drug , Guinea Pigs , Ileum/drug effects , Ileum/physiology , In Vitro Techniques , Male , Morphine Dependence , Muscle, Smooth/drug effects , Muscle, Smooth/physiopathology , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Phenethylamines/pharmacology , Purinergic P2 Receptor Agonists , Purinergic P2 Receptor Antagonists , Quinidine/pharmacology , Theobromine/analogs & derivatives , Theobromine/pharmacology , Theophylline/analogs & derivatives , Theophylline/pharmacology
18.
Lett Appl Microbiol ; 48(1): 19-24, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19018970

ABSTRACT

AIM: The biotransformation of pentoxifylline (PTX), propentofylline (PPT) and their racemic hydroxy metabolites ((+/-)-OHPTX and (+/-)-OHPPT) by using the fungus Cunninghamella echinulata NRRL 1384. METHODS AND RESULTS: A fungus Cunninghamella echinulata NRRL 1384 was used to catalyse the (S)-selective oxidation of the racemic hydroxy metabolites: (+/-)-OHPTX and (+/-)-OHPPT and for reduction of PTX and PPT. The first oxidation step appears to be selective and relatively fast while the second reduction step is slower and more selective with PTX. Modifications involving supplementing the bioconversion with glucose give yields and enantiomeric excess (ee) values similar to those obtained without glucose. CONCLUSIONS: The bioconversion of (+/-)-OHPTX gave an (R)-enantiomer (LSF-lisofylline) with a higher enantiopurity (maximum approximately 93% ee) compared to the bioconversion of (+/-)-OHPPT, when the maximum ee value for (R)-OHPPT was recorded at 83%. SIGNIFICANCE AND IMPACT OF THE STUDY: The conversion of (+/-)-OHPTX and (+/-)-OHPPT using Cunninghamella echinulata can be recognized as a process, which may be recommended as an alternative to the methods used to obtain (R)-OHPTX and (R)-OHPPT.


Subject(s)
Cunninghamella/metabolism , Theobromine/analogs & derivatives , Theobromine/metabolism , Biotransformation , Glucose/metabolism , Oxidation-Reduction , Pentoxifylline/metabolism , Stereoisomerism , Xanthines/metabolism
19.
Am J Physiol Cell Physiol ; 295(5): C1123-32, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18768924

ABSTRACT

We examined the effects of supernatants of culture media of MDA-MB-231 and MCF-7 cells on the expression of adhesion molecules on human lymphatic endothelial cells (LECs) and evaluated whether the overexpression of adhesion molecules facilitated the attachment of carcinoma cells to LECs. The 48-h stimulation of MDA-MB-231, but not MCF-7, supernatant produced a significant expression of ICAM-1 on human LECs but little or no expression of E-selectin. Chemical treatment with dialyzed substances of <1,000 molecular weight (MW) caused a complete reduction of the supernatant-mediated response. In contrast, pretreatment with heating, digestion with protease, or chemical treatment with dialyzed substances of <500 MW produced no significant effect on the supernatant-mediated response. ATP (10(-7) M) caused overexpression of ICAM-1 on human LECs similar to that produced by the supernatant of MDA-MB-231. The ATP- and MDA-MB-231 supernatant-mediated responses were significantly reduced by treatment with 10(-6) M suramin (a purinergic P2X and P2Y receptor antagonist). In attachment assays, 10(-7) M ATP or MDA-MB-231 supernatant produced a significant increase in the attachment of carcinoma cells to human LECs. The treatment with 10(-6) M suramin caused a significant reduction of ATP- and supernatant-mediated facilitation of the attachment responses. Additional treatment with anti-ICAM-1 antibody also caused a significant reduction of ATP- and supernatant-mediated facilitation of the attachment responses. The experimental findings suggest that MDA-MB-231 may release or leak ATP, which produces the overexpression of ICAM-1 on human LECs through activation of purinergic P2X and/or P2Y receptors and then facilitates ICAM-1-mediated attachment of carcinoma cells to LECs.


Subject(s)
Adenosine Triphosphate/metabolism , Breast Neoplasms/metabolism , Cell Adhesion , Endothelial Cells/metabolism , Endothelium, Lymphatic/metabolism , Intercellular Adhesion Molecule-1/metabolism , Paracrine Communication , Breast Neoplasms/pathology , Cell Adhesion/drug effects , Cell Line, Tumor , Culture Media, Conditioned/metabolism , Endothelial Cells/drug effects , Endothelium, Lymphatic/drug effects , Female , Humans , Lymphatic Metastasis , Paracrine Communication/drug effects , Protein Denaturation , Receptors, Purinergic P2/metabolism , Suramin/pharmacology , Theobromine/analogs & derivatives , Theobromine/pharmacology , Time Factors , Xanthines/pharmacology
20.
Eur J Pharmacol ; 584(1): 125-36, 2008 Apr 14.
Article in English | MEDLINE | ID: mdl-18313046

ABSTRACT

In the present study, we investigated the cardiovascular effects of intravenously injected uridine or cytidine, and the role of adenosine receptors in mediating these effects, in conscious normotensive rats. Intravenous (i.v.) administration of uridine (124, 250, 500 mg/kg) dose-dependently decreased arterial pressure and heart rate. Cytidine (124, 250, 500 mg/kg; i.v.) produced slight dose-related hypotension without changing heart rate. Plasma uridine and cytidine concentrations increased time- and dose-dependently while plasma adenosine levels did not change after injection of the respective nucleosides. Pretreatment with intravenous caffeine (20 mg/kg), 8-phenyltheophylline (8-PT) (1 mg/kg), nonselective adenosine receptor antagonists, or 8-p-sulfophenyltheophylline (8-SPT) (20 mg/kg), a nonselective adenosine receptor antagonist which does not cross the blood-brain barrier, abolished the cardiovascular effects of uridine (250 mg/kg; i.v.) or cytidine (250 mg/kg; i.v.). Intracerebroventricular (i.c.v.) caffeine (200 microg) or 8-SPT (50 microg) pretreatment did not change the magnitude of the cardiovascular responses induced by nucleosides. Intravenous 8-cyclopenthyl-1,3-dipropylxanthine (DPCPX) (5 mg/kg), a selective adenosine A(1) receptor antagonist, greatly attenuated the cardiovascular responses to uridine and cytidine. Pretreatment with 3,7,-dimethyl-1-propargylxanthine (DMPX) (2 mg/kg), an adenosine A(1)/A(2) receptor antagonist, attenuated hypotension induced by uridine and blocked the arterial pressure decrease in response to cytidine. Uridine-induced bradycardia was blocked by DMPX. 4-(2-[7-amino-2-(2-furyl[1,2,4]-triazolo[2,3-a[1,3,5]triazin-5-yl-aminoethyl)phenol (ZM241385) (1 mg/kg; i.v.), a selective adenosine A(2A) receptor antagonist, pretreatment produced an only very small blockade in the first minute of the hypotensive effects of uridine without affecting the bradycardia. ZM241385 pretreatment completely blocked cytidine's hypotensive effect. In Langendorff-perfused rat heart preparation, uridine (10(-3) M), but not cytidine, decreased the heart rate. Our results show that intravenously injected uridine or cytidine is able to decrease arterial pressure by activating peripheral adenosine receptors. The data also implicates that the mainly adenosine A(1) receptor activation is involved in the uridine-induced cardiovascular effects, while both adenosine A(1) and A(2A) receptor activations mediate the cytidine's effects.


Subject(s)
Adenosine A1 Receptor Agonists , Adenosine A2 Receptor Agonists , Antihypertensive Agents/administration & dosage , Blood Pressure/drug effects , Carotid Arteries/drug effects , Cytidine/administration & dosage , Hypotension/chemically induced , Uridine/administration & dosage , Adenosine/blood , Animals , Antihypertensive Agents/adverse effects , Antihypertensive Agents/blood , Caffeine/administration & dosage , Carotid Arteries/metabolism , Carotid Arteries/physiopathology , Consciousness , Cytidine/adverse effects , Cytidine/blood , Dose-Response Relationship, Drug , Heart Rate/drug effects , Hypotension/metabolism , Hypotension/physiopathology , Injections, Intravenous , Injections, Intraventricular , Male , Rats , Rats, Wistar , Receptor, Adenosine A1/metabolism , Receptor, Adenosine A2A/metabolism , Theobromine/administration & dosage , Theobromine/analogs & derivatives , Theophylline/administration & dosage , Theophylline/analogs & derivatives , Time Factors , Triazines/administration & dosage , Triazoles/administration & dosage , Uridine/adverse effects , Uridine/blood , Ventricular Function, Left/drug effects , Ventricular Pressure/drug effects , Xanthines/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...