Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Int Immunopharmacol ; 97: 107680, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33932698

ABSTRACT

Peripheral tramadol's delivery in the temporomandibular joint (TMJ) leads to significant analgesic outcomes and inflammatory process's resolvent actions. Mechanistically, these properties are apart from the opioid system. Nevertheless, the molecular mechanisms behind these effects are still unclear. Therefore, the present study investigated the hypothesis that adenosine A1 receptors are involved in the tramadol-induced analgesic and anti-inflammatory effects in the TMJ. Animals were pretreated with an intra-TMJ injection of DPCPX (antagonist of A1 receptor) or tramadol and subsequent nociceptive challenge with an intra-TMJ injection of 1.5% formalin. For over 45 min, the nociceptive behavior was quantitated, and by the end of this assessment, the animals were euthanized, and the periarticular tissue was collected. Lastly, an in vitro assay of BMDM (Bone Marrow-Derived Macrophages) was performed to investigate tramadol activity in macrophages. The intra-TMJ injection of tramadol ameliorates formalin-induced hypernociception along with inhibiting leukocyte migration. The tramadol's peripheral anti-inflammatory effect was mediated by the adenosine A1 receptor and was associated with increased protein expression of α2a-adrenoceptor in the periarticular tissues (p < 0.05: ANOVA, Tukey's test). Also, tramadol inhibits formalin-induced leukocyte migration and protein expression of P2X7 receptors in the periarticular tissue (p < 0.05); however, DPCPX did not alter this effect (p > 0.05). Moreover, DPCPX significantly reduced the protein expression of the M2 macrophage marker, MRC1. In BMDM, tramadol significantly reduces inflammatory cytokines release, and DPCPX abrogated this effect (p < 0.05). We identify tramadol's peripheral effect is mediated by adenosine A1 receptor, possibly expressed in macrophages in the TMJ tissue. We also determined an important discovery related to the activation of A1R/α2a receptors in the tramadol action.


Subject(s)
Adenosine A1 Receptor Agonists/administration & dosage , Arthralgia/drug therapy , Receptor, Adenosine A1/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Tramadol/administration & dosage , Analgesics, Opioid/administration & dosage , Animals , Anti-Inflammatory Agents/administration & dosage , Arthralgia/chemically induced , Arthralgia/immunology , Arthralgia/pathology , Disease Models, Animal , Formaldehyde/administration & dosage , Formaldehyde/toxicity , Humans , Injections, Intra-Articular , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Male , Nociception/drug effects , Rats , Temporomandibular Joint/drug effects , Temporomandibular Joint/immunology , Temporomandibular Joint/pathology , Xanthines/administration & dosage , Xanthines/toxicity
2.
Toxicol Appl Pharmacol ; 419: 115515, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33798593

ABSTRACT

Exposure to organophosphorus nerve agents (NAs) like sarin (GB) and soman (GD) can lead to sustained seizure activity, or status epilepticus (SE). Previous research has shown that activation of A1 adenosine receptors (A1ARs) can inhibit neuronal excitability, which could aid in SE termination. Two A1AR agonists, 2-Chloro-N6-cyclopentyladenosine (CCPA) and N-Bicyclo(2.2.1)hept-2-yl-5'-chloro-5'-deoxyadenosine (ENBA), were effective in terminating GD-induced SE in rats when administered via intraperitoneal (IP) injection. However, IP injection is not a clinically relevant route of administration. This study evaluated the efficacy of these agonists in terminating NA-induced SE when administered via intramuscular (IM) route. Adult male rats were exposed subcutaneously (SC) to either GB (150 µg/kg) or GD (90 µg/kg) and were treated with ENBA or CCPA at 15, 30, or 60 min after seizure onset or left untreated. Up to 7 days after exposure, deeply anesthetized rats were euthanized and perfused brains were removed for histologic assessment of neuropathology (i.e., neuronal damage) in six brain regions (amygdala, cerebral cortex, piriform cortex, thalamus, dorsal hippocampus, and ventral hippocampus). A total neuropathy score (0-24) was determined for each rat by adding the scores from each of the six regions. The higher the total score the more severe the neuropathology. With the GB model and 60 min treatment delay, ENBA-treated rats experienced 78.6% seizure termination (N = 14) and reduced neuropathology (11.6 ± 2.6, N = 5), CCPA-treated rats experienced 85.7% seizure termination (N = 14) and slightly reduced neuropathology (20.7 ± 1.8, N = 6), and untreated rats experienced no seizure termination (N = 13) and severe neuropathology (22.3 ± 1.0, N = 4). With the GD model and 60 min treatment delay, ENBA-treated rats experienced 92.9% seizure termination (N = 14) and reduced neuropathology (13.96 ± 1.8, N = 9), CCPA-treated rats experienced 78.6% seizure termination (N = 14) and slightly reduced neuropathology (22.0 ± 0.9, N = 10); and untreated rats experienced 16.7% seizure termination (N = 12) and severe neuropathology (22.0 ± 1.8, N = 5). While ENBA and CCPA both demonstrate a clear ability to terminate SE when administered up to 60 min after seizure onset, ENBA offers more neuroprotection, making it a promising candidate for NA-induced SE.


Subject(s)
Adenosine A1 Receptor Agonists/administration & dosage , Adenosine/analogs & derivatives , Anticonvulsants/administration & dosage , Brain/drug effects , Deoxyadenosines/administration & dosage , Neuroprotective Agents/administration & dosage , Norbornanes/administration & dosage , Sarin , Soman , Status Epilepticus/prevention & control , Adenosine/administration & dosage , Animals , Brain/metabolism , Brain/pathology , Brain/physiopathology , Disease Models, Animal , Drug Administration Schedule , Injections, Intramuscular , Male , Rats, Sprague-Dawley , Status Epilepticus/chemically induced , Status Epilepticus/metabolism , Status Epilepticus/pathology , Time Factors
3.
Epilepsy Behav ; 106: 107034, 2020 05.
Article in English | MEDLINE | ID: mdl-32208337

ABSTRACT

PURPOSES: This study was to further explore the adenosine dysfunction in refractory epilepsy in Sturge-Weber Syndrome (SWS), to evaluate the neuronal-level effect of the A1 receptor (A1R) agonist on both excitatory pyramidal neurons and inhibitory interneurons, to discuss the possibility of adenosine augmentation therapy (AAT) using A1R agonist for treating refractory epilepsy in SWS. MATERIALS AND METHODS: The intrinsic excitatory properties of pyramidal cells (PCs) and fast-spiking (FS) interneurons from human brain tissues with SWS cases and malformations of cortical development (MCD) cases were compared using electrophysiology. With application of either A1R agonist or antagonist, the neuronal-level effect of A1R agonist was evaluated in vitro in PCs and FS interneurons from SWS cases and MCD cases. RESULTS: No significant difference of passive excitatory properties of PCs and FS interneurons was found between SWS cases and MCD cases. In terms of the neuronal-level effect of A1R agonist, with 22.88 ±â€¯1.12% percentage of decreased frequency, FS interneurons showed relatively highest sensitivity of A1R agonist application, compared with PCs from SWS cases and FS interneurons and PCs from MCD cases. CONCLUSION: Our results supported the potential of AATs using A1R agonist to be a novel therapy for reducing life burden from patients with refractory epilepsy in SWS, with application to epileptic generation region but not propagation region.


Subject(s)
Adenosine A1 Receptor Agonists/administration & dosage , Adenosine/administration & dosage , Drug Resistant Epilepsy/drug therapy , Electroencephalography , Interneurons/drug effects , Sturge-Weber Syndrome/drug therapy , Adenosine A1 Receptor Antagonists/administration & dosage , Adolescent , Adult , Animals , Child , Child, Preschool , Drug Resistant Epilepsy/physiopathology , Electroencephalography/methods , Electrophysiological Phenomena/physiology , Female , Humans , Infant , Interneurons/physiology , Male , Neurons/drug effects , Neurons/physiology , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , Sturge-Weber Syndrome/physiopathology , Young Adult
4.
Neurol Res ; 42(3): 189-208, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32013788

ABSTRACT

Objective: Trans-resveratrol has been shown to have neuroprotective effects and could be a promising therapeutic agent in the treatment of intracerebral haemorrhage (ICH). This study aimed to investigate the involvement of the adenosine A1 receptor (A1R) in trans-resveratrol-induced neuroprotection in rats with collagenase-induced ICH.Methods: Sixty male Sprague-Dawley rats weighing 330-380 g were randomly divided into five groups (n = 12): (i) control, (ii) sham-operated rats, (iii) ICH rats pretreated with vehicle (0.1% DMSO saline, i.c.v.), (iv) ICH rats pretreated with trans-resveratrol (0.9 µg, i.c.v.) and (v) ICH rats pretreated with trans-resveratrol (0.9 µg) and the A1R antagonist, DPCPX (2.5 µg, i.c.v.). Thirty minutes after pretreatment, ICH was induced by intrastriatal injection of collagenase (0.04 U). Forty-eight hours after ICH, the rats were assessed using a variety of neurobehavioural tests. Subsequently, rats were sacrificed and brains were subjected to gross morphological examination of the haematoma area and histological examination of the damaged area.Results: Severe neurobehavioural deficits and haematoma with diffuse oedema were observed after intrastriatal collagenase injection. Pretreatment with trans-resveratrol partially restored general locomotor activity, muscle strength and coordination, which was accompanied with reduction of haematoma volume by 73.22% (P < 0.05) and damaged area by 60.77% (P < 0.05) in comparison to the vehicle-pretreated ICH group. The trans-resveratrol-induced improvement in neurobehavioural outcomes and morphological features of brain tissues was inhibited by DPCPX pretreatment.Conclusion: This study demonstrates that the A1R activation is possibly the mechanism underlying the trans-resveratrol-induced neurological and neurobehavioural protection in rats with ICH.


Subject(s)
Adenosine A1 Receptor Agonists/administration & dosage , Behavior, Animal/drug effects , Brain/drug effects , Cerebral Hemorrhage/pathology , Cerebral Hemorrhage/psychology , Collagenases/administration & dosage , Neuroprotective Agents/administration & dosage , Resveratrol/administration & dosage , Animals , Brain/pathology , Cerebral Hemorrhage/chemically induced , Disease Models, Animal , Male , Rats, Sprague-Dawley
5.
Pharmacol Biochem Behav ; 181: 110-116, 2019 06.
Article in English | MEDLINE | ID: mdl-31054946

ABSTRACT

Repetitive behaviors are diagnostic for autism spectrum disorder (ASD) and commonly observed in other neurodevelopmental disorders. Currently, there are no effective pharmacological treatments for repetitive behavior in these clinical conditions. This is due to the lack of information about the specific neural circuitry that mediates the development and expression of repetitive behavior. Our previous work in mouse models has linked repetitive behavior to decreased activation of the subthalamic nucleus, a brain region in the indirect and hyperdirect pathways in the basal ganglia circuitry. The present experiments were designed to further test our hypothesis that pharmacological activation of the indirect pathway would reduce repetitive behavior. We used a combination of adenosine A1 and A2A receptor agonists that have been shown to alter the firing frequency of dorsal striatal neurons within the indirect pathway of the basal ganglia. This drug combination markedly and selectively reduced repetitive behavior in both male and female C58 mice over a six-hour period, an effect that required both A1 and A2A agonists as neither alone reduced repetitive behavior. The adenosine A1 and A2A receptor agonist combination also significantly increased the number of Fos transcripts and Fos positive cells in dorsal striatum. Fos induction was found in both direct and indirect pathway neurons suggesting that the drug combination restored the balance of activation across these complementary basal ganglia pathways. The adenosine A1 and A2A receptor agonist combination also maintained its effectiveness in reducing repetitive behavior over a 7-day period. These findings point to novel potential therapeutic targets for development of drug therapies for repetitive behavior in clinical disorders.


Subject(s)
Adenosine A1 Receptor Agonists/therapeutic use , Adenosine A2 Receptor Agonists/therapeutic use , Adenosine/analogs & derivatives , Compulsive Behavior/drug therapy , Phenethylamines/therapeutic use , Stereotyped Behavior/drug effects , Adenosine/administration & dosage , Adenosine/chemistry , Adenosine/therapeutic use , Adenosine A1 Receptor Agonists/administration & dosage , Adenosine A1 Receptor Agonists/chemistry , Adenosine A2 Receptor Agonists/administration & dosage , Adenosine A2 Receptor Agonists/chemistry , Analysis of Variance , Animals , Autism Spectrum Disorder/drug therapy , Autism Spectrum Disorder/metabolism , Behavior, Animal/drug effects , Corpus Striatum/cytology , Drug Therapy, Combination , Female , Male , Mice , Mice, Inbred C57BL , Models, Animal , Neurons/metabolism , Peanut Oil/chemistry , Peanut Oil/pharmacology , Phenethylamines/administration & dosage , Phenethylamines/chemistry , Phenotype , Proto-Oncogene Proteins c-fos/metabolism
6.
Biochem Pharmacol ; 164: 45-52, 2019 06.
Article in English | MEDLINE | ID: mdl-30905656

ABSTRACT

Elevated circulating free fatty acid (FFA) level is closely linked to the pathogenesis of insulin resistance and type 2 diabetes mellitus. Activation of the adenosine A1 receptor (A1R) inhibits lipolysis in adipocytes and hence reduces the concentration of FFA, which represents a potential target for the development of antilipolytic agents. We aimed to assess the binding affinity as well as target binding kinetics of A1R agonists and further delineate a possible relationship with their antilipolytic effect in adipocytes. Radioligand binding assays were performed to determine the affinity and kinetics of three representative A1R agonists, namely CPA, LUF6944 and LUF6941, on the rat A1R. Functional responses to these agonists were examined in both a recombinant cell system and physiologically relevant rat adipocytes. The three A1R agonists displayed similar affinity while divergent target binding kinetics on the rat A1R. Irrespective of equilibrium binding affinity, temporal analysis of receptor signaling demonstrated persistent functional responses for the long residence time agonist, despite removal of excess agonist, in both a recombinant cell system and in rat adipocytes. By contrast, such effect was less pronounced or even lost for agonists with medium or short receptor residence time, respectively. Our results indicate that ligand receptor binding kinetics rather than their affinity or potency play an essential role in regulating cellular responses. The long residence time A1R agonist produces a sustained wash-resistant antilipolytic effect in rat adipocytes and thus may represent a potential antilipolytic alternative for further investigation.


Subject(s)
Adenosine A1 Receptor Agonists/administration & dosage , Adenosine A1 Receptor Agonists/metabolism , Adipocytes/drug effects , Adipocytes/metabolism , Lipolysis/drug effects , Animals , CHO Cells , Cricetinae , Cricetulus , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/metabolism , Dose-Response Relationship, Drug , Lipolysis/physiology , Rats
7.
Eur J Heart Fail ; 20(11): 1601-1610, 2018 11.
Article in English | MEDLINE | ID: mdl-30225882

ABSTRACT

Despite major advances in the treatment of chronic heart failure (HF) with reduced ejection fraction (HFrEF), morbidity and mortality associated with the condition remain high, suggesting the need for additional treatment options, particularly haemodynamically neutral treatments that do not alter blood pressure, heart rate, or renal function. HF with preserved ejection fraction (HFpEF) is also associated with high morbidity and mortality and adequate treatment options are limited; thus there is a critical unmet need for the development of novel therapies for HFpEF. Chronic HFrEF and HFpEF are both systemic disorders that affect not only the heart but several other tissues and organs including skeletal muscle, leading to exercise intolerance and dyspnoea. Partial adenosine A1-receptor agonists represent a novel potential therapy for HF regardless of underlying ejection fraction given their minimal effect on heart rate and blood pressure, and preclinical data demonstrate several possible beneficial mechanisms, including improved mitochondrial function and sarcoplasmic reticulum Ca2+ -ATPase (SERCA2a) activity, enhanced energy substrate utilization, reverse ventricular remodelling, and anti-ischemic, cardioprotective properties. However, data on this class of drugs in humans are scarce, and the optimal dose of the partial adenosine A1 receptor, neladenoson bialanate, has not been defined. Here we describe the design and rationale of two randomized, double-blind, placebo-controlled, parallel-group, dose-finding phase 2b trials, PANTHEON (HFrEF) and PANACHE (HFpEF), that will advance our understanding of the potential benefit and optimal dose of neladenoson bialanate and provide critical information for the planning of future phase 3 trials.


Subject(s)
Adenosine A1 Receptor Agonists/administration & dosage , Dipeptides/administration & dosage , Heart Failure/drug therapy , Pyridines/administration & dosage , Stroke Volume/physiology , Ventricular Function, Left/physiology , Adolescent , Adult , Aged , Dose-Response Relationship, Drug , Double-Blind Method , Female , Follow-Up Studies , Heart Failure/physiopathology , Heart Rate/drug effects , Heart Rate/physiology , Humans , Male , Middle Aged , Stroke Volume/drug effects , Treatment Outcome , Young Adult
8.
J Biol Rhythms ; 33(5): 523-534, 2018 10.
Article in English | MEDLINE | ID: mdl-30033847

ABSTRACT

Caffeine is widely used to reduce sedation and increase alertness. However, long-term caffeine use may disrupt circadian (daily, 24-h) rhythms and thereby negatively affect health. Here, we examined the effect of caffeine on photic regulation of circadian activity rhythms in mice. We found that entrainment to a standard 12-h light, 12-h dark (LD) photocycle was delayed during oral self-administration of caffeine. Both acute, high-dose caffeine and chronic, oral caffeine exposure potentiated photic phase-delays in mice, suggesting a possible mechanism by which entrainment to LD was delayed. The effect of caffeine on photic phase-resetting was mimicked by administration of adenosine A1, but not A2A, receptor antagonist in mice. Our results support the hypothesis that caffeine interferes with the ability of the circadian clock to respond normally to light.


Subject(s)
Caffeine/administration & dosage , Circadian Rhythm/drug effects , Light , Photoperiod , Adenosine A1 Receptor Agonists/administration & dosage , Adenosine A2 Receptor Agonists/administration & dosage , Animals , Caffeine/adverse effects , Circadian Clocks/drug effects , Circadian Clocks/radiation effects , Male , Mice , Mice, Inbred C57BL , Motor Activity
9.
PLoS One ; 13(6): e0198838, 2018.
Article in English | MEDLINE | ID: mdl-29912966

ABSTRACT

Light induced retinal degeneration (LIRD) is a useful model that resembles human retinal degenerative diseases. The modulation of adenosine A1 receptor is neuroprotective in different models of retinal injury. The aim of this work was to evaluate the potential neuroprotective effect of the modulation of A1 receptor in LIRD. The eyes of rats intravitreally injected with N6-cyclopentyladenosine (CPA), an A1 agonist, which were later subjected to continuous illumination (CI) for 24 h, showed retinas with a lower number of apoptotic nuclei and a decrease of Glial Fibrillary Acidic Protein (GFAP) immunoreactive area than controls. Lower levels of activated Caspase 3 and GFAP were demonstrated by Western Blot (WB) in treated animals. Also a decrease of iNOS, TNFα and GFAP mRNA was demonstrated by RT-PCR. A decrease of Iba 1+/MHC-II+ reactive microglial cells was shown by immunohistochemistry. Electroretinograms (ERG) showed higher amplitudes of a-wave, b-wave and oscillatory potentials after CI compared to controls. Conversely, the eyes of rats intravitreally injected with dipropylcyclopentylxanthine (DPCPX), an A1 antagonist, and subjected to CI for 24 h, showed retinas with a higher number of apoptotic nuclei and an increase of GFAP immunoreactive area compared to controls. Also, higher levels of activated Caspase 3 and GFAP were demonstrated by Western Blot. The mRNA levels of iNOS, nNOS and inflammatory cytokines (IL-1ß and TNFα) were not modified by DPCPX treatment. An increase of Iba 1+/MHC-II+ reactive microglial cells was shown by immunohistochemistry. ERG showed that the amplitudes of a-wave, b-wave, and oscillatory potentials after CI were similar to control values. A single pharmacological intervention prior illumination stress was able to swing retinal fate in opposite directions: CPA was neuroprotective, while DPCPX worsened retinal damage. In summary, A1 receptor agonism is a plausible neuroprotective strategy in LIRD.


Subject(s)
Adenosine A1 Receptor Agonists/therapeutic use , Adenosine/analogs & derivatives , Receptor, Adenosine A1/drug effects , Retinal Degeneration/drug therapy , Adenosine/administration & dosage , Adenosine/therapeutic use , Adenosine A1 Receptor Agonists/administration & dosage , Animals , Blotting, Western , Caspase 3/metabolism , Disease Models, Animal , Electroretinography , Glial Fibrillary Acidic Protein/metabolism , Intravitreal Injections , Male , Nitric Oxide Synthase Type II/metabolism , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Receptor, Adenosine A1/physiology , Retina/drug effects , Retina/radiation effects , Tumor Necrosis Factor-alpha/metabolism
10.
Science ; 356(6345): 1352-1356, 2017 06 30.
Article in English | MEDLINE | ID: mdl-28663494

ABSTRACT

Circuits in the auditory cortex are highly susceptible to acoustic influences during an early postnatal critical period. The auditory cortex selectively expands neural representations of enriched acoustic stimuli, a process important for human language acquisition. Adults lack this plasticity. Here we show in the murine auditory cortex that juvenile plasticity can be reestablished in adulthood if acoustic stimuli are paired with disruption of ecto-5'-nucleotidase-dependent adenosine production or A1-adenosine receptor signaling in the auditory thalamus. This plasticity occurs at the level of cortical maps and individual neurons in the auditory cortex of awake adult mice and is associated with long-term improvement of tone-discrimination abilities. We conclude that, in adult mice, disrupting adenosine signaling in the thalamus rejuvenates plasticity in the auditory cortex and improves auditory perception.


Subject(s)
Adenosine/metabolism , Auditory Cortex/metabolism , Signal Transduction , 5'-Nucleotidase/metabolism , Adenosine/administration & dosage , Adenosine/analogs & derivatives , Adenosine A1 Receptor Agonists/administration & dosage , Adenosine A1 Receptor Antagonists/administration & dosage , Animals , Auditory Perception , GPI-Linked Proteins/metabolism , Mice , Neuronal Plasticity , Piperidines/administration & dosage , Pyridazines/administration & dosage , Receptor, Adenosine A1/metabolism , Thalamus/metabolism
11.
ChemMedChem ; 12(10): 728-737, 2017 05 22.
Article in English | MEDLINE | ID: mdl-28488817

ABSTRACT

Adenosine is known to be released under a variety of physiological and pathophysiological conditions to facilitate the protection and regeneration of injured ischemic tissues. The activation of myocardial adenosine A1 receptors (A1 Rs) has been shown to inhibit myocardial pathologies associated with ischemia and reperfusion injury, suggesting several options for new cardiovascular therapies. When full A1 R agonists are used, the desired protective and regenerative cardiovascular effects are usually overshadowed by unintended pharmacological effects such as induction of bradycardia, atrioventricular (AV) blocks, and sedation. These unwanted effects can be overcome by using partial A1 R agonists. Starting from previously reported capadenoson we evaluated options to tailor A1 R agonists to a specific partiality range, thereby optimizing the therapeutic window. This led to the identification of the potent and selective agonist neladenoson, which shows the desired partial response on the A1 R, resulting in cardioprotection without sedative effects or cardiac AV blocks. To circumvent solubility and formulation issues for neladenoson, a prodrug approach was pursued. The dipeptide ester neladenoson bialanate hydrochloride showed significantly improved solubility and exposure after oral administration. Neladenoson bialanate hydrochloride is currently being evaluated in clinical trials for the treatment of heart failure.


Subject(s)
Adenosine A1 Receptor Agonists/pharmacology , Dipeptides/pharmacology , Heart Diseases/drug therapy , Prodrugs/pharmacology , Pyridines/pharmacology , Receptor, Adenosine A1/metabolism , Adenosine A1 Receptor Agonists/administration & dosage , Adenosine A1 Receptor Agonists/chemistry , Administration, Oral , Animals , Chronic Disease , Dipeptides/administration & dosage , Dipeptides/chemistry , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Injections, Intravenous , Molecular Structure , Prodrugs/administration & dosage , Prodrugs/chemistry , Pyridines/administration & dosage , Pyridines/chemistry , Rats , Solubility , Structure-Activity Relationship
12.
Neuropharmacology ; 114: 101-113, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27914963

ABSTRACT

Small mammals have the ability to enter torpor, a hypothermic, hypometabolic state, allowing impressive energy conservation. Administration of adenosine or adenosine 5'-monophosphate (AMP) can trigger a hypothermic, torpor-like state. We investigated the mechanisms for hypothermia using telemetric monitoring of body temperature in wild type and receptor knock out (Adora1-/-, Adora3-/-) mice. Confirming prior data, stimulation of the A3 adenosine receptor (AR) induced hypothermia via peripheral mast cell degranulation, histamine release, and activation of central histamine H1 receptors. In contrast, A1AR agonists and AMP both acted centrally to cause hypothermia. Commonly used, selective A1AR agonists, including N6-cyclopentyladenosine (CPA), N6-cyclohexyladenosine (CHA), and MRS5474, caused hypothermia via both A1AR and A3AR when given intraperitoneally. Intracerebroventricular dosing, low peripheral doses of Cl-ENBA [(±)-5'-chloro-5'-deoxy-N6-endo-norbornyladenosine], or using Adora3-/- mice allowed selective stimulation of A1AR. AMP-stimulated hypothermia can occur independently of A1AR, A3AR, and mast cells. A1AR and A3AR agonists and AMP cause regulated hypothermia that was characterized by a drop in total energy expenditure, physical inactivity, and preference for cooler environmental temperatures, indicating a reduced body temperature set point. Neither A1AR nor A3AR was required for fasting-induced torpor. A1AR and A3AR agonists and AMP trigger regulated hypothermia via three distinct mechanisms.


Subject(s)
Adenosine A1 Receptor Agonists/administration & dosage , Adenosine A3 Receptor Agonists/administration & dosage , Adenosine Monophosphate/physiology , Fever/chemically induced , Receptor, Adenosine A1/physiology , Receptor, Adenosine A3/physiology , Torpor , Adenosine/administration & dosage , Adenosine/analogs & derivatives , Animals , Brain/drug effects , Brain/physiology , Histamine/metabolism , Injections, Intraventricular , Male , Mast Cells/drug effects , Mast Cells/metabolism , Mice , Mice, Inbred C57BL , Receptor, Adenosine A1/genetics , Receptor, Adenosine A3/genetics , Receptors, Histamine H1/physiology
13.
J Clin Pharmacol ; 57(4): 440-451, 2017 04.
Article in English | MEDLINE | ID: mdl-27624622

ABSTRACT

We studied safety and tolerability of neladenoson bialanate, a novel oral selective partial adenosine A1 receptor agonist that maintains the cardioprotective effects of adenosine without the undesired side effects of a full agonist, in 2 pilot studies in patients with heart failure with reduced ejection fraction (HFrEF). The ß-blocker interaction study was a single-blind, placebo-controlled study on the effects of a 30-mg single dose of neladenoson bialanate on atrioventricular (AV) conduction in 11 patients with HFrEF treated with ß-blockers. The PARSiFAL pilot study was a double-blind, placebo-controlled study on the effects of a 7-day treatment with 10 or 20 mg neladenoson bialanate or placebo in 31 patients with HFrEF on ß-blocker therapy. In the ß-blocker interaction study with 11 HFrEF patients, no second- or third-degree AV block was detected on 48-hour Holter monitoring. In the 31 HFrEF patients included in the PARSiFAL pilot study, no second- or third-degree AV blocks were observed during 24-hour Holter monitoring, and no effects were seen on heart rate and blood pressure. Median absolute changes in LVEF, measured by cardiac magnetic resonance, were 1.9% (interquartile range -1.1 to 4.3), 0.3% (-1.4 to 2.7), and 2.2% (0.4 to 4.5), in the placebo, 10-mg, and 20-mg groups, respectively. Treatment of HFrEF patients with the novel partial adenosine A1 agonist neladenoson bialanate appeared to be safe in 2 small pilot studies, and no atrioventricular conduction disorders or neurological side effects were observed. No significant early changes in cardiac function were detected.


Subject(s)
Adenosine A1 Receptor Agonists/administration & dosage , Drug Partial Agonism , Heart Failure/diagnosis , Heart Failure/drug therapy , Administration, Oral , Aged , Blood Pressure/drug effects , Blood Pressure/physiology , Chronic Disease , Double-Blind Method , Electrocardiography/drug effects , Electrocardiography/methods , Female , Follow-Up Studies , Heart Conduction System/drug effects , Heart Conduction System/physiology , Heart Failure/physiopathology , Heart Rate/drug effects , Heart Rate/physiology , Humans , Male , Middle Aged , Pilot Projects , Single-Blind Method
14.
Neuropharmacology ; 111: 283-292, 2016 12.
Article in English | MEDLINE | ID: mdl-27639989

ABSTRACT

Activation of A1 adenosine receptors (ARs) has been associated with anxiolytic-like effects in different behavioral tests, but development of A1AR agonists for therapeutic use has been hampered, most likely due to the presence of side effects. With the aim to identify a safer approach for the treatment of anxiety, we investigated, in mice, the anxiolytic-like properties of a novel A1AR positive allosteric modulator, TRR469. Acute administration of TRR469 (0.3-3 mg/kg) resulted in robust anxiolytic-like effects in the elevated plus maze, the dark/light box, the open field and the marble burying tests. The magnitude of the anxiolytic action of TRR469 was comparable to that obtained with benzodiazepine diazepam (1 mg/kg). The use of the A1AR antagonist DPCPX (3 mg/kg) suggested that the effects of TRR469 were mediated by this receptor subtype. In contrast to diazepam, the novel positive allosteric modulator did not potentiate the sedative effect of ethanol (3.5 g/kg) evaluated by the loss of righting reflex. While diazepam produced motor coordination impairment in the rotarod test, this effect being enhanced by the presence of ethanol (1.5 g/kg), TRR469 did not elicit locomotor disturbances either when administered alone or in the presence of ethanol. In vitro, TRR469 was able to increase the number of A1AR recognizable by the agonist radioligand [3H]-CCPA in mouse brain regions involved in emotional processes. TRR469 markedly increased the affinity of the agonist CCPA, suggesting the capability, in vivo, to increase the affinity of endogenous adenosine. Taken together, these findings indicate that the positive allosteric modulation of A1AR may represent a promising approach for the treatment of anxiety-related disorders.


Subject(s)
Adenosine A1 Receptor Agonists/administration & dosage , Anti-Anxiety Agents/administration & dosage , Anxiety Disorders/drug therapy , Piperazines/administration & dosage , Receptor, Adenosine A1/metabolism , Thiophenes/administration & dosage , Adenosine/administration & dosage , Adenosine/analogs & derivatives , Adenosine A1 Receptor Antagonists/administration & dosage , Allosteric Regulation , Amygdala/drug effects , Amygdala/metabolism , Animals , Diazepam/administration & dosage , Ethanol/administration & dosage , Exploratory Behavior/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice , Motor Activity/drug effects , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Reflex, Righting/drug effects , Rotarod Performance Test , Xanthines/administration & dosage
15.
J Neurosci ; 36(14): 3962-77, 2016 Apr 06.
Article in English | MEDLINE | ID: mdl-27053204

ABSTRACT

Cisplatin is a commonly used antineoplastic agent that produces ototoxicity that is mediated in part by increasing levels of reactive oxygen species (ROS) via the NOX3 NADPH oxidase pathway in the cochlea. Recent studies implicate ROS generation in mediating inflammatory and apoptotic processes and hearing loss by activating signal transducer and activator of transcription (STAT1). In this study, we show that the adenosine A1 receptor (A1AR) protects against cisplatin ototoxicity by suppressing an inflammatory response initiated by ROS generation via NOX3 NADPH oxidase, leading to inhibition of STAT1. Trans-tympanic administration of the A1AR agonist R-phenylisopropyladenosine (R-PIA) inhibited cisplatin-induced ototoxicity, as measured by auditory brainstem responses and scanning electron microscopy in male Wistar rats. This was associated with reduced NOX3 expression, STAT1 activation, tumor necrosis factor-α (TNF-α) levels, and apoptosis in the cochlea. In vitro studies in UB/OC-1 cells, an organ of Corti immortalized cell line, showed that R-PIA reduced cisplatin-induced phosphorylation of STAT1 Ser(727) (but not Tyr(701)) and STAT1 luciferase activity by suppressing the ERK1/2, p38, and JNK mitogen-activated protein kinase (MAPK) pathways.R-PIA also decreased the expression of STAT1 target genes, such as TNF-α, inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) and reduced cisplatin-mediated apoptosis. These data suggest that the A1AR provides otoprotection by suppressing NOX3 and inflammation in the cochlea and could serve as an ideal target for otoprotective drug therapy. SIGNIFICANCE STATEMENT: Cisplatin is a widely used chemotherapeutic agent for the treatment of solid tumors. Its use results in significant and permanent hearing loss, for which no US Food and Drug Administration-approved treatment is currently available. In this study, we targeted the cochlear adenosine A1 receptor (A1AR) by trans-tympanic injections of the agonist R-phenylisopropyladenosine (R-PIA) and showed that it reduced cisplatin-induced inflammation and apoptosis in the rat cochlea and preserved hearing. The mechanism of protection involves suppression of the NOX3 NADPH oxidase enzyme, a major target of cisplatin-induced reactive oxygen species (ROS) generation in the cochlea. ROS initiates an inflammatory and apoptotic cascade in the cochlea by activating STAT1 transcription factor, which is attenuated byR-PIA. Therefore, trans-tympanic delivery of A1AR agonists could effectively treat cisplatin ototoxicity.


Subject(s)
Antineoplastic Agents/toxicity , Cisplatin/toxicity , Cochlea/drug effects , Inflammation/physiopathology , NADPH Oxidases/drug effects , NADPH Oxidases/genetics , Receptor, Adenosine A1/drug effects , STAT1 Transcription Factor/drug effects , STAT1 Transcription Factor/genetics , Adenosine A1 Receptor Agonists/administration & dosage , Adenosine A1 Receptor Agonists/pharmacology , Adenosine A1 Receptor Antagonists/administration & dosage , Adenosine A1 Receptor Antagonists/pharmacology , Animals , Cell Line , Evoked Potentials, Auditory, Brain Stem/drug effects , Hair Cells, Auditory/drug effects , Hearing Disorders/chemically induced , Hearing Disorders/physiopathology , MAP Kinase Signaling System/drug effects , Male , Rats , Rats, Wistar , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism
17.
Neuroscience ; 285: 312-23, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25451280

ABSTRACT

Diabetic peripheral neuropathy is a common complication of diabetes mellitus, and a significant proportion of individuals suffer debilitating pain that significantly affects their quality of life. Unfortunately, symptomatic treatment options have limited efficacy, and often carry significant risk of systemic adverse effects. Activation of the adenosine A1 receptor (A1R) by the analgesic small molecule adenosine has been shown to have antinociceptive benefits in models of inflammatory and neuropathic pain. The current study used a mouse model of painful diabetic neuropathy to determine the effect of diabetes on endogenous adenosine production, and if central or peripheral delivery of adenosine receptor agonists could alleviate signs of mechanical allodynia in diabetic mice. Diabetes was induced using streptozocin in male A/J mice. Mechanical withdrawal thresholds were measured weekly to characterize neuropathy phenotype. Hydrolysis of AMP into adenosine by ectonucleotidases was determined in the dorsal root ganglia (DRG) and spinal cord at 8 weeks post-induction of diabetes. AMP, adenosine and the specific A1R agonist, N(6)-cyclopentyladenosine (CPA), were administered both centrally (intrathecal) and peripherally (intraplantar) to determine the effect of activation of adenosine receptors on mechanical allodynia in diabetic mice. Eight weeks post-induction, diabetic mice displayed significantly decreased hydrolysis of extracellular AMP in the DRG; at this same time, diabetic mice displayed significantly decreased mechanical withdrawal thresholds compared to nondiabetic controls. Central delivery AMP, adenosine and CPA significantly improved mechanical withdrawal thresholds in diabetic mice. Surprisingly, peripheral delivery of CPA also improved mechanical allodynia in diabetic mice. This study provides new evidence that diabetes significantly affects endogenous AMP hydrolysis, suggesting that altered adenosine production could contribute to the development of painful diabetic neuropathy. Moreover, central and peripheral activation of A1R significantly improved mechanical sensitivity, warranting further investigation into this important antinociceptive pathway as a novel therapeutic option for the treatment of painful diabetic neuropathy.


Subject(s)
Adenosine A1 Receptor Agonists/administration & dosage , Analgesics, Non-Narcotic/administration & dosage , Diabetes Mellitus, Experimental/physiopathology , Diabetic Neuropathies/drug therapy , Hyperalgesia/drug therapy , Adenosine/administration & dosage , Adenosine/analogs & derivatives , Adenosine/metabolism , Adenosine Monophosphate/administration & dosage , Adenosine Monophosphate/metabolism , Animals , Diabetes Mellitus, Experimental/pathology , Diabetic Neuropathies/pathology , Diabetic Neuropathies/physiopathology , Foot , Ganglia, Spinal/drug effects , Ganglia, Spinal/pathology , Ganglia, Spinal/physiopathology , Hindlimb , Hydrolysis/drug effects , Hyperalgesia/pathology , Hyperalgesia/physiopathology , Injections, Spinal , Male , Mice , Neurons/drug effects , Neurons/pathology , Neurons/physiology , Receptor, Adenosine A1/metabolism , Spinal Cord/drug effects , Spinal Cord/physiopathology , Touch
18.
J Clin Pharmacol ; 53(4): 385-92, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23427000

ABSTRACT

GS-9667, a new selective, partial agonist of the A(1) adenosine receptor (AR), may represent an effective therapy for Type 2 diabetes (T2DM) and dyslipidemia via lowering of free fatty acids (FFA). The objectives of the studies were to evaluate the effects of single and multiple doses of GS-9667 on plasma FFA concentrations, its pharmacokinetics (PK) and safety/tolerability. Two studies were conducted. In the single ascending dose study, healthy, non-obese, and obese subjects received a single oral dose of GS-9667 (30-1,800 mg). In the multiple, ascending dose study, healthy, obese subjects received GS-9667 (600-2,400 mg QD, 1,200 mg BID, or 600 mg QID) for 14 days. Blood and urine samples were collected for lipid profiling and PK analyses. The ECG, vital signs, and subject tolerability were monitored. Doses of GS-9667 ≥300 mg caused dose-dependent reductions in FFA levels that were reproducible over 14 days without evidence of desensitization or rebound. All doses were well tolerated. GS-9667 was rapidly absorbed and distributed; Steady-state concentrations were achieved within 3-5 days. The A(1) AR partial agonist GS-9667 reduced plasma FFA, exhibited linear kinetics, and was well-tolerated in healthy non-obese and obese subjects.


Subject(s)
Adenosine A1 Receptor Agonists/administration & dosage , Adenosine/analogs & derivatives , Adenosine/administration & dosage , Adenosine/blood , Adenosine/pharmacokinetics , Adenosine/urine , Adenosine A1 Receptor Agonists/blood , Adenosine A1 Receptor Agonists/pharmacokinetics , Adenosine A1 Receptor Agonists/urine , Adolescent , Adult , Fatty Acids, Nonesterified/blood , Female , Humans , Male , Middle Aged , Obesity/blood , Obesity/urine , Young Adult
19.
Molecules ; 17(12): 13712-26, 2012 Nov 22.
Article in English | MEDLINE | ID: mdl-23174891

ABSTRACT

This study was undertaken in order to investigate the effect of chronic treatment with 5′-chloro-5′-deoxy-(±)-ENBA, a potent and highly selective agonist of human adenosine A(1) receptor, on thermal hyperalgesia and mechanical allodynia in a mouse model of neuropathic pain, the Spared Nerve Injury (SNI) of the sciatic nerve. Chronic systemic administration of 5′-chloro-5′-deoxy-(±)-ENBA (0.5 mg/kg, i.p.) reduced both mechanical allodynia and thermal hyperalgesia 3 and 7 days post-SNI, in a way prevented by DPCPX (3 mg/kg, i.p.), a selective A(1) adenosine receptor antagonist, without exerting any significant change on the motor coordination or arterial blood pressure. In addition, a single intraperitoneal injection of 5′-chloro-5′-deoxy-(±)-ENBA (0.5 mg/kg, i.p.) 7 days post-SNI also reduced both symptoms for at least two hours. SNI was associated with spinal changes in microglial activation ipsilaterally to the nerve injury. Activated, hypertrophic microglia were significantly reduced by 5′-chloro-5′-deoxy-(±)-ENBA chronic treatment. Our results demonstrated an involvement of adenosine A(1) receptor in the amplified nociceptive thresholds and in spinal glial and microglial changes occurred in neuropathic pain, without affecting motor coordination or blood pressure. Our data suggest a possible use of adenosine A(1) receptor agonist in neuropathic pain symptoms.


Subject(s)
Adenosine A1 Receptor Agonists/administration & dosage , Adenosine/analogs & derivatives , Neuralgia/drug therapy , Norbornanes/administration & dosage , Receptor, Adenosine A1/metabolism , Adenosine/administration & dosage , Animals , Cardiovascular System/drug effects , Humans , Hyperalgesia/drug therapy , Hyperalgesia/pathology , Mice , Microglia/metabolism , Microglia/pathology , Motor Activity/drug effects , Neuroglia/metabolism , Neuroglia/pathology , Sciatic Nerve/drug effects , Sciatic Nerve/injuries , Xanthines/administration & dosage
20.
Zhongguo Zhong Xi Yi Jie He Za Zhi ; 32(3): 390-3, 2012 Mar.
Article in Chinese | MEDLINE | ID: mdl-22686090

ABSTRACT

OBJECTIVE: To investigate the effects of injecting adenosine A1 receptor agonist (CCPA) into Baihui (GV20) on the cerebral cortex induced by the ischemia/reperfusion of middle cerebral artery occlusion (MCAO) in rats. METHODS: Twenty-four SD rats were randomly divided into four groups, i. e., the sham-operation group, the model group, the DMSO group, and the CCPA group. The MCAO model was established by thread embolism method. At the moment of ischemia/reperfusion, the rats in DMSO group and the CCPA group were injected with DMSO (20 microL) and CCPA (0.1 mmol) 20 microL into Baihui respectively. The rats' behavior, the histomorphology of ischemic penumbra in the cerebral cortex, the expressions of Bcl-2 protein, and the apoptosis rate of neurocytes were assessed. RESULTS: Compared with the model group and the DMSO group, the rats' behavior were markedly improved in the CCPA group (P<0.05). No obvious karyopyknosis and cytoplasm empty dye of neurons appeared. The Bcl-2 expressions in rats' cerebral cortex obviously increased (P<0.01). The apoptosis number of neurons obviously decreased (P<0.01). CONCLUSIONS: Injecting CCPA into Bahui improved the rats' behavior and histomorphology in the ischemic penumbra, elevated the expressions of Bcl-2 protein, and reduced the neurons apoptosis rate in the ischemic penumbra. It alleviated the cerebral ischemia-reperfusion injury. Therefore, it could be taken as a new treatment method.


Subject(s)
Adenosine A1 Receptor Agonists/pharmacology , Brain Ischemia/therapy , Cerebral Cortex/drug effects , Reperfusion Injury/therapy , Acupuncture Points , Adenosine A1 Receptor Agonists/administration & dosage , Adenosine A1 Receptor Agonists/therapeutic use , Animals , Brain Ischemia/metabolism , Cerebral Cortex/metabolism , Male , Rats , Rats, Sprague-Dawley , Reperfusion Injury/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...