Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
2.
Acta Neuropathol ; 147(1): 55, 2024 03 12.
Article in English | MEDLINE | ID: mdl-38472475

ABSTRACT

Inclusions comprised of microtubule-associated protein tau (tau) are implicated in a group of neurodegenerative diseases, collectively known as tauopathies, that include Alzheimer's disease (AD). The spreading of misfolded tau "seeds" along neuronal networks is thought to play a crucial role in the progression of tau pathology. Consequently, restricting the release or uptake of tau seeds may inhibit the spread of tau pathology and potentially halt the advancement of the disease. Previous studies have demonstrated that the Mammalian Suppressor of Tauopathy 2 (MSUT2), an RNA binding protein, modulates tau pathogenesis in a transgenic mouse model. In this study, we investigated the impact of MSUT2 on tau pathogenesis using tau seeding models. Our findings indicate that the loss of MSUT2 mitigates human tau seed-induced pathology in neuron cultures and mouse models. In addition, MSUT2 regulates many gene transcripts, including the Adenosine Receptor 1 (A1AR), and we show that down regulation or inhibition of A1AR modulates the activity of the "ArfGAP with SH3 Domain, Ankyrin Repeat, and PH Domain 1 protein" (ASAP1), thereby influencing the internalization of pathogenic tau seeds into neurons resulting in reduction of tau pathology.


Subject(s)
Alzheimer Disease , Tauopathies , Mice , Humans , Animals , Brain/pathology , tau Proteins/metabolism , Tauopathies/pathology , Alzheimer Disease/pathology , Neurons/pathology , Mice, Transgenic , Mammals/metabolism , Adaptor Proteins, Signal Transducing/metabolism
3.
Pharmaceuticals (Basel) ; 16(7)2023 Jun 22.
Article in English | MEDLINE | ID: mdl-37513829

ABSTRACT

Adenosine receptors (ARs) are being explored to generate non-opioid pain therapeutics. Vanilloid compounds, curcumin, capsaicin, and vanillin possess antinociceptive properties through their interactions with the transient receptor potential channel family. However, their binding with adenosine receptors has not been well studied. The hypothesis in this study was that a vanilloid compound, cis-trans curcumin (CTCUR), binds to each of the two Gi-linked AR subtypes (A1AR and A3AR). CTCUR was synthesized from curcumin (CUR) using the cavitand-mediated photoisomerization technique. The cell lines transfected with the specific receptor (A1AR or A3AR) were treated with CTCUR or CUR and the binding was analyzed using competitive assays, confocal microscopy, and docking. The binding assays and molecular docking indicated that CTCUR had Ki values of 306 nM (A1AR) and 400 nM (A3AR). These values suggest that CTCUR is selective for Gi-linked ARs (A1AR or A3AR) over Gs-linked ARs (A2AAR or A2BAR), based on our previous published research. In addition, the docking showed that CTCUR binds to the toggle switch domain of ARs. Curcumin (CUR) did not exhibit binding at any of these receptors. In summary, CTCUR and other modifications of CUR can be developed as novel therapeutic ligands for the Gi-linked ARs (A1AR and A3AR) involved with pain and cancer.

4.
Reprod Sci ; 29(2): 564-577, 2022 02.
Article in English | MEDLINE | ID: mdl-34582004

ABSTRACT

Heat shock protein alpha 8 (HSPA8) was found to be downregulated in the placentas of patients with hypertensive disorders in pregnancy (HDP). We aim to explore the underlying role and mechanism of HSPA8 in HDP progression. Herein, HSPA8 mRNA expression in placentas and peripheral blood of patients with HDP and normal pregnant controls was measured with RT-qPCR. We found that HSPA8 expression was downregulated in placentas and peripheral blood of patients with HDP. HTR8/SVneo human trophoblast cells were transfected with pcDNA-HSPA8 or si-HSPA8. HSPA8 overexpression promoted cell proliferation, migration, and MMP-2 and MMP-9 protein levels, and inhibited apoptosis, while HSPA8 silencing showed the opposite results. Co-immunoprecipitation assay validated the binding between HSPA8 and ß-arrestin1, as well as ß-arrestin1 and A1AR proteins. HSPA8 bound with ß-arrestin1 protein and promoted ß-arrestin1 expression. ß-arrestin1 bound with A1AR protein and inhibited A1AR expression. Then, HTR8/SVneo cells were transfected with pcDNA-HSPA8 alone or together with si-ß-arrestin1, as well as transfected with pcDNA-ß-arrestin1 alone or together with pcDNA-A1AR. ß-arrestin1 silencing reversed the effects of HSPA8 overexpression on HTR8/SVneo cell functions. ß-arrestin1 overexpression promoted cell proliferation migration, and MMP-2 and MMP-9 protein levels, and inhibited apoptosis, while these effects were reversed by A1AR overexpression. Lentivirus HSPA8 overexpression vector (Lv-HSPA8) was injected into a preeclampsia (PE) rat model, which attenuated blood pressure and fetal detrimental changes in PE rats. In conclusion, HSPA8 promoted proliferation and migration and inhibited apoptosis in trophoblast cells, and attenuated the symptoms of PE rats by modulating the ß-arrestin1/A1AR axis. Our study provided a novel theoretical evidence and potential strategy for HDP treatment.


Subject(s)
HSC70 Heat-Shock Proteins/physiology , Hypertension, Pregnancy-Induced/physiopathology , beta-Arrestin 1/metabolism , Adult , Animals , Apoptosis , Blotting, Western , Case-Control Studies , Female , HSC70 Heat-Shock Proteins/blood , HSC70 Heat-Shock Proteins/metabolism , Humans , Hypertension, Pregnancy-Induced/blood , Hypertension, Pregnancy-Induced/metabolism , Immunoprecipitation , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Placenta/metabolism , Pregnancy , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , beta-Arrestin 1/blood
5.
Neuroimage ; 245: 118695, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34732326

ABSTRACT

The neuromodulator adenosine and its receptors are mediators of sleep-wake regulation which is known to differ between sexes. We, therefore, investigated sex differences in A1 adenosine receptor (A1AR) availability in healthy human subjects under well-rested conditions using [18F]CPFPX and positron emission tomography (PET). [18F]CPFPX PET scans were acquired in 50 healthy human participants (20 females; mean age ± SD 28.0 ± 5.3 years). Mean binding potential (BPND; Logan's reference tissue model with cerebellum as reference region) and volume of distribution (VT) values were calculated in 12 and 15 grey matter brain regions, respectively. [18F]CPFPX BPND was higher in females compared to males in all investigated brain regions (p < 0.025). The largest differences were found in the pallidum and anterior cingulate cortex, where mean BPND values were higher by 29% in females than in males. In females, sleep efficiency correlated positively and sleep latency negatively with BPND in most brain regions. VT values did not differ between sexes. Sleep efficiency correlated positively with VT in most brain regions in female participants. In conclusion, our analysis gives a first indication for potential sex differences in A1AR availability even under well-rested conditions. A1AR availability as measured by [18F]CPFPX BPND is higher in females compared to males. Considering the involvement of adenosine in sleep-wake control, this finding might partially explain the known sex differences in sleep efficiency and sleep latency.


Subject(s)
Brain Mapping/methods , Positron-Emission Tomography , Receptor, Adenosine A1/metabolism , Sleep , Adult , Female , Fluorodeoxyglucose F18 , Healthy Volunteers , Humans , Male , Radiopharmaceuticals , Sex Factors
7.
Front Oncol ; 11: 636373, 2021.
Article in English | MEDLINE | ID: mdl-33777791

ABSTRACT

Personized treatment of breast cancer is still a challenge, and more treatment options for breast cancer are warranted. Combination therapies have been a highly appreciated strategy for breast cancer treatment in recent years, and the development of new combination therapies could improve patient outcomes. Adenosine and polyamines are both endogenous metabolites with indispensable biological functions. Adenosine binds with the A1 adenosine receptor (A1AR) to downregulate cAMP concentration, and both low cAMP content and high polyamine levels stimulate the growth and proliferation of cancer cells. In this work, we initially used a polyamine synthesis inhibitor, DFMO (α-difluoromethylornithine), and an A1AR inhibitor, DPCPX (8-cyclopentyl-1,3-dipropylxanthine) to investigate if simultaneously inhibiting A1AR and polyamine synthesis has synergistical antitumor effects. Next, we investigated a dual inhibitor (ODC-MPI-2) of A1AR and ODC (ornithine decarboxylase 1), the rate-limiting enzyme in polyamine biosynthesis. We investigated if ODC-MPI-2 could inhibit the proliferation and growth of breast cancer cells. Our data showed that DFMO and DPCPX synergistically inhibit the growth and proliferation of MCF-7 cells. We also demonstrated that ODC-MPI-2 reduces cellular polyamine levels and elevates cAMP concentration. We further showed that ODC-MPI-2 inhibits the growth, proliferation, and migration/invasion of MCF-7 cells. Finally, ODC-MPI-2 showed a preference for inhibiting triple-negative breast cancer cells. The dual inhibition of ODC and A1AR is a new combination therapy strategy for treating breast cancer, and dual inhibitors of ODC and A1AR may be effective future drugs for treating breast cancer.

8.
Molecules ; 26(4)2021 Feb 23.
Article in English | MEDLINE | ID: mdl-33672225

ABSTRACT

Alzheimer's, Parkinson's, and multiple sclerosis are neurodegenerative diseases related by neuronal degeneration and death in specific areas of the central nervous system. These pathologies are associated with neuroinflammation, which is involved in disease progression, and halting this process represents a potential therapeutic strategy. Evidence suggests that microglia function is regulated by A1 and A2A adenosine receptors (AR), which are considered as neuroprotective and neurodegenerative receptors, respectively. The manuscript's aim is to elucidate the role of these receptors in neuroinflammation modulation through potent and selective A1AR agonists (N6-cyclopentyl-2'- or 3'-deoxyadenosine substituted or unsubstituted in 2 position) and A2AAR antagonists (9-ethyl-adenine substituted in 8 and/or in 2 position), synthesized in house, using N13 microglial cells. In addition, the combined therapy of A1AR agonists and A2AAR antagonists to modulate neuroinflammation was evaluated. Results showed that A1AR agonists were able, to varying degrees, to prevent the inflammatory effect induced by cytokine cocktail (tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, and interferon (IFN)-γ), while A2AAR antagonists showed a good ability to counteract neuroinflammation. Moreover, the effect achieved by combining the two most effective compounds (1 and 6) in doses previously found to be non-effective was greater than the treatment effect of each of the two compounds used separately at maximal dose.


Subject(s)
Adenosine A1 Receptor Agonists/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Inflammation/drug therapy , Receptor, Adenosine A1/metabolism , Receptors, Adenosine A2/metabolism , Animals , Cells, Cultured , Inflammation/metabolism , Mice
9.
Med Chem Res ; 30(2): 353-370, 2021.
Article in English | MEDLINE | ID: mdl-33519168

ABSTRACT

The ocular drug discovery arena has undergone a significant improvement in the last few years culminating in the FDA approvals of 8 new drugs. However, despite a large number of drugs, generics, and combination products available, it remains an urgent need to find breakthrough strategies and therapies for tackling ocular diseases. Targeting the adenosinergic system may represent an innovative strategy for discovering new ocular therapeutics. This review focused on the recent advance in the field and described the numerous nucleoside and non-nucleoside modulators of the four adenosine receptors (ARs) used as potential tools or clinical drug candidates.

10.
Cells ; 9(7)2020 07 21.
Article in English | MEDLINE | ID: mdl-32708189

ABSTRACT

The pathological condition of neuroinflammation is caused by the activation of the neuroimmune cells astrocytes and microglia. The autacoid adenosine seems to be an important neuromodulator in this condition. Its main receptors involved in the neuroinflammation modulation are A1AR and A2AAR. Evidence suggests that A1AR activation produces a neuroprotective effect and A2AARs block prevents neuroinflammation. The aim of this work is to elucidate the effects of these receptors in neuroinflammation using the partial agonist 2'-dCCPA (2-chloro-N6-cyclopentyl-2'-deoxyadenosine) (C1 KiA1AR = 550 nM, KiA2AAR = 24,800 nM, and KiA3AR = 5560 nM, α = 0.70, EC50A1AR = 832 nM) and the newly synthesized in house compound 8-chloro-9-ethyl-2-phenethoxyadenine (C2 KiA2AAR = 0.75 nM; KiA1AR = 17 nM and KiA3AR = 227 nM, IC50A2AAR = 251 nM unpublished results). The experiments were performed in in vitro and in in vivo models of neuroinflammation. Results showed that C1 was able to prevent the inflammatory effect induced by cytokine cocktail (TNF-α, IL-1ß, and IFN-γ) while C2 possess both anti-inflammatory and antioxidant properties, counteracting both neuroinflammation in mixed glial cells and in an animal model of neuroinflammation. In conclusion, C2 is a potential candidate for neuroinflammation therapy.


Subject(s)
Adenosine A1 Receptor Agonists/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Inflammation/pathology , Neurons/pathology , Receptors, Purinergic P1/metabolism , Adenosine/analogs & derivatives , Animals , Cells, Cultured , Male , Neuroglia/metabolism , Neurons/drug effects , Neurons/metabolism , Rats, Wistar , Triazines/pharmacology , Triazoles/pharmacology
11.
Biomolecules ; 10(5)2020 05 07.
Article in English | MEDLINE | ID: mdl-32392873

ABSTRACT

Adenosine receptors (ARs), like many otherGprotein-coupledreceptors (GPCRs), are targets of primary interest indrug design. However, one of the main limits for the development of drugs for this class of GPCRs is the complex selectivity profile usually displayed by ligands. Numerous efforts have been madefor clarifying the selectivity of ARs, leading to the development of many ligand-based models. The structure of the AR subtype A1 (A1AR) has been recently solved,providing important structural insights. In the present work, we rationalized the selectivity profile of two selective A1AR and A2AAR antagonists, investigating their recognition trajectories obtained by Supervised Molecular Dynamics from an unbound state and monitoring the role of the water molecules in the binding site.


Subject(s)
Adenosine A1 Receptor Antagonists/chemistry , Molecular Dynamics Simulation , Receptor, Adenosine A1/chemistry , Adenosine A1 Receptor Antagonists/pharmacology , Binding Sites , Humans , Molecular Docking Simulation/methods , Protein Binding , Receptor, Adenosine A1/metabolism , Supervised Machine Learning
12.
J Comput Aided Mol Des ; 34(6): 697-707, 2020 06.
Article in English | MEDLINE | ID: mdl-32112287

ABSTRACT

Among still comparatively few G protein-coupled receptors, the adenosine A2A receptor has been co-crystallized with several ligands, agonists as well as antagonists. It can thus serve as a template with a well-described orthosteric ligand binding region for adenosine receptors. As not all subtypes have been crystallized yet, and in order to investigate the usability of homology models in this context, multiple adenosine A1 receptor (A1AR) homology models had been previously obtained and a library of lead-like compounds had been docked. As a result, a number of potent and one selective ligand toward the intended target have been identified. However, in in vitro experimental verification studies, many ligands also bound to the A2AAR and the A3AR subtypes. In this work we asked the question whether a classification of the ligands according to their selectivity was possible based on docking scores. Therefore, we built an A3AR homology model and docked all previously found ligands to all three receptor subtypes. As a metric, we employed an in vitro/in silico selectivity ranking system based on taxicab geometry and obtained a classification model with reasonable separation. In the next step, the method was validated with an external library of, selective ligands with similarly good performance. This classification system might also be useful in further screens.


Subject(s)
Protein Conformation , Receptor, Adenosine A1/chemistry , Receptor, Adenosine A2A/chemistry , Receptor, Adenosine A3/chemistry , Adenosine A1 Receptor Agonists/chemistry , Adenosine A1 Receptor Antagonists/chemistry , Binding Sites/drug effects , Humans , Ligands , Models, Molecular , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Protein Binding/drug effects , Protein Conformation/drug effects , Receptor, Adenosine A1/ultrastructure , Receptor, Adenosine A2A/ultrastructure , Receptor, Adenosine A3/ultrastructure , Structure-Activity Relationship
13.
Curr Pharm Des ; 25(25): 2697-2715, 2019.
Article in English | MEDLINE | ID: mdl-31333094

ABSTRACT

Adenosine is a purine nucleoside, responsible for the regulation of a wide range of physiological and pathophysiological conditions by binding with four G-protein-coupled receptors (GPCRs), namely A1, A2A, A2B and A3 adenosine receptors (ARs). In particular, A1 AR is ubiquitously present, mediating a variety of physiological processes throughout the body, thus represents a promising drug target for the management of various pathological conditions. Agonists of A1 AR are found to be useful for the treatment of atrial arrhythmia, angina, type-2 diabetes, glaucoma, neuropathic pain, epilepsy, depression and Huntington's disease, whereas antagonists are being investigated for the treatment of diuresis, congestive heart failure, asthma, COPD, anxiety and dementia. However, treatment with full A1 AR agonists has been associated with numerous challenges like cardiovascular side effects, off-target activation as well as desensitization of A1 AR leading to tachyphylaxis. In this regard, partial agonists of A1 AR have been found to be beneficial in enhancing insulin sensitivity and subsequently reducing blood glucose level, while avoiding severe CVS side effects and tachyphylaxis. Allosteric enhancer of A1 AR is found to be potent for the treatment of neuropathic pain, culminating the side effects related to off-target tissue activation of A1 AR. This review provides an overview of the medicinal chemistry and therapeutic potential of various agonists/partial agonists, antagonists and allosteric modulators of A1 AR, with a particular emphasis on their current status and future perspectives in clinical settings.


Subject(s)
Chemistry, Pharmaceutical , Purinergic P1 Receptor Agonists/pharmacology , Purinergic P1 Receptor Antagonists/pharmacology , Receptors, Purinergic P1/metabolism , Allosteric Regulation , Humans
14.
Eur J Pharmacol ; 842: 111-117, 2019 Jan 05.
Article in English | MEDLINE | ID: mdl-30347181

ABSTRACT

L-NAME-induced hypertension is commonly used to study endothelial dysfunction and related vascular effects. It has been reported that genetic deletion of A1 adenosine receptor (AR) reduces blood pressure (BP) increases in mice and thus, suggesting the involvement of A1AR. Thus, we sought to determine whether A1AR-induced vascular responses were altered in this mouse model of hypertension. L-NAME (1 mg/ml) was given in the drinking water for 28 days to mice. The BP was monitored using non-invasive tail-cuff system. Muscle tension studies were performed using DMT for mesenteric arteries (MAs) and organ bath for aorta. Protein expression was analyzed by western blot. Significantly, higher systolic and mean arterial blood pressure was noted in L-NAME mice. In MAs, higher 2-Chloro-N6-cyclopentyladenosine (CCPA, selective A1AR agonist) induced contractions in hypertensive mice were observed. This enhanced contraction was inhibited by HET0016 (Cytochrome 450 4A inhibitor, 10 µM, 15 min). Contrary, 5'-(N-Ethylcarboxamido) adenosine (NECA, non-selective AR agonist) induced vascular responses were comparable in both groups. Pinacidil (KATP channel opener) induced relaxation was significantly increased in hypertensive mice. In aorta, CCPA-induced contractions were enhanced and inhibited by HET0016 in hypertensive mice. Notably, NECA-induced contractions in aorta were enhanced in hypertensive mice. Higher expressions of A1AR and Cyp4A were noted in MAs of hypertensive mice. In addition, in aorta, higher A1AR and comparable Cyp4A levels were observed in hypertensive mice. A1AR-induced vascular contractions were enhanced in hypertensive mice aorta and MAs. Cyp4A plays a role in altered vascular responses in MAs.


Subject(s)
Aorta/physiopathology , Hypertension/physiopathology , Mesenteric Arteries/physiopathology , NG-Nitroarginine Methyl Ester/pharmacology , Receptor, Adenosine A1/metabolism , Vasoconstriction/drug effects , Vasodilation/drug effects , Animals , Aorta/drug effects , Blood Pressure/drug effects , Cytochrome P-450 CYP4A/metabolism , Disease Models, Animal , Gene Expression Regulation, Enzymologic/drug effects , Male , Mesenteric Arteries/drug effects , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type III/metabolism
15.
Ther Hypothermia Temp Manag ; 8(2): 108-116, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29480748

ABSTRACT

Targeted temperature management is standard of care for cardiac arrest and is in clinical trials for stroke. N6-cyclohexyladenosine (CHA), an A1 adenosine receptor (A1AR) agonist, inhibits thermogenesis and induces onset of hibernation in hibernating species. Despite promising thermolytic efficacy of CHA, prior work has failed to achieve and maintain a prescribed target core body temperature (Tb) between 32°C and 34°C for 24 hours. We instrumented Sprague-Dawley rats (n = 19) with indwelling arterial and venous cannulae and a transmitter for monitoring Tb and ECG, then administered CHA via continuous IV infusion or intraperitoneal (IP) injection. In the first experiment (n = 11), we modulated ambient temperature and increased the dose of CHA in an attempt to manage Tb. In the second experiment (n = 8), we administered CHA (0.25 mg/[kg·h]) via continuous IV infusion and modulated cage surface temperature to control Tb. We rewarmed animals by increasing surface temperature at 1°C h-1 and discontinued CHA after Tb reached 36.5°C. Tb, brain temperature (Tbrain), heart rate, blood gas, and electrolytes were also monitored. Results show that titrating dose to adjust for individual variation in response to CHA led to tolerance and failed to manage a prescribed Tb. Starting with a dose (0.25 mg/[kg·h]) and modulating surface temperature to prevent overcooling proved to be an effective means to achieve and maintain Tb between 32°C and 34°C for 24 hours. Increasing surface temperature to 37°C during CHA administration brought Tb back to normothermic levels. All animals treated in this way rewarmed without incident. During the initiation of cooling, we observed bradycardia within 30 minutes of the start of IV infusion, transient hyperglycemia, and a mild hypercapnia; the latter normalized via metabolic compensation. In conclusion, we describe an intravenous delivery protocol for CHA at 0.25 mg/(kg·h) that, when coupled with conductive cooling, achieves and maintains a prescribed and consistent target Tb between 32°C and 34°C for 24 hours.


Subject(s)
Adenosine/analogs & derivatives , Hypothermia, Induced/methods , Adenosine/administration & dosage , Animals , Body Temperature , Drug Evaluation, Preclinical , Electrocardiography , Female , Hyperglycemia/blood , Hyperglycemia/etiology , Hypothermia, Induced/adverse effects , Male , Rats, Sprague-Dawley , Telemetry
16.
Adv Exp Med Biol ; 1051: 193-232, 2017.
Article in English | MEDLINE | ID: mdl-28676923

ABSTRACT

Adenosine, the purine nucleoside, mediates its effects through activation of four G-protein coupled adenosine receptors (ARs) named as A1, A2A, A2B and A3. In particular, A1ARs are distributed through the body, primarily inhibitory in the regulation of adenylyl cyclase activity and able to reduce the cyclic AMP levels. Considerable advances have been made in the pharmacological and molecular characterization of A1ARs, which had been proposed as targets for the discovery and drug design of antagonists, agonists and allosteric enhancers. Several lines of evidence indicate that adenosine interacting with A1ARs may be an endogenous protective agent in the human body since it prevents the damage caused by various pathological conditions, such as in ischemia/hypoxia, epileptic seizures, excitotoxic neuronal injury and cardiac arrhythmias in cardiovascular system. It has also been reported that one of the most promising targets for the development of new anxiolytic drugs could be A1ARs, and that their activation may reduce pain signaling in the spinal cord. A1AR antagonists induce diuresis and natriuresis in various experimental models, mediating the inhibition of A1ARs in the proximal tubule which is primarily responsible for reabsorption and fluid uptake. In addition, the results of various studies indicate that adenosine is present within pancreatic islets and is implicated through A1ARs in the regulation of insulin secretion and in glucose concentrations. In the present paper it will become apparent that A1ARs could be implicated in the pharmacological treatment of several pathologies with an important influence on human health.


Subject(s)
Adenosine A1 Receptor Agonists/therapeutic use , Adenosine A1 Receptor Antagonists/therapeutic use , Arrhythmias, Cardiac , Drug Discovery , Ischemia , Pain , Receptor, Adenosine A1/metabolism , Seizures , Animals , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/metabolism , Humans , Ischemia/drug therapy , Ischemia/metabolism , Pain/drug therapy , Pain/metabolism , Seizures/drug therapy , Seizures/metabolism
17.
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
18.
Toxicology ; 370: 106-115, 2016 Aug 31.
Article in English | MEDLINE | ID: mdl-27702592

ABSTRACT

Alpha-naphthylisothiocyanate (ANIT) is widely used to induce cholestasis in basic researches. Although direct damage induced by ANIT to bile duct epithelial cells has been documented in previous studies, few works investigated ANIT-induced effects on hepatocytes. Our previous study indicated that activated AMP-activated protein kinase (AMPK) inhibited farnesoid X receptor (FXR) expression and further participated in the pathogenesis of estrogen-induced cholestasis. However, whether ANIT has effects on bile acid homeostasis in hepatocytes, and the role of AMPK-FXR pathway played in these effects remain unclear. In this study, our results showed that ANIT induced intracellular bile acid accumulation without obvious cellular toxicity in sandwich cultured rat primary hepatocytes (SCRHs), accompanied with significant decreased expression of FXR and bile acid transporters. AMPK activation via ERK1/2-LKB1 pathway was critical for ANIT-induced effects on hepatocytes. Compound C, specific AMPK inhibitor, blocked ANIT-regulated gene expression, decreased bile acid accumulation and recovered bile canalicular structure both in vitro and in vivo. Furthermore, the expression of A1 adenosine receptor (A1AR), a potential cholestatic target, was relatively low in hepatocytes compared with expression in rat whole livers. Consistent with these findings, DPCPX, a classic antagonist of A1AR, had no effect on ANIT-induced hepatocytes injury. In summary, our results indicate that AMPK-FXR signaling is critical for ANIT-induced toxic effects on hepatocytes, provide new insights into the pathogenesis of ANIT-induced cholestasis, and suggest AMPK-FXR pathway as a potential therapeutic target for cholestasis.


Subject(s)
1-Naphthylisothiocyanate/toxicity , AMP-Activated Protein Kinases/antagonists & inhibitors , Bile Acids and Salts/metabolism , Homeostasis/drug effects , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Adenosine A1 Receptor Antagonists/pharmacology , Animals , Bile Ducts/cytology , Bile Ducts/drug effects , Bile Ducts/metabolism , Cell Survival/drug effects , Cholestasis/chemically induced , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Hepatocytes/drug effects , Liver/cytology , Liver/drug effects , Liver/metabolism , Male , Phosphorylation , Rats , Rats, Sprague-Dawley , Receptor, Adenosine A1/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Xanthines/pharmacology
19.
Mol Cell Biochem ; 422(1-2): 197-206, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27629787

ABSTRACT

Soluble epoxide hydrolase (sEH) converts epoxyeicosatrienoic acids that are endothelium-derived hyperpolarizing factors into less active dihydroxyeicosatrienoic acids. Previously, we reported a decrease in adenosine A1 receptor (A1AR) protein levels in sEH knockout (sEH-/-) and an increase in sEH and A1AR protein levels in A2AAR-/- mice. Additionally, KATP channels are involved in adenosine receptor (AR)-dependent vascular relaxation. Thus, we hypothesize that a potential relationship may exist among sEH over-expression, A1AR upregulation, inactivation of KATP channels, and increased in vascular tone. We performed DMT myograph muscle tension measurements and western blot analysis in isolated mouse mesenteric arteries (MAs) from wild-type (WT) and endothelial over-expression of sEH (Tie2-sEH Tr) mice. Our data revealed that NECA (a non-selective adenosine receptors agonist)-induced relaxation was significantly reduced in Tie2-sEH Tr mice, and CCPA (A1AR agonist)-induced contraction was increased in Tie2-sEH Tr mice. A1AR-dependent contraction in Tie2-sEH Tr mice was significantly attenuated by pharmacological inhibition of CYP4A (HET0016, 10 µM), PKCα (GO6976, 1 µM), and ERK1/2 (PD58059, 1 µM). Our western blot analysis revealed significantly higher basal protein expression of CYP4A, A1AR, and reduced p-ERK in MAs of Tie2-sEH Tr mice. Notably, pinacidil (KATP channel opener)-induced relaxation was also significantly reduced in MAs of Tie2-sEH Tr mice. Furthermore, KATP channel-dependent relaxation in MAs was enhanced by inhibition of PKCα and ERK1/2 in WT but not Tie2-sEH Tr mice. In conclusion, our data suggest that over-expression of sEH enhances A1AR-dependent contraction and reduces KATP channel-dependent relaxation in MAs. These results suggest a possible interaction between sEH, A1AR, and KATP channels in regulating vascular tone.


Subject(s)
Endothelial Cells/metabolism , Epoxide Hydrolases/biosynthesis , KATP Channels/metabolism , Mesenteric Arteries/enzymology , Receptor, Adenosine A1/metabolism , Vasoconstriction , Adenosine A1 Receptor Agonists/pharmacology , Animals , Cytochrome P-450 CYP4A/antagonists & inhibitors , Cytochrome P-450 CYP4A/genetics , Cytochrome P-450 CYP4A/metabolism , Enzyme Inhibitors/pharmacology , Epoxide Hydrolases/genetics , KATP Channels/genetics , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase C-alpha/genetics , Protein Kinase C-alpha/metabolism , Receptor, Adenosine A1/genetics , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism
20.
Chem Biol Drug Des ; 88(5): 724-729, 2016 11.
Article in English | MEDLINE | ID: mdl-27282729

ABSTRACT

Three 4-amino-6-alkyloxy-2-alkylthiopyrimidine derivatives (4-6) were investigated as potential non-nucleoside agonists at human adenosine receptors (ARs). When tested in competition binding experiments, these compounds exhibited low micromolar affinity (Ki values comprised between 1.2 and 1.9 µm) for the A1 AR and no appreciable affinity for the A2A and A3 ARs. Evaluation of their efficacy profiles by measurement of intracellular cAMP levels revealed that 4 and 5 behave as non-nucleoside agonists of the A1 AR with EC50 values of 0.47 and 0.87 µm, respectively. No clear concentration-response curves could be instead obtained for 6, probably because this compound modulates one or more additional targets, thus masking the putative effects exerted by its activation of A1 AR. The three compounds were not able to modulate A2B AR-mediated cAMP accumulation induced by the non-selective AR agonist NECA, thus demonstrating no affinity toward this receptor.


Subject(s)
Adenosine A1 Receptor Agonists/chemistry , Pyrimidines/chemistry , Receptor, Adenosine A1/metabolism , Adenosine A1 Receptor Agonists/chemical synthesis , Adenosine A1 Receptor Agonists/metabolism , Adenosine A1 Receptor Agonists/pharmacology , Animals , CHO Cells , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Humans , Protein Binding , Protein Isoforms/agonists , Protein Isoforms/genetics , Protein Isoforms/metabolism , Pyrimidines/chemical synthesis , Pyrimidines/metabolism , Pyrimidines/pharmacology , Receptor, Adenosine A1/chemistry , Receptor, Adenosine A1/genetics , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL