Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 31
1.
Pharmacol Res Perspect ; 10(1): e00924, 2022 02.
Article En | MEDLINE | ID: mdl-35106949

Gefapixant (MK-7264) is a first-in-class, selective antagonist of the P2X3 purinergic receptor currently being investigated as a therapeutic agent for the treatment of refractory or unexplained chronic cough. In non-clinical studies, gefapixant was eliminated primarily by renal excretion of the parent drug. The objective of this study was to assess the disposition of gefapixant in humans. The absorption, metabolism, and excretion profiles of gefapixant were assessed after oral administration of a single dose of [14 C]gefapixant to six healthy adult males. Following a single-oral [14 C]gefapixant dose to healthy adult males, the mass balance was achieved, with 98.9% of the administered radioactivity recovered in urine and feces. Elimination of gefapixant occurred primarily via renal excretion of the intact drug (64%); metabolism was a minor pathway of elimination of gefapixant (12% and 2% recovered in urine and feces, respectively). Single-dose administration of [14 C]gefapixant 50 mg was generally well tolerated in healthy adult males. The fraction of the anticipated therapeutic oral dose of gefapixant absorbed is estimated to be at least 78%. Gefapixant is expected to be the major circulating drug-related material in plasma, and the majority of the dosed drug will be excreted unchanged in urine.


Purinergic P2X Receptor Antagonists/pharmacokinetics , Pyrimidines/pharmacokinetics , Sulfonamides/pharmacokinetics , Administration, Oral , Adult , Carbon Radioisotopes , Humans , Male , Purinergic P2X Receptor Antagonists/administration & dosage , Purinergic P2X Receptor Antagonists/adverse effects , Pyrimidines/administration & dosage , Pyrimidines/adverse effects , Receptors, Purinergic P2X3/drug effects , Sulfonamides/administration & dosage , Sulfonamides/adverse effects , Young Adult
2.
CNS Neurosci Ther ; 28(2): 289-297, 2022 02.
Article En | MEDLINE | ID: mdl-34862748

AIMS: The α2 -adrenergic receptor (α2 -AR) agonists have been shown to be effective in the treatment of various pain. For example, dexmedetomidine (DEX), a selective α2A -AR agonist, can be used for peripheral analgesia. However, it is not yet fully elucidated for the precise molecular mechanisms. P2X3 receptor is a major receptor processing nociceptive information in primary sensory neurons. Herein, we show that a functional interaction of α2A -ARs and P2X3 receptors in dorsal root ganglia (DRG) neurons could contribute to peripheral analgesia of DEX. METHODS: Electrophysiological recordings were carried out on rat DRG neurons, and nociceptive behavior was quantified in rats. RESULTS: The activation of α2A -ARs by DEX suppressed P2X3 receptor-mediated and α,ß-methylene-ATP (α,ß-meATP)-evoked inward currents in a concentration-dependent and voltage-independent manner. Pre-application of DEX shifted the α,ß-meATP concentration-response curve downwards, with a decrease of 50.43 ± 4.75% in the maximal current response of P2X3 receptors to α,ß-meATP in the presence of DEX. Suppression of α,ß-meATP-evoked currents by DEX was blocked by the α2A -AR antagonist BRL44408 and prevented by intracellular application of the Gi/o protein inhibitor pertussis toxin, the adenylate cyclase activator forskolin, and the cAMP analog 8-Br-cAMP. DEX also suppressed α,ß-meATP-evoked action potentials through α2A -ARs in rat DRG neurons. Finally, the activation of peripheral α2A -ARs by DEX had an analgesic effect on the α,ß-meATP-induced nociception. CONCLUSIONS: These results suggested that activation of α2A -ARs by DEX suppressed P2X3 receptor-mediated electrophysiological and behavioral activity via a Gi/o proteins and cAMP signaling pathway, which was a novel potential mechanism underlying analgesia of peripheral α2A -AR agonists.


Adrenergic alpha-2 Receptor Agonists/pharmacology , Electrophysiological Phenomena/drug effects , Ganglia, Spinal/drug effects , Nociception/drug effects , Receptors, Adrenergic, alpha-2/drug effects , Receptors, Purinergic P2X3/drug effects , Animals , Behavior, Animal/drug effects , Dexmedetomidine/pharmacology , Male , Rats , Rats, Sprague-Dawley
3.
Neuropharmacology ; 181: 108356, 2020 12 15.
Article En | MEDLINE | ID: mdl-33069757

Endothelin-1 (ET-1), an endogenous vasoconstrictor, has been known as a pro-nociceptive agent involved in multitude of pain. ET-1 acts on endothelin receptors on vascular endothelial cells, sensitizes release of ATP, which then acts on P2X3 receptors on nociceptors and results in mechanical hyperalgesia. Both endothelin receptors and P2X3 receptors are present in primary sensory neuron, where it remains unclear whether there is an interaction between them. Herein, we reported that ET-1 potentiated the electrophysiological activity of P2X3 receptors in rat dorsal root ganglia (DRG) neurons. ET-1 concentration-dependently increased α,ß-methylene-ATP (α,ß-meATP)-evoked inward currents, which were mediated by P2X3 receptors. ET-1 shifted the α,ß-meATP concentration-response curve upwards, with an increase of 34.38 ± 4.72% in the maximal current response to α,ß-meATP in the presence of ET-1. ET-1 potentiation of α,ß-meATP-evoked currents was voltage-independent. ET-1 potentiated P2X3 receptor-mediated currents through endothelin-A receptors (ETAR), but not endothelin-B receptors (ETBR). ET-1 potentiation was supressed by blockade of intracellular G-protein or protein kinase C (PKC) signaling. Moreover, there is a synergistic effect on mechanical allodynia induced by intraplantar injection of ET-1 and α,ß-meATP in rats. Pharmacological blockade of P2X3 receptors also alleviated ET-1-induced mechanical allodynia. These results suggested that ET-1 sensitized P2X3 receptors in primary sensory neurons via an ETAR and PKC signaling pathway. Our data provide evidence that cutaneous ET-1 induced mechanical allodynia not only by increasing the release of ATP from vascular endothelial cells, but also by sensitizing P2X3 receptors on nociceptive DRG neurons.


Endothelin-1/pharmacology , Ganglia, Spinal/drug effects , Neurons/drug effects , Purinergic P2X Receptor Agonists/pharmacology , Receptors, Purinergic P2X3/drug effects , Adenosine Triphosphate/analogs & derivatives , Animals , Behavior, Animal , Electrophysiological Phenomena , GTP-Binding Proteins/drug effects , Ganglia, Spinal/cytology , Hyperalgesia/chemically induced , Male , Protein Kinase C/drug effects , Rats , Rats, Sprague-Dawley , Receptor, Endothelin A/drug effects , Sensory Receptor Cells/drug effects , Signal Transduction/drug effects
4.
Brain Res Bull ; 157: 90-99, 2020 04.
Article En | MEDLINE | ID: mdl-32017970

Diabetic cardiac autonomic neuropathy (DCAN) is a serious complication of diabetes mellitus, which often leads to cardiac dysfunction and even threatens patients' life. Osthole, a natural coumarin derivative, has anti-inflammatory, anti-oxidant and antihypertensive effects. The P2X3 receptor is related to DCAN. The objective of this study will investigate whether osthole relieves DCAN associated with the P2X3 receptor in the stellate ganglia of diabetic rats. A type 2 diabetes mellitus rat model was induced by a combination of diet and streptozotocin. Our results showed that osthole improved the abnormal changes of blood pressure, heart rate, and heart rate variability in diabetic rats and significantly reduced the up-regulated expression levels of the P2X3 receptor, tumor necrosis factor-α and interleukin-1ß in stellate ganglia of diabetic rats. Meanwhile, osthole significantly decreased the elevated serum adrenaline concentration and phosphorylation level of extracellular regulated protein kinase 1/2. In addition, the molecular docking result indicated that osthole was a perfect fit for interacting with the P2X3 receptor. Overall, osthole alleviates the sympathetic relative excitation via inhibiting the expression of P2X3 receptors in the stellate ganglia, to achieve a balance between sympathetic and parasympathetic nerves, relieves the DCAN.


Coumarins/pharmacology , Diabetic Neuropathies/drug therapy , Ganglia/drug effects , Receptors, Purinergic P2X3/drug effects , Animals , Blood Pressure/drug effects , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/pathology , Diabetic Neuropathies/pathology , Ganglia/pathology , Male , Molecular Docking Simulation , Rats, Sprague-Dawley , Receptors, Purinergic P2X3/metabolism , Streptozocin/pharmacology
5.
Sleep Breath ; 24(1): 329-337, 2020 Mar.
Article En | MEDLINE | ID: mdl-31898190

OBJECTIVE: The objective of this study was to explore the effect of Alpiniae oxyphyllae Fructus (AOF) on a rat model of chronic intermittent hypoxia (CIH)-induced enuresis. Findings of this study may help identify therapeutic targets in children with nocturnal enuresis (NE). METHODS: Female rats were randomly divided into a control group (saline gavage, 4 weeks of normal air), CIH group (saline gavage, 4 weeks of CIH), and AOF group (AOF gavage, 4 weeks of CIH). The variables measured in this study included water intake, urine output, bladder leak point pressure (BLPP), malondialdehyde (MDA) levels, and superoxide dismutase (SOD) activity. The expression levels of the purinergic P2X3 receptor, muscarinic M3 receptor, and ß3-adrenergic receptor (ß3-AR) in the bladder were also measured. The bladder was subjected to haematoxylin and eosin (HE) and Weigert staining, and histological changes were observed under a light microscope to evaluate the morphological changes in the bladder in each group. RESULTS: Compared with the control group, urine output was increased, and the BLPP was decreased in the CIH group, but AOF administration decreased urine output and increased BLPP. In addition, the serum MDA level increased and the SOD activity decreased in the CIH group compared with the control group. Administration of AOF decreased the MDA level and increased the SOD activity. Additionally, compared with the control group, HE and Weigert staining in the CIH group showed that the bladder detrusor muscle bundles were disordered and loose, some muscle bundles were broken, the content of collagen fibres in the gap was reduced, and the gap was significantly widened. However, following the administration of AOF, the bladder detrusor muscle bundles were neatly arranged, and the content of collagen fibres in the gap was increased. Furthermore, compared with the control group, the purinergic P2X3 receptor and muscarinic M3 receptor were expressed at higher levels, and ß3-AR was expressed at lower levels in the CIH group, but AOF administration decreased the expression of the purinergic P2X3 receptor and muscarinic M3 receptor and increased the expression of the ß3-AR. CONCLUSIONS: AOF improves enuresis by inhibiting oxidative stress and regulating the expression of the purinergic P2X3 receptor, muscarinic M3 receptor, and ß3 adrenergic receptor.


Disease Models, Animal , Enuresis/prevention & control , Hypoxia/complications , Plant Extracts/pharmacology , Alpinia , Animals , Enuresis/blood , Female , Hypoxia/blood , Malondialdehyde/blood , Oxidative Stress/drug effects , Rats , Receptor, Muscarinic M3/drug effects , Receptors, Adrenergic, beta-3/drug effects , Receptors, Purinergic P2X3/drug effects , Superoxide Dismutase/blood , Urinary Bladder/drug effects , Urination/drug effects
6.
Expert Opin Ther Pat ; 29(12): 943-963, 2019 Dec.
Article En | MEDLINE | ID: mdl-31726893

Introduction: Purinergic P2X3-P2X2/3 receptors are placed in nociceptive neurons' strategic location and show unique desensitization properties; hence, they represent an attractive target for many pain-related diseases. Therefore, a broad interest from academic and pharmaceutical scientists has focused on the search for P2X3 and P2X2/3 receptor ligands and has led to the discovery of numerous new selective antagonists. Some of them have been studied in clinical trials for the treatment of pathological conditions such as bladder disorders, gastrointestinal and chronic obstructive pulmonary diseases.Areas covered: This review provides a summary of the patents concerning the discovery of P2X3 and/or P2X2/3 receptor antagonists published between 2015 and 2019 and their potential clinical use. Thus, the structures and biological data of the most representative molecules are reported.Expert opinion: The 2016 publication of the crystallographic structure of the human P2X3 receptor subtype gave an improvement of published patents in 2017. Hence, a great number of small molecules with dual antagonist activity on P2X3-P2X2/3 receptors, a favorable pharmacokinetic profile, and reasonable oral bioavailability was discovered. The most promising compounds are the phenoxy-diaminopyrimidines including gefapixant (AF-219), and the imidazo-pyridines like BLU-5937, which are in phase III and phase II clinical trials, respectively, for refractory chronic cough.


Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X2/drug effects , Receptors, Purinergic P2X3/drug effects , Animals , Cough/drug therapy , Cough/pathology , Drug Discovery , Humans , Pain/drug therapy , Pain/pathology , Patents as Topic , Purinergic P2X Receptor Antagonists/pharmacokinetics , Receptors, Purinergic P2X2/metabolism , Receptors, Purinergic P2X3/metabolism
7.
Ther Adv Respir Dis ; 13: 1753466619877960, 2019.
Article En | MEDLINE | ID: mdl-31558105

BACKGROUND: Extracellular adenosine 5'-triphosphate (ATP) plays important mechanistic roles in pulmonary disorders in general and chronic obstructive pulmonary disease (COPD) and cough in particular. The effects of ATP in the lungs are mediated to a large extent by P2X2/3 receptors (P2X2/3R) localized on vagal sensory nerve terminals (both C and Aδ fibers). The activation of these receptors by ATP triggers a pulmonary-pulmonary central reflex, which results in bronchoconstriction and cough, and is also proinflammatory due to the release of neuropeptides from these nerve terminals via the axon reflex. These actions of ATP in the lungs constitute a strong rationale for the development of a new class of drugs targeting P2X2/3R. DT-0111 is a novel, small, water-soluble molecule that acts as an antagonist at P2X2/3R sites. METHODS: Experiments using receptor-binding functional assays, rat nodose ganglionic cells, perfused innervated guinea pig lung preparation ex vivo, and anesthetized and conscious guinea pigs in vivo were performed. RESULTS: DT-0111 acted as a selective and effective antagonist at P2X2/3R, that is, it did not activate or block P2YR; markedly inhibited the activation by ATP of nodose pulmonary vagal afferents in vitro; and, given as an aerosol, inhibited aerosolized ATP-induced bronchoconstriction and cough in vivo. CONCLUSIONS: These results indicate that DT-0111 is an attractive drug-candidate for the treatment of COPD and chronic cough, both of which still constitute major unmet clinical needs. The reviews of this paper are available via the supplementary material section.


Cough/drug therapy , Lung/innervation , Neurons/drug effects , Nodose Ganglion/drug effects , Pulmonary Disease, Chronic Obstructive/drug therapy , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X2/drug effects , Receptors, Purinergic P2X3/drug effects , Action Potentials , Adenosine Triphosphate/metabolism , Administration, Inhalation , Aerosols , Animals , Bronchoconstriction/drug effects , Cough/metabolism , Cough/physiopathology , Guinea Pigs , Male , Neurons/metabolism , Nodose Ganglion/metabolism , Nodose Ganglion/physiopathology , Proof of Concept Study , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/physiopathology , Purinergic P2X Receptor Antagonists/administration & dosage , Rats , Receptors, Purinergic P2X2/metabolism , Receptors, Purinergic P2X3/metabolism , Signal Transduction
8.
Neuropharmacology ; 158: 107749, 2019 11 01.
Article En | MEDLINE | ID: mdl-31461640

The homotrimeric P2X3 receptor, one of the seven members of the ATP-gated P2X receptor family, plays a crucial role in sensory neurotransmission. P2X3 receptor antagonists have been identified as promising drugs to treat chronic cough and are suggested to offer pain relief in chronic pain such as neuropathic pain. Here, we analysed whether compounds affect P2X3 receptor activity by high-throughput screening of the Spectrum Collection of 2000 approved drugs, natural products and bioactive substances. We identified aurintricarboxylic acid (ATA) as a nanomolar-potency antagonist of P2X3 receptor-mediated responses. Two-electrode voltage clamp electrophysiology-based concentration-response analysis and selectivity profiling revealed that ATA strongly inhibits the rP2X1 and rP2X3 receptors (with IC50 values of 8.6 nM and 72.9 nM, respectively) and more weakly inhibits P2X2/3, P2X2, P2X4 or P2X7 receptors (IC50 values of 0.76 µM, 22 µM, 763 µM or 118 µM, respectively). Patch-clamp analysis of mouse DRG neurons revealed that ATA inhibited native P2X3 and P2X2/3 receptors to a similar extent than rat P2X3 and P2X2/3 receptors expressed in Xenopus oocytes. In a radioligand binding assay, up to 30 µM ATA did not compete with [3H]-ATP for rP2X3 receptor binding, indicating a non-competitive mechanism of action. Molecular docking studies, site-directed mutagenesis and concentration-response analysis revealed that ATA binds to the negative allosteric site of the hP2X3 receptor. In summary, ATA as a drug-like pharmacological tool compound is a nanomolar-potency, allosteric antagonist with selectivity towards αß-methylene-ATP-sensitive P2X1 and P2X3 receptors.


Aurintricarboxylic Acid/pharmacology , Neurons/drug effects , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X1/drug effects , Receptors, Purinergic P2X3/drug effects , Allosteric Regulation , Allosteric Site , Animals , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , High-Throughput Screening Assays , Humans , Mice , Molecular Docking Simulation , Neurons/metabolism , Oocytes , Patch-Clamp Techniques , Rats , Receptors, Purinergic P2X1/metabolism , Receptors, Purinergic P2X3/metabolism , Xenopus laevis
9.
Cephalalgia ; 39(11): 1421-1434, 2019 Oct.
Article En | MEDLINE | ID: mdl-31104506

BACKGROUND: The current understanding of mechanisms behind migraine pain has been greatly enhanced with the recent therapies targeting calcitonin gene-related peptide and its receptor. The clinical efficacy of calcitonin gene-related peptide-blocking drugs indicates that, at least in a considerable proportion of patients, calcitonin gene-related peptide is a key molecule in migraine pain. There are several receptors and molecular pathways that can affect the release of and response to calcitonin gene-related peptide. One of these could be purinergic receptors that are involved in nociception, but these are greatly understudied with respect to migraine. OBJECTIVE: We aimed to explore purinergic receptors as potential anti-migraine targets. METHODS: We used the human middle meningeal artery as a proxy for the trigeminal system to screen for possible anti-migraine candidates. The human findings were followed by intravital microscopy and calcitonin gene-related peptide release measurements in rodents. RESULTS: We show that the purinergic P2Y13 receptor fulfills all the features of a potential anti-migraine target. The P2Y13 receptor is expressed in both the human trigeminal ganglion and middle meningeal artery and activation of this receptor causes: a) middle meningeal artery contraction in vitro; b) reduced dural artery dilation following periarterial electrical stimulation in vivo and c) a reduction of CGRP release from both the dura and the trigeminal ganglion in situ. Furthermore, we show that P2X3 receptor activation of the trigeminal ganglion causes calcitonin gene-related peptide release and middle meningeal artery dilation. CONCLUSION: Both an agonist directed at the P2Y13 receptor and an antagonist of the P2X3 receptor seem to be viable potential anti-migraine therapies.


Meningeal Arteries/drug effects , Migraine Disorders/metabolism , Purinergic P2 Receptor Agonists/pharmacology , Purinergic P2 Receptor Antagonists/pharmacology , Aged , Aged, 80 and over , Animals , Calcitonin Gene-Related Peptide/drug effects , Calcitonin Gene-Related Peptide/metabolism , Disease Models, Animal , Female , Humans , Male , Meningeal Arteries/metabolism , Middle Aged , Rats, Sprague-Dawley , Receptors, Purinergic P2/drug effects , Receptors, Purinergic P2/metabolism , Receptors, Purinergic P2X3/drug effects , Receptors, Purinergic P2X3/metabolism , Trigeminal Ganglion/drug effects , Trigeminal Ganglion/metabolism
10.
Pulm Pharmacol Ther ; 56: 75-78, 2019 06.
Article En | MEDLINE | ID: mdl-30880151

Chronic cough, or cough lasting >8 weeks, is often associated with underlying medical conditions (ie, asthma, gastroesophageal reflux disease, nonasthmatic eosinophilic bronchitis, and upper-airway cough syndrome). In some patients with chronic cough, treatment of these underlying conditions does not resolve the cough (refractory chronic cough [RCC]), or none of these conditions are present (unexplained chronic cough [UCC]). Despite appropriate medical evaluation, patients with RCC or UCC frequently experience cough persisting for many years, as there are currently no targeted pharmacological approaches approved for the treatment of these conditions. However, the adenosine triphosphate (ATP)-gated P2X3 receptor, a key modulator of the activation of sensory neurons central to the cough reflex, has recently garnered attention as a potential therapeutic target for the treatment of chronic cough. Gefapixant, a first-in-class, non-narcotic, selective antagonist of the P2X3 receptor, recently demonstrated efficacy and was generally well tolerated in phase 2 clinical trials in patients with RCC, validating the utility of targeting this receptor in patients with chronic cough. On the basis of these data, 2 global phase 3 trials, with combined anticipated enrolment exceeding 2000 patients and with treatment durations of up to 1 year, have been initiated. Together, these trials will further evaluate efficacy and safety of gefapixant in the control of cough in patients with RCC or UCC.


Cough/drug therapy , Purinergic P2X Receptor Antagonists/pharmacology , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Animals , Antitussive Agents/adverse effects , Antitussive Agents/pharmacology , Chronic Disease , Cough/etiology , Cough/physiopathology , Humans , Purinergic P2X Receptor Antagonists/adverse effects , Pyrimidines/adverse effects , Receptors, Purinergic P2X3/drug effects , Receptors, Purinergic P2X3/metabolism , Sulfonamides/adverse effects
11.
Pulm Pharmacol Ther ; 56: 56-62, 2019 06.
Article En | MEDLINE | ID: mdl-30902655

BLU-5937 is a small molecule that was shown to be a potent, selective and non-competitive P2X3 homotrimeric receptor antagonist. P2X3 receptors are ATP ion-gated channels located on primary afferent neurons. ATP released from damaged or inflamed tissues in the airways acts on P2X3 receptors of primary afferent neurons, triggering depolarization and action potentials that are transmitted centrally and interpreted as urge to cough. There are strong preclinical and clinical evidence supporting the role of P2X3 receptors in hypersensitization of the cough reflex, leading to chronic cough. By inhibiting P2X3 receptors on the primary sensory neurons, BLU-5937 would inhibit the hypersensitization of the cough reflex and, hence, the exaggerated cough experienced in chronic cough patients. BLU-5937 is being developed for the treatment of unexplained, refractory chronic cough. The high potency and selectivity of BLU-5937 for P2X3 homotrimeric receptors was demonstrated in vitro by inhibiting αß-meATP-evoked P2X3 or P2X2/3 receptor activity in cloned human hP2X3 and hP2X2/3 channels expressed in mammalian cells. The IC50 of BLU-5937 for hP2X3 homotrimeric and hP2X2/3 heterotrimeric receptors was established at 25 nM and >24 µM, respectively. Furthermore, BLU-5937 (500 nM) was able to block αß-meATP-induced sensitization and firing activity of isolated primary nociceptors in rat dorsal root ganglions (DRGs), through P2X3 homotrimeric receptor antagonism. In a guinea pig cough model, BLU-5937 (0.3, 3 and 30 mg/kg, oral) significantly reduced, in a dose-dependent fashion, the histamine-induced enhancement in the number of citric acid-induced coughs. BLU-5937 (3 and 30 mg/kg, oral) was also shown to reduce significantly and dose-dependently the ATP-induced enhancement of citric acid-induced coughs in the guinea pig. These anti-tussive effects were obtained at a plasma concentration known to block P2X3 homotrimeric receptors, but at concentration 50-fold lower than that required to block P2X2/3 heterotrimeric receptors. These results indicate that the anti-tussive effect of BLU-5937 is primarily mediated through the inhibition of P2X3 homotrimeric receptors. In a rat behavioral taste model, BLU-5937 (10-20 mg/kg, IP) did not alter taste perception as compared to control animals. In the same experiment, N-00588 (10-20 mg/kg, IP), a weakly selective antagonist for P2X3 versus P2X2/3 receptors, had a significant inhibitory effect on taste perception. Pharmacokinetic analysis of drug plasma concentrations showed that BLU-5937 did not affect taste function at concentrations up to 30 times the IC50 for P2X3. These results suggest that N-00588 achieved systemic concentration that blocked P2X3 and P2X2/3 receptors expressed on gustatory nerve ending innervating taste buds. The lack of effect of BLU-5937, even at high doses, on taste perception may be attributed to its higher selectivity for the P2X3 versus P2X2/3 receptors on the taste buds. The safety, tolerability and pharmacokinetic profile of BLU-5937 was assessed in a battery of preclinical studies and have revealed that BLU-5937 exhibits excellent drug-like characteristics, including good oral bioavailability, low predicted clearance in human, no blood-brain barrier permeability and high safety margin versus human predicted efficacious exposure. BLU-5937 is currently in clinical phase I development stage. In conclusion, BLU-5937 was selected as a drug candidate for the treatment of chronic cough due to its high potency and selectivity for P2X3 homotrimeric receptors, strong anti-tussive effects, excellent tolerability and predicted pharmacokinetic properties in humans.


Antitussive Agents/administration & dosage , Cough/drug therapy , Imidazoles/administration & dosage , Piperidines/administration & dosage , Purinergic P2X Receptor Antagonists/administration & dosage , Pyridines/administration & dosage , Receptors, Purinergic P2X3/drug effects , Animals , Antitussive Agents/adverse effects , Antitussive Agents/pharmacology , Chronic Disease , Disease Models, Animal , Dose-Response Relationship, Drug , Guinea Pigs , Humans , Imidazoles/adverse effects , Imidazoles/pharmacology , Inhibitory Concentration 50 , Male , Piperidines/adverse effects , Piperidines/pharmacology , Purinergic P2X Receptor Antagonists/adverse effects , Purinergic P2X Receptor Antagonists/pharmacology , Pyridines/adverse effects , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2X3/metabolism , Taste Perception/drug effects
12.
Bioorg Med Chem Lett ; 29(5): 688-693, 2019 03 01.
Article En | MEDLINE | ID: mdl-30728111

Some P2X3 receptor antagonists have been developed as new therapeutic drugs for pain. We discovered a novel chemotype of P2X3 receptor antagonists with a pyrrolinone skeleton. Because of SAR studies to improve bioavailability of lead compound 2, compound (R)-24 was identified, which showed an analgesic effect against neuropathic pain by oral administration. We constructed a human P2X3 homology model as a template for the zebrafish P2X4 receptor, which agreed with SAR studies of pyrrolinone derivatives.


Purinergic P2X Receptor Antagonists/pharmacology , Pyrroles/pharmacology , Receptors, Purinergic P2X3/drug effects , Administration, Oral , Biological Availability , Drug Discovery , High-Throughput Screening Assays , Humans , Inhibitory Concentration 50 , Neuralgia/drug therapy , Purinergic P2X Receptor Antagonists/administration & dosage , Purinergic P2X Receptor Antagonists/pharmacokinetics , Purinergic P2X Receptor Antagonists/therapeutic use , Structure-Activity Relationship
13.
Int J Neurosci ; 129(8): 784-793, 2019 Aug.
Article En | MEDLINE | ID: mdl-30621504

Aim: This study investigated whether the neuronal P2X3 receptor in rat dorsal root ganglia (DRG) mediated the effects of hesperidin on neuropathic pain. Materials and methods: The chronic constriction injury (CCI) model was used as a model of neuropathic pain. The mechanical withdrawal threshold (MWT) and thermal withdrawal latency (TWL) were measured. The mRNA and protein expression levels were assayed by real-time RT-PCR and Western blotting. Results: The results showed that mechanical and thermal hyperalgesia in the CCI rats were increased as compared to those in the sham group. The expression levels of P2X3 mRNA and protein in CCI rats were higher than those in the sham group. Dual-labelling immunofluorescence showed that the elevated P2X3 receptor was co-expressed with the neuronal marker NeuN in the DRG of CCI rats. Hesperidin treatment decreased both the mechanical and thermal hyperalgesia, and upregulated P2X3 expression in the CCI rats. Hesperidin treatment also reduced the ERK1/2 phosphorylation in the DRG of CCI rats. Moreover, hesperidin inhibited the P2X3 agonist ATP-induced currents in HEK293 cells transfected with the P2X3 plasmid. Therefore, hesperidin treatment could reverse the elevated expression of neuronal P2X3 receptor and reduce the activation of ERK1/2 in the DRG of CCI rats. Conclusions: Our findings suggested that hesperidin inhibited the nociceptive transmission mediated by the P2X3 receptor in neurons of DRG, and thus, relieved the mechanical and thermal hyperalgesia in CCI rats.


Ganglia, Spinal , Hesperidin/pharmacology , Hyperalgesia , Neuralgia , Nociception/drug effects , Receptors, Purinergic P2X3 , Animals , Disease Models, Animal , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , HEK293 Cells , Humans , Hyperalgesia/drug therapy , Hyperalgesia/metabolism , Male , Neuralgia/drug therapy , Neuralgia/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2X3/drug effects , Receptors, Purinergic P2X3/metabolism
14.
Purinergic Signal ; 14(4): 359-369, 2018 12.
Article En | MEDLINE | ID: mdl-30084084

Painful diabetic neuropathy (PDN) is a common and troublesome diabetes complication. Protein kinase C (PKC)-mediated dorsal root ganglia (DRG) P2X3 receptor upregulation is one important mechanism underlying PDN. Accumulating evidence demonstrated that electroacupuncture (EA) at low frequency could effectively attenuate neuropathic pain. Our previous study showed that 2-Hz EA could relieve pain well in PDN. The study aimed to investigate whether 2-Hz EA relieves pain in PDN through suppressing PKC-mediated DRG P2X3 receptor upregulation. A 7-week feeding of high-fat and high-sugar diet plus a single injection of streptozotocin (STZ) in a dose of 35 mg/kg after a 5-week feeding of the diet successfully induced type 2 PDN in rats as revealed by the elevated body weight, fasting blood glucose, fasting insulin and insulin resistance, and the reduced paw withdrawal threshold (PWT), as well as the destructive ultrastructural change of sciatic nerve. DRG plasma membrane P2X3 receptor level and DRG PKC expression were elevated. Two-hertz EA failed to improve peripheral neuropathy; however, it reduced PWT, DRG plasma membrane P2X3 receptor level, and DRG PKC expression in PDN rats. Intraperitoneal administration of P2X3 receptor agonist αß-meATP or PKC activator phorbol 12-myristate 13-acetate (PMA) blocked 2-Hz EA analgesia. Furthermore, PMA administration increased DRG plasma membrane P2X3 receptor level in PDN rats subject to 2-Hz EA treatment. These findings together indicated that the analgesic effect of EA in PDN is mediated by suppressing PKC-dependent membrane P2X3 upregulation in DRG. EA at low frequency is a valuable approach for PDN control.


Ganglia, Spinal/metabolism , Neuralgia/metabolism , Receptors for Activated C Kinase/metabolism , Receptors, Purinergic P2X3/metabolism , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetic Neuropathies/metabolism , Purinergic P2X Receptor Antagonists/pharmacology , Rats, Sprague-Dawley , Receptors for Activated C Kinase/drug effects , Receptors, Purinergic P2X3/drug effects , Up-Regulation
15.
Brain Res Bull ; 135: 53-61, 2017 Oct.
Article En | MEDLINE | ID: mdl-28962965

HIV-1 envelope glycoprotein (Glycoprotein 120, gp120) can directly stimulate primary sensory afferent neurons and cause chronic neuropathic pain. The P2X3 receptor in the dorsal root ganglia (DRG) is associated with the transmission of neuropathic pain. Curcumin isolated from the herb Curcuma rhizome has anti-inflammatory and anti-tumor effects. The water solubility, targeting and bioavailability of curcumin can be improved by nanoparticle encapsulation. In this study, we sought to explore the effects of nanoparticle-encapsulated curcumin (nano curcumin) on HIV-gp120-induced neuropathic pain mediated by the P2X3 receptor in DRG neurons. The results showed that mechanical and thermal hyperalgesia in rats treated with gp120 were increased compared to those in the control group. The expression levels of P2X3 mRNA and protein in rats treated with gp120 were higher than those in the control group. Nano curcumin treatment decreased mechanical hyperalgesia and thermal hyperalgesia and upregulated the expression levels of P2X3 mRNA and protein in rats treated with gp120. Nano curcumin treatment also reduced the ERK1/2 phosphorylation levels in gp120-treated rat DRG. In addition, P2X3 agonist α,ß-methylene ATP (α,ß-meATP)-induced currents in DRG neurons cultured with gp120 significantly decreased after co-treatment with nano curcumin. Therefore, nano curcumin treatment may inhibit P2X3 activation, decrease the sensitizing DRG primary afferents and relieve mechanical hyperalgesia and thermal hyperalgesia in gp120-treated rats.


Curcumin/therapeutic use , Neuralgia/drug therapy , Receptors, Purinergic P2X3/drug effects , Animals , Curcumin/administration & dosage , Curcumin/metabolism , Drug Delivery Systems/methods , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , HIV Envelope Protein gp120/drug effects , HIV Envelope Protein gp120/metabolism , HIV Infections/complications , Hyperalgesia/metabolism , Male , Nanoparticles/therapeutic use , Neuralgia/metabolism , Neurons/drug effects , Pain Threshold/drug effects , Phosphorylation , Rats , Rats, Sprague-Dawley
16.
Purinergic Signal ; 13(2): 227-235, 2017 06.
Article En | MEDLINE | ID: mdl-28054206

Type 2 diabetes mellitus (T2DM) accounts for more than 90% of all cases of diabetes mellitus (DM). Diabetic neuropathic pain (DNP) is a common complication of T2DM. Sinomenine is a natural bioactive component extracted from the Sinomenium acutum and has anti-inflammatory effects. The aim of our study was to investigate the effects of sinomenine on DNP mediated by the P2X3 receptor in dorsal root ganglia (DRG). The mechanical withdrawal threshold (MWT) and thermal withdrawal latency (TWL) in T2DM rats were lower than those of control rats. MWT and TWL in T2DM rats treated with sinomenine were higher compared with those in T2DM rats. The expression levels of the P2X3 protein and mRNA in T2DM rat DRG were higher compared with those of the control, while those in T2DM rats treated with sinomenine were significantly lower compared with those of the T2DM rats. Sinomenine significantly inhibited P2X3 agonist ATP-activated currents in HEK293 cells transfected with the P2X3 receptor. Sinomenine decreased the phosphorylation and activation of P38MAPK in T2DM DRG. Therefore, sinomenine treatment may suppress the up-regulated expression and activation of the P2X3 receptor and relieve the hyperalgesia potentiated by the activation of P38MAPK in T2DM rats.


Diabetic Neuropathies , Morphinans/pharmacology , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X3/drug effects , Animals , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2/complications , Ganglia, Spinal/drug effects , HEK293 Cells , Humans , Hyperalgesia , Male , Pain Threshold/drug effects , Rats , Rats, Sprague-Dawley
17.
J Neurochem ; 138(4): 587-97, 2016 08.
Article En | MEDLINE | ID: mdl-27217099

P2X3 receptors, gated by extracellular ATP, are expressed by sensory neurons and are involved in peripheral nociception and pain sensitization. The ability of P2X3 receptors to transduce extracellular stimuli into neuronal signals critically depends on the dynamic molecular partnership with the calcium/calmodulin-dependent serine protein kinase (CASK). The present work used trigeminal sensory neurons to study the impact that activation of P2X3 receptors (evoked by the agonist α,ß-meATP) has on the release of endogenous ATP and how CASK modulates this phenomenon. P2X3 receptor function was followed by ATP efflux via Pannexin1 (Panx1) hemichannels, a mechanism that was blocked by the P2X3 receptor antagonist A-317491, and by P2X3 silencing. ATP efflux was enhanced by nerve growth factor, a treatment known to potentiate P2X3 receptor function. Basal ATP efflux was not controlled by CASK, and carbenoxolone or Pannexin silencing reduced ATP release upon P2X3 receptor function. CASK-controlled ATP efflux followed P2X3 receptor activity, but not depolarization-evoked ATP release. Molecular biology experiments showed that CASK was essential for the transactivation of Panx1 upon P2X3 receptor activation. These data suggest that P2X3 receptor function controls a new type of feed-forward purinergic signaling on surrounding cells, with consequences at peripheral and spinal cord level. Thus, P2X3 receptor-mediated ATP efflux may be considered for the future development of pharmacological strategies aimed at containing neuronal sensitization. P2X3 receptors are involved in sensory transduction and associate to CASK. We have studied in primary sensory neurons the molecular mechanisms downstream P2X3 receptor activation, namely ATP release and partnership with CASK or Panx1. Our data suggest that CASK and P2X3 receptors are part of an ATP keeper complex, with important feed-forward consequences at peripheral and central level.


Adenosine Triphosphate/metabolism , Calcium/metabolism , Calmodulin/metabolism , Ganglia, Sensory/metabolism , Receptors, Purinergic P2X3/metabolism , Sensory Receptor Cells/metabolism , Animals , Ganglia, Sensory/drug effects , Mice, Inbred C57BL , Phenols/pharmacology , Polycyclic Compounds/pharmacology , Receptors, Purinergic P2X3/drug effects , Sensory Receptor Cells/drug effects , Signal Transduction/drug effects , Trigeminal Ganglion/cytology , Trigeminal Ganglion/drug effects
18.
Eur J Pharmacol ; 767: 24-9, 2015 Nov 15.
Article En | MEDLINE | ID: mdl-26435025

Prokineticin 2 (PK2), a new chemokine, causes mechanical hypersensitivity in the rat hind paw, but little is known about the molecular mechanism. Here, we have found that ionotropic P2X receptor is essential to mechanical allodynia induced by PK2. First, intraplantar injection of high dose (3 or 10 pmol) of PK2 significantly increased paw withdrawal response frequency (%) to innocuous mechanical stimuli (mechanical allodynia). And the mechanical allodynia induced by PK2 was prevented by co-administration of TNP-ATP, a selective P2X receptor antagonist. Second, although low dose (0.3 or 1 pmol) of PK2 itself did not produce an allodynic response, it significantly facilitated the mechanical allodynia evoked by intraplantar injection of α,ß-methylene ATP (α,ß-meATP). Third, PK2 concentration-dependently potentiated α,ß-meATP-activated currents in rat dorsal root ganglion (DRG) neurons. Finally, PK2 receptors and intracellular signal transduction were involved in PK2 potentiation of α,ß-meATP-induced mechanical allodynia and α,ß-meATP-activated currents, since the potentiation were blocked by PK2 receptor antagonist PKRA and selective PKC inhibitor GF 109203X. These results suggested that PK2 facilitated mechanical allodynia induced by α,ß-meATP through a mechanism involved in sensitization of cutaneous P2X receptors expressed by nociceptive nerve endings.


Adenosine Triphosphate/analogs & derivatives , Gastrointestinal Hormones/pharmacology , Hyperalgesia/chemically induced , Neuropeptides/pharmacology , Adenosine Triphosphate/adverse effects , Adenosine Triphosphate/pharmacology , Animals , Drug Synergism , Ganglia, Spinal/drug effects , Ganglia, Spinal/physiology , Gastrointestinal Hormones/antagonists & inhibitors , Hyperalgesia/physiopathology , Indoles/pharmacology , Male , Maleimides/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neuropeptides/antagonists & inhibitors , Protein Kinase C/antagonists & inhibitors , Purinergic P2X Receptor Antagonists/pharmacology , Rats , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, Peptide/antagonists & inhibitors , Receptors, Purinergic P2X3/drug effects , Receptors, Purinergic P2X3/physiology
19.
PLoS One ; 10(8): e0134803, 2015.
Article En | MEDLINE | ID: mdl-26241848

OBJECTIVES: To evaluate whether botulinum toxin A (BoNT-A) injection and Lipotoxin (liposomes with 200 U of BoNT-A) instillation target different proteins, including P2X3, synaptic vesicle glycoprotein 2A, and SNAP-25, in the bladder mucosa, leading to different treatment outcomes. MATERIALS AND METHODS: This was a retrospective study performed in a tertiary teaching hospital. We evaluated the clinical results of 27 OAB patients treated with intravesical BoNT-A injection (n = 16) or Lipotoxin instillation (n = 11). Seven controls were treated with saline. Patients were injected with 100 U of BoNT-A or Lipotoxinin a single intravesical instillation. The patients enrolled in this study all had bladder biopsies performed at baseline and one month after BoNT-A therapy. Treatment outcome was measured by the decreases in urgency and frequency episodes at 1 month. The functional protein expressions in the urothelium were measured at baseline and after 1 month. The Wilcoxon signed-rank test and ordinal logistic regression were used to compare the treatment outcomes. RESULTS: Both BoNT-A injection and Lipotoxin instillation treatments effectively decreased the frequency of urgency episodes in OAB patients. Lipotoxin instillation did not increase post-void residual volume. BoNT-A injection effectively cleaved SNAP-25 (p < 0.01). Liposome encapsulated BoNT-A decreased urothelial P2X3 expression in the five responders (p = 0.04), while SNAP-25 was not significantly cleaved. CONCLUSIONS: The results of this study provide a possible mechanism for the therapeutic effects of BoNT-A for the treatment of OAB via different treatment forms. BoNT-A and Lipotoxin treatments effectively decreased the frequency of urgency episodes in patients with OAB.


Botulinum Toxins, Type A/therapeutic use , Urinary Bladder, Overactive/drug therapy , Urinary Bladder/drug effects , Urothelium/drug effects , Administration, Intravesical , Biopsy , Botulinum Toxins, Type A/administration & dosage , Botulinum Toxins, Type A/pharmacology , Drug Carriers , Gene Expression , Humans , Liposomes , Membrane Glycoproteins/drug effects , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Receptors, Purinergic P2X3/drug effects , Receptors, Purinergic P2X3/genetics , Receptors, Purinergic P2X3/physiology , Retrospective Studies , Synaptosomal-Associated Protein 25/drug effects , Synaptosomal-Associated Protein 25/genetics , Synaptosomal-Associated Protein 25/metabolism , Treatment Outcome , Urinary Bladder/metabolism , Urinary Bladder/pathology , Urinary Bladder, Overactive/metabolism , Urothelium/metabolism , Urothelium/pathology
20.
Anesth Analg ; 120(3): 597-605, 2015 Mar.
Article En | MEDLINE | ID: mdl-25695577

BACKGROUND: Lidocaine has been widely used to relieve acute pain and chronic refractory pain effectively by both systemic and local administration. Numerous studies reported that lidocaine affects several pain signaling pathways as well as voltage-gated sodium channels, suggesting the existence of multiple mechanisms underlying pain relief by lidocaine. Some extracellular adenosine triphosphate (ATP) receptor subunits are thought to play a role in chronic pain mechanisms, but there have been few studies on the effects of lidocaine on ATP receptors. We studied the effects of lidocaine on purinergic P2X3, P2X4, and P2X7 receptors to explore the mechanisms underlying pain-relieving effects of lidocaine. METHODS: We investigated the effects of lidocaine on ATP-induced currents in ATP receptor subunits, P2X3, P2X4, and P2X7 expressed in Xenopus oocytes, by using whole-cell, two-electrode, voltage-clamp techniques. RESULTS: Lidocaine inhibited ATP-induced currents in P2X7, but not in P2X3 or P2X4 subunits, in a concentration-dependent manner. The half maximal inhibitory concentration for lidocaine inhibition was 282 ± 45 µmol/L. By contrast, mepivacaine, ropivacaine, and bupivacaine exerted only limited effects on the P2X7 receptor. Lidocaine inhibited the ATP concentration-response curve for the P2X7 receptor via noncompetitive inhibition. Intracellular and extracellular N-(2,6-dimethylphenylcarbamoylmethyl) triethylammonium bromide (QX-314) and benzocaine suppressed ATP-induced currents in the P2X7 receptor in a concentration-dependent manner. In addition, repetitive ATP treatments at 5-minute intervals in the continuous presence of lidocaine revealed that lidocaine inhibition was use-dependent. Finally, the selective P2X7 receptor antagonists Brilliant Blue G and AZ11645373 did not affect the inhibitory actions of lidocaine on the P2X7 receptor. CONCLUSIONS: Lidocaine selectively inhibited the function of the P2X7 receptor expressed in Xenopus oocytes. This effect may be caused by acting on sites in the ion channel pore both extracellularly and intracellularly. These results help to understand the mechanisms underlying the analgesic effects of lidocaine when it is administered locally at least.


Anesthetics, Local/pharmacology , Lidocaine/pharmacology , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X7/drug effects , Adenosine Triphosphate/metabolism , Anesthetics, Local/metabolism , Animals , Binding Sites , Binding, Competitive , Dose-Response Relationship, Drug , Female , Humans , Lidocaine/metabolism , Membrane Potentials , Purinergic P2X Receptor Antagonists/metabolism , Receptors, Purinergic P2X3/drug effects , Receptors, Purinergic P2X3/metabolism , Receptors, Purinergic P2X4/drug effects , Receptors, Purinergic P2X4/metabolism , Receptors, Purinergic P2X7/metabolism , Time Factors , Xenopus laevis
...