Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 483
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Physiol Rev ; 101(2): 545-567, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33124941

ABSTRACT

Evolving information has identified disease mechanisms and dysregulation of host biology that might be targeted therapeutically in coronavirus disease 2019 (COVID-19). Thrombosis and coagulopathy, associated with pulmonary injury and inflammation, are emerging clinical features of COVID-19. We present a framework for mechanisms of thrombosis in COVID-19 that initially derive from interaction of SARS-CoV-2 with ACE2, resulting in dysregulation of angiotensin signaling and subsequent inflammation and tissue injury. These responses result in increased signaling by thrombin (proteinase-activated) and purinergic receptors, which promote platelet activation and exert pathological effects on other cell types (e.g., endothelial cells, epithelial cells, and fibroblasts), further enhancing inflammation and injury. Inhibitors of thrombin and purinergic receptors may, thus, have therapeutic effects by blunting platelet-mediated thromboinflammation and dysfunction in other cell types. Such inhibitors include agents (e.g., anti-platelet drugs) approved for other indications, and that could be repurposed to treat, and potentially improve the outcome of, COVID-19 patients. COVID-19, caused by the SARS-CoV-2 virus, drives dysregulation of angiotensin signaling, which, in turn, increases thrombin-mediated and purinergic-mediated activation of platelets and increase in inflammation. This thromboinflammation impacts the lungs and can also have systemic effects. Inhibitors of receptors that drive platelet activation or inhibitors of the coagulation cascade provide opportunities to treat COVID-19 thromboinflammation.


Subject(s)
COVID-19/complications , Inflammation/etiology , Receptors, Proteinase-Activated/metabolism , Receptors, Purinergic/metabolism , SARS-CoV-2 , Thrombosis/etiology , Humans , Inflammation/drug therapy , Purinergic Antagonists/pharmacology , Receptors, Proteinase-Activated/antagonists & inhibitors , Receptors, Proteinase-Activated/genetics , Receptors, Purinergic/genetics , Thrombosis/prevention & control
2.
Kidney Int ; 106(3): 392-399, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38821448

ABSTRACT

Platelets are anucleated cells that circulate in the bloodstream. Historically, platelets were thought to perform a singular function-stop bleeding via clotting. Although platelets do play a key role in hemostasis and thrombosis, recent studies indicate that platelets also modulate inflammation, and this platelet-induced inflammation contributes to the pathophysiology of various diseases such as atherosclerosis and diabetes mellitus. Thus, in recent years, our understanding of platelet function has broadened. In this review, we revisit the classic role of platelets in hemostasis and thrombosis and describe the newly recognized function of platelets in modulating inflammation. We cover the potential use of purinergic receptor antagonists to prevent platelet-modulated inflammation, particularly in patients with chronic kidney disease, and finally, we define key questions that must be addressed to understand how platelet-modulated inflammation contributes to the pathophysiology of chronic kidney disease.


Subject(s)
Blood Platelets , Inflammation , Receptors, Purinergic , Renal Insufficiency, Chronic , Humans , Blood Platelets/metabolism , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Inflammation/blood , Inflammation/metabolism , Receptors, Purinergic/metabolism , Animals , Purinergic Antagonists , Signal Transduction , Hemostasis/physiology , Thrombosis/blood , Thrombosis/metabolism
3.
Pharmacol Rev ; 71(3): 345-382, 2019 07.
Article in English | MEDLINE | ID: mdl-31235653

ABSTRACT

Immune-mediated inflammatory diseases (IMIDs) encompass a wide range of seemingly unrelated conditions, such as multiple sclerosis, rheumatoid arthritis, psoriasis, inflammatory bowel diseases, asthma, chronic obstructive pulmonary disease, and systemic lupus erythematosus. Despite differing etiologies, these diseases share common inflammatory pathways, which lead to damage in primary target organs and frequently to a plethora of systemic effects as well. The purinergic signaling complex comprising extracellular nucleotides and nucleosides and their receptors, the P2 and P1 purinergic receptors, respectively, as well as catabolic enzymes and nucleoside transporters is a major regulatory system in the body. The purinergic signaling complex can regulate the development and course of IMIDs. Here we provide a comprehensive review on the role of purinergic signaling in controlling immunity, inflammation, and organ function in IMIDs. In addition, we discuss the possible therapeutic applications of drugs acting on purinergic pathways, which have been entering clinical development, to manage patients suffering from IMIDs.


Subject(s)
Inflammation/drug therapy , Inflammation/immunology , Purinergic Agonists/pharmacology , Purinergic Antagonists/pharmacology , Purines/metabolism , Receptors, Purinergic/metabolism , Animals , Humans , Inflammation/metabolism , Molecular Targeted Therapy , Purines/immunology , Receptors, Purinergic/immunology , Signal Transduction/drug effects
4.
Am J Physiol Heart Circ Physiol ; 320(4): H1699-H1711, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33606585

ABSTRACT

We sought to determine if a pannexin/purinergic-dependent intravascular communication pathway exists in skeletal muscle microvasculature that facilitates capillary communication with upstream arterioles that control their perfusion. Using the hamster cremaster muscle and intravital microscopy, we locally stimulated capillaries and observed the vasodilatory response in the associated upstream 4A arteriole. We stimulated capillaries with vasodilators relevant to muscle contraction: 10-6 M S-nitroso-N-acetyl-dl-penicillamine (SNAP; nitric oxide donor), 10-6 M adenosine, 10 mM potassium chloride, 10-5 M pinacidil, as well as a known initiator of gap-junction-dependent intravascular communication, acetylcholine (10-5 M), in the absence and the presence of the purinergic membrane receptor blocker suramin (10-5 M), pannexin blocker mefloquine (2 × 10-5 M), or probenecid (5 × 10-6 M) and gap-junction inhibitor halothane (0.07%) applied in the transmission pathway, between the capillary stimulation site and the upstream 4A observation site. Potassium chloride, SNAP, and adenosine-induced upstream vasodilations were significantly inhibited by suramin, mefloquine, and probenecid but not halothane, indicating the involvement of a pannexin/purinergic-dependent signaling pathway. Conversely, SNAP-induced upstream vasodilation was only inhibited by halothane indicating that communication was facilitated by gap junctions. Both pinacidil and acetylcholine were inhibited by suramin but only acetylcholine was inhibited by halothane. These data demonstrate the presence of a pannexin/purinergic-dependent communication pathway between capillaries and upstream arterioles controlling their perfusion. This pathway adds to the gap-junction-dependent pathway that exists at this vascular level as well. Given that vasodilators relevant to muscle contraction can use both of these pathways, our data implicate the involvement of both pathways in the coordination of skeletal muscle blood flow.NEW & NOTEWORTHY Blood flow control during increased metabolic demand in skeletal muscle is not fully understood. Capillaries have been implicated in controlling blood flow to active skeletal muscle, but how capillaries communicate to the arteriolar vascular network is not clear. Our study uncovers a novel pathway through which capillaries can communicate to upstream arterioles to cause vasodilation and therefore control perfusion. This work implicates a new vascular communication pathway in blood flow control in skeletal muscle.


Subject(s)
Abdominal Muscles/blood supply , Arterioles/metabolism , Capillaries/metabolism , Cell Communication , Connexins/metabolism , Purines/metabolism , Receptors, Purinergic/metabolism , Vasodilation , Animals , Capillaries/drug effects , Connexins/antagonists & inhibitors , Gap Junctions/metabolism , Male , Mesocricetus , Muscle Contraction , Purinergic Agonists/pharmacology , Purinergic Antagonists/pharmacology , Regional Blood Flow , Signal Transduction , Vasodilation/drug effects , Vasodilator Agents/pharmacology
5.
Cell Mol Neurobiol ; 41(5): 1085-1101, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33216235

ABSTRACT

Many G protein-coupled receptors (GPCRs) signal through more than one subtype of heterotrimeric G proteins. For example, the C-C chemokine receptor type 5 (CCR5), which serves as a co-receptor to facilitate cellular entry of human immunodeficiency virus 1 (HIV-1), normally signals through the heterotrimeric G protein, Gi. However, CCR5 also exhibits G protein signaling bias and certain chemokine analogs can cause a switch to Gq pathways to induce Ca2+ signaling. We want to understand how much of the Ca2+ signaling from Gi-coupled receptors is due to G protein promiscuity and how much is due to transactivation and crosstalk with other receptors. We propose a possible mechanism underlying the apparent switching between different G protein signaling pathways. We show that chemokine-mediated Ca2+ flux in HEK293T cells expressing CCR5 can be primed and enhanced by ATP pretreatment. In addition, agonist-dependent lysosomal exocytosis results in the release of ATP to the extracellular milieu, which amplifies cellular signaling networks. ATP is quickly degraded via ADP and AMP to adenosine. ATP, ADP and adenosine activate different cell surface purinergic receptors. Endogenous Gq-coupled purinergic P2Y receptors amplify Ca2+ signaling and allow for Gi- and Gq-coupled receptor signaling pathways to converge. Associated secretory release of GPCR ligands, such as chemokines, opioids, and monoamines, should also lead to concomitant release of ATP with a synergistic effect on Ca2+ signaling. Our results suggest that crosstalk between ATP-activated purinergic receptors and other Gi-coupled GPCRs is an important cooperative mechanism to amplify the intracellular Ca2+ signaling response.


Subject(s)
Calcium Signaling/physiology , Receptor Cross-Talk/physiology , Receptors, CCR5/agonists , Receptors, CCR5/metabolism , Receptors, Purinergic/metabolism , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Calcium Signaling/drug effects , Chemokine CCL5/metabolism , Chemokine CCL5/pharmacology , HEK293 Cells , Humans , Purinergic Agonists/metabolism , Purinergic Agonists/pharmacology , Purinergic Antagonists/metabolism , Purinergic Antagonists/pharmacology , Receptor Cross-Talk/drug effects , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Suramin/metabolism , Suramin/pharmacology
6.
Bioorg Chem ; 116: 105378, 2021 11.
Article in English | MEDLINE | ID: mdl-34601296

ABSTRACT

G-protein-coupled receptors for extracellular nucleotides are known as P2Y receptors and are made up of eight members that are encoded by distinct genes and can be classified into two classes based on their affinity for specific G-proteins. P2Y receptor modulators have been studied extensively, but only a few small-molecule P2Y receptor antagonists have been discovered so far and approved by drug agencies. Derivatives of indole carboxamide have been identified as P2Y12 and P2X7 antagonist, as a result, we developed and tested a series of indole derivatives4a-lhaving thiourea moiety as P2Y receptor antagonist by using a fluorescence-based assay to measure the inhibition of intracellular calcium release in 1321N1 astrocytoma cells that had been stably transfected with the P2Y1, P2Y2, P2Y4 and P2Y6 receptors. Most of the compounds exhibited moderate to excellent inhibition activity against P2Y1 receptor subtype. The series most potent compound, 4h exhibited an IC50 value of 0.36 ± 0.01 µM selectivity against other subtypes of P2Y receptor. To investigate the ligand-receptor interactions, the molecular docking studies were carried out. Compound 4h is the most potent P2Y1 receptor antagonist due to interaction with an important amino acid residue Pro105, in addition to Ile108, Phe119, and Leu102.


Subject(s)
Indomethacin/pharmacology , Purinergic Antagonists/pharmacology , Receptors, Purinergic/metabolism , Thiourea/pharmacology , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Indomethacin/chemical synthesis , Indomethacin/chemistry , Molecular Structure , Purinergic Antagonists/chemical synthesis , Purinergic Antagonists/chemistry , Structure-Activity Relationship , Thiourea/chemistry
7.
Immunopharmacol Immunotoxicol ; 43(6): 633-643, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34647511

ABSTRACT

The coronavirus disease-19 (COVID-19), at first, was reported in Wuhan, China, and then rapidly became pandemic throughout the world. Cytokine storm syndrome (CSS) in COVID-19 patients is associated with high levels of cytokines and chemokines that cause multiple organ failure, systemic inflammation, and hemodynamic instabilities. Acute respiratory distress syndrome (ARDS), a common complication of COVID-19, is a consequence of cytokine storm. In this regard, several drugs have been being investigated to suppress this inflammatory condition. Purinergic signaling receptors comprising of P1 adenosine and P2 purinoceptors play a critical role in inflammation. Therefore, activation or inhibition of some subtypes of these kinds of receptors is most likely to be beneficial to attenuate cytokine storm. This article summarizes suggested therapeutic drugs with potential anti-inflammatory effects through purinergic receptors.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , COVID-19 Drug Treatment , Cytokine Release Syndrome/prevention & control , Cytokines/blood , Purinergic Antagonists/therapeutic use , Receptors, Purinergic/drug effects , SARS-CoV-2/drug effects , Animals , Anti-Inflammatory Agents/adverse effects , Biomarkers/blood , COVID-19/blood , COVID-19/immunology , COVID-19/virology , Cytokine Release Syndrome/blood , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/virology , Host-Pathogen Interactions , Humans , Ligands , Molecular Targeted Therapy , Multiple Organ Failure/immunology , Multiple Organ Failure/prevention & control , Multiple Organ Failure/virology , Purinergic Antagonists/adverse effects , Receptors, Purinergic/metabolism , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity , Signal Transduction
8.
Int J Mol Sci ; 22(13)2021 Jun 25.
Article in English | MEDLINE | ID: mdl-34202215

ABSTRACT

Cytokines are important neuroinflammatory modulators in neurodegenerative brain disorders including traumatic brain injury (TBI) and stroke. However, their temporal effects on the physiological properties of microglia and neurons during the recovery period have been unclear. Here, using an ATP-induced cortical injury model, we characterized selective effects of ATP injection compared to needle-control. In the damaged region, the fluorescent intensity of CX3CR1-GFP (+) cells, as well as the cell density, was increased and the maturation of newborn BrdU (+) cells continued until 28 day-post-injection (dpi) of ATP. The excitability and synaptic E/I balance of neurons and the inward and outward membrane currents of microglia were increased at 3 dpi, when expressions of tumor necrosis factor (TNF)-α/interleukin (IL)-1ß and IL-10/IL-4 were also enhanced. These changes of both cells at 3 dpi were mostly decayed at 7 dpi and were suppressed by any of IL-10, IL-4, suramin (P2 receptor inhibitor) and 4-AP (K+ channel blocker). Acute ATP application alone induced only small effects from both naïve neurons and microglial cells in brain slice. However, TNF-α alone effectively increased the excitability of naïve neurons, which was blocked by suramin or 4-AP. TNF-α and IL-1ß increased and decreased membrane currents of naïve microglia, respectively. Our results suggest that ATP and TNF-α dominantly induce the physiological activities of 3 dpi neurons and microglia, and IL-10 effectively suppresses such changes of both activated cells in K+ channel- and P2 receptor-dependent manner, while IL-4 suppresses neurons preferentially.


Subject(s)
Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Membrane Potentials , Microglia/physiology , Adenosine Triphosphate/metabolism , Animals , Brain Injuries, Traumatic/diagnosis , Brain Injuries, Traumatic/etiology , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Gene Expression , Genes, Reporter , Mice , Neurons/drug effects , Neurons/metabolism , Purinergic Antagonists/pharmacology
9.
Int J Mol Sci ; 22(2)2021 Jan 12.
Article in English | MEDLINE | ID: mdl-33445804

ABSTRACT

Neural progenitor cells (NPCs) are self-renewing and multipotent cells that persist in the postnatal and adult brain in the subventricular zone and the hippocampus. NPCs can be expanded in vitro to be used in cell therapy. However, expansion is limited, since the survival and proliferation of adult NPCs decrease with serial passages. Many signaling pathways control NPC survival and renewal. Among these, purinergic receptor activation exerts differential effects on the biology of adult NPCs depending on the cellular context. In this study, we sought to analyze the effect of a general blockade of purinergic receptors with suramin on the proliferation and survival of NPCs isolated from the subventricular zone of postnatal rats, which are cultured as neurospheres. Treatment of neurospheres with suramin induced a significant increase in neurosphere diameter and in NPC number attributed to a decrease in apoptosis. Proliferation and multipotency were not affected. Suramin also induced an increase in the gap junction protein connexin43 and in vascular endothelial growth factor, which might be involved in the anti-apoptotic effect. Our results offer a valuable tool for increasing NPC survival before implantation in the lesioned brain and open the possibility of using this drug as adjunctive therapy to NPC transplantation.


Subject(s)
Cell Survival/drug effects , Neural Stem Cells/drug effects , Purinergic Antagonists/pharmacology , Receptors, Purinergic/metabolism , Stem Cells/drug effects , Suramin/pharmacology , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Female , Hippocampus/drug effects , Hippocampus/metabolism , Lateral Ventricles/drug effects , Lateral Ventricles/metabolism , Male , Neural Stem Cells/metabolism , Rats , Rats, Wistar , Signal Transduction/drug effects , Stem Cells/metabolism , Vascular Endothelial Growth Factor A/metabolism
10.
Am J Physiol Heart Circ Physiol ; 319(4): H723-H729, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32822211

ABSTRACT

Cardiovascular disease is a major cause of morbidity and mortality worldwide. Innovative new treatment options for this cardiovascular pandemic are urgently needed. Activation of purinergic receptors (PRs) is critically involved in the development and progression of cardiovascular disease including atherosclerosis, ischemic heart disease, hypertension, and diabetes. PRs have been targeted for the treatment of several cardiovascular diseases in a clinical setting. The P2Y12R antagonists such as clopidogrel, ticagrelor, and others are the most successful class of purinergic drugs targeting platelets for the treatment of acute coronary syndrome. In addition to targeting platelets, ticagrelor may exert P2Y12R-independent effect by targeting erythrocyte-mediated purinergic activation. The partial A1R agonist neladenoson and the A2AR agonist regadenoson have been applied in cardiovascular medicine. In experimental studies, many other PRs have been shown to play a significant role in the development and progression of cardiovascular diseases, and targeting these receptors have resulted in promising outcomes. Therefore, many of these PRs including A2BR, A3R, P2X3R, P2X4R, P2X7R, P2Y1R, P2Y4R, P2Y6R, and P2Y11R can be considered as therapeutic targets. However, the multitude of PR subtypes expressed in different cells of the cardiovascular system may constitute a challenge whether single or multiple receptors should be targeted at the same time for the best efficacy. The present review discusses the promising purinergic drugs used in clinical studies for the treatment of cardiovascular disease. We also update experimental evidence for many other PRs that can be considered as therapeutic targets for future drug development.


Subject(s)
Cardiovascular Agents/therapeutic use , Cardiovascular Diseases/drug therapy , Purinergic Agonists/therapeutic use , Purinergic Antagonists/therapeutic use , Receptors, Purinergic/drug effects , Animals , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/physiopathology , Drug Development , Drug Discovery , Humans , Molecular Targeted Therapy , Receptors, Purinergic/metabolism , Signal Transduction
11.
Int J Mol Sci ; 21(8)2020 Apr 15.
Article in English | MEDLINE | ID: mdl-32326617

ABSTRACT

The purinergic (P2) receptor P2Y14 is the only P2 receptor that is stimulated by uridine diphosphate (UDP)-sugars and its role in bone formation is unknown. We confirmed P2Y14 expression in primary murine osteoblasts (CB-Ob) and the C2C12-BMP2 osteoblastic cell line (C2-Ob). UDP-glucose (UDPG) had undiscernible effects on cAMP levels, however, induced dose-dependent elevations in the cytosolic free calcium concentration ([Ca2+]i) in CB-Ob, but not C2-Ob cells. To antagonize the P2Y14 function, we used the P2Y14 inhibitor PPTN or generated CRISPR-Cas9-mediated P2Y14 knockout C2-Ob clones (Y14KO). P2Y14 inhibition facilitated calcium signalling and altered basal cAMP levels in both models of osteoblasts. Importantly, P2Y14 inhibition augmented Ca2+ signalling in response to ATP, ADP and mechanical stimulation. P2Y14 knockout or inhibition reduced osteoblast proliferation and decreased ERK1/2 phosphorylation and increased AMPKα phosphorylation. During in vitro osteogenic differentiation, P2Y14 inhibition modulated the timing of osteogenic gene expression, collagen deposition, and mineralization, but did not significantly affect differentiation status by day 28. Of interest, while P2ry14-/- mice from the International Mouse Phenotyping Consortium were similar to wild-type controls in bone mineral density, their tibia length was significantly increased. We conclude that P2Y14 in osteoblasts reduces cell responsiveness to mechanical stimulation and mechanotransductive signalling and modulates osteoblast differentiation.


Subject(s)
Cell Proliferation/genetics , Osteoblasts/metabolism , Osteogenesis/genetics , Purinergic Antagonists/pharmacology , Receptors, Purinergic P2Y/metabolism , Signal Transduction/genetics , Uridine Diphosphate Sugars/metabolism , Adenosine Diphosphate/pharmacology , Adenosine Triphosphate/pharmacology , Animals , Bone Density/genetics , CRISPR-Cas Systems , Calcium/metabolism , Cell Line , Cell Proliferation/drug effects , Cells, Cultured , Cyclic AMP/metabolism , Gene Knockout Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Osteogenesis/drug effects , Phosphorylation , Purinergic Antagonists/metabolism , Receptors, Purinergic P2Y/genetics , Signal Transduction/drug effects , Uridine Diphosphate Glucose/metabolism , Uridine Diphosphate Glucose/pharmacology , Uridine Diphosphate Sugars/pharmacology
12.
J Physiol ; 597(7): 1957-1973, 2019 04.
Article in English | MEDLINE | ID: mdl-30746715

ABSTRACT

KEY POINTS: In the epididymis, elaborate communication networks between epithelial cells are important with respect to establishing an optimal acidic luminal environment for the maturation and storage of spermatozoa, which is essential for male fertility. Proton secretion by epididymal clear cells is achieved via the proton pumping V-ATPase located in their apical membrane. In the present study, we dissect the molecular mechanisms by which clear cells respond to luminal ATP and adenosine to modulate their acidifying activity via the adenosine receptor ADORA2B and the pH-sensitive ATP receptor P2X4. We demonstrate that the hydrolysis of ATP to produce adenosine by ectonucleotidases plays a key role in V-ATPase-dependent proton secretion, and is part of a feedback loop that ensures acidification of the luminal compartment These results help us better understand how professional proton-secreting cells respond to extracellular cues to modulate their functions, and how they communicate with neighbouring cells. ABSTRACT: Cell-cell cross-talk is crucial for the dynamic function of epithelia, although how epithelial cells detect and respond to variations in extracellular stimuli to modulate their environment remains incompletely understood. In the present study, we used the epididymis as a model system to investigate epithelial cell regulation by luminal factors. In the epididymis, elaborate communication networks between the different epithelial cell types are important for establishing an optimal acidic luminal environment for the maturation and storage of spermatozoa. In particular, clear cells (CCs) secrete protons into the lumen via the proton pumping V-ATPase located in their apical membrane, a process that is activated by luminal alkalinization. However, how CCs detect luminal pH variations to modulate their function remains uncharacterized. Purinergic regulation of epithelial transport is modulated by extracellular pH in other tissues. In the present study, functional analysis of the mouse cauda epididymis perfused in vivo showed that luminal ATP and adenosine modulate the acidifying activity of CCs via the purinergic ADORA2B and P2X4 receptors, and that luminal adenosine content is itself regulated by luminal pH. Altogether, our observations illustrate mechanisms by which CCs are activated by pH sensitive P2X4 receptor and ectonucleotidases, providing a feedback mechanism for the maintenance of luminal pH. These novel mechanisms by which professional proton-secreting cells respond to extracellular cues to modulate their functions, as well as how they communicate with neighbouring cells, might be translatable to other acidifying epithelia.


Subject(s)
Adenosine Triphosphate/pharmacology , Adenosine/pharmacology , Epididymis/physiology , Purinergic Agents , Purinergic Agonists/pharmacology , Vacuolar Proton-Translocating ATPases/metabolism , Animals , Epididymis/drug effects , Gene Expression Regulation , Hydrogen-Ion Concentration , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Purinergic Antagonists/pharmacology , Receptor, Adenosine A2B/genetics , Receptor, Adenosine A2B/metabolism , Receptors, Purinergic P2X4/genetics , Receptors, Purinergic P2X4/metabolism , Vacuolar Proton-Translocating ATPases/genetics
13.
J Cell Biochem ; 120(10): 17123-17130, 2019 10.
Article in English | MEDLINE | ID: mdl-31106899

ABSTRACT

AIM: To explore the role of P2Y6 receptors in the maintenance of neuropathic pain and progression of oxidative stress, we investigated the efficacy of the selective P2Y6 receptors antagonist MRS2578 on the antiallodynic effects and improvement of pathological neuropathic pain-induced oxidative stress, thereby finding a potential therapeutic target in neurological disease. MATERIALS AND METHODS: The mechanical allodynia in the ipsilateral spinal dorsal horn (SDH) of rats was observed in rats after chronic constriction injury (CCI). Meanwhile, the messenger RNA (mRNA) levels of biological parameters, including superoxide dismutase (SOD), glutathione (GSH), and heme oxygenase-1 (HO-1) in the SDH of rats were measured by real-time polymerase chain reaction (RT-PCR). In addition, the mRNA expression and protein levels of P2Y6 were measured by RT-PCR and Western blot assay, respectively. Next, the rats subjected to CCI were intrathecally infused with MRS2578 to block the expression of P2Y6 receptors. The positive expression of P2Y6 receptors was examined by immunohistochemistry. RESULTS: In the present study, the results revealed that the P2Y6 expression in the ipsilateral SDH of CCI rats was significantly upregulated. In addition, inhibition of the P2Y6 receptor in SDH increased CCI-induced tactile allodynia. Furthermore, the levels of SOD, GSH, and HO-1 which were correlated with oxidative stress produced by CCI were also decreased. CONCLUSION: The results demonstrated that inhibition of the P2Y6 receptor can generate antiallodynic effects and improved the pathological neuropathic pain-induced oxidative stress. Thus, this study provides a potential approach for the therapy of neurological disease.


Subject(s)
Analgesics/pharmacology , Hyperalgesia/drug therapy , Isothiocyanates/pharmacology , Neuralgia/drug therapy , Purinergic Antagonists/pharmacology , Receptors, Purinergic P2/genetics , Thiourea/analogs & derivatives , Animals , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , Glutathione/metabolism , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Hyperalgesia/genetics , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Injections, Spinal , Ligation , Neuralgia/genetics , Neuralgia/metabolism , Neuralgia/physiopathology , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2/metabolism , Sciatic Nerve/injuries , Sciatic Nerve/metabolism , Spinal Cord Dorsal Horn/drug effects , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Thiourea/pharmacology
14.
Adv Exp Med Biol ; 1201: 275-353, 2019.
Article in English | MEDLINE | ID: mdl-31898792

ABSTRACT

Extracellular purines exert several functions in physiological and pathophysiological mechanisms. ATP acts through P2 receptors as a neurotransmitter and neuromodulator and modulates heart contractility, while adenosine participates in neurotransmission, blood pressure, and many other mechanisms. Because of their capability to differentiate into mature cell types, they provide a unique therapeutic strategy for regenerating damaged tissue, such as in cardiovascular and neurodegenerative diseases. Purinergic signaling is pivotal for controlling stem cell differentiation and phenotype determination. Proliferation, differentiation, and apoptosis of stem cells of various origins are regulated by purinergic receptors. In this chapter, we selected neurodegenerative and cardiovascular diseases with clinical trials using cell therapy and purinergic receptor targeting. We discuss these approaches as therapeutic alternatives to neurodegenerative and cardiovascular diseases. For instance, promising results were demonstrated in the utilization of mesenchymal stem cells and bone marrow mononuclear cells in vascular regeneration. Regarding neurodegenerative diseases, in general, P2X7 and A2A receptors mostly worsen the degenerative state. Stem cell-based therapy, mainly through mesenchymal and hematopoietic stem cells, showed promising results in improving symptoms caused by neurodegeneration. We propose that purinergic receptor activity regulation combined with stem cells could enhance proliferative and differentiation rates as well as cell engraftment.


Subject(s)
Cardiovascular Diseases/therapy , Neurodegenerative Diseases/therapy , Purinergic Antagonists/therapeutic use , Receptors, Purinergic/metabolism , Signal Transduction/drug effects , Stem Cell Transplantation , Adenosine/metabolism , Adenosine Triphosphate/metabolism , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Humans , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Purinergic Antagonists/pharmacology
16.
J Immunol ; 195(2): 651-60, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26078273

ABSTRACT

High concentrations of extracellular ATP (eATP) resulting from cell damage may be found during an ischemia/reperfusion (I/R) episode at the site of injury. eATP activates purinergic receptors in dendritic cells (DCs) and may inhibit inflammation. This immunosuppressive activity could be of interest in the field of I/R, which is an inflammatory condition involved in myocardial infarction, stroke, and solid organ transplantation. However, the specific purinergic receptor responsible for this effect remains to be identified. In this study, we report that eATP induced maturation of human monocyte-derived DCs. Additionally, eATP inhibited IL-12 production whereas IL-10 levels remained unchanged in activated DCs. These effects were prevented by the P2Y11R antagonist NF340. Interestingly, a 5-h hypoxia prevented the effects of eATP on cytokine production whereas a 1-h hypoxia did not affect the eATP-mediated decrease of IL-12 and IL-6. We showed a time-dependent downregulation of P2Y11R at both mRNA and protein levels that was prevented by knocking down hypoxia-inducible factor-1α. In this study, we showed an immunosuppressive role of P2Y11R in human DCs. Additionally, we demonstrated that the time-dependent downregulation of P2Y11R by hypoxia orientates DCs toward a proinflammatory phenotype that may be involved in post-I/R injuries as observed after organ transplantation.


Subject(s)
Dendritic Cells/immunology , Oxygen/pharmacology , Receptors, Purinergic P2/immunology , Adenosine Triphosphate/pharmacology , Cell Hypoxia , Dendritic Cells/cytology , Dendritic Cells/drug effects , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , Immunosuppression Therapy , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-12/antagonists & inhibitors , Interleukin-12/genetics , Interleukin-12/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Primary Cell Culture , Purinergic Antagonists/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , Receptors, Purinergic P2/genetics , Signal Transduction , Time Factors , Transcription, Genetic
17.
Adv Exp Med Biol ; 1051: 1-6, 2017.
Article in English | MEDLINE | ID: mdl-28224484

ABSTRACT

In this Introduction to the series of papers that follow about purinergic receptors, there is a brief history of the discovery of purinergic signalling, the identity of purinoceptors and the current recognition of P1, P2X and P2Y subtypes. An account of key functions mediated by purinoceptors follows, including examples of both short-term and long-term (trophic) signalling and a table showing the selective agonists and antagonists for the purinoceptor subtypes. References to evolution and roles of purinoceptors in pathological conditions are also presented.


Subject(s)
Purinergic Agonists/therapeutic use , Purinergic Antagonists/therapeutic use , Receptors, Purinergic/metabolism , Signal Transduction/drug effects , Animals , Humans
18.
Adv Exp Med Biol ; 906: 325-350, 2017.
Article in English | MEDLINE | ID: mdl-27628008

ABSTRACT

Aspirin and P2Y12 receptor antagonists are widely used across the spectrum of cardiovascular diseases. Upper gastrointestinal complications, including ulcer and bleeding, are relatively common during antiplatelet treatment and, therefore, concomitant proton pump inhibitor (PPI) treatment is often prescribed.PPIs provide gastroprotection by changing the intragastric milieu, essentially by raising intragastric pH. In recent years, it has been heavily discussed whether PPIs may reduce the cardiovascular protection by aspirin and, even more so, clopidogrel. Pharmacodynamic and pharmacokinetic studies suggested an interaction between PPIs and clopidogrel, and subsequent clinical studies were conducted to evaluate the clinical impact of this interaction. More recently, it was reported that PPIs may also attenuate the antiplatelet effect of aspirin. This may be clinically important, because a fixed combination of aspirin and a PPI (esomeprazole) has recently been approved and because aspirin is the most widely used drug in patients with cardiovascular disease. The antiplatelet effect of the new P2Y12 receptor antagonists, ticagrelor and prasugrel, seems less influenced by PPI co-treatment.Given the large number of patients treated with antithrombotic drugs and PPIs, even a minor reduction of platelet inhibition potentially carries considerable clinical impact. The present book chapter summarizes the evidence regarding the widespread use of platelet inhibitors and PPIs in combination. Moreover, it outlines current evidence supporting or opposing drug interactions between these drugs and discusses clinical implications.


Subject(s)
Aspirin/pharmacokinetics , Cardiovascular Diseases/drug therapy , Esomeprazole/pharmacokinetics , Proton Pump Inhibitors/pharmacokinetics , Purinergic Antagonists/pharmacokinetics , Ticlopidine/analogs & derivatives , Adenosine/analogs & derivatives , Adenosine/therapeutic use , Aspirin/blood , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Clopidogrel , Drug Administration Schedule , Drug Dosage Calculations , Drug Interactions , Esomeprazole/blood , Gastrointestinal Hemorrhage/chemically induced , Gastrointestinal Hemorrhage/metabolism , Gastrointestinal Hemorrhage/pathology , Gene Expression , Humans , Hydrogen-Ion Concentration/drug effects , Peptic Ulcer/chemically induced , Peptic Ulcer/metabolism , Peptic Ulcer/pathology , Prasugrel Hydrochloride/therapeutic use , Proton Pump Inhibitors/blood , Purinergic Antagonists/blood , Receptors, Purinergic P2Y12/genetics , Receptors, Purinergic P2Y12/metabolism , Ticagrelor , Ticlopidine/blood , Ticlopidine/pharmacokinetics
19.
Article in English | MEDLINE | ID: mdl-28943320

ABSTRACT

The chemoreceptors involved in oxygen sensing in teleost fish are neuroepithelial cells (NECs) in the gills, and are analogous to glomus cells in the mammalian carotid body. Purinergic signalling mechanisms involving the neurotransmitters, ATP and adenosine, have been identified in mediating hypoxic signalling in the carotid body, but these pathways are not well understood in the fish gill. The present study used a behavioural assay to screen for the effects of drugs, that target purinergic and adenosine receptors, on the hyperventilatory response to hypoxia in larval zebrafish (Danio rerio) in order to determine if the receptors on which these drugs act may be involved in hypoxic signalling. The purinergic receptor antagonist, PPADS, targets purinergic P2X2/3 receptors and inhibited the hyperventilatory response to hypoxia (IC50=18.9µM). The broad-spectrum purinergic agonist, ATPγS, elicited a hyperventilatory response (EC50=168µM). The non-specific adenosine receptor antagonist, caffeine, inhibited the hyperventilatory response to hypoxia, as did the specific A2a receptor antagonist, SCH58261 (IC50=220nM). These results suggest that P2X2/3 and A2a receptors are candidates for mediating hypoxic hyperventilation in zebrafish. This study highlights the potential of applying chemical screening to ventilatory behaviour in zebrafish to further our understanding of the pathways involved in signalling by gill NECs and oxygen sensing in vertebrates.


Subject(s)
Gills/drug effects , Hyperventilation/prevention & control , Hypoxia/physiopathology , Purinergic Antagonists/pharmacology , Signal Transduction/drug effects , Zebrafish Proteins/antagonists & inhibitors , Zebrafish/metabolism , Animals , Behavior, Animal/drug effects , Biological Assay , Drug Evaluation, Preclinical , Gills/growth & development , Gills/metabolism , Hyperventilation/etiology , Hyperventilation/metabolism , Kinetics , Larva/drug effects , Larva/growth & development , Larva/metabolism , Microscopy, Video , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Pulmonary Ventilation/drug effects , Receptor, Adenosine A2A/chemistry , Receptor, Adenosine A2A/metabolism , Receptors, Purinergic P2X2/chemistry , Receptors, Purinergic P2X2/metabolism , Receptors, Purinergic P2X3/chemistry , Receptors, Purinergic P2X3/metabolism , Reproducibility of Results , Zebrafish/embryology , Zebrafish Proteins/metabolism
20.
Molecules ; 22(4)2017 Apr 23.
Article in English | MEDLINE | ID: mdl-28441750

ABSTRACT

The inhibitory adenosine A1 receptor (A1R) and excitatory A2A receptor (A2AR) are predominantly expressed in the brain. Whereas the A2AR has been implicated in normal aging and enhancing neurotoxicity in multiple neurodegenerative diseases, the inhibitory A1R has traditionally been ascribed to have a neuroprotective function in various brain insults. This review provides a summary of the emerging role of prolonged A1R signaling and its potential cross-talk with A2AR in the cellular basis for increased neurotoxicity in neurodegenerative disorders. This A1R signaling enhances A2AR-mediated neurodegeneration, and provides a platform for future development of neuroprotective agents in stroke, Parkinson's disease and epilepsy.


Subject(s)
Brain/metabolism , Neurodegenerative Diseases/metabolism , Receptor, Adenosine A1/physiology , Receptor, Adenosine A2A/physiology , Animals , Brain/pathology , Humans , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/pharmacology , Purinergic Agonists/pharmacology , Purinergic Antagonists/pharmacology , Receptor Cross-Talk
SELECTION OF CITATIONS
SEARCH DETAIL