Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Purinergic Signal ; 20(1): 83-89, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37074620

ABSTRACT

ATP is a ubiquitous extracellular messenger released in a wide number of pathophysiological conditions. ATP is known to be present in minute amounts in the extracellular space in healthy tissues and in the blood, and to modulate a multiplicity of cell responses. Cell culture systems are widely used to explore purinergic signaling. We show here that currently used fetal bovine sera contain ATP in the 300-1300 pmol/L range. Serum ATP is associated with albumin as well as with microparticle/microvesicle fraction. Serum microparticles/microvesicles affect in vitro cell responses due to their content of miRNAs, growth factors, and other bioactive molecules. ATP is likely to be one of these bioactive factors found in a variable amount in sera of different commercial sources. ATP in serum supports ATP-dependent biochemical reactions such as the hexokinase-dependent phosphorylation of glucose to glucose 6-phosphate, and affects purinergic signaling. These findings show that cells growing in vitro in serum-supplemented media are exposed to varying levels of extracellular ATP, and thus to varying degrees of purinergic stimulation.


Subject(s)
Extracellular Space , Serum Albumin, Bovine , Cells, Cultured , Extracellular Space/metabolism , Adenosine Triphosphate/metabolism , Glucose
2.
Int J Mol Sci ; 24(11)2023 May 30.
Article in English | MEDLINE | ID: mdl-37298415

ABSTRACT

The discovery of the P2X7 receptor (P2X7R, originally named P2Z) in immune cells, its cloning, and the identification of its role in a multiplicity of immune-mediated diseases raised great hopes for the development of novel and more potent anti-inflammatory medicaments. Unfortunately, such hopes were partially deluded by the unsatisfactory results of most early clinical trials. This failure substantially reduced the interest of the pharmaceutical and biotech industries in the clinical development of P2X7R-targeted therapies. However, recent findings ushered in a second life for the P2X7R in diagnostic medicine. New P2X7R radioligands proved to be very reliable tools for the diagnosis of neuroinflammation in preclinical and clinical studies, and detection and measurement of free P2X7 receptor (or P2X7 subunit) in human blood suggested its potential use as a circulating marker of inflammation. Here we provide a brief review of these novel developments.


Subject(s)
Inflammation , Receptors, Purinergic P2X7 , Humans , Inflammation/diagnosis , Inflammation/drug therapy , Anti-Inflammatory Agents/therapeutic use , Radiopharmaceuticals/therapeutic use
3.
Front Cell Dev Biol ; 11: 1180774, 2023.
Article in English | MEDLINE | ID: mdl-37215083

ABSTRACT

Identifying the subcellular localization of a protein within a cell is often an essential step in understanding its function. The main objective of this report was to determine the presence of the P2X7 receptor (P2X7R) in healthy human cells of skeletal system, specifically osteoblasts (OBs), chondrocytes (Chs) and intervertebral disc (IVD) cells. This receptor is a member of the ATP-gated ion channel family, known to be a main sensor of extracellular ATP, the prototype of the danger signal released at sites of tissue damage, and a ubiquitous player in inflammation and cancer, including bone and cartilaginous tissues. Despite overwhelming data supporting a role in immune cell responses and tumor growth and progression, a complete picture of the pathophysiological functions of P2X7R, especially when expressed by non-immune cells, is lacking. Here we show that human wild-type P2X7R (P2X7A) was expressed in different samples of human osteoblasts, chondrocytes and intervertebral disc cells. By fluorescence microscopy (LM) and immunogold transmission electron microscopy we localized P2X7R not only in the canonical sites (plasma membrane and cytoplasm), but also in the nucleus of all the 3 cell types, especially IVD cells and OBs. P2X7R mitochondrial immunoreactivity was predominantly detected in OBs and IVD cells, but not in Chs. Evidence of subcellular localization of P2X7R may help to i. understand the participation of P2X7R in as yet unidentified signaling pathways in the joint and bone microenvironment, ii. identify pathologies associated with P2X7R mislocalization and iii. design specific targeted therapies.

4.
Front Immunol ; 14: 1182454, 2023.
Article in English | MEDLINE | ID: mdl-37215142

ABSTRACT

Introduction: The pathophysiology of the Corona Virus Disease 2019 (COVID-19) is incompletely known. A robust inflammatory response caused by viral replication is a main cause of the acute lung and multiorgan injury observed in critical patients. Inflammasomes are likely players in COVID-19 pathogenesis. The P2X7 receptor (P2X7R), a plasma membrane ATP-gated ion channel, is a main activator of the NLRP3 inflammasome, of the ensuing release of inflammatory cytokines and of cell death by pyroptosis. The P2X7R has been implicated in COVID-19-dependent hyperinflammation and in the associated multiorgan damage. Shed P2X7R (sP2X7R) and shed NLRP3 (sNLRP3) have been detected in plasma and other body fluids, especially during infection and inflammation. Methods: Blood samples from 96 patients with confirmed SARS-CoV-2 infection with various degrees of disease severity were tested at the time of diagnosis at hospital admission. Standard haematological parameters and IL-6, IL-10, IL-1ß, sP2X7R and sNLRP3 levels were measured, compared to reference values, statistically validated, and correlated to clinical outcome. Results: Most COVID-19 patients included in this study had lymphopenia, eosinopenia, neutrophilia, increased inflammatory and coagulation indexes, and augmented sNLRP3, IL-6 and IL-10 levels. Blood concentration of sP2X7R was also increased, and significantly positively correlated with lymphopenia, procalcitonin (PCT), IL-10, and alanine transaminase (ALT). Patients with increased sP2X7R levels at diagnosis also showed fever and respiratory symptoms, were more often transferred to Pneumology division, required mechanical ventilation, and had a higher likelihood to die during hospitalization. Conclusion: Blood sP2X7R was elevated in the early phases of COVID-19 and predicted an adverse clinical outcome. It is suggested that sP2X7R might be a useful marker of disease progression.


Subject(s)
COVID-19 , Lymphopenia , Humans , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Interleukin-10/metabolism , Receptors, Purinergic P2X7 , Interleukin-6/metabolism , SARS-CoV-2/metabolism , Inflammasomes/metabolism
5.
Neuropharmacology ; 224: 109333, 2023 02 15.
Article in English | MEDLINE | ID: mdl-36400278

ABSTRACT

Nucleotides play a crucial role in extracellular signaling across species boundaries. All the three kingdoms of life (Bacteria, Archea and Eukariota) are responsive to extracellular ATP (eATP) and many release this and other nucleotides. Thus, eATP fulfills different functions, many related to danger-sensing or avoidance reactions. Basically all living organisms have evolved sensors for eATP and other nucleotides with very different affinity and selectivity, thus conferring a remarkable plasticity to this signaling system. Likewise, different intracellular transduction systems were associated during evolution to different receptors for eATP. In mammalian evolution, control of intracellular ATP (iATP) and eATP homeostasis has been closely intertwined with that of Ca2+, whether in the extracellular milieu or in the cytoplasm, establishing an inverse reciprocal relationship, i.e. high extracellular Ca2+ levels are associated to negligible eATP, while low intracellular Ca2+ levels are associated to high eATP concentrations. This inverse relationship is crucial for the messenger functions of both molecules. Extracellular ATP is sensed by specific plasma membrane receptors of widely different affinity named P2 receptors (P2Rs) of which 17 subtypes are known. This confers a remarkable plasticity to P2R signaling. The central nervous system (CNS) is a privileged site for purinergic signaling as all brain cell types express P2Rs. Accruing evidence suggests that eATP, in addition to participating in synaptic transmission, also plays a crucial homeostatic role by fine tuning microglia, astroglia and oligodendroglia responses. Drugs modulating the eATP concentration in the CNS are likely to be the new frontier in the therapy of neuroinflammation. This article is part of the Special Issue on 'Purinergic Signaling: 50 years'.


Subject(s)
Adenosine Triphosphate , Brain , Animals , Adenosine Triphosphate/metabolism , Cell Membrane/metabolism , Brain/metabolism , Nucleotides , Inflammation Mediators , Mammals/metabolism
6.
Methods Mol Biol ; 2510: 53-63, 2022.
Article in English | MEDLINE | ID: mdl-35776319

ABSTRACT

For many years the P2X7 receptor (P2X7R) was considered the prototypic cytolytic receptor due to its ability to cause dramatic changes in plasma membrane permeability, eventually leading to cell death. However, later studies revealed that controlled P2X7R activation has beneficial effects on cell metabolism and nowadays our perception of the physiological role of this receptor has radically changed. Some of the biochemical pathways underlying the trophic effect of the P2X7R are being unveiled, thus disclosing an unanticipated role of P2X7Rs in mitochondrial and glycolytic metabolism. We provide here an update of the effects of the P2X7R on cell energy metabolism.


Subject(s)
Glycolysis , Receptors, Purinergic P2X7 , Cell Death/physiology , Cell Membrane Permeability , Mitochondria , Receptors, Purinergic P2X7/genetics
7.
Biomolecules ; 12(7)2022 07 14.
Article in English | MEDLINE | ID: mdl-35883539

ABSTRACT

Extracellular ATP (eATP) and P2 receptors are novel emerging regulators of T-lymphocyte responses. Cellular ATP is released via multiple pathways and accumulates at sites of tissue damage and inflammation. P2 receptor expression and function are affected by numerous single nucleotide polymorphisms (SNPs) associated with diverse disease conditions. Stimulation by released nucleotides (purinergic signalling) modulates several T-lymphocyte functions, among which energy metabolism. Energy metabolism, whether oxidative or glycolytic, in turn deeply affects T-cell activation, differentiation and effector responses. Specific P2R subtypes, among which the P2X7 receptor (P2X7R), are either up- or down-regulated during T-cell activation and differentiation; thus, they can be considered indexes of activation/quiescence, reporters of T-cell metabolic status and, in principle, markers of immune-mediated disease conditions.


Subject(s)
Adenosine Triphosphate , Signal Transduction , Adenosine Triphosphate/metabolism , Humans , Inflammation/metabolism , Lymphocyte Activation , T-Lymphocytes
8.
Molecules ; 27(6)2022 Mar 11.
Article in English | MEDLINE | ID: mdl-35335211

ABSTRACT

Adenosine triphosphate (ATP) is the key energy intermediate of cellular metabolic processes and a ubiquitous extracellular messenger. As an extracellular messenger, ATP acts at plasma membrane P2 receptors (P2Rs). The levels of extracellular ATP (eATP) are set by both passive and active release mechanisms and degradation processes. Under physiological conditions, eATP concentration is in the low nanomolar range but can rise to tens or even hundreds of micromoles/L at inflammatory sites. A dysregulated eATP homeostasis is a pathogenic factor in several chronic inflammatory diseases, including type 2 diabetes mellitus (T2DM). T2DM is characterized by peripheral insulin resistance and impairment of insulin production from pancreatic ß-cells in a landscape of systemic inflammation. Although various hypoglycemic drugs are currently available, an effective treatment for T2DM and its complications is not available. However, counteracting systemic inflammation is anticipated to be beneficial. The postulated eATP increase in T2DM is understood to be a driver of inflammation via P2X7 receptor (P2X7R) activation and the release of inflammatory cytokines. Furthermore, P2X7R stimulation is thought to trigger apoptosis of pancreatic ß-cells, thus further aggravating hyperglycemia. Targeting eATP and the P2X7R might be an appealing novel approach to T2DM therapy.


Subject(s)
Diabetes Mellitus, Type 2 , Adenosine Triphosphate/metabolism , Cytokines , Humans , Inflammation/metabolism , Signal Transduction
9.
Biochim Biophys Acta Mol Cell Res ; 1869(5): 119237, 2022 05.
Article in English | MEDLINE | ID: mdl-35150807

ABSTRACT

Nucleotides are released from all cells through regulated pathways or as a result of plasma membrane damage or cell death. Outside the cell, nucleotides act as signalling molecules triggering multiple responses via specific plasma membrane receptors of the P2 family. In the nervous system, purinergic signalling has a key function in neurotransmission. Outside the nervous system, purinergic signalling is one of the major modulators of basal tissue homeostasis, while its dysregulation contributes to the pathogenesis of various disease, including inflammation and cancer. Pre-clinical and clinical evidence shows that selective P2 agonists or antagonists are effective treatments for many pathologies, thus highlighting the relevance of extracellular nucleotides and P2 receptors as therapeutic targets.


Subject(s)
Nucleotides/metabolism , Signal Transduction , Animals , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Cell Membrane/metabolism , Humans , Lung Diseases/metabolism , Lung Diseases/pathology , Neoplasms/metabolism , Neoplasms/pathology , Nucleotides/analysis , Receptors, Purinergic P2X/metabolism , Receptors, Purinergic P2Y/metabolism
10.
Theranostics ; 12(2): 859-874, 2022.
Article in English | MEDLINE | ID: mdl-34976217

ABSTRACT

Rationale: Caloric restriction improves the efficacy of anti-cancer therapy. This effect is largely dependent on the increase of the extracellular ATP concentration in the tumor microenvironment (TME). Pathways for ATP release triggered by nutrient deprivation are largely unknown. Methods: The extracellular ATP (eATP) concentration was in vivo measured in the tumor microenvironment of B16F10-inoculated C57Bl/6 mice with the pmeLuc probe. Alternatively, the pmeLuc-TG-mouse was used. Caloric restriction was in vivo induced with hydroxycitrate (HC). B16F10 melanoma cells or CT26 colon carcinoma cells were in vitro exposed to serum starvation to mimic nutrient deprivation. Energy metabolism was monitored by Seahorse. Microparticle release was measured by ultracentrifugation and by Nanosight. Results: Nutrient deprivation increases eATP release despite the dramatic inhibition of intracellular energy synthesis. Under these conditions oxidative phosphorylation was dramatically impaired, mitochondria fragmented and glycolysis and lactic acid release were enhanced. Nutrient deprivation stimulated a P2X7-dependent release of ATP-loaded, mitochondria-containing, microparticles as well as of naked mitochondria. Conclusions: Nutrient deprivation promotes a striking accumulation of eATP paralleled by a large release of ATP-laden microparticles and of naked mitochondria. This is likely to be a main mechanism driving the accumulation of eATP into the TME.


Subject(s)
Adenosine Triphosphate/metabolism , Cell-Derived Microparticles/metabolism , Neoplasms/metabolism , Animals , Caloric Restriction , Cell-Derived Microparticles/drug effects , Citrates/pharmacology , Colonic Neoplasms/metabolism , Extracellular Space/metabolism , Male , Melanoma/metabolism , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Nutrients , Tumor Cells, Cultured
12.
Function (Oxf) ; 2(2): zqab005, 2021.
Article in English | MEDLINE | ID: mdl-35330818

ABSTRACT

Basal expression of the P2X7 receptor (P2X7R) improves mitochondrial metabolism, Adenosine 5'-triphosphate (ATP) synthesis, and overall fitness of immune and non-immune cells. We investigated P2X7R contribution to energy metabolism and subcellular localization in fibroblasts (mouse embryo fibroblasts and HEK293 human fibroblasts), mouse microglia (primary brain microglia, and the N13 microglia cell line), and heart tissue. The P2X7R localizes to mitochondria, and its lack (1) decreases basal respiratory rate, ATP-coupled respiration, maximal uncoupled respiration, resting mitochondrial potential, mitochondrial matrix Ca2+ level, (2) modifies expression pattern of oxidative phosphorylation enzymes, and (3) severely affects cardiac performance. Hearts from P2rx7-deleted versus wild-type mice are larger, heart mitochondria smaller, and stroke volume, ejection fraction, fractional shortening, and cardiac output, are significantly decreased. Accordingly, the physical fitness of P2X7R-null mice is severely reduced. Thus, the P2X7R is a key modulator of mitochondrial energy metabolism and a determinant of physical fitness.


Subject(s)
Adenosine Triphosphate , Receptors, Purinergic P2X7 , Animals , Humans , Mice , Energy Metabolism , HEK293 Cells , Physical Functional Performance , Receptors, Purinergic P2X7/genetics
13.
Biochem Pharmacol ; 187: 114350, 2021 05.
Article in English | MEDLINE | ID: mdl-33253643

ABSTRACT

It is increasingly appreciated that ion channels have a crucial role in tumors, either as promoters of cancer cell growth, or modulators of immune cell functions, or both. Among ion channels, P2X receptors have a special status because they are gated by ATP, a common and abundant component of the tumor microenvironment. Furthermore, one P2X receptor, i.e. P2X7, may also function as a conduit for ATP release, thus fuelling the increased extracellular ATP level in the tumor interstitium. These findings show that P2X receptors and extracellular ATP are indissoluble partners and key regulators of tumor growth, and suggest the exploitation of the extracellular ATP-P2X partnership to develop innovative therapeutic approaches to cancer.


Subject(s)
Disease Progression , Neoplasms/metabolism , Receptors, Purinergic P2X/metabolism , Tumor Microenvironment/physiology , Adenosine Triphosphate/immunology , Adenosine Triphosphate/metabolism , Animals , Humans , Neoplasms/drug therapy , Neoplasms/immunology , Purinergic P2X Receptor Agonists/administration & dosage , Purinergic P2X Receptor Antagonists/administration & dosage , Receptors, Purinergic P2X/immunology , Tumor Microenvironment/drug effects
14.
Cells ; 9(11)2020 11 17.
Article in English | MEDLINE | ID: mdl-33212982

ABSTRACT

Adenosine triphosphate (ATP) is one of the main biochemical components of the tumor microenvironment (TME), where it can promote tumor progression or tumor suppression depending on its concentration and on the specific ecto-nucleotidases and receptors expressed by immune and cancer cells. ATP can be released from cells via both specific and nonspecific pathways. A non-regulated release occurs from dying and damaged cells, whereas active release involves exocytotic granules, plasma membrane-derived microvesicles, specific ATP-binding cassette (ABC) transporters and membrane channels (connexin hemichannels, pannexin 1 (PANX1), calcium homeostasis modulator 1 (CALHM1), volume-regulated anion channels (VRACs) and maxi-anion channels (MACs)). Extracellular ATP acts at P2 purinergic receptors, among which P2X7R is a key mediator of the final ATP-dependent biological effects. Over the years, P2 receptor- or ecto-nucleotidase-targeting for cancer therapy has been proposed and actively investigated, while comparatively fewer studies have explored the suitability of TME ATP as a target. In this review, we briefly summarize the available evidence suggesting that TME ATP has a central role in determining tumor fate and is, therefore, a suitable target for cancer therapy.


Subject(s)
Adenosine Triphosphate/therapeutic use , Neoplasms/therapy , Adenosine Triphosphate/pharmacology , Humans , Signal Transduction , Tumor Microenvironment
15.
Methods Enzymol ; 629: 115-150, 2019.
Article in English | MEDLINE | ID: mdl-31727237

ABSTRACT

The P2X7 receptor [P2X7R or P2RX7 in National Center for Biotechnology Information (NCBI) gene nomenclature] is a member of the P2X receptor (P2XR) subfamily of P2 receptors (P2Rs). The P2X7R is an extracellular ATP-gated ion channel with peculiar permeability properties expressed by most cell types, mainly in the immune system, where it has a leading role in cytokine release, oxygen radical generation, T lymphocyte differentiation and proliferation. A role in cancer cell growth and tumor progression has also been demonstrated. These features make the P2X7R an appealing target for drug development in inflammation and cancer. The functional P2X7R, recently (partially) crystallized and 3-D solved, is formed by the assembly of three identical subunits (homotrimer). The P2X7R is preferentially permeable to small cations (Ca2+, Na+, K+), and in most (but not all) cell types also to large positively charged molecules of molecular mass up to 900Da. Permeability to negatively charged species of comparable molecular mass (e.g., Lucifer yellow) is debated. Several highly selective P2X7R pharmacological blockers have been developed over the years, thus providing powerful tools for P2X7R studies. Biophysical properties and coupling to several different physiological responses make the P2X7R amenable to investigation by electrophysiology and cell biology techniques, which allow its identification and characterization in many different cell types and tissues. A careful description of the physiological features of the P2X7R is a prerequisite for an effective therapeutic development. Here we describe the most common techniques to asses P2X7R functions, including patch-clamp, intracellular calcium measurements, and membrane permeabilization to large fluorescent dyes in a selection of different cell types. In addition, we also describe common toxicity assays used to verify the effects of P2X7R stimulation on cell viability.


Subject(s)
Drug Screening Assays, Antitumor/methods , Receptors, Purinergic P2X7/analysis , Adenosine Triphosphate/immunology , Adenosine Triphosphate/metabolism , Allosteric Regulation , Animals , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/immunology , Cell Membrane/metabolism , Cell Membrane Permeability/drug effects , Cell Membrane Permeability/immunology , Drug Design , Fluorescent Dyes/chemistry , Fluorescent Dyes/pharmacology , Gene Knockout Techniques/instrumentation , Gene Knockout Techniques/methods , HEK293 Cells , Humans , Microscopy, Fluorescence/methods , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/pathology , Patch-Clamp Techniques/instrumentation , Patch-Clamp Techniques/methods , Primary Cell Culture , Purinergic P2X Receptor Antagonists/pharmacology , Purinergic P2X Receptor Antagonists/therapeutic use , Receptors, Purinergic P2X7/chemistry , Receptors, Purinergic P2X7/genetics , Receptors, Purinergic P2X7/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Single-Cell Analysis/methods , Structure-Activity Relationship
16.
Sci Rep ; 9(1): 6475, 2019 04 24.
Article in English | MEDLINE | ID: mdl-31019207

ABSTRACT

Previous data from our laboratory show that expression of the P2X7 receptor (P2X7R) is needed for amyloid ß (Aß)-stimulated microglia activation and IL-1ß release in vitro and in vivo. We also showed that Aß-dependent stimulation is inhibited by the dihydropyridine nimodipine at an intracellular site distal to the P2X7R. In the present study, we used the N13 microglia cell line and mouse primary microglia from wt and P2rx7-deleted mice to test the effect of nimodipine on amyloid ß (Aß)-dependent NLRP3 inflammasome expression and function, and on mitochondrial energy metabolism. Our data show that in microglia Aß causes P2X7R-dependent a) NFκB activation; b) NLRP3 inflammasome expression and function; c) mitochondria toxicity; and these changes are fully inhibited by nimodipine. Our study shows that nimodipine is a powerful blocker of cell damage caused by monomeric and oligomeric Aß, points to the mitochondria as a crucial target, and underlines the permissive role of the P2X7R.


Subject(s)
Amyloid beta-Peptides/pharmacology , Microglia/drug effects , Mitochondria/drug effects , Nimodipine/pharmacology , Receptors, Purinergic P2X7/metabolism , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Animals , Calcium Channel Blockers/pharmacology , Cell Line , Cells, Cultured , Female , Inflammasomes/drug effects , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Mitochondria/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Receptors, Purinergic P2X7/genetics
17.
Front Immunol ; 10: 793, 2019.
Article in English | MEDLINE | ID: mdl-31031771

ABSTRACT

The P2X7 receptor (P2X7R) is a key pro-inflammatory plasma membrane receptor responsible for NLRP3 inflammasome activation and IL-1ß release. Various inflammatory plasma membrane receptors (e.g., IL-1 type I receptor, TNF type I and II receptors, IL-2 receptor) are shed under different pathophysiological conditions. In the present study, we show that the full length P2X7R is released into circulation in patients as well as in healthy subjects. Blood levels of shed P2X7R (sP2X7R) correlate to those of the inflammatory marker C reactive protein (CRP). Blood sP2X7R ranged from 16.74 to 82.17 ng/L, mean ± SE 40.97 ± 3.82 (n = 26) in healthy subjects, from 33.1 to 484.0 ng/L, mean ± SE 114.78 ± 12.22 (n = 45) in patients with CRP <3 mg/L, and from 63.65 to 1092.3 ng/L, mean ± SE 204.2 ± 30.94 (n = 42) in patients with CRP >3 mg/L. sP2X7R in plasma was largely associated to microvesicles/microparticles. Peripheral blood monocytes from healthy subjects released sP2X7R upon stimulation with the semi-selective P2X7R agonist benzoyl ATP. These data show that the P2X7R can be released into circulation, and that its blood levels increase in various disease conditions.


Subject(s)
Receptors, Purinergic P2X7/blood , Adenosine Triphosphate/metabolism , Adult , Aged , Blood Vessels/metabolism , C-Reactive Protein/metabolism , Cytokines/metabolism , Female , Humans , Macrophages/immunology , Macrophages/metabolism , Male , Middle Aged , Receptors, Purinergic P2X7/metabolism , Reproducibility of Results
18.
Front Pharmacol ; 9: 39, 2018.
Article in English | MEDLINE | ID: mdl-29449813

ABSTRACT

The P2X7 receptor (P2X7R) is a ligand-gated plasma membrane ion channel belonging to the P2X receptor subfamily activated by extracellular nucleotides. General consensus holds that the physiological (and maybe the only) agonist is ATP. However, scattered evidence generated over the last several years suggests that ATP might not be the only agonist, especially at inflammatory sites. Solid data show that NAD+ covalently modifies the P2X7R of mouse T lymphocytes, thus lowering the ATP threshold for activation. Other structurally unrelated agents have been reported to activate the P2X7R via a poorly understood mechanism of action: (a) the antibiotic polymyxin B, possibly a positive allosteric P2X7R modulator, (b) the bactericidal peptide LL-37, (c) the amyloidogenic ß peptide, and (d) serum amyloid A. Some agents, such as Alu-RNA, have been suggested to activate the P2X7R acting on the intracellular N- or C-terminal domains. Mode of P2X7R activation by these non-nucleotide ligands is as yet unknown; however, these observations raise the intriguing question of how these different non-nucleotide ligands may co-operate with ATP at inflammatory or tumor sites. New information obtained from the cloning and characterization of the P2X7R from exotic mammalian species (e.g., giant panda) and data from recent patch-clamp studies are strongly accelerating our understanding of P2X7R mode of operation, and may provide hints to the mechanism of activation of P2X7R by non-nucleotide ligands.

SELECTION OF CITATIONS
SEARCH DETAIL
...