17 research outputs found

    Unorthodox localization of P2X7 receptor in subcellular compartments of skeletal system cells

    Get PDF
    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

    The shed P2X7 receptor is an index of adverse clinical outcome in COVID-19 patients

    Get PDF
    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-1β, 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

    Diagnostic and prognostic microRNAs in the serum of breast cancer patients measured by droplet digital PCR

    Get PDF
    Background: Breast cancer circulating biomarkers include carcinoembryonic antigen and carbohydrate antigen 15-3, which are used for patient follow-up. Since sensitivity and specificity are low, novel and more useful biomarkers are needed. The presence of stable circulating microRNAs (miRNAs) in serum or plasma suggested a promising role for these tiny RNAs as cancer biomarkers. To acquire an absolute concentration of circulating miRNAs and reduce the impact of preanalytical and analytical variables, we used the droplet digital PCR (ddPCR) technique. Results: We investigated a panel of five miRNAs in the sera of two independent cohorts of breast cancer patients and disease-free controls. The study showed that miR-148b-3p and miR-652-3p levels were significantly lower in the serum of breast cancer patients than that in controls in both cohorts. For these two miRNAs, the stratification of breast cancer patients versus controls was confirmed by receiver operating characteristic curve analyses. In addition, we showed that higher levels of serum miR-10b-5p were associated with clinicobiological markers of poor prognosis. Conclusions: The study revealed the usefulness of the ddPCR approach for the quantification of circulating miRNAs. The use of the ddPCR quantitative approach revealed very good agreement between two independent cohorts in terms of comparable absolute miRNA concentrations and consistent trends of dysregulation in breast cancer patients versus controls. Overall, this study supports the use of the quantitative ddPCR approach for monitoring the absolute levels of diagnostic and prognostic tumor-specific circulating miRNAs

    Extracellular ATP is increased by release of ATP-loaded microparticles triggered by nutrient deprivation

    Get PDF
    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

    P2X7R:role in the generation of an ATP-rich TME driven by nutrient deprivation

    Get PDF
    Extracellular ATP, one of the major constituent of the tumor microenvironment (TME), promotes immunosuppression or anti-tumor immunity. It has been shown that caloric restriction improves the efficacy of anti-cancer therapy by increasing the ATP concentration in the TME. In vivo administration of the caloric restriction mimetic (CRM) hydroxycitrate reduced B16F10 melanoma tumor size and at the same time increased the ATP levels in the TME. We mimicked the in vivo CRM activity by in vitro exposing melanoma cells to serum starvation. Chronic incubation in the absence of serum inhibited melanoma cell growth and increased the extracellular ATP concentration in the absence of autophagy or cell death. The P2X7 receptor (P2X7R) is a main sensor of extracellular ATP and a promoter of cell proliferation or cell death depending on the level of activation. Serum starvation severely affected P2X7R-dependent responses, despite P2X7R expression was unchanged. Cytoplasmic Ca2+ levels and intracellular ATP concentration were strikingly reduced in serum-starved versus serum-supplemented cells. In the absence of serum, mitochondria were fragmented and depolarized, oxidative phosphorylation was dramatically impaired and Complex I and II of the mitochondrial respiratory chain were down-regulated. In parallel glycolysis and release of lactic acid were increased. Melanoma cells are known to release exosomes/microparticles, collectively referred to as extracellular vesicles (EVs). We show that serum-starved cells released a higher amount of EVs compared to serum-supplemented cells. EVs contained a high amount of ATP as well as mitochondria. Moreover P2X7R silencing reduced EV and extracellular ATP release, suggesting that an efficient ATP release mechanism required functional P2X7R expression. Our findings suggest that serum starvation causes an impairment of cancer cell metabolism and support a role for the P2X7R in the mechanism by which nutrient deprivation drives the establishment of an ATP-rich tumor microenvironmentL’ ATP extracellulare, uno dei maggiori componenti del microambiente tumorale, svolge un ruolo fondamentale sia nell’attivazione dell’immunità anti-tumorale che nell’attività immunosoppressiva. La restrizione calorica si è rivelata un mezzo efficace per migliorare l’effetto delle terapie anti-tumorali, grazie alla sua capacità di incrementare la concentrazione di ATP nel microambiente tumorale. La somministrazione in vivo di idrossicitrato, un composto naturale che mima la restrizione calorica, riduce significativamente la dimensione della massa tumorale di melanoma B16F10 e causa l’aumento dei livelli di ATP nel microambiente tumorale. Abbiamo ricreato la restrizione calorica in vitro mantenendo le cellule di melanoma murino in assenza di nutrienti (serum starvation). La deprivazione cronica di nutrienti riduce significativamente la proliferazione cellulare e determina un aumento significativo dei livelli di ATP extracellulare, in assenza di segni evidenti di sofferenza o morte cellulare. L’ATP è il principale ligando del recettore P2X7, un importante recettore purinergico coinvolto nella regolazione della proliferazione e morte cellulare. La deprivazione di nutrienti riduce notevolmente la funzionalità del recettore P2X7 anche se l’espressione proteica di quest’ultimo resta invariata. I livelli di calcio e di ATP intracellulari sono inoltre significativamente ridotti e il metabolismo energetico profondamente compromesso. Nelle cellule mantenute in assenza di nutrienti, i mitocondri appaiono frammentati e depolarizzati, la fosforilazione ossidativa è significativamente inibita e i complessi I e II della catena respiratoria sono down-regolati. In parallelo la glicolisi aerobia e il rilascio di lattato risultano aumentati. Le cellule di melanoma rilasciano esosomi/microparticelle, comunemente denominati vescicole extracellulari (EVs). Abbiamo dimostrato che le cellule di melanoma mantenute in assenza di nutrienti rilasciano una quantità di vescicole maggiore rispetto a quella liberata dalle cellule mantenute in normali condizioni colturali. Queste vescicole contengono un’elevata concentrazione di ATP e mitocondri. Inoltre, il silenziamento del recettore P2X7 riduce significativamente la quantità di vescicole e di ATP extracellulare liberati, suggerendo che un efficiente meccanismo di rilascio di ATP richieda l’espressione del recettore P2X7. I dati ottenuti suggeriscono che la deprivazione di nutrienti causi un alterazione del metabolismo energetico tumorale e che il recettore P2X7 sia coinvolto nella formazione di un microambiente tumorale ricco di ATP guidata dalla deprivazione di nutrienti

    P2 Receptors: Novel Disease Markers and Metabolic Checkpoints in Immune Cells

    Get PDF
    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

    Extracellular ATP: A Feasible Target for Cancer Therapy

    No full text
    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

    Non-nucleotide agonists triggering P2X7 receptor activation and pore formation

    Get PDF
    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 β 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

    Extracellular ATP: A powerful inflammatory mediator in the central nervous system

    No full text
    : 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'

    The Coming of Age of the P2X7 Receptor in Diagnostic Medicine

    No full text
    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
    corecore