Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Function (Oxf) ; 5(4)2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38984997

ABSTRACT

Microparticles (MPs) are secreted by all cells, where they play a key role in intercellular communication, differentiation, inflammation, and cell energy transfer. P2X7 receptor (P2X7R) activation by extracellular ATP (eATP) causes a large MP release and affects their contents in a cell-specific fashion. We investigated MP release and functional impact in microglial cells from P2X7R-WT or P2X7R-KO mice, as well as mouse microglial cell lines characterized for high (N13-P2X7RHigh) or low (N13-P2X7RLow) P2X7R expression. P2X7R stimulation promoted release of a mixed MP population enriched with naked mitochondria. Released mitochondria were taken up and incorporated into the mitochondrial network of the recipient cells in a P2X7R-dependent fashion. NLRP3 and the P2X7R itself were also delivered to the recipient cells. Microparticle transfer increased the energy level of the recipient cells and conferred a pro-inflammatory phenotype. These data show that the P2X7R is a master regulator of intercellular organelle and MP trafficking in immune cells.


Subject(s)
Cell-Derived Microparticles , Mice, Knockout , Microglia , Mitochondria , Receptors, Purinergic P2X7 , Receptors, Purinergic P2X7/metabolism , Receptors, Purinergic P2X7/genetics , Animals , Microglia/metabolism , Mitochondria/metabolism , Mice , Cell-Derived Microparticles/metabolism , Adenosine Triphosphate/metabolism , Cell Line , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics
2.
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
3.
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
4.
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
5.
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
6.
Oncotarget ; 7(31): 49664-49676, 2016 Aug 02.
Article in English | MEDLINE | ID: mdl-27391069

ABSTRACT

Malignant pleural mesothelioma (MPM) is an aggressive tumor refractory to anti-blastic therapy. MPM cells show several genetic and biochemical defects, e.g. overexpression of oncogenes, downregulation of onco-suppressor genes, dysregulation of microRNA, or alteration of intracellular Ca2+ homeostasis and of apoptosis. No information is as yet available on purinergic signalling in this tumor. Signalling via the P2X7 (P2RX7 or P2X7R) purinergic receptor is attracting increasing attention as a pathway involved in cancer cell death or proliferation. In this report we show that the P2X7R is expressed by three MPM cell lines established from MPM patients but not by mesothelial cells from healthy subjects (healthy mesothelial cells, HMCs). MPM cell proliferation was inhibited by in vitro incubation in the presence of selective P2X7R antagonists, as well as by stimulation with the P2X7R agonist BzATP. Systemic administration of the selective P2X7R blocker AZ10606120 inhibited in vivo growth of MPM tumors whether implanted subcutaneously (s.c.) or intraperitoneally (i.p.). Our findings suggest that the P2X7R might be a novel target for the therapy of mesothelioma.


Subject(s)
Lung Neoplasms/metabolism , Mesothelioma/metabolism , Pleural Neoplasms/metabolism , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X7/metabolism , Adult , Animals , Apoptosis , Biopsy , Cell Line, Tumor , Cell Proliferation , Cytoplasm/metabolism , Humans , Male , Mesothelioma, Malignant , Mice , Mice, Nude , MicroRNAs/metabolism , Signal Transduction
7.
FASEB J ; 29(6): 2450-61, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25690658

ABSTRACT

The P2X7 receptor (P2X7R) is a known and powerful activator of the NOD-like receptor (NLR)P3 inflammasome; however, the underlying pathways are poorly understood. Thus, we investigated the molecular mechanisms involved. The effect of P2X7R expression and activation on NLRP3 expression and recruitment was investigated by Western blot, RT-PCR, coimmunoprecipitation, and confocal microscopy in microglial mouse cell lines selected for reduced P2X7R expression and in primary cells from P2X7R(-/-) C57BL/6 mice. We show here that P2X7R activation by ATP (EC50 = 1 mM) or benzoyl-ATP (EC50 = 300 µM) and P2X7R down-modulation caused a 2- to 8-fold up-regulation of NLRP3 mRNA in mouse N13 microglial cells. Moreover, NLRP3 mRNA was also up-regulated in primary microglial and macrophage cells from P2X7R(-/-) mice. Confocal microscopy and immunoprecipitation assays showed that P2X7R and NLRP3 closely interacted at discrete subplasmalemmal sites. Finally, P2X7R stimulation caused a transient (3-4 min) cytoplasmic Ca(2+) increase localized to small (2-3 µm wide) discrete subplasmalemmal regions. The Ca(2+) increase drove P2X7R recruitment and a 4-fold increase in P2X7R/NLRP3 association within 1-2 min. These data show a close P2X7R and NLRP3 interaction and highlight the role of P2X7R in the localized cytoplasmic ion changes responsible for both NLRP3 recruitment and activation.


Subject(s)
Carrier Proteins/genetics , Gene Expression Regulation , Inflammasomes/metabolism , Receptors, Purinergic P2X7/genetics , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Animals , Blotting, Western , Calcium/metabolism , Carrier Proteins/metabolism , Cell Line , Cells, Cultured , Cytoplasm/drug effects , Cytoplasm/metabolism , HEK293 Cells , Humans , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Microglia/cytology , Microglia/drug effects , Microglia/metabolism , Microscopy, Confocal , NLR Family, Pyrin Domain-Containing 3 Protein , Protein Binding , Receptors, Purinergic P2X7/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation/drug effects
8.
Cancer Res ; 72(12): 2957-69, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22505653

ABSTRACT

The P2X7 receptor is an ATP-gated ion channel known for its cytotoxic activity. However, recent evidence suggests a role for P2X7 in cell proliferation. Here, we found that P2X7 exhibits significant growth-promoting effects in vivo. Human embryonic kidney cells expressing P2X7 exhibited a more tumorigenic and anaplastic phenotype than control cells in vivo, and the growth rate and size of these tumors were significantly reduced by intratumoral injection of the P2X7 inhibitor-oxidized ATP. The accelerated growth of P2X7-expressing tumors was characterized by increased proliferation, reduced apoptosis, and a high level of activated transcription factor NFATc1. These tumors also showed a more developed vascular network than control tumors and secreted elevated amounts of VEGF. The growth and neoangiogenesis of P2X7-expressing tumors was blocked by intratumoral injection of the VEGF-blocking antibody Avastin (bevacizumab), pharmacologic P2X7 blockade, or P2X7 silencing in vivo. Immunohistochemistry revealed strong P2X7 positivity in several human cancers. Together, our findings provide direct evidence that P2X7 promotes tumor growth in vivo.


Subject(s)
Cell Transformation, Neoplastic , Neoplasms, Experimental/pathology , Neovascularization, Pathologic , Receptors, Purinergic P2X7/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Apoptosis , Bevacizumab , Biomarkers, Tumor , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , NFATC Transcription Factors/biosynthesis , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/metabolism , Receptors, Purinergic P2X7/biosynthesis , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism
9.
FASEB J ; 24(9): 3393-404, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20453110

ABSTRACT

P2X7 is the largest member of the P2X subfamily of purinergic receptors. A typical feature is the carboxyl tail, which allows formation of a large pore. Recently a naturally occurring truncated P2X7 splice variant, isoform B (P2X7B), has been identified. Here we show that P2X7B expression in HEK293 cells, a cell type lacking endogenous P2X receptors, mediated ATP-stimulated channel activity but not plasma membrane permeabilization, raised endoplasmic reticulum Ca(2+) content, activated the transcription factor NFATc1, increased the cellular ATP content, and stimulated growth. In addition, P2X7B-transfected HEK293 cells (HEK293-P2X7B), like most tumor cells, showed strong soft agar-infiltrating ability. When coexpressed with full-length P2X7 (P2X7A), P2X7B coassembled with P2X7A into a heterotrimer and potentiated all known responses mediated by this latter receptor. P2X7B mRNA was found to be widely distributed in human tissues, especially in the immune and nervous systems, and to a much higher level than P2X7A. Finally, P2X7B expression was increased on mitogenic stimulation of peripheral blood lymphocyte. Altogether, these data show that P2X7B is widely expressed in several human tissues, modulates P2X7A functions, participates in the control of cell growth, and may help understand the role of the P2X7 receptor in the control of normal and cancer cell proliferation.


Subject(s)
Receptors, Purinergic P2/chemistry , Receptors, Purinergic P2/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Cell Line , Cell Membrane/metabolism , Fluorescent Antibody Technique , Humans , Membrane Potentials/genetics , Membrane Potentials/physiology , Molecular Sequence Data , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2X7 , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid
10.
J Immunol ; 182(7): 4378-85, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19299738

ABSTRACT

Extracellular ATP is a mediator of intercellular communication and a danger signal. Release of this and other nucleotides modulates microglia responses via P2Y and P2X receptors, among which the P2X(7) subtype stands out for its proinflammatory activity and for up-regulation in a transgenic model of Alzheimer disease and in brains from Alzheimer disease patients. Here we show that amyloid beta (Abeta) triggered increases in intracellular Ca(2+) ([Ca(2+)](i)), ATP release, IL-1beta secretion, and plasma membrane permeabilization in microglia from wild-type but not from P2X(7)-deleted mice. Likewise, intra-hippocampal injection of Abeta caused a large accumulation of IL-1beta in wild-type but not in P2X(7)(-/-) mice. These observations suggest that Abeta activates a purinergic autocrine/paracrine stimulatory loop of which the P2X(7) receptor is an obligate component. Identification of the P2X(7) receptor as a non-dispensable factor of Abeta-mediated microglia stimulation may open new avenues for the treatment of Alzheimer disease.


Subject(s)
Amyloid beta-Peptides/metabolism , Interleukin-1beta/metabolism , Microglia/metabolism , Receptors, Purinergic P2/metabolism , Adenosine Triphosphate/metabolism , Animals , Calcium/metabolism , Cell Membrane Permeability/physiology , Mice , Mice, Knockout , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2X7
11.
Am J Physiol Renal Physiol ; 292(5): F1537-47, 2007 May.
Article in English | MEDLINE | ID: mdl-17264311

ABSTRACT

Apoptosis, a normal event in renal tissue homeostasis, has been considered as a major mechanism for either resolution of glomerular hypercellularity in glomerulonephritis or loss of cellularity and progression to glomerulosclerosis in chronic renal disease. This study was aimed at investigating the role of extracellular ATP (eATP) in mediating apoptosis in human mesangial cells (HMC) and identifying the subtype(s) of purinergic receptors involved. eATP, but not uridin-5'-triphosphate (UTP), caused dose-dependent modifications of cellular morphology, as assessed by contrast-phase microscopy, and late apoptosis, as measured by Annexin V/propidium iodide-based flow cytometry and caspase-3 activation. Both phenomena were prevented by the P2X antagonist oxidized-ATP. 2', 3'-O-(4-benzoylbenzoyl)adenosine 5'-triphosphate (BzATP) was less effective than ATP, whereas 1[N,O-bis (5-isoquinolinesulfonyl)-N-methyl-L-tyrosyl] -4-phenylpiperazine (KN62), a selective inhibitor of human P2X(7), prevented morphological changes but potentiated apoptosis induced by BzATP. P2X(7) was barely expressed in HMC and showed a relatively scarce functional activity, as assessed by monitoring nucleotide-induced intracellular calcium surge and plasma membrane depolarization by Fura-2/AM and bis[1,3-diethylthiobarbiturate]trimethineoxonal uptake, respectively. These data indicated a negligible role of P2X(7) in eATP-mediated apoptosis and pointed to the involvement of other P2X receptor(s). Molecular and inhibitor studies suggested a main role for P2X(4) receptor in nucleotide-induced apoptosis in HMC, indicating a relevant role for purinergic signaling in regulating death rate in these cells.


Subject(s)
Apoptosis/physiology , Mesangial Cells/physiology , Receptors, Purinergic P2/physiology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Adenosine Triphosphate/administration & dosage , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Adenosine Triphosphate/physiology , Apoptosis/drug effects , Calcium/metabolism , Caspase 3/metabolism , Cell Membrane/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Drug Synergism , Electrophysiology , Enzyme Activation , Extracellular Fluid/metabolism , Flow Cytometry , Humans , Intracellular Membranes/metabolism , Mesangial Cells/cytology , Mesangial Cells/drug effects , Microscopy, Phase-Contrast , Purinergic P2 Receptor Antagonists , Receptors, Purinergic P2X4 , Receptors, Purinergic P2X7 , Time Factors
12.
Blood ; 109(9): 3856-64, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17192399

ABSTRACT

Dendritic cells (DCs) are professional antigen-presenting cells that initiate the immune response by activating T lymphocytes. DCs express plasma membrane receptors for extracellular nucleotides named P2 receptors (P2Rs). Stimulation of P2Rs in these cells is known to cause chemotaxis, cytokine release, and cell death and to modulate LPS-dependent differentiation. Here we show that stimulation of the P2X(7) receptor subtype (P2X(7)R) causes fast microvesicle shedding from DC plasma membrane. Vesicle release occurs from both immature and mature DCs; however, only vesicles from mature DCs, due to their previous exposure to LPS, contain IL-1beta. Microvesicles, whether from immature or mature DCs, also contain caspase-1 and -3 and cathepsin D. They also express the P2X(7)R in addition to other P2Rs and known markers of immune cells such as major histocompatibility complex II (MHC II) and CD39. Activation of the P2X(7)R by extracellular ATP causes IL-1beta release from the vesicle lumen. Previous studies demonstrated that high extracellular K(+) inhibits IL-1beta processing and release; here we show that high ionic strength reduces microvesicle shedding when compared with a low ionic strength medium but strongly increases microvesicle IL-1beta loading.


Subject(s)
Adenosine Triphosphate/pharmacology , Dendritic Cells/metabolism , Interleukin-1beta/metabolism , Receptors, Purinergic P2/metabolism , Secretory Vesicles/metabolism , Caspase 1/immunology , Caspase 1/metabolism , Caspase 3/immunology , Caspase 3/metabolism , Cathepsin D/immunology , Cathepsin D/metabolism , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/immunology , Humans , Interleukin-1beta/immunology , Lipopolysaccharides/pharmacology , Potassium/immunology , Potassium/metabolism , Purinergic P2 Receptor Agonists , Receptors, Purinergic P2/immunology , Receptors, Purinergic P2X7 , Secretory Vesicles/immunology
13.
J Immunol ; 177(10): 7257-65, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17082644

ABSTRACT

Multinucleated giant cells (MGC), a hallmark of chronic inflammatory reactions, remain an enigma of cell biology. There is evidence implicating the purinergic P2X7 receptor in the fusion process leading to MGC. To investigate this, we used HEK 293 cells stably transfected with either 1) the full-length rat P2X7 receptor (P2X7 cells), 2) a rat P2X7 receptor lacking the C-terminal domain (P2X7TC), or 3) a mock vector, and rat alveolar macrophages (MA) expressing the native receptor. P2X7 cells cultured in serum-free medium formed increased numbers of MGC and displayed a higher fusion index compared with mock transfectants. Stimulation of P2X7 pore-forming activity in P2X7 cells by polymyxin B (PMB) further increased significantly the formation of MGC. Conversely, blockers of P2X-receptors including oxidized ATP, brilliant blue G, and pyridoxal phosphate-6-azophenyl-2'-4'-disulfonic acid inhibited significantly MGC formation in both unstimulated and PMB-stimulated P2X7-transfected cells. In contrast, cells transfected with the truncated P2X7TC were devoid of pore-forming activity, did not respond to PMB stimulation, and failed to form enhanced numbers of MGC, thus behaving as mock transfectants. As found for P2X7-transfected cells, PMB also potentiated dose-dependently the formation of multinucleated MA by rat alveolar MA. Pretreatment with oxidized ATP abrogated the PMB stimulatory effects. Together, these data demonstrate unequivocally the participation of P2X7 receptor in the process of MGC formation. Our study also provides evidence suggesting that stimulation of the P2X7 receptor pathway in MA may mediate increased formation of MGC during chronic inflammatory reactions.


Subject(s)
Cell Differentiation/physiology , Giant Cells/cytology , Giant Cells/metabolism , Receptors, Purinergic P2/physiology , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Animals , Cell Differentiation/drug effects , Cell Line , Cells, Cultured , Drug Synergism , Giant Cells/drug effects , Growth Inhibitors/pharmacology , Humans , Macrophages, Alveolar/cytology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Male , Polymyxin B/pharmacology , Purinergic P2 Receptor Antagonists , Rats , Rats, Wistar , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2X7 , Transfection
14.
Cancer Res ; 66(2): 907-14, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16424024

ABSTRACT

P2X(7) is a receptor for extracellular nucleotides expressed by different normal cell types. P2X(7) triggering may result in stimulation of cell proliferation or induction of apoptosis depending on the level of activation. P2X(7) expression and function in B-cell chronic lymphocytic leukemia has been shown to correlate with disease severity. Here, we have asked the question of whether P2X(7) is expressed and functional in neuroblastoma, a pediatric tumor of neuroectodermal origin. P2X(7) was detected both in primary neuroblastoma tumors and in neuroblastoma cell lines. In the latter cells, P2X(7) stimulation by ATP was found to trigger (a) increased intracellular calcium fluxes, (b) plasma membrane depolarization, and (c) formation of a nonselective plasma membrane permeable pore. In contrast to the usual response typically observed in the majority of cell types, P2X(7) in vitro stimulation did not induce caspase-3 activation or apoptosis of neuroblastoma cells but rather supported their proliferation. Growth stimulation was partially due to substance P release from nucleotide-activated neuroblastoma cells. Therefore, neuroblastoma cells seem to have molded P2X(7) function to their advantage in two ways (i.e., by silencing P2X(7) proapoptotic activity and by coupling P2X(7) stimulation to release of locally acting trophic factors).


Subject(s)
Neuroblastoma/genetics , Neuroblastoma/pathology , Receptors, Purinergic P2/physiology , Substance P/metabolism , Apoptosis , Cell Proliferation , Flow Cytometry , Gene Expression Profiling , Gene Silencing , Humans , Receptors, Purinergic P2/biosynthesis , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2X7 , Tumor Cells, Cultured
15.
Arterioscler Thromb Vasc Biol ; 25(11): 2392-7, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16151016

ABSTRACT

OBJECTIVE: Insulin activates several processes potentially dangerous for the arterial wall and hyperinsulinemia might be atherogenic. However, other insulin effects are protective for the vessel wall and thus anti-atherogenic. Aim of this study was to investigate whether insulin effects on potentially pro-atherogenic and anti-atherogenic processes were differently affected in cells from insulin-resistant individuals. METHODS AND RESULTS: We determined insulin effect on nitric oxide (NO) production and plasminogen activator inhibitor (PAI)-1 synthesis in 12 fibroblast strains obtained from skin biopsy samples of 6 insulin-sensitive (IS) (clamp M >7 mg/kg body weight per minute) and 6 insulin-resistant (IR) (clamp M <5 mg/kg body weight per minute) healthy volunteers. Insulin effects on NO release and Akt phosphorylation were significantly impaired in fibroblasts from IR as compared with IS individuals. Conversely, there was not any difference between IR and IS strains in insulin ability to increase PAI-1 antigen levels and, after 24-hour insulin incubation, PAI-1 mRNA increase in IR strains was only slightly less than in IS strains. Insulin ability to induce MAPK activation was also comparable in IR and IS cells. CONCLUSIONS: We conclude that in cells from IR individuals, insulin action on anti-atherogenic processes, such as NO release, is impaired, whereas the hormone ability to stimulate atherogenic processes, such as PAI-1 release, is preserved.


Subject(s)
Atherosclerosis/metabolism , Hyperinsulinism/metabolism , Insulin Resistance , Nitric Oxide/metabolism , Plasminogen Activator Inhibitor 1/genetics , Adult , Cells, Cultured , Culture Media , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Glucose/pharmacokinetics , Glycogen/biosynthesis , Humans , Insulin/pharmacology , MAP Kinase Signaling System/physiology , Male , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , Plasminogen Activator Inhibitor 1/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Serine/metabolism
16.
Kidney Int ; 67(3): 875-85, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15698427

ABSTRACT

BACKGROUND: Extracellular adenosine triphosphate (ATP) (eATP) mediates several biologic activities via purinergic P2 receptors (P2Rs). This study aimed at (1) evaluating the role of the purinergic system in modulating mesangial extracellular matrix (ECM) and transforming growth factor-beta (TGF-beta) production and (2) its contribution to diabetes-induced mesangial ECM accumulation. METHODS: Rat mesangial cells were grown in normal glucose (5.5 mmol/L) or high glucose (30 mmol/L) containing media and probed with purinergic agonists and antagonists for the assessment of the expression pattern and function of P2Rs; release of ATP and activity of ectoATPases; and changes in ECM and TGF-beta expression. RESULTS: Cells cultured in normal glucose and high glucose expressed similar amounts of functional P2Rs of the P2X(2), P2X(3), P2X(4), P2X(5), P2X(7), P2Y(1), P2Y(2), P2Y(4), and P2Y(6) subtypes. Levels of eATP were higher in high glucose vs. normal glucose, with unchanged ectoATPase activity. The ATP-hydrolyzing enzymes hexokinase or apyrase reduced ECM and TGF-beta production from cells grown in high glucose, but not normal glucose. Under both normal glucose and high glucose conditions, ATP and the P2X(7) agonist benzoylbenzoylATP increased dose-dependently ECM and TGF-beta production, whereas the P2Y agonist uridine triphosphate (UTP) produced the opposite effect. The P2X(7) inhibitor oxidized ATP attenuated the ECM and TGF-beta up-regulation induced by ATP and, to a lesser extent, that caused by high glucose. A TGF-beta neutralizing antibody also prevented ATP-induced ECM up-regulation. CONCLUSION: These data indicate a role for eATP in regulating ECM production via TGF-beta and suggest that P2XRs and P2YRs differentially modulate this process. An increased ATP release induced by hyperglycemia might contribute to mesangial matrix expansion occurring in diabetes.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Extracellular Matrix Proteins/biosynthesis , Glomerular Mesangium/metabolism , Kidney Diseases/metabolism , Receptors, Purinergic P2/physiology , Adenosine Triphosphatases , Adenosine Triphosphate/physiology , Animals , Cells, Cultured , Rats , Transforming Growth Factor beta/biosynthesis
17.
Blood ; 104(6): 1662-70, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15161674

ABSTRACT

Although extracellular nucleotides support a wide range of biologic responses of mature blood cells, little is known about their effect on blood cell progenitor cells. In this study, we assessed whether receptors for extracellular nucleotides (P2 receptors [P2Rs]) are expressed on human hematopoietic stem cells (HSCs), and whether activation by their natural ligands, adenosine triphosphate (ATP) and uridine triphosphate (UTP), induces HSC proliferation in vitro and in vivo. Our results demonstrated that CD34(+) HSCs express functional P2XRs and P2YRs of several subtypes. Furthermore, stimulation of CD34(+) cells with extracellular nucleotides caused a fast release of Ca(2+) from intracellular stores and an increase in ion fluxes across the plasma membrane. Functionally, ATP and, to a higher extent, UTP acted as potent early acting growth factors for HSCs, in vitro, because they strongly enhanced the stimulatory activity of several cytokines on clonogenic CD34(+) and lineage-negative CD34(-) progenitors and expanded more primitive CD34(+)-derived long-term culture-initiating cells. Furthermore, xenogenic transplantation studies showed that short-term preincubation with UTP significantly expanded the number of marrow-repopulating HSCs in nonobese diabetic/severe combined immunodeficiency mice. Our data suggest that extracellular nucleotides may provide a novel and powerful tool to modulate HSC functions.


Subject(s)
Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Nucleotides/pharmacology , Antigens, CD34/immunology , Antigens, CD34/metabolism , Cell Division/drug effects , Cells, Cultured , Clone Cells/cytology , Clone Cells/drug effects , Clone Cells/metabolism , Colony-Stimulating Factors/pharmacology , Culture Media, Conditioned/pharmacology , Hematopoietic Stem Cells/metabolism , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Purinergic P2/immunology , Uridine Triphosphate/pharmacology
18.
Arterioscler Thromb Vasc Biol ; 24(7): 1240-5, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15155383

ABSTRACT

OBJECTIVE: We have investigated expression and function of the P2X7 receptor in fibroblasts from healthy subjects and patients with type 2 diabetes. METHODS AND RESULTS: Fibroblasts were isolated from skin biopsies. P2X7 receptor expression in both cell populations was measured by functional assays, RT-PCR, fluorescence-activated cell sorter, and immunoblotting. We found that fibroblasts from diabetic subjects are characterized by enhanced P2X7-mediated responses as indicated by increased shape changes, microvesiculation, enhanced fibronectin and interleukin 6 secretion, and accelerated apoptosis. These responses were blocked by preincubation with the P2X blockers KN-62, oxidized ATP, or pyridoxal phosphate-6-azo(benzene-2,4-disulfonic acid). Furthermore, we also found a higher level of spontaneous fibronectin secretion and of apoptosis in fibroblasts from diabetic compared with healthy subjects. Both higher basal level of fibronectin secretion and spontaneous rate of apoptosis were likely attributable to the increased pericellular concentration of ATP because fibroblasts from diabetic subjects released 3x as much ATP into the supernatants compared with fibroblasts from healthy subjects. CONCLUSIONS: We conclude that fibroblasts from type 2 diabetes patients are characterized by a hyperactive purinergic loop based either on a higher level of ATP release or on increased P2X7 reactivity.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Adenosine Triphosphate/analogs & derivatives , Diabetes Mellitus, Type 2/pathology , Diabetic Angiopathies/etiology , Fibroblasts/pathology , Pyridoxal Phosphate/analogs & derivatives , Receptors, Purinergic P2/physiology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Adenosine Diphosphate/pharmacology , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Apoptosis/drug effects , Apyrase/pharmacology , Autocrine Communication , Cell Shape/drug effects , Cytidine Triphosphate/pharmacology , Fibroblasts/metabolism , Fibronectins/metabolism , Gene Expression Regulation , Humans , Interleukin-6/metabolism , Membrane Potentials/drug effects , Paracrine Communication , Pyridoxal Phosphate/pharmacology , Receptors, Purinergic P2/biosynthesis , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2X7 , Uridine Diphosphate/pharmacology , Uridine Triphosphate/pharmacology
19.
J Cell Physiol ; 197(3): 435-44, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14566973

ABSTRACT

Extracellular ATP is an ubiquitous mediator that regulates several cellular functions via specific P2 plasma membrane receptors (P2Rs), for which a role in modulating intracellular glucose metabolism has been recently suggested. We have investigated glucose uptake in response to P2Rs stimulation in fibroblasts from type 2 diabetic (T2D) patients and control subjects. P2Rs expression was evaluated by RT-PCR; intracellular calcium release by fluorometry; glucose transporter (GLUT1) translocation by immunoblotting and chemiluminescence; glucose uptake was measured with 2-deoxy-D-[1-(3)H]glucose (2-DOG) and ATP by luminometry. Cells from T2D patients, in contrast to those from healthy controls, showed no increase in glucose uptake after ATP stimulation; extracellular ATP caused, however, a similar GLUT1 recruitment to the plasma membrane in both groups. P2Rs expression did not differ between fibroblasts from diabetic and healthy subjects, but while plasma membrane depolarization, a P2X-mediated response was similar in both groups, no evident intracellular calcium increase was detectable in the cells from the former group. The calcium response in fibroblasts from diabetics was restored by co-incubation with apyrase or hexokinase, suggesting that P2YRs in those cells were normally expressed but chronically desensitised. In support to this finding, fibroblasts from T2D subjects secreted a two-fold larger amount of ATP compared to controls. Pre-treatment with apyrase or hexokinase also restored ATP stimulated glucose uptake in fibroblasts from diabetic subjects. These results suggest that extracellular ATP plays a role in the modulation of glucose transport via GLUT1, and that the P2Y-dependent GLUT1 activation is deficient in fibroblasts from T2D individuals. Our observations may point to additional therapeutic targets for improving glucose utilization in diabetes.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Monosaccharide Transport Proteins/metabolism , Receptors, Purinergic P2/genetics , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Apyrase/metabolism , Biological Transport/physiology , Calcium/metabolism , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Energy Metabolism/physiology , Extracellular Fluid/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Glucose Transporter Type 1 , Hexokinase/metabolism , Humans , Insulin Resistance/physiology , Membrane Potentials/drug effects , Membrane Potentials/physiology , RNA, Messenger/metabolism , Receptors, Purinergic P2Y1
20.
Mol Biol Cell ; 14(7): 2655-64, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12857854

ABSTRACT

The P2X7 ATP receptor mediates the cytotoxic effect of extracellular ATP. P2X7-dependent cell death is heralded by dramatic plasma membrane bleb formation. Membrane blebbing is a complex phenomenon involving as yet poorly characterized intracellular pathways. We have investigated the effect of extracellular ATP on HEK293 cells transfected with the cytotoxic/pore-forming P2X7 receptor. Addition of ATP to P2X7-transfected, but not to wt P2X7-less, HEK293 cells caused massive membrane blebbing within 1-2 min. UTP, a nucleotide incapable of activating P2X7, had no early effects on cell shape and bleb formation. Bleb formation triggered by ATP was reversible and required extracellular Ca2+ and an intact cytoskeleton. Furthermore, it was completely prevented by preincubation with the P2X blocker oxidized ATP. It was recently observed that the ROCK protein is a key determinant of bleb formation. Preincubation of HEK293-P2X7 cells with the ROCK blocker Y-27632 completely prevented P2X7-dependent blebbing. Although ATP triggered cleavage of the ROCK I isoform in P2X7-transfected HEK293 cells, the wide range caspase inhibitor z-VAD-fluoromethylketone had no effect. These observations suggest that P2X7-dependent plasma membrane blebbing depends on the activation of the serine/threonine kinase ROCK I.


Subject(s)
Adenosine Triphosphate/metabolism , Cytoskeleton/physiology , Protein Serine-Threonine Kinases/metabolism , Receptors, Purinergic P2/metabolism , Adenosine Triphosphate/physiology , Amides/pharmacology , Amino Acid Chloromethyl Ketones/pharmacology , Calcium/metabolism , Caspase Inhibitors , Cell Surface Extensions/metabolism , Cell Surface Extensions/physiology , Cells, Cultured , Cytoskeleton/metabolism , Enzyme Inhibitors/pharmacology , Extracellular Space/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/physiology , Pyridines/pharmacology , Receptors, Purinergic P2/physiology , Receptors, Purinergic P2X7 , Transfection , rho-Associated Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...