Activation of endothelial NO synthase and P2X7 receptor modification mediates the cholinergic control of ATP-induced interleukin-1β release by mononuclear phagocytes

Objective The pro-inflammatory cytokine interleukin-1β (IL-1β) plays a central role in host defense against infections. High systemic IL-1β levels, however, promote the pathogenesis of inflammatory disorders. Therefore, mechanisms controlling IL-1β release are of substantial clinical interest. Recently, we identified a cholinergic mechanism inhibiting the ATP-mediated IL-1β release by human monocytes via nicotinic acetylcholine receptor (nAChR) subunits α7, α9 and/or α10. We also discovered novel nAChR agonists that trigger this inhibitory function in monocytic cells without eliciting ionotropic functions at conventional nAChRs. Here, we investigate the ion flux-independent signaling pathway that links nAChR activation to the inhibition of the ATP-sensitive P2X7 receptor (P2X7R). Methods Different human and murine mononuclear phagocytes were primed with lipopolysaccharide and stimulated with the P2X7R agonist BzATP in the presence or absence of nAChR agonists, endothelial NO synthase (eNOS) inhibitors, and NO donors. IL-1β was measured in cell culture supernatants. Patch-clamp and intracellular Ca2+ imaging experiments were performed on HEK cells overexpressing human P2X7R or P2X7R with point mutations at cysteine residues in the cytoplasmic C-terminal domain. Results The inhibitory effect of nAChR agonists on the BzATP-induced IL-1β release was reversed in the presence of eNOS inhibitors (L-NIO, L-NAME) as well as in U937 cells after silencing of eNOS expression. In peripheral blood mononuclear leukocytes from eNOS gene-deficient mice, the inhibitory effect of nAChR agonists was absent, suggesting that nAChRs signal via eNOS to inhibit the BzATP-induced IL-1β release. Moreover, NO donors (SNAP, S-nitroso-N-acetyl-DL-penicillamine; SIN-1) inhibited the BzATP-induced IL-1β release by mononuclear phagocytes. The BzATP-induced ionotropic activity of the P2X7R was abolished in the presence of SIN-1 in both, Xenopus laevis oocytes and HEK cells over-expressing the human P2X7R. This inhibitory effect of SIN-1 was absent in HEK cells expressing P2X7R, in which C377 was mutated to alanine, indicating the importance of C377 for the regulation of the P2X7R function by protein modification. Conclusion We provide first evidence that ion flux-independent, metabotropic signaling of monocytic nAChRs involves eNOS activation and P2X7R modification, resulting in an inhibition of ATP signaling and ATP-mediated IL-1β release. This signaling pathway might be an interesting target for the treatment of inflammatory disorders.

[1]  W. Padberg,et al.  Negative regulation of ATP-induced inflammasome activation and cytokine secretion by acute-phase proteins: A mini review , 2022, Frontiers in Pharmacology.

[2]  R. Papke,et al.  Comparison of the Anti-inflammatory Properties of Two Nicotinic Acetylcholine Receptor Ligands, Phosphocholine and pCF3-diEPP , 2022, Frontiers in Cellular Neuroscience.

[3]  A. Viola,et al.  Reactive Oxygen Species in Macrophages: Sources and Targets , 2021, Frontiers in Immunology.

[4]  A. Mishra,et al.  P2X7 receptor in multifaceted cellular signaling and its relevance as a potential therapeutic target in different diseases. , 2021, European journal of pharmacology.

[5]  M. Morishima,et al.  Nitric oxide down-regulates voltage-gated Na+ channel in cardiomyocytes possibly through S-nitrosylation-mediated signaling , 2021, Scientific Reports.

[6]  S. Muench,et al.  Structural basis for the functional properties of the P2X7 receptor for extracellular ATP , 2021, Purinergic Signalling.

[7]  V. Lemos,et al.  Role of the α7 Nicotinic Acetylcholine Receptor in the Pathophysiology of Atherosclerosis , 2020, Frontiers in Physiology.

[8]  F. Di Virgilio,et al.  Update of P2X receptor properties and their pharmacology: IUPHAR Review 30 , 2020, British journal of pharmacology.

[9]  M. Tsuda,et al.  Nociceptive signaling mediated by P2X3, P2X4 and P2X7 receptors. , 2020, Biochemical pharmacology.

[10]  E. Schiffrin,et al.  P2X7: An Untapped Target for the Management of Cardiovascular Disease. , 2020, Arteriosclerosis, thrombosis, and vascular biology.

[11]  W. Padberg,et al.  Amyloid Beta Peptide (Aβ1-42) Reverses the Cholinergic Control of Monocytic IL-1β Release , 2020, Journal of clinical medicine.

[12]  A. Nicke,et al.  Design, Synthesis, and in vitro Evaluation of P2X7 Antagonists , 2020, ChemMedChem.

[13]  F. Di Virgilio,et al.  Purinergic signalling, DAMPs and inflammation. , 2020, American journal of physiology. Cell physiology.

[14]  C. Yoshioka,et al.  Full-Length P2X7 Structures Reveal How Palmitoylation Prevents Channel Desensitization , 2019, Cell.

[15]  S. Furuta,et al.  S-Nitrosylation: An Emerging Paradigm of Redox Signaling , 2019, Antioxidants.

[16]  Robin Kopp,et al.  P2X7 Interactions and Signaling – Making Head or Tail of It , 2019, Front. Mol. Neurosci..

[17]  F. Koch-Nolte,et al.  Purine Release, Metabolism, and Signaling in the Inflammatory Response. , 2019, Annual review of immunology.

[18]  R. Tikkanen,et al.  SLPI Inhibits ATP-Mediated Maturation of IL-1β in Human Monocytic Leukocytes: A Novel Function of an Old Player , 2019, Front. Immunol..

[19]  J. McIntosh,et al.  Conopeptides [V11L;V16D]ArIB and RgIA4: Powerful Tools for the Identification of Novel Nicotinic Acetylcholine Receptors in Monocytes , 2019, Front. Pharmacol..

[20]  M. Haque,et al.  Nitric oxide synthase enzymology in the 20 years after the Nobel Prize , 2018, British journal of pharmacology.

[21]  Mushfiquddin Khan,et al.  S-Nitrosylation in Regulation of Inflammation and Cell Damage. , 2018, Current drug targets.

[22]  S. Dervish,et al.  Characterization of Human Monocyte Subsets by Whole Blood Flow Cytometry Analysis , 2018, Journal of visualized experiments : JoVE.

[23]  A. Hise Faculty Opinions recommendation of Inflammasomes: mechanism of assembly, regulation and signalling. , 2018, Faculty Opinions – Post-Publication Peer Review of the Biomedical Literature.

[24]  Perrine Bortolotti,et al.  Inflammasomes in Tissue Damages and Immune Disorders After Trauma , 2018, Front. Immunol..

[25]  W. Padberg,et al.  Phosphocholine-Modified Lipooligosaccharides of Haemophilus influenzae Inhibit ATP-Induced IL-1β Release by Pulmonary Epithelial Cells , 2018, Molecules.

[26]  I. König,et al.  C-Reactive Protein Stimulates Nicotinic Acetylcholine Receptors to Control ATP-Mediated Monocytic Inflammasome Activation , 2018, Front. Immunol..

[27]  W. Padberg,et al.  Alpha-1 Antitrypsin Inhibits ATP-Mediated Release of Interleukin-1β via CD36 and Nicotinic Acetylcholine Receptors , 2018, Front. Immunol..

[28]  L. Joosten,et al.  OLT1177, a β-sulfonyl nitrile compound, safe in humans, inhibits the NLRP3 inflammasome and reverses the metabolic cost of inflammation , 2018, Proceedings of the National Academy of Sciences.

[29]  T. Kawate,et al.  The P2X7 receptor forms a dye-permeable pore independent of its intracellular domain but dependent on membrane lipid composition , 2017, eLife.

[30]  W. Padberg,et al.  Canonical and Novel Non-Canonical Cholinergic Agonists Inhibit ATP-Induced Release of Monocytic Interleukin-1β via Different Combinations of Nicotinic Acetylcholine Receptor Subunits α7, α9 and α10 , 2017, Front. Cell. Neurosci..

[31]  C. Ruppert,et al.  Surfactant inhibits ATP-induced release of interleukin-1β via nicotinic acetylcholine receptors[S] , 2017, Journal of Lipid Research.

[32]  G. Schmalzing,et al.  Localization of the gate and selectivity filter of the full-length P2X7 receptor , 2017, Proceedings of the National Academy of Sciences.

[33]  W. Padberg,et al.  Phosphocholine – an agonist of metabotropic but not of ionotropic functions of α9-containing nicotinic acetylcholine receptors , 2016, Scientific Reports.

[34]  V. Dixit,et al.  Inflammasomes: mechanism of assembly, regulation and signalling , 2016, Nature Reviews Immunology.

[35]  B. O'Brien,et al.  The choice of phorbol 12-myristate 13-acetate differentiation protocol influences the response of THP-1 macrophages to a pro-inflammatory stimulus. , 2016, Journal of immunological methods.

[36]  W. Padberg,et al.  Phosphocholine-Modified Macromolecules and Canonical Nicotinic Agonists Inhibit ATP-Induced IL-1β Release , 2015, The Journal of Immunology.

[37]  F. Guimarães,et al.  Antidepressant- and anticompulsive-like effects of purinergic receptor blockade: Involvement of nitric oxide , 2013, European Neuropsychopharmacology.

[38]  Xiaotian Li,et al.  Roles of Apolipoprotein E (ApoE) and Inducible Nitric Oxide Synthase (iNOS) in Inflammation and Apoptosis in Preeclampsia Pathogenesis and Progression , 2013, PloS one.

[39]  E. Lien,et al.  Inflammasomes and host defenses against bacterial infections. , 2013, Current opinion in microbiology.

[40]  S. Akira,et al.  Cutting Edge: Nitric Oxide Inhibits the NLRP3 Inflammasome , 2012, The Journal of Immunology.

[41]  Hardy Kornfeld,et al.  Nitric oxide controls the immunopathology of tuberculosis by inhibiting NLRP3 inflammasome–dependent processing of IL-1β , 2012, Nature Immunology.

[42]  D. Bayliss,et al.  S-Nitrosylation Inhibits Pannexin 1 Channel Function* , 2012, The Journal of Biological Chemistry.

[43]  L. Jones Chemistry and biology of biomolecule nitration. , 2012, Chemistry & biology.

[44]  J. Tschopp,et al.  The inflammasome: an integrated view , 2011, Immunological reviews.

[45]  S. Snyder,et al.  S-Nitrosylation and S-Palmitoylation Reciprocally Regulate Synaptic Targeting of PSD-95 , 2011, Neuron.

[46]  H. M. Costa-Junior,et al.  C terminus of the P2X7 receptor: treasure hunting , 2011, Purinergic Signalling.

[47]  A. Abdel‐Rahman,et al.  Estrogen dependence of the renal vasodilatory effect of nicotine in rats: role of α7 nicotinic cholinergic receptor/eNOS signaling. , 2011, Life sciences.

[48]  J. Greaves,et al.  The intracellular dynamic of protein palmitoylation , 2010, The Journal of cell biology.

[49]  Silvano Sozzani,et al.  Nomenclature of monocytes and dendritic cells in blood. , 2010, Blood.

[50]  J. Joseph,et al.  Peroxynitrite Is the Major Species Formed from Different Flux Ratios of Co-generated Nitric Oxide and Superoxide , 2010, The Journal of Biological Chemistry.

[51]  T. Michel,et al.  Cellular signaling and NO production , 2010, Pflügers Archiv - European Journal of Physiology.

[52]  J. Cooke,et al.  Cholinergic modulation of angiogenesis: Role of the 7 nicotinic acetylcholine receptor , 2009, Journal of cellular biochemistry.

[53]  G. Schmalzing,et al.  The P2X7 Carboxyl Tail Is a Regulatory Module of P2X7 Receptor Channel Activity* , 2008, Journal of Biological Chemistry.

[54]  F. Martinon,et al.  Activation of the NALP3 inflammasome is triggered by low intracellular potassium concentration , 2007, Cell Death and Differentiation.

[55]  Eric A. Barnard,et al.  International Union of Pharmacology LVIII: Update on the P2Y G Protein-Coupled Nucleotide Receptors: From Molecular Mechanisms and Pathophysiology to Therapy , 2006, Pharmacological Reviews.

[56]  W. Kummer,et al.  Nicotinic receptor mediated stimulation of NO-generation in neurons of rat thoracic dorsal root ganglia , 2004, Neuroscience Letters.

[57]  Wenzheng Zhang,et al.  Protein interactions with nitric oxide synthases: controlling the right time, the right place, and the right amount of nitric oxide. , 2003, American journal of physiology. Renal physiology.

[58]  W. Kummer,et al.  Nicotinic receptor alpha7-subunits are coupled to the stimulation of nitric oxide synthase in rat dorsal root ganglion neurons , 2003, Histochemistry and Cell Biology.

[59]  R. Freedman,et al.  Inhibition of nitric oxide synthase prevents α7 nicotinic receptor-mediated restoration of inhibitory auditory gating in rat hippocampus , 2000, Brain Research.

[60]  J. Buss,et al.  S-Nitrosocysteine Increases Palmitate Turnover on Ha-Ras in NIH 3T3 Cells* , 2000, The Journal of Biological Chemistry.

[61]  R. Busse,et al.  Signal transduction of eNOS activation. , 1999, Cardiovascular research.

[62]  B. Mayer,et al.  Enzymatic function of nitric oxide synthases. , 1999, Cardiovascular research.

[63]  J. Boucher,et al.  Nitric oxide biosynthesis, nitric oxide synthase inhibitors and arginase competition for L-arginine utilization , 1999, Cellular and Molecular Life Sciences CMLS.

[64]  S. Matalon,et al.  Peroxynitrite inhibits amiloride-sensitive Na+ currents in Xenopus oocytes expressing αβγ-rENaC. , 1998, American journal of physiology. Cell physiology.

[65]  S. Matalon,et al.  Peroxynitrite inhibits amiloride-sensitive Na+ currents in Xenopus oocytes expressing alpha beta gamma-rENaC. , 1998, American Journal of Physiology.

[66]  H. Bidmon,et al.  Coronary hemodynamics in endothelial NO synthase knockout mice. , 1998, Circulation research.

[67]  M. Marletta,et al.  Potent and selective inhibition of neuronal nitric oxide synthase by N(ω)-propyl-L-arginine , 1997 .

[68]  P. Moore,et al.  Selective inhibitors of neuronal nitric oxide synthase--is no NOS really good NOS for the nervous system? , 1997, Trends in pharmacological sciences.

[69]  Richard Graham Knowles,et al.  1400W Is a Slow, Tight Binding, and Highly Selective Inhibitor of Inducible Nitric-oxide Synthase in Vitro and in Vivo* , 1997, The Journal of Biological Chemistry.

[70]  B. Mayer,et al.  Inhibition of nitric oxide synthesis by NG‐nitro‐L‐arginine methyl ester (L‐NAME): requirement for bioactivation to the free acid, NG‐nitro‐L‐arginine , 1996, British journal of pharmacology.

[71]  S Moncada,et al.  Nitric oxide synthases in mammals. , 1994, The Biochemical journal.

[72]  D. Hess,et al.  Neuronal growth cone collapse and inhibition of protein fatty acylation by nitric oxide , 1993, Nature.

[73]  S. Moncada,et al.  Characterization of three inhibitors of endothelial nitric oxide synthase in vitro and in vivo , 1990, British journal of pharmacology.

[74]  Ruth E. Hartley,et al.  An integrated view. , 1973 .

[75]  G. Schmalzing,et al.  Established Protocols for cRNA Expression and Voltage-Clamp Characterization of the P2X7 Receptor in Xenopus laevis Oocytes. , 2022, Methods in molecular biology.

[76]  F. Di Virgilio,et al.  Structure, function and techniques of investigation of the P2X7 receptor (P2X7R) in mammalian cells. , 2019, Methods in enzymology.

[77]  H. Eduardo Nitric oxide inhibits the NLRP3 inflammasome , 2013 .

[78]  W. Seeger,et al.  Nitric oxide inhibits highly selective sodium channels and the Na+/K+-ATPase in H441 cells. , 2011, American journal of respiratory cell and molecular biology.

[79]  C. Raggi,et al.  Mini review , 2004 .

[80]  W. Kummer,et al.  Nicotinic receptor alpha 7-subunits are coupled to the stimulation of nitric oxide synthase in rat dorsal root ganglion neurons. , 2003, Histochemistry and cell biology.

[81]  A. Greener,et al.  Site-directed mutagenesis using double-stranded plasmid DNA templates. , 1996, Methods in molecular biology.

[82]  B. Watkin Treasure hunting. , 1976, Nursing mirror and midwives journal.