ATP redirects cytokine trafficking and promotes novel membrane TNF signaling via microvesicles

Sanooj Soni, Kieran P O'Dea, Ying Ying Tan, Kahori Cho, Eiko Abe, Rosalba Romano, Jiang Cui, Daqing Ma, Padmini Sarathchandra, Michael R Wilson, Masao Takata, Sanooj Soni, Kieran P O'Dea, Ying Ying Tan, Kahori Cho, Eiko Abe, Rosalba Romano, Jiang Cui, Daqing Ma, Padmini Sarathchandra, Michael R Wilson, Masao Takata

Abstract

Cellular stress or injury induces release of endogenous danger signals such as ATP, which plays a central role in activating immune cells. ATP is essential for the release of nonclassically secreted cytokines such as IL-1β but, paradoxically, has been reported to inhibit the release of classically secreted cytokines such as TNF. Here, we reveal that ATP does switch off soluble TNF (17 kDa) release from LPS-treated macrophages, but rather than inhibiting the entire TNF secretion, ATP packages membrane TNF (26 kDa) within microvesicles (MVs). Secretion of membrane TNF within MVs bypasses the conventional endoplasmic reticulum- and Golgi transport-dependent pathway and is mediated by acid sphingomyelinase. These membrane TNF-carrying MVs are biologically more potent than soluble TNF in vivo, producing significant lung inflammation in mice. Thus, ATP critically alters TNF trafficking and secretion from macrophages, inducing novel unconventional membrane TNF signaling via MVs without direct cell-to-cell contact. These data have crucial implications for this key cytokine, particularly when therapeutically targeting TNF in acute inflammatory diseases.-Soni, S., O'Dea, K. P., Tan, Y. Y., Cho, K., Abe, E., Romano, R., Cui, J., Ma, D., Sarathchandra, P., Wilson, M. R., Takata, M. ATP redirects cytokine trafficking and promotes novel membrane TNF signaling via microvesicles.

Keywords: cellular communication; danger signals; extracellular vesicles; protein signalling.

Conflict of interest statement

This work was supported by grants from the Medical Research Council and British Journal of Anaesthesia (P54008) and the Chelsea and Westminster Health Charity. The authors declare no conflicts of interest.

Figures

Figure 1
Figure 1
ATP inhibits soluble TNF release. A) RAW 264.7 macrophages exposed either LPS alone (1 µg/ml, 2 h) or 2-hit injury model with LPS (1 µg/ml, 1 h) followed by ATP (3 mM, 1 h). LPS induced soluble TNF secretion into cell- and MV-depleted supernatants, which was almost entirely abolished by ATP treatment (left, n = 9–10). ATP reduced both surface [middle, represented as mean fluorescence intensity (MFI), n = 5–6] and total cellular TNF expression (right, n = 5–6). B) Confocal images clearly showed disappearance, rather than accumulation, of cellular TNF from the RAW macrophages upon ATP exposure (n = 3). C) To confirm these results in primary cells, we exposed mouse BMDMs to LPS (1 µg/ml, 1 h) followed by ATP (3 mM, 15 min). Similarly, ATP inhibited soluble TNF secretion (left, n = 6) and reduced total cellular TNF expression (middle and right, n = 6). D) Confocal images of BMDMs illustrating the reduction of cellular TNF expression following ATP treatment (n = 3). Cells were also checked for TNF expression under nonstimulated conditions (i.e., in PBS or medium) and, as expected, TNF was not detected in this sterile or noninflamed environment (Supplemental Fig. S1G). Parametric or nonparametric data are displayed as means ± sd or box-whisker plots showing the median, IQR, and minimum or maximum values, respectively. Scale bars, 10 µm. **P < 0.01, ***P < 0.001.
Figure 2
Figure 2
ATP preferentially packages membrane TNF within shed MVs. A) Scanning (left; scale bar, 1 µm) and transmission (right; scale bar, 0.2 µm) EM of RAW cells illustrates membrane blebbing and MV formation in response to ATP. B) RAW cell–derived MVs, identified as CD45+ or CD11b+ particles by flow cytometry (left and middle), significantly increased in response to ATP (right; n = 6–10). C) These MVs contained TNF, accounting for the above missing TNF from the cells but in the form of 26 kDa transmembrane pro-TNF isoform rather than 17 kDa soluble mature TNF (left and middle; n = 4–8). ATP packaged substantial amounts of pro-TNF within MVs (right; n = 4–8). D) To confirm these results in primary cells, we exposed mouse BMDMs to LPS (1 µg/ml, 1 h) followed by ATP (3 mM, 15 min). Similarly, ATP induced significant release of MVs (left; n = 6), which contained substantive amounts of pro-TNF (right; n = 6). E) Confocal microscopy illustrates that these BMDM-derived MVs, identified as CD11b+ (green, top left) particles negative for nuclear materials (DAPI−, top right), actually contained TNF (red, bottom left, colocalization shown in the bottom right combined image). Scale bars, 1 µm. F) Immune EM demonstrates transfer of pro-TNF (black dots, red arrows) from BMDMs to MVs during their formation, and these pro-TNF molecules gradually localized to MV membrane surface (hence, membrane TNF) (right). Scale bars, 0.2 µm. Parametric or nonparametric data are displayed as means ± sd or box-whisker plots showing the median, IQR, and minimum or maximum values, respectively. **P < 0.01, ***P < 0.001.
Figure 3
Figure 3
In vivo, ATP selectively packages membrane within alveolar macrophage–derived MVs while simultaneously switching off soluble TNF release. A) Mice were exposed to an in vivo 2-hit injury model: intratracheal LPS (50 ng, in 25 µl saline) instilled into murine lungs for 30 min followed by 5 mM ATP (in saline, 25 µl) for 30 min. LPS control mice consisted of intratracheal LPS followed by intratracheal saline. LPS induced soluble TNF secretion into alveoli (measured in cell- or MV-depleted BALF samples), which was substantively decreased by ATP (left; n = 5–6). ATP also caused a reduction in total cellular TNF content (measured by flow cytometry) in LPS-stimulated alveolar macrophages (middle and right), confirming that TNF was not internalized or did not accumulate within cells but disappeared from the cells (n = 5). B) In addition to inhibiting soluble TNF release, ATP significantly stimulated the release of MVs from alveolar macrophages, identified as CD45+CD11c+ particles by flow cytometry (n = 5). C) As in vitro, in vivo–derived MVs contained almost exclusively membrane TNF. Consequently, in vivo ATP preferentially packaged membrane TNF within MVs compared with LPS control MVs (n = 4). Parametric and nonparametric data are displayed as means ± sd or box-whisker plots showing the median, IQR, and minimum or maximum values, respectively. **P < 0.01.
Figure 4
Figure 4
ATP secretes membrane TNF within MVs via an unconventional, ER- and Golgi-independent pathway. A) RAW macrophages were pretreated with BFA (5 µg/ml), an inhibitor of classic cytokine secretory pathway, in order to block TNF secretion in our in vitro 2-hit model. As expected, BFA completely abolished LPS-induced, soluble-TNF release. However, even in the presence of BFA (which blocked ER- and Golgi-dependent protein transport), ATP caused a dramatic reduction in total cellular TNF content, demonstrating that TNF is still secreted in some way. TNF expression in BFA-treated cells incubated in PBS was undetectable, confirming that the accumulation of TNF within macrophages was because of LPS rather than an effect of BFA itself. B) MVs released from BFA-treated RAW cells were identified via flow cytometry as CD45+ and CD11b+ (left), and ATP caused a significant increase in MV production in these cells (right; n = 4). C) These MVs contained a substantial amount of membrane TNF (left and right; n = 4). Data are displayed as means ± sd. **P < 0.01, ***P < 0.001.
Figure 5
Figure 5
Switch of TNF secretion to nonclassic pathway is mediated by ASM. A) ASM activity was increased in RAW macrophages following ATP treatment (n = 3). B) LPS-primed RAW cells were treated with recombinant sphingomyelinase, which caused a marked increase in MV release compared with LPS alone or LPS- or ATP-treated RAW cells (n = 4–9). C) These ASM-generated MVs contain substantial amounts of membrane TNF compared with ATP-generated MVs (n = 4–7). D) Knockdown of ASM in RAW macrophages using siRNA prevented packaging of membrane TNF within MVs (n = 4). E) To confirm these results in primary cells, BMDMs were pretreated with the ASM inhibitor desipramine (25 mM) prior to exposure to LPS or LPS and ATP. Desipramine prevented ATP-induced reduction in soluble TNF release (left; n = 4), markedly attenuated ATP-induced MV release (middle; n = 4), and significantly reduced packaging of membrane TNF within MVs (right; n = 4–5). Parametric and nonparametric data are displayed as means ± sd or box-whisker plots showing the median, IQR, and minimum or maximum values respectively. NS, not significant. *P < 0.05, **P < 0.01, ***P < 0.001.
Figure 6
Figure 6
Membrane TNF is stable within macrophage-derived MVs. A) Membrane TNF is harbored in a stable, protected environment within MVs and was not spontaneously cleaved into the mature 17 kDa isoform despite being incubated at 37°C for 1 h. (n = 4). B) ATP also packages TACE into RAW cell–derived MVs (top) and in vivo–derived MVs (bottom). Confocal images of BMDM-derived MVs demonstrate the presence of TACE within BMDM-derived MVs. Scale bars, 1 μm. Data are displayed as means ± sd.
Figure 7
Figure 7
MVs provide a vehicle for membrane TNF signaling, causing significant inflammation in vivo. Membrane TNF containing BMDM-MVs from WT mice (TNF+ve MVs) were instilled intratracheally into the lungs of mice. Their biologic effects were compared with intratracheal instillation of BMDM-MVs taken from TNF−/−mice (TNF−ve MVs) and intratracheal high-dose recombinant (i.e., soluble) TNF (50 µl 100 ng/ml). TNF+ve MVs caused significant increases in infiltrating inflammatory monocytes in lung tissue (flow cytometry) (A), ICAM1 expression on alveolar epithelial cells (flow cytometry) (B), BALF CXCL1 levels (by ELISA) (C), and BALF protein (by Qubit assay) (D) compared with TNF−ve MVs and intratracheal soluble TNF. Data are displayed as means ± sd (n = 4). *P < 0.05, **P < 0.01.

References

    1. Arango Duque G., Descoteaux A. (2014) Macrophage cytokines: involvement in immunity and infectious diseases. Front. Immunol. 5, 491
    1. Lacy P. (2015) Editorial: secretion of cytokines and chemokines by innate immune cells. Front. Immunol. 6, 190
    1. Lacy P., Stow J. L. (2011) Cytokine release from innate immune cells: association with diverse membrane trafficking pathways. Blood 118, 9–18
    1. Murray R. Z., Kay J. G., Sangermani D. G., Stow J. L. (2005) A role for the phagosome in cytokine secretion. Science 310, 1492–1495
    1. Chiaruttini G., Piperno G. M., Jouve M., De Nardi F., Larghi P., Peden A. A., Baj G., Müller S., Valitutti S., Galli T., Benvenuti F. (2016) The SNARE VAMP7 regulates exocytic trafficking of interleukin-12 in dendritic cells. Cell Rep. 14, 2624–2636
    1. De Matteis M. A., Luini A. (2008) Exiting the Golgi complex. Nat. Rev. Mol. Cell Biol. 9, 273–284
    1. Afonina I. S., Müller C., Martin S. J., Beyaert R. (2015) Proteolytic processing of interleukin-1 family cytokines: variations on a common theme. Immunity 42, 991–1004
    1. Matzinger P. (1994) Tolerance, danger, and the extended family. Annu. Rev. Immunol. 12, 991–1045
    1. Trautmann A. (2009) Extracellular ATP in the immune system: more than just a “danger signal”. Sci. Signal. 2, pe6
    1. Cekic C., Linden J. (2016) Purinergic regulation of the immune system. Nat. Rev. Immunol. 16, 177–192
    1. Giuliani A. L., Sarti A. C., Falzoni S., Di Virgilio F. (2017) The P2X7 receptor-interleukin-1 liaison. Front. Pharmacol. 8, 123
    1. Rubartelli A., Cozzolino F., Talio M., Sitia R. (1990) A novel secretory pathway for interleukin-1 beta, a protein lacking a signal sequence. EMBO J. 9, 1503–1510
    1. Cohen H. B., Briggs K. T., Marino J. P., Ravid K., Robson S. C., Mosser D. M. (2013) TLR stimulation initiates a CD39-based autoregulatory mechanism that limits macrophage inflammatory responses. Blood 122, 1935–1945
    1. Dubois-Colas N., Petit-Jentreau L., Barreiro L. B., Durand S., Soubigou G., Lecointe C., Klibi J., Rezaï K., Lokiec F., Coppée J.-Y., Gicquel B., Tailleux L. (2014) Extracellular adenosine triphosphate affects the response of human macrophages infected with Mycobacterium tuberculosis. J. Infect. Dis. 210, 824–833
    1. Kawamura H., Kawamura T., Kanda Y., Kobayashi T., Abo T. (2012) Extracellular ATP-stimulated macrophages produce macrophage inflammatory protein-2 which is important for neutrophil migration. Immunology 136, 448–458
    1. Nalos M., Huang S., Sluyter R., Khan A., Santner-Nanan B., Nanan R., McLean A. S. (2008) “Host tissue damage” signal ATP impairs IL-12 and IFNgamma secretion in LPS stimulated whole human blood. Intensive Care Med. 34, 1891–1897
    1. Boucsein C., Zacharias R., Färber K., Pavlovic S., Hanisch U. K., Kettenmann H. (2003) Purinergic receptors on microglial cells: functional expression in acute brain slices and modulation of microglial activation in vitro. Eur. J. Neurosci. 17, 2267–2276
    1. Qiu P., Cui X., Sun J., Welsh J., Natanson C., Eichacker P. Q. (2013) Antitumor necrosis factor therapy is associated with improved survival in clinical sepsis trials: a meta-analysis. Crit. Care Med. 41, 2419–2429
    1. Manzanero S. (2012) Generation of mouse bone marrow-derived macrophages. Methods Mol. Biol. 844, 177–181
    1. Qu Y., Franchi L., Nunez G., Dubyak G. R. (2007) Nonclassical IL-1 beta secretion stimulated by P2X7 receptors is dependent on inflammasome activation and correlated with exosome release in murine macrophages. J. Immunol. 179, 1913–1925
    1. Soni S., Wilson M. R., O’dea K. P., Yoshida M., Katbeh U., Woods S. J., Takata M. (2016) Alveolar macrophage-derived microvesicles mediate acute lung injury. Thorax 71, 1020–1029
    1. Bianco F., Perrotta C., Novellino L., Francolini M., Riganti L., Menna E., Saglietti L., Schuchman E. H., Furlan R., Clementi E., Matteoli M., Verderio C. (2009) Acid sphingomyelinase activity triggers microparticle release from glial cells. EMBO J. 28, 1043–1054
    1. Thomas L. M., Salter R. D. (2010) Activation of macrophages by P2X7-induced microvesicles from myeloid cells is mediated by phospholipids and is partially dependent on TLR4. J. Immunol. 185, 3740–3749
    1. O’Dea K. P., Young A. J., Yamamoto H., Robotham J. L., Brennan F. M., Takata M. (2005) Lung-marginated monocytes modulate pulmonary microvascular injury during early endotoxemia. Am. J. Respir. Crit. Care Med. 172, 1119–1127
    1. Perry D. M., Newcomb B., Adada M., Wu B. X., Roddy P., Kitatani K., Siskind L., Obeid L. M., Hannun Y. A. (2014) Defining a role for acid sphingomyelinase in the p38/interleukin-6 pathway. J. Biol. Chem. 289, 22401–22412
    1. Shurety W., Pagan J. K., Prins J. B., Stow J. L. (2001) Endocytosis of uncleaved tumor necrosis factor-α in macrophages. Lab. Invest. 81, 107–117
    1. Patel B. V., Wilson M. R., O’Dea K. P., Takata M. (2013) TNF-induced death signaling triggers alveolar epithelial dysfunction in acute lung injury. J. Immunol. 190, 4274–4282
    1. Woods S. J., Waite A. A., O’Dea K. P., Halford P., Takata M., and Wilson M. R. (2015) Kinetic profiling of in vivo lung cellular inflammatory responses to mechanical ventilation. Am. J. Physiol. Lung Cell. Mol. Physiol. 308, L912–L921
    1. Shah D., Romero F., Stafstrom W., Duong M., Summer R. (2014) Extracellular ATP mediates the late phase of neutrophil recruitment to the lung in murine models of acute lung injury. Am. J. Physiol. Lung Cell. Mol. Physiol. 306, L152–L161
    1. Zhao H., Watts H. R., Chong M., Huang H., Tralau-Stewart C., Maxwell P. H., Maze M., George A. J., Ma D. (2013) Xenon treatment protects against cold ischemia associated delayed graft function and prolongs graft survival in rats. Am. J. Transplant. 13, 2006–2018
    1. Xuan W., Zhao H., Hankin J., Chen L., Yao S., Ma D. (2016) Local anesthetic bupivacaine induced ovarian and prostate cancer apoptotic cell death and underlying mechanisms in vitro. Sci. Rep. 6, 26277
    1. Pizzirani C., Ferrari D., Chiozzi P., Adinolfi E., Sandonà D., Savaglio E., Di Virgilio F. (2007) Stimulation of P2 receptors causes release of IL-1β-loaded microvesicles from human dendritic cells. Blood 109, 3856–3864
    1. MacKenzie A., Wilson H. L., Kiss-Toth E., Dower S. K., North R. A., Surprenant A. (2001) Rapid secretion of interleukin-1β by microvesicle shedding. Immunity 15, 825–835
    1. Gulinelli S., Salaro E., Vuerich M., Bozzato D., Pizzirani C., Bolognesi G., Idzko M., Di Virgilio F., Ferrari D. (2012) IL-18 associates to microvesicles shed from human macrophages by a LPS/TLR-4 independent mechanism in response to P2X receptor stimulation. Eur. J. Immunol. 42, 3334–3345
    1. Perregaux D., Gabel C. A. (1994) Interleukin-1 beta maturation and release in response to ATP and nigericin. Evidence that potassium depletion mediated by these agents is a necessary and common feature of their activity. J. Biol. Chem. 269, 15195–15203
    1. Laliberte R. E., Eggler J., Gabel C. A. (1999) ATP treatment of human monocytes promotes caspase-1 maturation and externalization. J. Biol. Chem. 274, 36944–36951
    1. Solle M., Labasi J., Perregaux D. G., Stam E., Petrushova N., Koller B. H., Griffiths R. J., Gabel C. A. (2001) Altered cytokine production in mice lacking P2X(7) receptors. J. Biol. Chem. 276, 125–132
    1. Kunder C. A., St John A. L., Li G., Leong K. W., Berwin B., Staats H. F., Abraham S. N. (2009) Mast cell-derived particles deliver peripheral signals to remote lymph nodes. J. Exp. Med. 206, 2455–2467
    1. Fehrenbach H., Zissel G., Goldmann T., Tschernig T., Vollmer E., Pabst R., Müller-Quernheim J. (2003) Alveolar macrophages are the main source for tumour necrosis factor-α in patients with sarcoidosis. Eur. Respir. J. 21, 421–428
    1. Wilson M. R., Choudhury S., Takata M. (2005) Pulmonary inflammation induced by high-stretch ventilation is mediated by tumor necrosis factor signaling in mice. Am. J. Physiol. Lung Cell. Mol. Physiol. 288, L599–L607
    1. Dinter A., Berger E. G. (1998) Golgi-disturbing agents. Histochem. Cell Biol. 109, 571–590
    1. Engele M., Stössel E., Castiglione K., Schwerdtner N., Wagner M., Bölcskei P., Röllinghoff M., Stenger S. (2002) Induction of TNF in human alveolar macrophages as a potential evasion mechanism of virulent Mycobacterium tuberculosis. J. Immunol. 168, 1328–1337
    1. Vicetti Miguel R. D., Maryak S. A., Cherpes T. L. (2012) Brefeldin A, but not monensin, enables flow cytometric detection of interleukin-4 within peripheral T cells responding to ex vivo stimulation with Chlamydia trachomatis. J. Immunol. Methods 384, 191–195
    1. Black R. A., Rauch C. T., Kozlosky C. J., Peschon J. J., Slack J. L., Wolfson M. F., Castner B. J., Stocking K. L., Reddy P., Srinivasan S., Nelson N., Boiani N., Schooley K. A., Gerhart M., Davis R., Fitzner J. N., Johnson R. S., Paxton R. J., March C. J., Cerretti D. P. (1997) A metalloproteinase disintegrin that releases tumour-necrosis factor-α from cells. Nature 385, 729–733
    1. Scott A. J., O’Dea K. P., O’Callaghan D., Williams L., Dokpesi J. O., Tatton L., Handy J. M., Hogg P. J., Takata M. (2011) Reactive oxygen species and p38 mitogen-activated protein kinase mediate tumor necrosis factor α-converting enzyme (TACE/ADAM-17) activation in primary human monocytes. J. Biol. Chem. 286, 35466–35476
    1. Patel B. V., Wilson M. R., Takata M. (2012) Resolution of acute lung injury and inflammation: a translational mouse model. Eur. Respir. J. 39, 1162–1170
    1. Rittirsch D., Flierl M. A., Day D. E., Nadeau B. A., McGuire S. R., Hoesel L. M., Ipaktchi K., Zetoune F. S., Sarma J. V., Leng L., Huber-Lang M. S., Neff T. A., Bucala R., Ward P. A. (2008) Acute lung injury induced by lipopolysaccharide is independent of complement activation. J. Immunol. 180, 7664–7672
    1. Dorr A. D., Wilson M. R., Wakabayashi K., Waite A. C., Patel B. V., van Rooijen N., O’Dea K. P., Takata M. (2011) Sources of alveolar soluble TNF receptors during acute lung injury of different etiologies. J. Appl. Physiol. 111, 177–184
    1. Mehta V. B., Hart J., Wewers M. D. (2001) ATP-stimulated release of interleukin (IL)-1β and IL-18 requires priming by lipopolysaccharide and is independent of caspase-1 cleavage. J. Biol. Chem. 276, 3820–3826
    1. Beckmann N., Sharma D., Gulbins E., Becker K. A., Edelmann B. (2014) Inhibition of acid sphingomyelinase by tricyclic antidepressants and analogons. Front. Physiol. 5, 331
    1. Horiuchi T., Mitoma H., Harashima S., Tsukamoto H., Shimoda T. (2010) Transmembrane TNF-alpha: structure, function and interaction with anti-TNF agents. Rheumatology (Oxford) 49, 1215–1228
    1. Perez C., Albert I., DeFay K., Zachariades N., Gooding L., Kriegler M. (1990) A nonsecretable cell surface mutant of tumor necrosis factor (TNF) kills by cell-to-cell contact. Cell 63, 251–258
    1. Salome R. G., McCoy D. M., Ryan A. J., Mallampalli R. K. (2000) Effects of intratracheal instillation of TNF-α on surfactant metabolism. J. Appl. Physiol. 88, 10–16
    1. Corti A., Fassina G., Marcucci F., Barbanti E., Cassani G. (1992) Oligomeric tumour necrosis factor α slowly converts into inactive forms at bioactive levels. Biochem. J. 284, 905–910
    1. Drago F., Lombardi M., Prada I., Gabrielli M., Joshi P., Cojoc D., Franck J., Fournier I., Vizioli J., Verderio C. (2017) ATP modifies the proteome of extracellular vesicles released by microglia and influences their action on astrocytes. Front. Pharmacol. 8, 910
    1. Zhang H.-G., Liu C., Su K., Yu S., Zhang L., Zhang S., Wang J., Cao X., Grizzle W., Kimberly R. P. (2006) A membrane form of TNF-alpha presented by exosomes delays T cell activation-induced cell death. J. Immunol. 176, 7385–7393; erratum: 177, 2025
    1. Van der Pol E., Böing A. N., Harrison P., Sturk A., Nieuwland R. (2012) Classification, functions, and clinical relevance of extracellular vesicles. Pharmacol. Rev. 64, 676–705
    1. Levin C., Koren A., Rebibo-Sabbah A., Koifman N., Brenner B., Aharon A. (2018) Extracellular vesicle characteristics in β-thalassemia as potential biomarkers for spleen functional status and ineffective erythropoiesis. Front. Physiol. 9, 1214
    1. Bourdonnay E., Zasłona Z., Penke L. R. K., Speth J. M., Schneider D. J., Przybranowski S., Swanson J. A., Mancuso P., Freeman C. M., Curtis J. L. (2015) Transcellular delivery of vesicular SOCS proteins from macrophages to epithelial cells blunts inflammatory signaling. J. Exp. Med. 212, 729–742
    1. Jiang H., Hampel H., Prvulovic D., Wallin A., Blennow K., Li R., Shen Y. (2011) Elevated CSF levels of TACE activity and soluble TNF receptors in subjects with mild cognitive impairment and patients with Alzheimer’s disease. Mol. Neurodegener. 6, 69
    1. Barberà-Cremades M., Gómez A. I., Baroja-Mazo A., Martínez-Alarcón L., Martínez C. M., de Torre-Minguela C., Pelegrín P. (2017) P2X7 receptor induces tumor necrosis Factor-α converting enzyme activation and release to boost TNF-α production. Front. Immunol. 8, 862

Source: PubMed

3
Abonnere