Microglia and Neuroinflammation: What Place for P2RY12?
Abstract
:1. Introduction
2. P2RY12
2.1. Purinergic Receptor Families
2.2. Structure
2.3. Expression
2.4. Functions
2.4.1. Platelet Aggregation
2.4.2. T Cell Activation
2.4.3. Vascular Effects
2.4.4. Bone Remodeling
2.4.5. Microglia Functions: Motility and Migration
3. Pharmacological and Genetic Tools for Studying P2RY12
4. Contribution of Microglial P2RY12 to Neuroinflammation
4.1. Cerebral Ischemia
4.2. Traumatic Brain Injury
4.3. Epilepsy
4.4. Glial Tumors
4.5. Alzheimer’s Disease
4.6. Multiple Sclerosis
4.7. Amyotrophic Lateral Sclerosis
5. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
Abbreviations
Aβ | Amyloid beta |
AC | Adenylate cyclase |
AD | Alzheimer’s disease |
ADP | Adenosine diphosphate |
ALS | Amyotrophic lateral sclerosis |
AR | Adenosine receptor |
ATP | Adenosine triphosphate |
BBB | Blood-brain barrier |
cAMP | Cyclic adenosine monophosphate |
CD39 | Cluster differentiation 39 |
CX3CL1 | C-X3-C motif chemokine ligand |
CX3CR1 | C-X3-C motif chemokine receptor 1 |
Cys | Cysteine |
EAE | Experimental autoimmune encephalomyelitis |
ERK | Extracellular signal-regulated kinase |
GPCR | G protein-coupled receptor |
IL | Interleukin |
KO | Knockout |
OGD | Oxygen-glucose deprivation |
MCP-1 | Monocyte chemoattractant protein-1 |
MPC | Microglial process convergence |
mRNA | Messenger ribonucleic acid |
MS | Multiple sclerosis |
NLRP3 | Nod-like receptor protein 3 |
PI3K | Phosphoinositide 3-kinase |
PKA | Protein kinase A |
PKC | Protein kinase C |
PLC | Phospholipase C |
siRNA | Silent ribonucleic acid |
SOD1 | Superoxide dismutase 1 |
TBI | Traumatic brain injury |
THIK-1 | TWIK-related halothane-inhibited K+ channel |
UDP | Uridine diphosphate |
UTP | Uridine triphosphate |
VASP | Vasodilator-stimulated phosphoprotein |
WT | Wild-type |
References
- Chagas, L.; Sandre, P.C.; Ribeiro E Ribeiro, N.; Marcondes, H.; Oliveira Silva, P.; Savino, W.; Serfaty, C.A. Environmental signals on microglial function during brain development, neuroplasticity, and disease. Int. J. Mol. Sci. 2020, 21, 2111. [Google Scholar] [CrossRef] [Green Version]
- Burnstock, G. Purinergic nerves. Pharmacol. Rev. 1972, 24, 509–581. [Google Scholar] [PubMed]
- Burnstock, G. A basis for distinguishing two types of purinergic receptor. In Cell Membrane Receptors for Drugs and Hormones: A Multidisciplinary Approach; Straub, R.W., Bolis, L., Eds.; Raven Press: New York, NY, USA, 1978; pp. 107–118. [Google Scholar]
- Cheffer, A.; Castillo, A.; Corrêa-Velloso, J.; Gonçalves, M.; Naaldijk, Y.; Nascimento, I.C.; Burnstock, G.; Ulrich, H. Purinergic system in psychiatric diseases. Mol. Psychiatry 2018, 23, 94–106. [Google Scholar] [CrossRef] [PubMed]
- Burnstock, G. Purine and purinergic receptors. Brain Neurosci. Adv. 2018, 2, 2398212818817494. [Google Scholar] [CrossRef] [Green Version]
- von Kügelgen, I. Pharmacology of P2Y receptors. Brain Res. Bull. 2019, 151, 12–24. [Google Scholar] [CrossRef]
- Zhang, J.; Zhang, K.; Gao, Z.G.; Paoletta, S.; Zhang, D.; Han, G.W.; Li, T.; Ma, L.; Zhang, W.; Müller, C.E.; et al. Agonist-bound structure of the human P2Y12 receptor. Nature 2014, 509, 119–122. [Google Scholar] [CrossRef]
- Zhang, K.; Zhang, J.; Gao, Z.G.; Zhang, D.; Zhu, L.; Han, G.W.; Moss, S.M.; Paoletta, S.; Kiselev, E.; Lu, W.; et al. Structure of the human P2Y12 receptor in complex with an antithrombotic drug. Nature 2014, 509, 115–118. [Google Scholar] [CrossRef] [Green Version]
- Gachet, C. P2Y(12) receptors in platelets and other hematopoietic and non-hematopoietic cells. Purinergic Signal. 2012, 8, 609–619. [Google Scholar] [CrossRef]
- Liverani, E.; Kilpatrick, L.E.; Tsygankov, A.Y.; Kunapuli, S.P. The role of P2Y12 receptor and activated platelets during inflammation. Curr. Drug Targets 2014, 15, 720–728. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jacobson, K.A.; Delicado, E.G.; Gachet, C.; Kennedy, C.; von Kügelgen, I.; Li, B.; Miras-Portugal, M.T.; Novak, I.; Schöneberg, T.; Perez-Sen, R.; et al. Update of P2Y receptor pharmacology: Iuphar Review 27. Br. J. Pharmacol. 2020, 177, 2413–2433. [Google Scholar] [CrossRef]
- Mansour, A.; Bachelot-Loza, C.; Nesseler, N.; Gaussem, P.; Gouin-Thibault, I. P2Y12 inhibition beyond thrombosis: Effects on inflammation. Int. J. Mol. Sci. 2020, 21, 1391. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Haynes, S.E.; Hollopeter, G.; Yang, G.; Kurpius, D.; Dailey, M.E.; Gan, W.B.; Julius, D. The P2Y12 receptor regulates microglial activation by extracellular nucleotides. Nat. Neurosci. 2006, 9, 1512–1519. [Google Scholar] [CrossRef]
- Moore, C.S.; Ase, A.R.; Kinsara, A.; Rao, V.T.; Michell-Robinson, M.; Leong, S.Y.; Butovsky, O.; Ludwin, S.K.; Séguéla, P.; Bar-Or, A.; et al. P2Y12 expression and function in alternatively activated human microglia. Neurol. Neuroimmunol. Neuroinflamm. 2015, 2, e80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mildner, A.; Huang, H.; Radke, J.; Stenzel, W.; Priller, J. P2Y12 receptor is expressed on human microglia under physiological conditions throughout development and is sensitive to neuroinflammatory diseases. Glia 2017, 65, 375–387. [Google Scholar] [CrossRef] [PubMed]
- Kronlage, M.; Song, J.; Sorokin, L.; Isfort, K.; Schwerdtle, T.; Leipziger, J.; Robaye, B.; Conley, P.B.; Kim, H.-C.; Sargin, S.; et al. Autocrine purinergic receptor signaling is essential for macrophage chemotaxis. Sci. Signal. 2010, 3, ra55. [Google Scholar] [CrossRef] [PubMed]
- Amadio, S.; Parisi, C.; Montilli, C.; Carrubba, A.S.; Apolloni, S.; Volonté, C. P2Y(12) receptor on the verge of a neuroinflammatory breakdown. Mediat. Inflamm. 2014, 2014, 975849. [Google Scholar] [CrossRef] [Green Version]
- Cserép, C.; Pósfai, B.; Lénárt, N.; Fekete, R.; László, Z.I.; Lele, Z.; Orsolits, B.; Molnár, G.; Heindl, S.; Schwarcz, A.D.; et al. Microglia monitor and protect neuronal function through specialized somatic purinergic junctions. Science 2020, 367, 528–537. [Google Scholar] [CrossRef] [PubMed]
- Butovsky, O.; Jedrychowski, M.P.; Moore, C.S.; Cialic, R.; Lanser, A.J.; Gabriely, G.; Koeglsperger, T.; Dake, B.; Wu, P.M.; Doykan, C.E.; et al. Identification of a unique TGF-β-dependent molecular and functional signature in microglia. Nat. Neurosci. 2014, 17, 131–143. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Y.; Chen, K.; Sloan, S.A.; Bennett, M.L.; Scholze, A.R.; O’Keeffe, S.; Phatnani, H.P.; Guarnieri, P.; Caneda, C.; Ruderisch, N.; et al. An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. J. Neurosci. 2014, 34, 11929–11947. [Google Scholar] [CrossRef]
- Lou, N.; Takano, T.; Pei, Y.; Xavier, A.L.; Goldman, S.A.; Nedergaard, M. Purinergic receptor P2RY12-dependent microglial closure of the injured blood-brain barrier. Proc. Natl. Acad. Sci. USA 2016, 113, 1074–1079. [Google Scholar] [CrossRef] [Green Version]
- Galatro, T.F.; Holtman, I.R.; Lerario, A.M.; Vainchtein, I.D.; Brouwer, N.; Sola, P.R.; Veras, M.M.; Pereira, T.F.; Leite, R.E.P.; Möller, T.; et al. Transcriptomic analysis of purified human cortical microglia reveals age-associated changes. Nat. Neurosci. 2017, 20, 1162–1171. [Google Scholar] [CrossRef]
- Crain, J.M.; Nikodemova, M.; Watters, J.J. Expression of P2 nucleotide receptors varies with age and sex in murine brain microglia. J. Neuroinflamm. 2009, 6, 24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Giossi, A.; Pezzini, A.; Del Zotto, E.; Volonghi, I.; Costa, P.; Ferrari, D.; Padovani, A. Advances in antiplatelet therapy for stroke prevention: The new P2Y12 antagonists. Curr. Drug Targets 2010, 11, 380–391. [Google Scholar] [CrossRef] [PubMed]
- Cattaneo, M. P2Y12 receptors: Structure and function. J. Thromb. Haemost. 2015, 13, S10–S16. [Google Scholar] [CrossRef] [PubMed]
- Rossaint, J.; Margraf, A.; Zarbock, A. Role of platelets in leukocyte recruitment and resolution of inflammation. Front. Immunol. 2018, 9, 2712. [Google Scholar] [CrossRef]
- Ben Addi, A.; Cammarata, D.; Conley, P.B.; Boeynaems, J.M.; Robaye, B. Role of the P2Y12 receptor in the modulation of murine dendritic cell function by ADP. J. Immunol. 2010, 185, 5900–5906. [Google Scholar] [CrossRef] [Green Version]
- Satonaka, H.; Nagata, D.; Takahashi, M.; Kiyosue, A.; Myojo, M.; Fujita, D.; Ishimitsu, T.; Nagano, T.; Nagai, R.; Hirata, Y. Involvement of P2Y12 receptor in vascular smooth muscle inflammatory changes via MCP-1 upregulation and monocyte adhesion. Am. J. Physiol. Heart Circ. Physiol. 2015, 308, H853–H861. [Google Scholar] [CrossRef] [Green Version]
- Colonna, M.; Butovsky, O. Microglia function in the central nervous system during health and neurodegeneration. Annu. Rev. Immunol. 2017, 35, 441–468. [Google Scholar] [CrossRef]
- Smolders, S.M.; Kessels, S.; Vangansewinkel, T.; Rigo, J.M.; Legendre, P.; Brône, B. Microglia: Brain cells on the move. Prog. Neurobiol. 2019, 178, 101612. [Google Scholar] [CrossRef]
- Wolf, S.A.; Boddeke, H.W.; Kettenmann, H. Microglia in physiology and disease. Annu. Rev. Physiol. 2017, 79, 619–643. [Google Scholar] [CrossRef]
- Askew, K.; Gomez-Nicola, D. A story of birth and death: Insights into the formation and dynamics of the microglial population. Brain Behav. Immun. 2018, 69, 9–17. [Google Scholar] [CrossRef] [PubMed]
- Kabba, J.A.; Xu, Y.; Christian, H.; Ruan, W.; Chenai, K.; Xiang, Y.; Zhang, L.; Saavedra, J.M.; Pang, T. Microglia: Housekeeper of the central nervous system. Cell. Mol. Neurobiol. 2018, 38, 53–71. [Google Scholar] [CrossRef] [PubMed]
- Szepesi, Z.; Manouchehrian, O.; Bachiller, S.; Deierborg, T. Bidirectional microglia-neuron communication in health and disease. Front. Cell. Neurosci. 2018, 12, 323. [Google Scholar] [CrossRef]
- Madry, C.; Kyrargyri, V.; Arancibia-Cárcamo, I.L.; Jolivet, R.; Kohsaka, S.; Bryan, R.M.; Attwell, D. Microglial ramification, surveillance, and interleukin-1β release are regulated by the two-pore domain K+ channel THIK-1. Neuron 2018, 97, 299–312.e6. [Google Scholar] [CrossRef]
- Sipe, G.O.; Lowery, R.L.; Tremblay, M.È.; Kelly, E.A.; Lamantia, C.E.; Majewska, A.K. Microglial P2Y12 is necessary for synaptic plasticity in mouse visual cortex. Nat. Commun. 2016, 7, 10905. [Google Scholar] [CrossRef]
- Illes, P.; Rubini, P.; Ulrich, H.; Zhao, Y.; Tang, Y. Regulation of microglial functions by purinergic mechanisms in the healthy and diseased CNS. Cells 2020, 9, 1108. [Google Scholar] [CrossRef]
- Rivera, A.; Vanzulli, I.; Butt, A.M. A central role for ATP signalling in glial interactions in the CNS. Curr. Drug Targets 2016, 17, 1829–1833. [Google Scholar] [CrossRef] [PubMed]
- Fekete, R.; Cserép, C.; Lénárt, N.; Tóth, K.; Orsolits, B.; Martinecz, B.; Méhes, E.; Szabó, B.; Németh, V.; Gönci, B.; et al. Microglia control the spread of neurotropic virus infection via P2Y12 signalling and recruit monocytes through P2Y12-independent mechanisms. Acta Neuropathol. 2018, 136, 461–482. [Google Scholar] [CrossRef] [Green Version]
- Fan, Y.; Xie, L.; Chung, C.Y. Signaling pathways controlling microglia chemotaxis. Mol. Cells 2017, 40, 163–168. [Google Scholar] [PubMed] [Green Version]
- Weiner, O.D.; Neilsen, P.O.; Prestwich, G.D.; Kirschner, M.W.; Cantley, L.C.; Bourne, H.R. A PtdInsP(3)- and Rho GTPase-mediated positive feedback loop regulates neutrophil polarity. Nat. Cell Biol. 2002, 4, 509–513. [Google Scholar] [CrossRef] [Green Version]
- Eyo, U.B.; Peng, J.; Murugan, M.; Mo, M.; Lalani, A.; Xie, P.; Xu, P.; Margolis, D.J.; Wu, L.J. Regulation of physical microglia-neuron interactions by fractalkine signaling after status epilepticus. eNeuro 2017, 3, 6. [Google Scholar] [CrossRef] [PubMed]
- Whitelaw, B.S. Microglia-mediated synaptic elimination in neuronal development and disease. J Neurophysiol. 2018, 119, 1–4. [Google Scholar] [CrossRef] [PubMed]
- Pósfai, B.; Cserép, C.; Orsolits, B.; Dénes, Á. New insights into microglia-neuron interactions: A neuron’s perspective. Neuroscience 2019, 405, 103–117. [Google Scholar] [CrossRef] [Green Version]
- Koizumi, S.; Ohsawa, K.; Inoue, K.; Kohsaka, S. Purinergic receptors in microglia: Functional modal shifts of microglia mediated by P2 and P1 receptors. Glia 2013, 61, 47–54. [Google Scholar] [CrossRef] [PubMed]
- Damani, M.R.; Zhao, L.; Fontainhas, A.M.; Amaral, J.; Fariss, R.N.; Wong, W.T. Age-related alterations in the dynamic behavior of microglia. Aging Cell 2011, 10, 263–276. [Google Scholar] [CrossRef] [Green Version]
- Calovi, S.; Mut-Arbona, P.; Sperlágh, B. Microglia and the purinergic signaling system. Neuroscience 2019, 405, 137–147. [Google Scholar] [CrossRef] [PubMed]
- Springthorpe, B.; Bailey, A.; Barton, P.; Birkinshaw, T.N.; Bonnert, R.V.; Brown, R.C.; Chapman, D.; Dixon, J.; Guile, S.D.; Humphries, R.G.; et al. From ATP to AZD6140: The discovery of an orally active reversible P2Y12 receptor antagonist for the prevention of thrombosis. Bioorg. Med. Chem. Lett. 2007, 17, 6013–6018. [Google Scholar] [CrossRef]
- Färber, K.; Markworth, S.; Pannasch, U.; Nolte, C.; Prinz, V.; Kronenberg, G.; Gertz, K.; Endres, M.; Bechmann, I.; Enjyoji, K.; et al. The ectonucleotidase cd39/ENTPDase1 modulates purinergic-mediated microglial migration. Glia 2008, 56, 331–341. [Google Scholar] [CrossRef]
- Matyash, M.; Zabiegalov, O.; Wendt, S.; Matyash, V.; Kettenmann, H. The adenosine generating enzymes CD39/CD73 control microglial processes ramification in the mouse brain. PLoS ONE 2017, 12, e0175012. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Braun, N.; Sévigny, J.; Robson, S.C.; Enjyoji, K.; Guckelberger, O.; Hammer, K.; Di Virgilio, F.; Zimmermann, H. Assignment of ecto-nucleoside triphosphate diphosphohydrolase-1/cd39 expression to microglia and vasculature of the brain. Eur. J. Neurosci. 2000, 12, 4357–4366. [Google Scholar]
- Högberg, C.; Gidlöf, O.; Deflorian, F.; Jacobson, K.A.; Abdelrahman, A.; Müller, C.E.; Olde, B.; Erlinge, D. Farnesyl pyrophosphate is an endogenous antagonist to ADP-stimulated P2Y₁₂ receptor-mediated platelet aggregation. Thromb. Haemost. 2012, 108, 119–132. [Google Scholar] [CrossRef] [Green Version]
- Garcia, C.; Maurel-Ribes, A.; Nauze, M.; N’Guyen, D.; Martinez, L.O.; Payrastre, B.; Sénard, J.-M.; Galés, C.; Pons, V. Deciphering biased inverse agonism of cangrelor and ticagrelor at P2Y12 receptor. Cell. Mol. Life Sci. 2019, 76, 561–576. [Google Scholar] [CrossRef]
- Cattaneo, M. New P2Y(12) inhibitors. Circulation 2010, 121, 171–179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- van Giezen, J.J.; Humphries, R.G. Preclinical and clinical studies with selective reversible direct P2Y12 antagonists. Semin. Thromb. Hemost. 2005, 31, 195–204. [Google Scholar] [CrossRef] [PubMed]
- Ford, N.F. The metabolism of clopidogrel: CYP2C19 is a minor pathway. J. Clin. Pharmacol. 2016, 56, 1474–1483. [Google Scholar] [CrossRef] [PubMed]
- Algaier, I.; Jakubowski, J.A.; Asai, F.; von Kügelgen, I. Interaction of the active metabolite of prasugrel, R-138727, with cysteine 97 and cysteine 175 of the human P2Y12 receptor. J. Thromb. Haemost. 2008, 6, 1908–1914. [Google Scholar] [CrossRef] [PubMed]
- Thebault, J.J.; Blatrix, C.E.; Blanchard, J.F.; Panak, E.A. Effects of ticlopidine, a new platelet aggregation inhibitor in man. Clin. Pharmacol. Ther. 1975, 18, 485–490. [Google Scholar] [CrossRef]
- Yoneda, K.; Iwamura, R.; Kishi, H.; Mizukami, Y.; Mogami, K.; Kobayashi, S. Identification of the active metabolite of ticlopidine from rat in vitro metabolites. Br. J. Pharmacol. 2004, 142, 551–557. [Google Scholar] [CrossRef] [Green Version]
- Remko, M.; Remková, A.; Broer, R. A comparative study of molecular structure, pKa, lipophilicity, solubility, absorption and polar surface area of some antiplatelet drugs. Int. J. Mol. Sci. 2016, 17, 388. [Google Scholar] [CrossRef] [Green Version]
- Trujillo, C.A.; Nery, A.A.; Martins, A.H.; Majumder, P.; Gonzalez, F.A.; Ulrich, H. Inhibition mechanism of the recombinant rat P2X(2) receptor in glial cells by suramin and TNP-ATP. Biochemistry 2006, 45, 224–233. [Google Scholar] [CrossRef]
- Von Kügelgen, I.; Hoffmann, K. Pharmacology and structure of P2Y receptors. Neuropharmacology 2016, 104, 50–61. [Google Scholar] [CrossRef]
- Yanachkov, I.B.; Chang, H.; Yanachkova, M.I.; Dix, E.J.; Berny-Lang, M.A.; Gremmel, T.; Michelson, A.D.; Wright, G.E.; Frelinger, A.L., 3rd. New highly active antiplatelet agents with dual specificity for platelet P2Y1 and P2Y12 adenosine diphosphate receptors. Eur. J. Med. Chem. 2016, 107, 204–218. [Google Scholar] [CrossRef] [Green Version]
- Gündüz, D.; Tanislav, C.; Sedding, D.; Parahuleva, M.; Santoso, S.; Troidl, C.; Hamm, C.W.; Aslam, M. Uridine triphosphate thio analogues inhibit platelet P2Y12 receptor and aggregation. Int. J. Mol. Sci. 2017, 18, 269. [Google Scholar] [CrossRef] [Green Version]
- Von Kügelgen, I. Pharmacological profiles of cloned mammalian P2Y-receptor subtypes. Pharmacol. Ther. 2006, 110, 415–432. [Google Scholar] [CrossRef] [PubMed]
- Le Duc, D.; Schulz, A.; Lede, V.; Schulze, A.; Thor, D.; Brüser, A.; Schöneberg, T. P2Y receptors in immune response and inflammation. Adv. Immunol. 2017, 136, 85–121. [Google Scholar]
- DiSabato, D.J.; Quan, N.; Godbout, J.P. Neuroinflammation: The devil is in the details. J. Neurochem. 2016, 139, 136–153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shabab, T.; Khanabdali, R.; Moghadamtousi, S.Z.; Kadir, H.A.; Mohan, G. Neuroinflammation pathways: A general review. Int. J. Neurosci. 2017, 127, 624–633. [Google Scholar] [CrossRef] [PubMed]
- Yong, H.; Rawji, K.S.; Ghorbani, S.; Xue, M.; Yong, V.W. The benefits of neuroinflammation for the repair of the injured central nervous system. Cell. Mol. Immunol. 2019, 16, 540–546. [Google Scholar] [CrossRef]
- Colton, C.A. Heterogeneity of microglial activation in the innate immune response in the brain. J. Neuroimmune Pharmacol. 2009, 4, 399–418. [Google Scholar] [CrossRef] [Green Version]
- Ransohoff, R.M. A polarizing question: Do M1 and M2 microglia exist? Nat. Neurosci. 2016, 19, 987–991. [Google Scholar] [CrossRef] [PubMed]
- Stratoulias, V.; Venero, J.L.; Tremblay, M.È.; Joseph, B. Microglial subtypes: Diversity within the microglial community. EMBO J. 2019, 38, e101997. [Google Scholar] [CrossRef] [PubMed]
- Beamer, E.; Gölöncsér, F.; Horváth, G.; Bekő, K.; Otrokocsi, L.; Koványi, B.; Sperlágh, B. Purinergic mechanisms in neuroinflammation: An update from molecules to behavior. Neuropharmacology 2016, 104, 94–104. [Google Scholar] [CrossRef] [Green Version]
- Voet, S.; Srinivasan, S.; Lamkanfi, M.; van Loo, G. Inflammasomes in neuroinflammatory and neurodegenerative diseases. EMBO Mol. Med. 2019, 11, e10248. [Google Scholar] [CrossRef] [PubMed]
- Song, L.; Pei, L.; Yao, S.; Wu, Y.; Shang, Y. NLRP3 inflammasome in neurological diseases, from functions to therapies. Front. Cell. Neurosci. 2017, 11, 63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Suzuki, T.; Kohyama, K.; Moriyama, K.; Ozaki, M.; Hasegawa, S.; Ueno, T.; Saitoe, M.; Morio, T.; Hayashi, M.; Sakuma, H. Extracellular ADP augments microglial inflammasome and NF-κB activation via the P2Y12 receptor. Eur. J. Immunol. 2020, 50, 205–219. [Google Scholar] [CrossRef] [PubMed]
- Rodrigues, R.J.; Tomé, A.R.; Cunha, R.A. ATP as a multi-target danger signal in the brain. Front. Neurosci. 2015, 9, 148. [Google Scholar] [CrossRef] [Green Version]
- Webster, C.M.; Hokari, M.; McManus, A.; Tang, X.N.; Ma, H.; Kacimi, R.; Yenari, M.A. Microglial P2Y12 deficiency/inhibition protects against brain ischemia. PLoS ONE 2013, 8, e70927. [Google Scholar] [CrossRef] [Green Version]
- Gelosa, P.; Lecca, D.; Fumagalli, M.; Wypych, D.; Pignieri, A.; Cimino, M.; Verderio, C.; Enerbäck, M.; Nikookhesal, E.; Tremoli, E.; et al. Microglia is a key player in the reduction of stroke damage promoted by the new antithrombotic agent ticagrelor. J. Cereb. Blood Flow Metab. 2014, 34, 979–988. [Google Scholar] [CrossRef] [Green Version]
- Taib, T.; Leconte, C.; Van Steenwinckel, J.; Cho, A.H.; Palmier, B.; Torsello, E.; Kuen, R.L.; Onyeomah, S.; Ecomard, K.; Benedetto, C.; et al. Neuroinflammation, myelin and behavior: Temporal patterns following mild traumatic brain injury in mice. PLoS ONE 2017, 12, e0184811. [Google Scholar]
- Kumar, A.; Stoica, B.A.; Loane, D.J.; Yang, M.; Abulwerdi, G.; Khan, N.; Kumar, A.; Thom, S.R.; Faden, A.I. Microglial-derived microparticles mediate neuroinflammation after traumatic brain injury. J. Neuroinflammation 2017, 14, 47. [Google Scholar] [CrossRef] [Green Version]
- Eyo, U.B.; Murugan, M.; Wu, L.J. Microglia-neuron communication in epilepsy. Glia 2017, 65, 5–18. [Google Scholar] [CrossRef] [Green Version]
- Avignone, E.; Ulmann, L.; Levavasseur, F.; Rassendren, F.; Audinat, E. Status epilepticus induces a particular microglial activation state characterized by enhanced purinergic signaling. J. Neurosci. 2008, 28, 9133–9144. [Google Scholar] [CrossRef] [Green Version]
- Eyo, U.B.; Peng, J.; Swiatkowski, P.; Mukherjee, A.; Bispo, A.; Wu, L.J. Neuronal hyperactivity recruits microglial processes via neuronal NMDA receptors and microglial P2Y12 receptors after status epilepticus. J. Neurosci. 2014, 34, 10528–10540. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Inoue, K. UDP facilitates microglial phagocytosis through P2Y6 receptors. Cell Adh. Migr. 2007, 1, 131–132. [Google Scholar] [CrossRef] [Green Version]
- Zhu, C.; Kros, J.M.; van der Weiden, M.; Zheng, P.; Cheng, C.; Mustafa, D.A. Expression site of P2RY12 in residential microglial cells in astrocytomas correlates with M1 and M2 marker expression and tumor grade. Acta Neuropathol. Commun. 2017, 5, 4. [Google Scholar] [CrossRef] [Green Version]
- Walker, D.G.; Tang, T.M.; Mendsaikhan, A.; Tooyama, I.; Serrano, G.E.; Sue, L.I.; Beach, T.G.; Lue, L.F. Patterns of expression of purinergic receptor P2RY12, a putative marker for non-activated microglia, in aged and Alzheimer’s disease brains. Int. J. Mol. Sci. 2020, 21, 678. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sanchez-Mejias, E.; Navarro, V.; Jimenez, S.; Sanchez-Mico, M.; Sanchez-Varo, R.; Nuñez-Diaz, C.; Trujillo-Estrada, L.; Davila, J.C.; Vizuete, M.; Gutierrez, A.; et al. Soluble phospho-tau from Alzheimer’s disease hippocampus drives microglial degeneration. Acta Neuropathol. 2016, 132, 897–916. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jay, T.R.; Miller, C.M.; Cheng, P.J.; Graham, L.C.; Bemiller, S.; Broihier, M.L.; Xu, G.; Margevicius, D.; Karlo, J.C.; Sousa, G.L.; et al. TREM2 deficiency eliminates TREM2+ inflammatory macrophages and ameliorates pathology in Alzheimer’s disease mouse models. J. Exp. Med. 2015, 212, 287–295. [Google Scholar] [CrossRef] [PubMed]
- van Wageningen, T.A.; Vlaar, E.; Kooij, G.; Jongenelen, C.; Geurts, J.; van Dam, A.M. Regulation of microglial TMEM119 and P2RY12 immunoreactivity in multiple sclerosis white and grey matter lesions is dependent on their inflammatory environment. Acta Neuropathol. Commun. 2019, 7, 206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zrzavy, T.; Hametner, S.; Wimmer, I.; Butovsky, O.; Weiner, H.L.; Lassmann, H. Loss of ‘homeostatic’ microglia and patterns of their activation in active multiple sclerosis. Brain 2017, 140, 1900–1913. [Google Scholar] [CrossRef]
- Amadio, S.; Montilli, C.; Magliozzi, R.; Bernardi, G.; Reynolds, R.; Volonté, C. P2Y12 receptor protein in cortical gray matter lesions in multiple sclerosis. Cereb. Cortex 2010, 20, 1263–1273. [Google Scholar] [CrossRef] [Green Version]
- Beaino, W.; Janssen, B.; Kooij, G.; van der Pol, S.; van Het Hof, B.; van Horssen, J.; Windhorst, A.D.; de Vries, H.E. Purinergic receptors P2Y12R and P2X7R: Potential targets for PET imaging of microglia phenotypes in multiple sclerosis. J. Neuroinflammation 2017, 14, 259. [Google Scholar] [CrossRef]
- Klein, B.; Mrowetz, H.; Barker, C.M.; Lange, S.; Rivera, F.J.; Aigner, L. Age influences microglial activation after cuprizone-induced demyelination. Front. Aging Neurosci. 2018, 10, 278. [Google Scholar] [CrossRef] [Green Version]
- Butovsky, O.; Jedrychowski, M.P.; Cialic, R.; Krasemann, S.; Murugaiyan, G.; Fanek, Z.; Greco, D.J.; Wu, P.M.; Doykan, C.E.; Kiner, O.; et al. Targeting miR-155 restores abnormal microglia and attenuates disease in SOD1 mice. Ann. Neurol. 2015, 77, 75–99. [Google Scholar] [CrossRef]
Antagonist | IC50 (µM) | Binding | References | |
---|---|---|---|---|
Natural antagonist | ATP | 0.0003 | Reversible | [48,55] |
Thienopiridines | 2-oxo-clopidogrel (clopidogrel metabolite) | 0.1 | Irreversible | [6,56] |
R-138727 1 (prasugrel metabolite) | 1 | [57] | ||
UR-4501 2 (ticlopidine metabolite) | 2 | [58,59,60] | ||
Nucleotide-Nucleoside derivatives | Ticagrelor | 0.003 | Reversible | [6] |
Cangrelor | 0.0008 | [6] | ||
Other molecules | Suramin | 3 | Reversible | [6,61] |
Reactive blue-2 | 0.025 | [6] | ||
PSB-0739 | 0.0002 | [6] | ||
AZD1283 | 0.01 | [62] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Gómez Morillas, A.; Besson, V.C.; Lerouet, D. Microglia and Neuroinflammation: What Place for P2RY12? Int. J. Mol. Sci. 2021, 22, 1636. https://doi.org/10.3390/ijms22041636
Gómez Morillas A, Besson VC, Lerouet D. Microglia and Neuroinflammation: What Place for P2RY12? International Journal of Molecular Sciences. 2021; 22(4):1636. https://doi.org/10.3390/ijms22041636
Chicago/Turabian StyleGómez Morillas, Albert, Valérie C. Besson, and Dominique Lerouet. 2021. "Microglia and Neuroinflammation: What Place for P2RY12?" International Journal of Molecular Sciences 22, no. 4: 1636. https://doi.org/10.3390/ijms22041636
APA StyleGómez Morillas, A., Besson, V. C., & Lerouet, D. (2021). Microglia and Neuroinflammation: What Place for P2RY12? International Journal of Molecular Sciences, 22(4), 1636. https://doi.org/10.3390/ijms22041636