Skip to main content

Advertisement

Log in

Glia and Mast Cells as Targets for Palmitoylethanolamide, an Anti-inflammatory and Neuroprotective Lipid Mediator

  • Published:
Molecular Neurobiology Aims and scope Submit manuscript

Abstract

Glia are key players in a number of nervous system disorders. Besides releasing glial and neuronal signaling molecules directed to cellular homeostasis, glia respond also to pro-inflammatory signals released from immune-related cells, with the mast cell being of particular interest. A proposed mast cell–glia communication may open new perspectives for designing therapies to target neuroinflammation by differentially modulating activation of non-neuronal cells normally controlling neuronal sensitization—both peripherally and centrally. Mast cells and glia possess endogenous homeostatic mechanisms/molecules that can be upregulated as a result of tissue damage or stimulation of inflammatory responses. Such molecules include the N-acylethanolamines, whose principal family members are the endocannabinoid N-arachidonoylethanolamine (anandamide), and its congeners N-stearoylethanolamine, N-oleoylethanolamine, and N-palmitoylethanolamine (PEA). A key role of PEA may be to maintain cellular homeostasis when faced with external stressors provoking, for example, inflammation: PEA is produced and hydrolyzed by microglia, it downmodulates mast cell activation, it increases in glutamate-treated neocortical neurons ex vivo and in injured cortex, and PEA levels increase in the spinal cord of mice with chronic relapsing experimental allergic encephalomyelitis. Applied exogenously, PEA has proven efficacious in mast cell-mediated experimental models of acute and neurogenic inflammation. This fatty acid amide possesses also neuroprotective effects, for example, in a model of spinal cord trauma, in a delayed post-glutamate paradigm of excitotoxic death, and against amyloid β-peptide-induced learning and memory impairment in mice. These actions may be mediated by PEA acting through “receptor pleiotropism,” i.e., both direct and indirect interactions of PEA with different receptor targets, e.g., cannabinoid CB2 and peroxisome proliferator-activated receptor-alpha.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3

Similar content being viewed by others

References

  1. Giovannini MG, Scali C, Prosperi C, Bellucci A, Vannucchi MG, Rosi S, Pepeu G, Casamenti F (2002) β-Amyloid-induced inflammation and cholinergic hypofunction in the rat brain in vivo. Involvement of the p38MAPK pathway. Neurobiol Dis 11:257–274. doi:10.1006/nbdi.2002.0538

    Article  PubMed  CAS  Google Scholar 

  2. Dauer W, Przedborski S (2003) Parkinson disease. Mechanisms and models. Neuron 39:889–909. doi:10.1016/S0896-6273(03)00568-3

    Article  PubMed  CAS  Google Scholar 

  3. Gao HM, Liu B, Zhang W, Hong JS (2003) Novel anti-inflammatory therapy for Parkinson disease. Trends Pharmacol Sci 24:395–401. doi:10.1016/S0165-6147(03)00176-7

    Article  PubMed  CAS  Google Scholar 

  4. Jantaratnotai N, Ryu JK, Kim SU, McLarnon JG (2003) Amyloid β peptide-induced corpus callosum damage and glial activation in vivo. Neuroreport 14:1429–1433. doi:10.1097/01.wnr.0000086097.47480.a0

    Article  PubMed  CAS  Google Scholar 

  5. Barnum CJ, Tansey MG (2010) Modeling neuroinflammatory pathogenesis of Parkinson disease. Prog Brain Res 184:113–132. doi:10.1016/S0079-6123(10)84006-3

    Article  PubMed  CAS  Google Scholar 

  6. Qureshi GA, Baig S, Bednar I, Södersten P, Forsberg G, Siden A (1995) Increased cerebrospinal fluid concentration of nitrite in Parkinson disease. Neuroreport 6:1642–1644

    Article  PubMed  CAS  Google Scholar 

  7. Dehmer T, Lindenau J, Haid S, Dichgans J, Schulz JB (2000) Deficiency of inducible nitric-oxide synthase protects against MPTP toxicity in vivo. J Neurochem 74:2213–2216. doi:10.1046/j.1471-4159.2000.0742213.x

    Article  PubMed  CAS  Google Scholar 

  8. Nagatsu T, Mogi M, Ichinose H, Togari A (2000) Changes in cytokines and neurotrophins in Parkinson disease. J Neural Transm Suppl 60:277–290

    PubMed  Google Scholar 

  9. Ghosh A, Roy A, Liu X, Kordower JH, Mufson EJ, Hartley DM, Ghosh S, Mosley RL, Gendelman HE, Pahan K (2007) Selective inhibition of NF-κB activation prevents dopaminergic neuronal loss in a mouse model of Parkinson disease. Proc Natl Acad Sci USA 104:18754–18759. doi:10.1073/pnas.0704908104

    Article  PubMed  CAS  Google Scholar 

  10. Mondal S, Roy A, Jana A, Ghosh S, Kordower JH, Pahan K (2012) Testing NF-κB-based therapy in hemiparkinsonian monkeys. J Neuroimmune Pharmacol 7:544–556. doi:10.1007/s11481-012-9377-9

    Article  PubMed  Google Scholar 

  11. Skaper SD, Giusti P (2009) P2X7 receptors as a transducer in the co-occurrence of neurological/psychiatric and cardiovascular aisorders: a hypothesis. Cardiovasc Psychiatry Neurol 2009:545263. doi:10.1155/2009/545263

    PubMed  Google Scholar 

  12. Rivat C, Becker C, Blugeot A, Zeau B, Mauborgne A, Pohl M, Benoliel JJ (2010) Chronic stress induces transient spinal neuroinflammation, triggering sensory hypersensitivity and long-lasting anxiety-induced hyperalgesia. Pain 150:358–368. doi:10.1016/j.pain.2010.05.031

    Article  PubMed  Google Scholar 

  13. Vichaya EG, Young EE, Frazier MA, Cook JL, Welsh CJ, Meagher MW (2011) Social disruption induced priming of CNS inflammatory response to Theiler's virus is dependent upon stress induced IL-6 release. J Neuroimmunol 239:44–52. doi:10.1016/j.jneuroim.2011.08.006

    Article  PubMed  CAS  Google Scholar 

  14. González-Scarano F, Baltuch G (1999) Microglia as mediators of inflammatory and degenerative diseases. Annu Rev Neurosci 22:219–240. doi:10.1146/annurev.neuro.22.1.219

    Article  PubMed  Google Scholar 

  15. Sailasuta N, Harris K, Tran T, Ross B (2011) Minimally invasive biomarker confirms glial activation present in Alzheimer's disease: a preliminary study. Neuropsychiatr Dis Treat 7:495–499. doi:10.2147/NDT.S23721

    Article  PubMed  CAS  Google Scholar 

  16. Barcia C, Ros CM, Annese V, Gómez A, Ros-Bernal F, Aguado-Yera D, Martínez-Pagán ME, de Pablos V, Fernandez-Villalba E, Herrero MT (2011) IFN-γ signaling, with the synergistic contribution of TNF-α, mediates cell specific microglial and astroglial activation in experimental models of Parkinson's disease. Cell Death Dis 2:e142. doi:10.1038/cddis.2011.17

    Article  PubMed  CAS  Google Scholar 

  17. Appel SH, Zhao W, Beers DR, Henkel JS (2011) The microglial-motoneuron dialogue in ALS. Acta Myol 30:4–8

    PubMed  CAS  Google Scholar 

  18. Mitterauer BJ (2011) Possible role of glia in cognitive impairment in schizophrenia. CNS Neurosci Ther 17:333–344. doi:10.1111/j.1755-5949.2009.00113.x

    Article  PubMed  Google Scholar 

  19. Hinwood M, Morandini J, Day TA, Walker FR (2012) Evidence that microglia mediate the neurobiological effects of chronic psychological stress on the medial prefrontal cortex. Cereb Cortex 22:1442–1454

    Article  PubMed  CAS  Google Scholar 

  20. Rosano C, Marsland AL, Gianaros PJ (2012) Maintaining brain health by monitoring inflammatory processes: a mechanism to promote successful aging. Aging Dis 3:16–33

    PubMed  Google Scholar 

  21. Jha MK, Jeon S, Suk K (2012) Glia as a link between neuroinflammation and neuropathic pain. Immune Netw 12:41–47. doi:10.4110/in.2012.12.2.41

    Article  PubMed  Google Scholar 

  22. Carson MJ (2002) Microglia as liaisons between the immune and central nervous systems. Functional implications for multiple sclerosis. Glia 40:218–231. doi:10.1002/glia.10145

    Article  PubMed  Google Scholar 

  23. Chen CC, Grimbaldeston MA, Tsai M, Weissman IL, Galli SJ (2005) Identification of mast cell progenitors in adult mice. Proc Natl Acad Sci USA 102:11408–11413. doi:10.1073/pnas.0504197102

    Article  PubMed  CAS  Google Scholar 

  24. Gilfillan AM, Austin SJ, Metcalfe DD (2011) Mast cell biology: introduction and overview. Adv Exp Med Biol 716:2–12. doi:10.1038/ni1158

    Article  PubMed  CAS  Google Scholar 

  25. Lambracht-Hall M, Dimitriadou V, Theoharides TC (1990) Migration of mast cells in the developing rat brain. Dev Brain Res 56:151–159. doi:10.1016/0165-3806(90)90077-C

    Article  CAS  Google Scholar 

  26. Silverman AJ, Sutherland AK, Wilhelm M, Silver R (2000) Mast cells migrate from blood to brain. J Neurosci 20:401–408

    PubMed  CAS  Google Scholar 

  27. Engler H, Doenlen R, Engler A, Riether C, Prager G, Niemi MB, Pacheco-López G, Krügel U, Schedlowski M (2011) Acute amygdaloid response to systemic inflammation. Brain Behav Immun 25:1384–1392. doi:10.1016/j.bbi.2011.04.005

    Article  PubMed  CAS  Google Scholar 

  28. Moreno B, Jukes JP, Vergara-Irigaray N, Errea O, Villoslada P, Perry VH, Newman TA (2011) Systemic inflammation induces axon injury during brain inflammation. Ann Neurol 70:932–942. doi:10.1002/ana.22550

    Article  PubMed  CAS  Google Scholar 

  29. Johnson D, Krenger W (1992) Interactions of mast cells with the nervous system—recent advances. Neurochem Res 17:939–951. doi:10.1007/BF00993271

    Article  PubMed  CAS  Google Scholar 

  30. Galli SJ, Nakae S, Tsai M (2005) Mast cells in the development of adaptive immune responses. Nat Immunol 6:135–142. doi:10.1038/ni1158

    Article  PubMed  CAS  Google Scholar 

  31. Wardlaw AJ, Moqbel R, Cromwell O, Kay AB (1986) Platelet-activating factor. A potent chemotactic and chemokinetic factor for human eosinophils. J Clin Invest 78:1701–1706. doi:10.1172/JCI112765

    Article  PubMed  CAS  Google Scholar 

  32. Perry VH, Andersson P-B, Gordon G (1993) Macrophages and inflammation in the central nervous system. Trends Neurosci 16:268–273. doi:10.1016/0166-2266(93)90180-T

    Article  PubMed  CAS  Google Scholar 

  33. Johnson D, Yasui D, Seeldayers P (1991) An analysis of mast cell frequency in the rodent nervous system: numbers vary between different strains and can be reconstituted in mast cell-deficient mice. J Neuropathol Exp Neurol 50:227–234. doi:10.1097/00005072-19910005-00005

    Article  PubMed  CAS  Google Scholar 

  34. Brenner T, Soffer D, Shalit M, Levi-Schaffer F (1994) Mast cells in experimental allergic encephalomyelitis: characterization, distribution in the CNS and in vitro activation by myelin basic protein and neuropeptides. J Neurol Sci 122:210–213. doi:10.1016/0022-510X(94)90300-X

    Article  PubMed  CAS  Google Scholar 

  35. Theoharides TC (1990) Mast cells: the immune gate to the brain. Life Sci 46:607–617. doi:10.1016/0024-3205(90)90129-F

    Article  PubMed  CAS  Google Scholar 

  36. Rozniecki JJ, Hauser SL, Stein M, Lincoln R, Theoharides TC (1995) Elevated mast cell tryptase in cerebrospinal fluid of multiple sclerosis patients. Ann Neurol 37:63–66. doi:10.1002/ana.410370112

    Article  PubMed  CAS  Google Scholar 

  37. Medic N, Vita F, Abbate R, Soranzo MR, Pacor S, Fabbretti E, Borelli V, Zabucchi G (2008) Mast cell activation by myelin through scavenger receptor. J Neuroimmunol 200:27–40. doi:10.1016/j.jneuroim.2008.05.019

    Article  PubMed  CAS  Google Scholar 

  38. Skaper SD, Facci L, Romanello S, Leon A (1996) Mast cell activation causes delayed neurodegeneration in mixed hippocampal cultures via the nitric oxide pathway. J Neurochem 66:1157–1166. doi:10.1046/j.1471-4159.1996.66031157.x

    Article  PubMed  CAS  Google Scholar 

  39. Theoharides TC, Baloyannis SJ, Manolidis LS (1991) Activated rat peritoneal mast cells can cause syngeneic brain demyelination in vitro. Int J Immunopathol Pharmacol 4:137–144

    Google Scholar 

  40. Nautiyal KM, Ribeiro AC, Pfaff DW, Silver R (2008) Brain mast cells link the immune system to anxiety-like behavior. Proc Natl Acad Sci USA 105:18053–18057. doi:10.1073/pnas.0809479105

    Article  PubMed  CAS  Google Scholar 

  41. DeLeo JA, Yezierski RP (2001) The role of neuroinflammation and neuroimmune activation in persistent pain. Pain 90:1–6. doi:10.1016/S0304-3959(0)00490-5

    Article  PubMed  CAS  Google Scholar 

  42. Watkins LR, Milligan ED, Maier SF (2001) Spinal cord glia: new players in pain. Pain 93:201–205. doi:10.1016/S0304-3959(01)00359-1

    Article  PubMed  CAS  Google Scholar 

  43. Watkins LR, Milligan ED, Maier SF (2003) Glial proinflammatory cytokines mediate exaggerated pain states: implications for clinical pain. Adv Exp Med Biol 521:1–21

    PubMed  CAS  Google Scholar 

  44. Tozaki-Saitoh H, Tsuda M, Miyata H, Ueda K, Kohsaka S, Inoue K (2008) P2Y12 receptors in spinal microglia are required for neuropathic pain after peripheral nerve injury. J Neurosci 28:4949–4956. doi:10.1523/JNEUROSCI.0323-08.2008

    Article  PubMed  CAS  Google Scholar 

  45. Burnstock G, Krügel U, Abbracchio MP, Illes P (2011) Purinergic signalling: from normal behaviour to pathological brain function. Prog Neurobiol 95:229–274. doi:10.1016/j.pneurobio.2011.08.006

    Article  PubMed  CAS  Google Scholar 

  46. Chessell IP, Hatcher JP, Bountra C, Michel AD, Hughes JP, Green P, Egerton J, Murfin M, Richardson J, Peck WL, Grahames CB, Casula MA, Yiangou Y, Birch R, Anand P, Buell GN (2005) Disruption of the P2X7 purinoceptor gene abolishes chronic inflammatory and neuropathic pain. Pain 114:386–396. doi:10.1016/j.pain.2005.01.002

    Article  PubMed  CAS  Google Scholar 

  47. Tsuda M, Kuboyama K, Inoue T, Nagata K, Tozaki-Saitoh H, Inoue K (2009) Behavioral phenotypes of mice lacking purinergic P2X4 receptors in acute and chronic pain assays. Mol Pain 5:28. doi:10.1186/1744-8069-5-28

    Article  PubMed  CAS  Google Scholar 

  48. Ji RR, Gereau RW 4th, Malcangio M, Strichartz GR (2009) MAP kinase and pain. Brain Res Rev 60:135–148. doi:10.1016/j.brainresrev.2008.12.011

    Article  PubMed  CAS  Google Scholar 

  49. Xanthos DN, Gaderer S, Drdla R, Nuro E, Abramova A, Ellmeier W, Sandkühler J (2011) Central nervous system mast cells in peripheral inflammatory nociception. Mol Pain 7:42. doi:10.1186/1744-8069-7-42

    Article  PubMed  Google Scholar 

  50. Zuo Y, Perkins NM, Tracey DJ, Geczy CL (2003) Inflammation and hyperalgesia induced by nerve injury in the rat: a key role of mast cells. Pain 105:467–479. doi:10.1016/S0304-3959(03)00261-6

    Article  PubMed  Google Scholar 

  51. Levy D, Kainz V, Burstein R, Strassman AM (2012) Mast cell degranulation distinctly activates trigemino-cervical and lumbosacral pain pathways and elicits widespread tactile pain hypersensitivity. Brain Behav Immun 26:311–317. doi:10.1016/j.bbi.2011.09.016

    Article  PubMed  Google Scholar 

  52. Koda H, Mizumura K (2002) Sensitization to mechanical stimulation by inflammatory mediators and by mild burn in canine visceral nociceptors in vitro. J Neurophysiol 87:2043–2051. doi:10.1152/jn.00593.2001

    PubMed  Google Scholar 

  53. Leon A, Buriani A, Dal Toso R, Fabris M, Romanello S, Aloe L, Levi-Montalcini R (1994) Mast cells synthesize, store, and release nerve growth factor. Proc Natl Acad Sci USA 91:3739–3743. doi:10.1073/pnas.91.9.3739

    Article  PubMed  CAS  Google Scholar 

  54. Levi-Montalcini R, Skaper SD, Dal Toso R, Petrelli L, Leon A (1996) Nerve growth factor: from neurotrophin to neurokine. Trends Neurosci 19:514–520. doi:10.1016/S0166-2236(96)10058-8

    Article  PubMed  CAS  Google Scholar 

  55. Vallières L, Rivest S (1997) Regulation of the genes encoding interleukin-6, its receptor, and gp130 in the rat brain in response to the immune activator lipopolysaccharide and the proinflammatory cytokine interleukin-1β. J Neurochem 69:1668–1683. doi:10.1046/j.1471-4159.1997.69041668.x

    Article  PubMed  Google Scholar 

  56. Leal-Berumen I, Conlon P, Marshall JS (1994) IL-6 production by rat peritoneal mast cells is not necessarily preceded by histamine release and can be induced by bacterial lipopolysaccharide. J Immunol 152:5468–5476

    PubMed  CAS  Google Scholar 

  57. Hayashi R, Xiao W, Kawamato M, Yuge O, Bennett GJ (2011) Systemic glucocorticoid therapy reduces pain and the number of endoneurial tumor necrosis factor-alpha (TNFα)-positive mast cells in rats with a painful peripheral neuropathy. J Pharmacol Sci 106:559–565. doi:10.1254/jphs.FP0072181

    Article  CAS  Google Scholar 

  58. Wang Q, Tang XN, Yenari MA (2007) The inflammatory response in stroke. J Neuroimmunol 184:53–68. doi:10.1016/j.jneuroim.2006.11.014

    Article  PubMed  CAS  Google Scholar 

  59. Hanisch U-K, Kettenmann H (2007) Microglia: active sensor and versatile effector cells in the normal and pathologic brain. Nat Neurosci 10:1387–1394. doi:10.1038/nn1997

    Article  PubMed  CAS  Google Scholar 

  60. Silver R, Silverman A, Vitkovic L, Lederhendler I (1996) Mast cells in the brain: evidence and functional significance. Trends Neurosci 19:25–31. doi:10.1016/0166-2236(96)81863-7

    Article  PubMed  CAS  Google Scholar 

  61. Michaloudi H, Grivas I, Batzios C, Chiotelli M, Papadopoulos G (2003) Parallel development of blood vessels and mast cells in the lateral geniculate nuclei. Brain Res Develop Brain Res 140:269–276. doi:10.1016/S0165-3806(02)00613-2

    Article  CAS  Google Scholar 

  62. Chew LJ, Takanohashi A, Bell M (2006) Microglia and inflammation: impact on developmental brain injuries. Ment Retard Dev Disabil Res Rev 12:105–112. doi:10.1002/mrdd.20102

    Article  PubMed  Google Scholar 

  63. Jin Y, Silverman AJ, Vannucci SJ (2009) Mast cells are early responders after hypoxia-ischemia in immature rat brain. Stroke 40:3107–3112. doi:10.1161/STROKEAHA.109.549691

    Article  PubMed  CAS  Google Scholar 

  64. Gordon JR, Galli SJ (1991) Release of both preformed and newly synthesized tumor necrosis factor alpha (TNF-alpha)/cachectin by mouse mast cells stimulated via the Fc epsilon RI. A mechanism for the sustained action of mast cell-derived TNF-alpha during IgE-dependent biological responses. J Exp Med 174:103–107. doi:10.1084/174.1.103

    Article  PubMed  CAS  Google Scholar 

  65. Gregersen R, Lambertsen K, Finsen B (2000) Microglia and macrophages are the major source of tumor necrosis factor in permanent middle cerebral artery occlusion in mice. J Cereb Blood Flow Metab 20:53–65. doi:10.1097/00004647-200001000-00009

    Article  PubMed  CAS  Google Scholar 

  66. Hallenbeck JM (2002) The many faces of tumor necrosis factor in stroke. Nat Med 8:1363–1368. doi:10.1038/nm1202-1363

    Article  PubMed  CAS  Google Scholar 

  67. Jin Y, Silverman AJ, Vannucci SJ (2007) Mast cell stabilization limits hypoxic-ischemic brain damage in the immature rat. Dev Neurosci 29:373–384. doi:10.1159/000105478

    Article  PubMed  CAS  Google Scholar 

  68. Strbian D, Karjalainen-Lindsberg ML, Tatlisumak T, Lindsberg PJ (2006) Cerebral mast cells regulate early ischemic brain swelling and neutrophil accumulation. J Cereb Blood Flow Metab 26:605–612. doi:10.1038/sj.jcbfm.9600228

    Article  PubMed  Google Scholar 

  69. Biran V, Cochois V, Karroubi A, Arrang JM, Charriaut-Marlangue C, Heron A (2008) Stroke induces histamine accumulation and mast cell degranulation in the neonatal rat brain. Brain Pathol 18:1–9. doi:10.1111/j.1750-3639.2007.00092.x

    Article  PubMed  CAS  Google Scholar 

  70. Lozada A, Maegele M, Stark H, Neugebauer EM, Panula P (2005) Traumatic brain injury results in mast cell increase and changes in regulation of central histamine receptors. Neuropathol Appl Neurobiol 31:150–162. doi:10.1111/j.1365-2990.2004.00622.x

    Article  PubMed  CAS  Google Scholar 

  71. Lindsberg PJ, Strbian D, Karjalainen-Lindsberg ML (2010) Mast cells as early responders in the regulation of acute blood–brain barrier changes after cerebral ischemia and hemorrhage. J Cereb Blood Flow Metab 30:689–702. doi:10.1038/jcbfm.2009.282

    Article  PubMed  Google Scholar 

  72. Mattila OS, Strbian D, Saksi J, Pikkarainen TO, Rantanen V, Tatlisumak T, Lindsberg PJ (2011) Cerebral mast cells mediate blood–brain barrier disruption in acute experimental ischemic stroke through perivascular gelatinase activation. Stroke 42:3600–3605. doi:10.1161/STROKEAHA.111.632224

    Article  PubMed  CAS  Google Scholar 

  73. Kim SU, de Vellis J (2005) Microglia in health and disease. J Neurosci Res 81:302–313. doi:10.1002/jnr.20562

    Article  PubMed  CAS  Google Scholar 

  74. Heppner FL, Greter M, Marino D, Falsig J, Raivich G, Hövelmeyer N, Waisman A, Rülicke T, Prinz M, Priller J, Becher B, Aguzzi A (2005) Experimental autoimmune encephalomyelitis repressed by microglial paralysis. Nat Med 11:146–152. doi:10.1038/nm1177

    Article  PubMed  CAS  Google Scholar 

  75. Filbin MT (2003) Myelin-associated inhibitors of axonal regeneration in the adult mammalian CNS. Nat Rev Neurosci 4:703–713. doi:10.1038/nrn1195

    Article  PubMed  CAS  Google Scholar 

  76. Olah M, Amor S, Brouwer N, Vinet J, Eggen B, Biber K, Boddeke HW (2012) Identification of a microglia phenotype supportive of remyelination. Glia 60:306–321. doi:10.1002/glia.21266

    Article  PubMed  Google Scholar 

  77. Secor VH, Secor WE, Gutekunst CA, Brown MA (2000) Mast cells are essential for early onset and severe disease in a murine model of multiple sclerosis. J Exp Med 191:813–822. doi:10.1084/jem.191.5.813

    Article  PubMed  CAS  Google Scholar 

  78. Tanzola MB, Robbie-Ryan M, Gutekunst CA, Brown MA (2003) Mast cells exert effects outside the central nervous system to influence experimental allergic encephalomyelitis disease course. J Immunol 171:4385–4391

    PubMed  CAS  Google Scholar 

  79. Bennett JL, Blanchet MR, Zhao L, Zbytnuik L, Antignano F, Gold M, Kubes P, McNagny KM (2009) Bone marrow-derived mast cells accumulate in the central nervous system during inflammation but are dispensable for experimental autoimmune encephalomyelitis pathogenesis. J Immunol 182:5507–5514. doi:10.4049/jimmunol.0801485

    Article  PubMed  CAS  Google Scholar 

  80. Wyss-Coray T (2006) Inflammation in Alzheimer disease: driving force, bystander or beneficial response? Nat Med 12:1005–1015. doi:10.1038/nm1474

    PubMed  CAS  Google Scholar 

  81. Martín-Moreno AM, Reigada D, Ramírez BG, Mechoulam R, Innamorato N, Cuadrado A, de Ceballos ML (2011) Cannabidiol and other cannabinoids reduce microglial activation in vitro and in vivo: relevance to Alzheimer's disease. Mol Pharmacol 79:964–973. doi:10.1124/mol.111.071290

    Article  PubMed  CAS  Google Scholar 

  82. Fan R, Xu F, Previti ML, Davis J, Grande AM, Robinson JK, Van Nostrand WE (2007) Minocycline reduces microglial activation and improves behavioral deficits in a transgenic model of cerebral microvascular amyloid. J Neurosci 27:3057–3063. doi:10.1523/JNEUROSCI.4371-06.2007

    Article  PubMed  CAS  Google Scholar 

  83. Grathwohl SA, Kälin RE, Bolmont T, Prokop S, Winkelmann G, Kaeser SA, Odenthal J, Radde R, Eldh T, Gandy S, Aguzzi A, Staufenbiel M, Mathews PM, Wolburg H, Heppner FL, Jucker M (2009) Formation and maintenance of Alzheimer's disease β-amyloid plaques in the absence of microglia. Nat Neurosci 12:1361–1363. doi:10.1038/nn.2432

    Article  PubMed  CAS  Google Scholar 

  84. Lalancette-Hébert M, Gowing G, Simard A, Weng YC, Kriz J (2007) Selective ablation of proliferating microglial cells exacerbates ischemic injury in the brain. J Neurosci 27:2596–2605. doi:10.1523/JNEUROSCI.5360-06.2007

    Article  PubMed  CAS  Google Scholar 

  85. Imai F, Suzuki H, Oda J, Ninomiya T, Ono K, Sano H, Sawada M (2007) Neuroprotective effect of exogenous microglia in global brain ischemia. J Cereb Blood Flow Metab 27:488–500. doi:10.1038/sj.jcbfm.9600362

    Article  PubMed  CAS  Google Scholar 

  86. Vinet J, van Weering HR, Heinrich A, Kälin RE, Wegner A, Brouwer N, Heppner FL, van Rooijen N, Boddeke HW, Biber K (2012) Neuroprotective function for ramified microglia in hippocampal excitotoxicity. J Neuroinflammation 9:27. doi:10.1186/1742-2094-9-27

    Article  PubMed  CAS  Google Scholar 

  87. Tremblay ME, Lowery RL, Majewska AK (2010) Microglial interactions with synapses are modulated by visual experience. PLoS Biol 8:e1000527. doi:10.1371/journal.pbio.1000527

    Article  PubMed  CAS  Google Scholar 

  88. Lazarini F, Gabellec M-M, Torquet N, Lledo PM (2012) Early activation of microglia triggers long-lasting impairment of adult neurogenesis in the olfactory bulb. J Neurosci 32:3652–3664. doi:10.1523/JNEUROSCI.6394-11.2012

    Article  PubMed  CAS  Google Scholar 

  89. Skaper SD, Giusti P, Facci F (2012) Microglia and mast cells: two tracks on the road to neuroinflammation. FASEB J 26:3103–3117. doi:10.1096/fj.11-197194

    Article  PubMed  CAS  Google Scholar 

  90. Hösli L, Hösli E, Schneider U, Wiget W (1984) Evidence for the existence of histamine H1- and H2-receptors on astrocytes of cultured rat central nervous system. Neurosci Lett 48:287–291. doi:10.1016/0304-3940(84)90052-1

    Article  PubMed  Google Scholar 

  91. Dong Y, Benveniste EN (2001) Immune function of astrocytes. Glia 36:180–190. doi:10.1002/glia.1107

    Article  PubMed  CAS  Google Scholar 

  92. Kim DY, Jeoung D, Ro JY (2010) Signaling pathways in the activation of mast cells cocultured with astrocytes and colocalization of both cells in experimental allergic encephalomyelitis. J Immunol 185:273–283. doi:10.4049/jimmunol.1000991

    Article  PubMed  CAS  Google Scholar 

  93. Kim DY, Hong GU, Ro JY (2011) Signal pathways in astrocytes activated by cross-talk between of astrocytes and mast cells through CD40-CD40L. J Neuroinflammation 8:25. doi:10.1186/1742-2094-8-25

    Article  PubMed  CAS  Google Scholar 

  94. Gosselin RD, Suter MR, Ji RR, Decosterd I (2010) Glial cells and chronic pain. Neuroscientist 16:519. doi:10.1177/1073858409360822

    Article  PubMed  CAS  Google Scholar 

  95. Suk K, Ock J (2011) Chemical genetics of neuroinflammation: natural and synthetic compounds as microglial inhibitors. Inflammopharmacology 20:151. doi:10.1007/s10787-011-0108-2

    Article  PubMed  CAS  Google Scholar 

  96. Oliveira SM, Drewes CC, Silva CR, Trevisan G, Boschen SL, Moreira CG, de Almeida CD, Da Cunha C, Ferreira J (2011) Involvement of mast cells in a mouse model of postoperative pain. Eur J Pharmacol 672:88–95. doi:10.1016/j.ejphar.2011.10.001

    Article  PubMed  CAS  Google Scholar 

  97. Ralay Ranaivo H, Craft JM, Hu W, Guo L, Wing LK, Van Eldik LJ, Watterson DM (2006) Glia as a therapeutic target: selective suppression of human amyloid-beta-induced upregulation of brain proinflammatory cytokine production attenuates neurodegeneration. J Neurosci 26:662–670. doi:10.1523/JNEUROSCI.4652-05.2006

    Article  PubMed  CAS  Google Scholar 

  98. Pacher P, Bátkai S, Kunos G (2006) The endocannabinoid system as an emerging target of pharmacotherapy. Pharmacol Rev 58:389–462. doi:10.1124/pr.58.3.2

    Article  PubMed  CAS  Google Scholar 

  99. Sepe N, De Petrocellis L, Montanaro F, Cimino G, Di Marzo V (1998) Bioactive long chain N-acylethanolamines in five species of edible bivalve molluscs. Possible implications for mollusc physiology and sea food industry. Biochim Biophys Acta 1389:101–111. doi:10.1016/S0005-2760(97)00132-X

    Article  PubMed  CAS  Google Scholar 

  100. Bisogno T, Ventriglia M, Milone A, Mosca M, Cimino G, Di Marzo V (1997) Occurrence and metabolism of anandamide and related acyl-ethanolamides in ovaries of the sea urchin Paracentrotus lividus. Biochim Biophys Acta 1345:338–348. doi:10.1016/S0005-2760(97)00009-X

    Article  PubMed  Google Scholar 

  101. Matias I, Bisogno T, Melck D, Vandenbulcke F, Verger-Bocquet M, De Petrocellis L, Sergheraert C, Breton C, Di Marzo V, Salzet M (2001) Evidence for an endocannabinoid system in the central nervous system of the leech Hirudo medicinalis. Mol Brain Res 87:145–159. doi:10.1016/S0169-328X(00)00290-4

    Article  PubMed  CAS  Google Scholar 

  102. Okamoto Y, Morishita J, Tsuboi K, Tonai T, Ueda N (2004) Molecular characterization of a phospholipase D generating anandamide and its congeners. J Biol Chem 279:5298–5305. doi:10.1074/jbc.M306642200

    Article  PubMed  CAS  Google Scholar 

  103. Coburn AF, Moore LV (1943) Nutrition as conditioning factor in the rheumatic state. Am J Dis Child 65:744–756

    Google Scholar 

  104. Coburn AF, Graham CE, Hahinger J (1954) Effect of egg yolk in diets on anaphylactic arthritis (passive Arthus phenomenon) in the guinea pig. J Exp Med 100:425–435. doi:10.1084/jem.100.5.425

    Article  PubMed  CAS  Google Scholar 

  105. Kuehl FA Jr, Jacob TA, Ganley OH, Ormond RE, Meisinger MAP (1957) The identification of N-(2-hydroxyethyl)-palmitamide as a naturally occurring anti-inflammatory agent. J Am Chem Soc 79:5577–5578. doi:10.1021/ja01577a066

    Article  CAS  Google Scholar 

  106. Mazzari S, Canella R, Petrelli L, Marcolongo G, Leon L (1996) N-(2-hydroxyethyl)hexadecanamide is orally active in reducing edema formation and inflammatory hyperalgesia by downmodulating mast cell activation. Eur J Pharmacol 300:227–236. doi:10.1016/0014-2999(96)00015-5

    Article  PubMed  CAS  Google Scholar 

  107. Calignano A, La Rana G, Giuffrida A, Piomelli D (1998) Control of pain initiation by endogenous cannabinoids. Nature 394:277–281. doi:10.1038/28393

    Article  PubMed  CAS  Google Scholar 

  108. Lambert DM, Vandevoorde S, Diependaele G, Govaerts SJ, Robert AR (2001) Anticonvulsant activity of N-palmitoylethanolamide, a putative endocannabinoid, in mice. Epilepsia 42:321–327. doi:10.1046/j.1528-1157.2001.41499.x

    Article  PubMed  CAS  Google Scholar 

  109. Petrosino S, Iuvone T, Di Marzo V (2010) N-palmitoyl-ethanolamine: biochemistry and new therapeutic opportunities. Biochimie 92:724–727. doi:10.1016/j.biochi.2010.01.006

    Article  PubMed  CAS  Google Scholar 

  110. Skaper SD (2012) Conference report: 1st workshop on “Palmitoylethanolamide: biochemistry, pharmacology and therapeutic use of a pleiotropic anti-inflammatory lipid mediator”. CNS Neurol Disord Drug Targets 11:191. doi:10.2174/187152712800672427

    Article  PubMed  CAS  Google Scholar 

  111. Muccioli GG, Stella N (2008) Microglia produce and hydrolyze palmitoylethanolamide. Neuropharmacology 54:16–22. doi:10.1016/j.neuropharm.2007.05.015

    Article  PubMed  CAS  Google Scholar 

  112. Facci L, Dal Toso R, Romanello S, Buriani A, Skaper SD, Leon A (1995) Mast cells express a peripheral cannabinoid receptor with differential sensitivity to anandamide and palmitoylethanolamide. Proc Natl Acad Sci USA 92:3376–3380. doi:10.1073/pnas.92.8.3376

    Article  PubMed  CAS  Google Scholar 

  113. Cerrato S, Brazis P, della Valle MF, Miolo A, Puigdemont A (2010) Effects of palmitoylethanolamide on immunologically induced histamine, PGD2 and TNFα release from canine skin mast cells. Vet Immunol Immunopathol 133:9–15. doi:10.1016/j.vetimm.2009.06.011

    Article  PubMed  CAS  Google Scholar 

  114. Hansen HS, Lauritzen L, Strand AM, Vinggaard AM, Frandsen A, Schousboe A (1997) Characterization of glutamate-induced formation of N-acylphosphatidylethanolamine and N-acylethanolamine in cultured neocortical neurons. J Neurochem 69:753–761. doi:10.1061/j.1471-4159.1997.69020753x

    Article  PubMed  CAS  Google Scholar 

  115. Franklin A, Parmentier-Batteur S, Walter L, Greenberg DA, Stella N (2003) Palmitoylethanolamide increases after focal cerebral ischemia and potentiates microglial cell motility. J Neurosci 23:7767–7775

    PubMed  CAS  Google Scholar 

  116. Berger C, Schmid PC, Schabitz WR, Wolf M, Schwab S, Schmid HH (2004) Massive accumulation of N-acylethanolamines after stroke. Cell signalling in acute cerebral ischemia? J Neurochem 88:1159–1167. doi:10.1046/j.1471-4159.2003.02244.x

    Article  PubMed  CAS  Google Scholar 

  117. Schäbitz WR, Giuffrida A, Berger C, Aschoff A, Schwaninger M, Schwab S, Piomelli D (2002) Release of fatty acid amides in a patient with hemispheric stroke: a microdialysis study. Stroke 33:2112–2114. doi:10.1161/01.STR.0000023491.63693.18

    Article  PubMed  CAS  Google Scholar 

  118. Ghafouri N, Ghafouri B, Larsson B, Turkina MV, Karlsson L, Fowler CJ, Gerdle B (2011) High levels of N-palmitoylethanolamide and N-stearoylethanolamide in microdialysate samples from myalgic trapezius muscle in women. PLoS One 6:e27257. doi:10.1371/journal.pone.0027257

    Article  PubMed  CAS  Google Scholar 

  119. Baker D, Pryce G, Croxford JL, Brown P, Pertwee RG, Makriyannis A, Khanolkar A, Layward L, Fezza F, Bisogno T, Di Marzo V (2001) Endocannabinoids control spasticity in a multiple sclerosis model. FASEB J 15:300–302. doi:10.1096/fj.00-0399fje

    PubMed  CAS  Google Scholar 

  120. Conti S, Costa B, Colleoni M, Parolaro D, Giagnoni G (2002) Antiinflammatory action of endocannabinoid palmitoylethanolamide and the synthetic cannabinoid nabilone in a model of acute inflammation in the rat. Brit J Pharmacol 135:181–187. doi:10.1038/sj.bjp.0704466

    Article  CAS  Google Scholar 

  121. Jaggar SI, Hasnie FS, Sellaturay S, Rice AS (1998) The antihyperalgesic actions of the cannabinoid anandamide and the putative CB2 receptor agonist palmitoylethanolamide in visceral and somatic inflammatory pain. Pain 76:189–199. doi:10.1016/S0304-3959(98)00041-4

    Article  PubMed  CAS  Google Scholar 

  122. Calignano A, La Rana G, Piomelli D (2001) Antinociceptive activity of the endogenous fatty acid amide, palmitylethanolamide. Eur J Pharmacol 419:191–198. doi:10.1016/S0014-2999(01)00988-8

    Article  PubMed  CAS  Google Scholar 

  123. Costa B, Conti S, Giagnoni G, Colleoni M (2002) Therapeutic effect of the endogenous fatty acid amide, palmitoylethanolamide, in rat acute inflammation: inhibition of nitric oxide and cyclo-oxygenase systems. Br J Pharmacol 137:413–420. doi:10.1038/sj.bjp.0704900

    Article  PubMed  CAS  Google Scholar 

  124. Luongo L, Guida F, Boccella S, Bellini G, Gatta L, Rossi F, de Novellis V, Maione S (2013) Palmitoylethanolamide reduces formalin-induced neuropathic-like behaviour through spinal glial/microglial phenotypical changes in mice. CNS Neurol Disord Drug Targets 12:45–54

    Article  PubMed  CAS  Google Scholar 

  125. D'Agostino G, La Rana G, Russo R, Sasso O, Iacono A, Esposito E, Mattace Raso G, Cuzzocrea S, Loverme J, Piomelli D, Meli R, Calignano A (2009) Central administration of palmitoylethanolamide reduces hyperalgesia in mice via inhibition of NF-κB nuclear signalling in dorsal root ganglia. Eur J Pharmacol 613:54–59. doi:10.1016/j.ejphar.2009.04.022

    Article  PubMed  CAS  Google Scholar 

  126. De Filippis D, Luongo L, Cipriano M, Palazzo E, Cinelli MP, de Novellis V, Maione S, Iuvone T (2011) Palmitoylethanolamide reduces granuloma-induced hyperalgesia by modulation of mast cell activation in rats. Mol Pain 10:3. doi:10.1186/1744-8069-7-3

    Article  CAS  Google Scholar 

  127. Helyes Z, Németh J, Thán M, Bölcskei K, Pintér E, Szolcsányi J (2003) Inhibitory effect of anandamide on resiniferatoxin-induced sensory neuropeptide release in vivo and neuropathic hyperalgesia in the rat. Life Sci 73:2345–2353. doi:10.1016/S0024-3205(03)00651-9

    Article  PubMed  CAS  Google Scholar 

  128. Costa B, Comelli F, Bettoni I, Colleoni M, Giagnoni G (2008) The endogenous fatty acid amide, palmitoylethanolamide, has anti-allodynic and anti-hyperalgesic effects in a murine model of neuropathic pain: involvement of CB1, TRPV1 and PPARγ receptors and neurotrophic factors. Pain 139:541–550. doi:10.1016/j.pain.2008.06.003

    Article  PubMed  CAS  Google Scholar 

  129. Garcia-Ovejero D, Arevalo-Martin A, Petrosino S, Docagne F, Hagen C, Bisogno T, Watanabe M, Guaza C, Di Marzo V, Molina-Holgado E (2009) The endocannabinoid system is modulated in response to spinal cord injury in rats. Neurobiol Dis 33:57–71. doi:10.1016/j.nbd.2008.09.015

    Article  PubMed  CAS  Google Scholar 

  130. Esposito E, Paterniti I, Mazzon E, Genovese T, Di Paola R, Galuppo M, Cuzzocrea S (2011) Effects of palmitoylethanolamide on release of mast cell peptidases and neurotrophic factors after spinal cord injury. Brain Behav Immun 25:1099–1112. doi:10.1016/j.bbi.2011.02.006

    Article  PubMed  CAS  Google Scholar 

  131. Genovese T, Esposito E, Mazzon E, Di Paola R, Meli R, Bramanti P, Piomelli D, Calignano A, Cuzzocrea S (2008) Effects of palmitoylethanolamide on signaling pathways implicated in the development of spinal cord injury. J Pharmacol Exp Ther 326:12–23. doi:10.1124/jpet.108.136903

    Article  PubMed  CAS  Google Scholar 

  132. Skaper SD, Buriani A, Dal Toso R, Petrelli L, Romanello S, Facci L, Leon A (1996) The ALIAmide palmitoylethanolamide and cannabinoids, but not anandamide, are protective in a delayed postglutamate paradigm of excitotoxic death in cerebellar granule neurons. Proc Natl Acad Sci USA 93:3984–3989. doi:10.1073/pnas,93.9.3984

    Article  PubMed  CAS  Google Scholar 

  133. D'Agostino G, Russo R, Avagliano C, Cristiano C, Meli R, Calignano A (2012) Palmitoylethanolamide protects against the amyloid-β25-35-induced learning and memory impairment in mice, an experimental model of Alzheimer disease. Neuropsychopharmacology 37:1784–1792. doi:10.1038/npp.2012.25

    Article  PubMed  CAS  Google Scholar 

  134. Scuderi C, Valenza M, Stecca C, Esposito G, Carratù MR, Steardo L (2012) Palmitoylethanolamide exerts neuroprotective effects in mixed neuroglial cultures and organotypic hippocampal slices via peroxisome proliferator-activated receptor-α. J Neuroinflammation 9:49. doi:10.1186/1742-2094-9-21

    Article  PubMed  CAS  Google Scholar 

  135. Rees K, Stowe R, Patel S, Ives N, Breen K, Clarke CE, Ben-Shlomo Y (2011) Non-steroidal anti-inflammatory drugs as disease-modifying agents for Parkinson's disease: evidence from observational studies. Cochrane Database Syst Rev 11, CD008454. doi:10.1002/14651858.CD008454.pub2

    PubMed  Google Scholar 

  136. Driver JA, Logroscino G, Lu L, Gaziano JM, Kurth T (2011) Use of non-steroidal anti-inflammatory drugs and risk of Parkinson’s disease: nested case–control study. Brit Med J 342:d198. doi:10.1136/bmj.d198

    Article  PubMed  Google Scholar 

  137. Esposito E, Impellizzeri D, Mazzon E, Paterniti I, Cuzzocrea S (2012) Neuroprotective activities of palmitoylethanolamide in an animal model of Parkinson's disease. PLoS One 7(8):e41880. doi:10.1371/journal.pone.0041880

    Article  PubMed  CAS  Google Scholar 

  138. Ahmad A, Genovese T, Impellizzeri D, Crupi R, Velardi E, Marino A, Esposito E, Cuzzocrea S (2012) Reduction of ischemic brain injury by administration of palmitoylethanolamide after transient middle cerebral artery occlusion in rats. Brain Res 1477:45–58. doi:10.1016/j.brainres.2012.08.006

    Article  PubMed  CAS  Google Scholar 

  139. Ahmad A, Crupi R, Impellizzeri D, Campolo M, Marino A, Esposito E, Cuzzocrea S (2012) Administration of palmitoylethanolamide (PEA) protects the neurovascular unit and reduces secondary injury after traumatic brain injury in mice. Brain Behav Immun 26:1310–1321. doi:10.1016/j.bbi.2012.07.021

    Article  PubMed  CAS  Google Scholar 

  140. Lo Verme J, Fu J, Astarita G, La Rana G, Russo R, Calignano A, Piomelli D (2005) The nuclear receptor peroxisome proliferator-activated receptor-alpha mediates the anti-inflammatory actions of palmitoylethanolamide. Mol Pharmacol 67:15–19. doi:10.1124/mol.104.006353

    Article  PubMed  CAS  Google Scholar 

  141. Raso GM, Esposito E, Vitiello S, Iacono A, Santoro A, D'Agostino G, Sasso O, Russo R, Piazza PV, Calignano A, Meli R (2011) Palmitoylethanolamide stimulation induces allopregnanolone synthesis in C6 cells and primary astrocytes: involvement of peroxisome-proliferator activated receptor-α. J Neuroendocrinol 23:591–600. doi:10.1111/j.1365-2826.2011.02152.x

    Article  PubMed  CAS  Google Scholar 

  142. Scuderi C, Esposito G, Blasio A, Valenza M, Arietti P, Steardo L Jr, Carnuccio R, De Filippis D, Petrosino S, Iuvone T, Di Marzo V, Steardo L (2011) Palmitoylethanolamide counteracts reactive astrogliosis induced by β-amyloid peptide. J Cell Mol Med 15:2664–2674. doi:10.1111/j.1582-4934.2011.01267.x

    Article  PubMed  CAS  Google Scholar 

  143. D'Agostino G, La Rana G, Russo R, Sasso O, Iacono A, Esposito E, Raso GM, Cuzzocrea S, Lo Verme J, Piomelli D, Meli R, Calignano A (2007) Acute intracerebroventricular administration of palmitoylethanolamide, an endogenous peroxisome proliferator-activated receptor-alpha agonist, modulates carrageenan-induced paw edema in mice. J Pharmacol Exp Ther 322:1137–1143. doi:10.1124/jpet.107.123265

    Article  PubMed  CAS  Google Scholar 

  144. de Novellis V, Luongo L, Guida F, Cristino L, Palazzo E, Russo R, Marabese I, D'Agostino G, Calignano A, Rossi F, Di Marzo V, Maione S (2012) Effects of intra-ventrolateral periaqueductal grey palmitoylethanolamide on thermoceptive threshold and rostral ventromedial medulla cell activity. Eur J Pharmacol 676:41–50. doi:10.1016/j.ejphar.2011.11.034

    Article  PubMed  CAS  Google Scholar 

  145. Smart D, Jonsson KO, Vandevoorde S, Lambert DM, Fowler CJ (2002) ‘Entourage’ effects of N-acyl ethanolamines at human vanilloid receptors. Comparison of effects upon anandamide-induced vanilloid receptor activation and upon anandamide metabolism. Brit J Pharmacol 136:452–458. doi:10.1038/sj.bjp.0704732

    Article  CAS  Google Scholar 

  146. Petrocellis D, Davis JB, Di Marzo V (2001) Palmitoylethanolamide enhances anandamide stimulation of human vanilloid VR1 receptors. FEBS Lett 506:253–256. doi:10.1016/S0014-5793(01)02934-9)

    Article  PubMed  Google Scholar 

  147. Bíró T, Maurer M, Modarres S, Lewin E, Brodie C, Acs G, Acs P, Paus R, Blumberg PM (1998) Characterization of functional vanilloid receptors expressed by mast cells. Blood 91:1332–1340

    PubMed  Google Scholar 

  148. Kim SR, Kim SU, Oh U, Jin BK (2006) Transient receptor potential vanilloid subtype 1 mediates microglial cell death in vivo and in vitro via Ca2+-mediated mitochondrial damage and cytochrome c release. J Immunol 177:4322–4329

    PubMed  CAS  Google Scholar 

  149. Katsura H, Obata K, Mizushima T, Sakurai J, Kobayashi K, Yamanaka H, Dai Y, Fukuoka T, Sakagami M, Noguchi K (2006) Activation of Src-family kinases in spinal microglia contributes to mechanical hypersensitivity after nerve injury. J Neurosci 26:8680–8690. doi:10.1523/JNEUROSCI.1771-06.2006

    Article  PubMed  CAS  Google Scholar 

  150. Cravatt BF, Giang DK, Mayfield SP, Boger DL, Lerner RA, Gilula NB (1996) Molecular characterization of an enzyme that degrades neuromodulatory fatty-acid amides. Nature 384:83–87. doi:10.1038/384083a0

    Article  PubMed  CAS  Google Scholar 

  151. Ueda N, Yamanaka K, Yamamoto S (2001) Purification and characterization of an acid amidase selective for N-palmitoylethanolamine, a putative endogenous anti-inflammatory substance. J Biol Chem 276:35552–35557. doi:10.1074/jbc.M106261200

    Article  PubMed  CAS  Google Scholar 

  152. Solorzano C, Zhu C, Battista N, Astarita G, Lodola A, Rivara S, Mor M, Russo R, Maccarrone M, Antonietti F, Duranti A, Tontini A, Cuzzocrea S, Tarzia G, Piomelli D (2009) Selective N-acylethanolamine-hydrolyzing acid amidase inhibition reveals a key role for endogenous palmitoylethanolamide in inflammation. Proc Natl Acad Sci USA 106:20966–20971. doi:10.1073/pnas.0907417106

    Article  PubMed  CAS  Google Scholar 

  153. Saturnino C, Petrosino S, Ligresti A, Palladino C, De Martino G, Bisogno T, Di Marzo V (2010) Synthesis and biological evaluation of new potential inhibitors of N-acylethanolamine hydrolyzing acid amidase. Bioorg Med Chem Lett 20:1210–1213. doi:10.1016/j.bmcl.2009.11.134

    Article  PubMed  CAS  Google Scholar 

  154. Yamano Y, Tsuboi K, Hozaki Y, Takahashi K, Jin XH, Ueda N, Wada A (2012) Lipophilic amines as potent inhibitors of N-acylethanolamine-hydrolyzing acid amidase. Bioorg Med Chem 20:3658–3665. doi:10.1016/j.bmc.2012.03.065

    Article  PubMed  CAS  Google Scholar 

  155. Li Y, Yang L, Chen L, Zhu C, Huang R, Zheng X, Qiu Y, Fu J (2012) Design and synthesis of potent N-acylethanolamine-hydrolyzing acid amidase (NAAA) inhibitor as anti-inflammatory compounds. PLoS One 7:8. doi:10.1371/journal.pone.0043023

    Google Scholar 

  156. David S, Kroner A (2011) Repertoire of microglial and macrophage responses after spinal cord injury. Nat Rev Neurosci 12:388–399. doi:10.1038/nrn3053

    Article  PubMed  CAS  Google Scholar 

  157. Schäfer T, Starkl P, Allard C, Wolf RM, Schweighoffer T (2010) A granular variant of CD63 is a regulator of repeated human mast cell degranulation. Allergy 65:1242–1255. doi:10.1111/j.1398-9995.2010.02350.x

    Article  PubMed  CAS  Google Scholar 

  158. Truini A, Biasiotta A, Di Stefano G, La Cesa S, Leone C, Cartoni C, Federico V, Petrucci M, Cruccu G (2011) Palmitoylethanolamide restores myelinated-fibre function in patients with chemotherapy-induced painful neuropathy. CNS Neurol Disorders Drug Targets 10:916–920. doi:10.2174/187152711799219307

    Article  CAS  Google Scholar 

  159. Kopsky DJ, Hesselink JM (2012) Multimodal stepped care approach with acupuncture and PPAR-α agonist palmitoylethanolamide in the treatment of a patient with multiple sclerosis and central neuropathic pain. Acupunct Med 30:53–55. doi:10.1136/acupmed-2011-010119

    Article  PubMed  Google Scholar 

  160. Hesselink JM, Hekker TA (2012) Therapeutic utility of palmitoylethanolamide in the treatment of neuropathic pain associated with various pathological conditions: a case series. J Pain Res 5:437–442. doi:10.2147/JPR.S32143

    Article  PubMed  CAS  Google Scholar 

  161. Hesselink JMK (2012) New targets in pain, non-neuronal cells, and the role of palmitoylethanolamide. The Open Pain Journal 5:12–23

    Article  CAS  Google Scholar 

  162. Clemente S (2012) Amytrophic lateral sclerosis treatment with ultramicronized palmitoylethanolamide: a case report. CNS Neurol Disord Drug Targets 11:933–936. doi:10.2174/1871527311201070933

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

The authors wish to thank Stefano Lovison for excellent graphic design assistance. L. Facci was supported by Fondazione CARIPARO “Progetto Dottorati di Ricerca” Anno 2009.

Conflicts of interest

The authors report no conflicts of interest.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Stephen D. Skaper.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Skaper, S.D., Facci, L. & Giusti, P. Glia and Mast Cells as Targets for Palmitoylethanolamide, an Anti-inflammatory and Neuroprotective Lipid Mediator. Mol Neurobiol 48, 340–352 (2013). https://doi.org/10.1007/s12035-013-8487-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12035-013-8487-6

Keywords

Navigation