Microglial Phenotypes and Their Relationship to the Cannabinoid System: Therapeutic Implications for Parkinson’s Disease
Abstract
:1. Introduction
2. Microglia
2.1. The Origin of Microglia: A Historical Perspective
2.2. Microglial Phenotypes and Activation
2.3. Microglial Role in Acute Neuroinflammation
2.4. Microglial Role in Chronic Neuroinflammation and Neurodegeneration
2.5. Role of Microglia in Parkinson’s Disease Pathology
3. The Cannabinoid System
3.1. Overview of the Endocannabinoid System
3.2. The Cannabinoid System in Inflammation and Immune Modulation
3.3. The Cannabinoid System, Microglia, and Microglia Phenotypes
3.4. The Cannabinoid System in Neuroinflammation and Neurodegeneration
4. Cannabinoids in Parkinson’s Disease: Therapeutic Implications of Targeting Microglia
5. Summary and Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Lawson, L.J.; Perry, V.H.; Dri, P.; Gordon, S. Heterogeneity in the distribution and morphology of microglia in the normal adult mouse brain. Neuroscience 1990, 39, 151–170. [Google Scholar] [CrossRef]
- Mittelbronn, M.; Dietz, K.; Schluesener, H.J.; Meyermann, R. Local distribution of microglia in the normal adult human central nervous system differs by up to one order of magnitude. Acta Neuropathol. 2001, 101, 249–255. [Google Scholar] [CrossRef] [PubMed]
- Nimmerjahn, A.; Kirchhoff, F.; Helmchen, F. Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science 2005, 308, 1314–1318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Davalos, D.; Grutzendler, J.; Yang, G.; Kim, J.V.; Zuo, Y.; Jung, S.; Littman, D.R.; Dustin, M.L.; Gan, W.B. ATP mediates rapid microglial response to local brain injury in vivo. Nat. Neurosci. 2005, 8, 752–758. [Google Scholar] [CrossRef] [PubMed]
- Lehnardt, S. Innate immunity and neuroinflammation in the CNS: The role of microglia in Toll-like receptor-mediated neuronal injury. Glia 2010, 58, 253–263. [Google Scholar] [CrossRef] [PubMed]
- Bessis, A.; Bechade, C.; Bernard, D.; Roumier, A. Microglial control of neuronal death and synaptic properties. Glia 2007, 55, 233–238. [Google Scholar] [CrossRef] [PubMed]
- Stella, N. Cannabinoid and cannabinoid-like receptors in microglia, astrocytes, and astrocytomas. Glia 2010, 58, 1017–1030. [Google Scholar] [CrossRef] [Green Version]
- Rom, S.; Persidsky, Y. Cannabinoid Receptor 2: Potential Role in Immunomodulation and Neuroinflammation. J. Neuroimmune Pharm. 2013, 8, 608–620. [Google Scholar] [CrossRef] [Green Version]
- Nissl, F. Über einige Beziehungen zwischen Nervenzellerkrankungen und gliösen Erscheinungen bei verschiedenen Psychosen. Arch. Psychiatr. 1899, 32, 656–676. [Google Scholar]
- Ramón y Cajal, S. Contribución al conocimiento de la neuroglía del cerebro humano. Trab. Lab. Investig. Biol. Univer Madr. 1913, XI, 215–315. [Google Scholar]
- Río-Hortega, P. Estudios sobre la neuroglía. La microglía y su transformación en células en bastoncito y cuerpos granuloadiposos. Trab. Lab. Investig. Biol. 1920, 18, 37–82. [Google Scholar]
- Río-Hortega, P. The microglia. Lancet 1939, 233, 1023–1026. [Google Scholar] [CrossRef]
- Río-Hortega, P. El “tercer elemento” de los centros nerviosos. I. La microglıa en estado normal. Bol. Soc. Esp. Biol. 1919, VIII, 67–82. [Google Scholar]
- Río-Hortega, P. “Tercer elemento” de Los Centros Nerviosos. II. Intervencion de la microglia en los procesos patologicos (Cellulas en bastocito y cuerpos granulo-adiposos). Bol. Soc. Esp. Biol. 1919, 9, 91–103. [Google Scholar]
- Río-Hortega, P. El “tercer elemento” de los centros nerviosos. III. Naturaleza probable de la microglía. Bol. Soc. Esp. Biol. 1919, 8, 108–115. [Google Scholar]
- Río-Hortega, P. El “tercer elemento de los centros nerviosos”. IV. Poder fagocitario y movilidad de la microglía. Bol. Soc. Esp. Biol. 1919, VIII, 154–166. [Google Scholar]
- Ginhoux, F.; Lim, S.; Hoeffel, G.; Low, D.; Huber, T. Origin and differentiation of microglia. Front. Cell. Neurosci. 2013, 7, 45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fedoroff, S.; Zhai, R.; Novak, J.P. Microglia and astroglia have a common progenitor cell. J. Neurosci. Res. 1997, 50, 477–486. [Google Scholar] [CrossRef]
- Hao, C.; Richardson, A.; Fedoroff, S. Macrophage-like cells originate from neuroepithelium in culture: characterization and properties of the macrophage-like cells. Int. J. Dev. Neurosci. 1991, 9, 1–14. [Google Scholar] [CrossRef]
- Fujita, S.; Kitamura, T. Origin of brain macrophages and the nature of the so-called microglia. Acta Neuropathol. 1975, 6, 291–296. [Google Scholar]
- Ling, E.A. Some aspects of amoeboid microglia in the corpus callosum and neighbouring regions of neonatal rats. J. Anat. 1976, 121, 29–45. [Google Scholar] [PubMed]
- Kitamura, T.; Miyake, T.; Fujita, S. Genesis of resting microglia in the gray matter of mouse hippocampus. J. Comp. Neurol. 1984, 226, 421–433. [Google Scholar] [CrossRef] [PubMed]
- Skoff, R.P. The fine structure of pulse labeled (3-H-thymidine cells) in degenerating rat optic nerve. J. Comp. Neurol. 1975, 161, 595–611. [Google Scholar] [CrossRef] [PubMed]
- Eglitis, M.A.; Mezey, E. Hematopoietic cells differentiate into both microglia and macroglia in the brains of adult mice. Proc. Natl. Acad. Sci. USA 1997, 94, 4080–4085. [Google Scholar] [CrossRef] [Green Version]
- Simard, A.R.; Rivest, S. Bone marrow stem cells have the ability to populate the entire central nervous system into fully differentiated parenchymal microglia. FASEB J. 2004, 18, 998–1000. [Google Scholar] [CrossRef] [Green Version]
- Hickey, W.F.; Kimura, H. Perivascular microglial cells of the CNS are bone marrow-derived and present antigen in vivo. Science 1988, 239, 290–292. [Google Scholar] [CrossRef]
- Perry, V.H.; Hume, D.A.; Gordon, S. Immunohistochemical localization of macrophages and microglia in the adult and developing mouse brain. Neuroscience 1985, 15, 313–326. [Google Scholar] [CrossRef]
- Akiyama, H.; McGeer, P.L. Brain microglia constitutively express beta-2 integrins. J. Neuroimmunol. 1990, 30, 81–93. [Google Scholar] [CrossRef]
- Mildner, A.; Schmidt, H.; Nitsche, M.; Merkler, D.; Hanisch, U.K.; Mack, M.; Heikenwalder, M.; Bruck, W.; Priller, J.; Prinz, M. Microglia in the adult brain arise from Ly-6ChiCCR2+ monocytes only under defined host conditions. Nat. Neurosci. 2007, 10, 1544–1553. [Google Scholar] [CrossRef]
- Varvel, N.H.; Grathwohl, S.A.; Baumann, F.; Liebig, C.; Bosch, A.; Brawek, B.; Thal, D.R.; Charo, I.F.; Heppner, F.L.; Aguzzi, A.; et al. Microglial repopulation model reveals a robust homeostatic process for replacing CNS myeloid cells. Proc. Natl. Acad. Sci. USA 2012, 109, 18150–18155. [Google Scholar] [CrossRef] [Green Version]
- Wirenfeldt, M.; Dissing-Olesen, L.; Anne Babcock, A.; Nielsen, M.; Meldgaard, M.; Zimmer, J.; Azcoitia, I.; Leslie, R.G.; Dagnaes-Hansen, F.; Finsen, B. Population control of resident and immigrant microglia by mitosis and apoptosis. Am. J. Pathol. 2007, 171, 617–631. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Beck, H.; Voswinckel, R.; Wagner, S.; Ziegelhoeffer, T.; Heil, M.; Helisch, A.; Schaper, W.; Acker, T.; Hatzopoulos, A.K.; Plate, K.H. Participation of bone marrow-derived cells in long-term repair processes after experimental stroke. J. Cereb. Blood Flow Metab. 2003, 23, 709–717. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Flugel, A.; Bradl, M.; Kreutzberg, G.W.; Graeber, M.B. Transformation of donor-derived bone marrow precursors into host microglia during autoimmune CNS inflammation and during the retrograde response to axotomy. J. Neurosci. Res. 2001, 66, 74–82. [Google Scholar] [CrossRef]
- Priller, J.; Flugel, A.; Wehner, T.; Boentert, M.; Haas, C.A.; Prinz, M.; Fernandez-Klett, F.; Prass, K.; Bechmann, I.; de Boer, B.A.; et al. Targeting gene-modified hematopoietic cells to the central nervous system: Use of green fluorescent protein uncovers microglial engraftment. Nat. Med. 2001, 7, 1356–1361. [Google Scholar] [CrossRef] [PubMed]
- Huang, Y.; Xu, Z.; Xiong, S.; Sun, F.; Qin, G.; Hu, G.; Wang, J.; Zhao, L.; Liang, Y.X.; Wu, T.; et al. Repopulated microglia are solely derived from the proliferation of residual microglia after acute depletion. Nat. Neurosci. 2018, 21, 530–540. [Google Scholar] [CrossRef] [PubMed]
- Tay, T.L.; Mai, D.; Dautzenberg, J.; Fernandez-Klett, F.; Lin, G.; Sagar; Datta, M.; Drougard, A.; Stempfl, T.; Ardura-Fabregat, A.; et al. A new fate mapping system reveals context-dependent random or clonal expansion of microglia. Nat. Neurosci. 2017, 20, 793–803. [Google Scholar] [CrossRef] [PubMed]
- Ginhoux, F.; Garel, S. The mysterious origins of microglia. Nat. Neurosci. 2018, 21, 897–899. [Google Scholar] [CrossRef]
- Cuadros, M.A.; Martin, C.; Coltey, P.; Almendros, A.; Navascues, J. First appearance, distribution, and origin of macrophages in the early development of the avian central nervous system. J. Comp. Neurol. 1993, 330, 113–129. [Google Scholar] [CrossRef]
- Alliot, F.; Godin, I.; Pessac, B. Microglia derive from progenitors, originating from the yolk sac, and which proliferate in the brain. Brain Res. Dev. Brain Res. 1999, 117, 145–152. [Google Scholar] [CrossRef]
- Ginhoux, F.; Greter, M.; Leboeuf, M.; Nandi, S.; See, P.; Gokhan, S.; Mehler, M.F.; Conway, S.J.; Ng, L.G.; Stanley, E.R.; et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science 2010, 330, 841–845. [Google Scholar] [CrossRef] [Green Version]
- Schulz, C.; Gomez Perdiguero, E.; Chorro, L.; Szabo-Rogers, H.; Cagnard, N.; Kierdorf, K.; Prinz, M.; Wu, B.; Jacobsen, S.E.; Pollard, J.W.; et al. A lineage of myeloid cells independent of Myb and hematopoietic stem cells. Science 2012, 336, 86–90. [Google Scholar] [CrossRef] [Green Version]
- Bertrand, J.Y.; Jalil, A.; Klaine, M.; Jung, S.; Cumano, A.; Godin, I. Three pathways to mature macrophages in the early mouse yolk sac. Blood 2005, 106, 3004–3011. [Google Scholar] [CrossRef] [PubMed]
- Naito, M.; Yamamura, F.; Nishikawa, S.; Takahashi, K. Development, differentiation, and maturation of fetal mouse yolk sac macrophages in cultures. J. Leukoc. Biol. 1989, 46, 1–10. [Google Scholar] [CrossRef] [PubMed]
- Takahashi, K.; Yamamura, F.; Naito, M. Differentiation, maturation, and proliferation of macrophages in the mouse yolk sac: A light-microscopic, enzyme-cytochemical, immunohistochemical, and ultrastructural study. J. Leukoc. Biol. 1989, 45, 87–96. [Google Scholar] [CrossRef] [PubMed]
- Orkin, S.H.; Zon, L.I. Hematopoiesis: An evolving paradigm for stem cell biology. Cell 2008, 132, 631–644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Walls, J.R.; Coultas, L.; Rossant, J.; Henkelman, R.M. Three-dimensional analysis of vascular development in the mouse embryo. PLoS ONE 2008, 3, e2853. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Swinnen, N.; Smolders, S.; Avila, A.; Notelaers, K.; Paesen, R.; Ameloot, M.; Brone, B.; Legendre, P.; Rigo, J.M. Complex invasion pattern of the cerebral cortex bymicroglial cells during development of the mouse embryo. Glia 2013, 61, 150–163. [Google Scholar] [CrossRef] [PubMed]
- Schwarz, J.M.; Sholar, P.W.; Bilbo, S.D. Sex differences in microglial colonization of the developing rat brain. J. Neurochem. 2012, 120, 948–963. [Google Scholar] [CrossRef] [PubMed]
- Nayak, D.; Roth, T.L.; McGavern, D.B. Microglia development and function. Annu. Rev. Immunol. 2014, 32, 367–402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Paolicelli, R.C.; Bolasco, G.; Pagani, F.; Maggi, L.; Scianni, M.; Panzanelli, P.; Giustetto, M.; Ferreira, T.A.; Guiducci, E.; Dumas, L.; et al. Synaptic pruning by microglia is necessary for normal brain development. Science 2011, 333, 1456–1458. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sierra, A.; Encinas, J.M.; Deudero, J.J.; Chancey, J.H.; Enikolopov, G.; Overstreet-Wadiche, L.S.; Tsirka, S.E.; Maletic-Savatic, M. Microglia shape adult hippocampal neurogenesis through apoptosis-coupled phagocytosis. Cell Stem. Cell 2010, 7, 483–495. [Google Scholar] [CrossRef] [Green Version]
- Walton, N.M.; Sutter, B.M.; Laywell, E.D.; Levkoff, L.H.; Kearns, S.M.; Marshall, G.P.; Scheffler, B.; Steindler, D.A. Microglia instruct subventricular zone neurogenesis. Glia 2006, 54, 815–825. [Google Scholar] [CrossRef] [PubMed]
- Parkhurst, C.N.; Yang, G.; Ninan, I.; Savas, J.N.; Yates, J.R., 3rd; Lafaille, J.J.; Hempstead, B.L.; Littman, D.R.; Gan, W.B. Microglia promote learning-dependent synapse formation through brain-derived neurotrophic factor. Cell 2013, 155, 1596–1609. [Google Scholar] [CrossRef] [Green Version]
- Galloway, D.A.; Phillips, A.E.M.; Owen, D.R.J.; Moore, C.S. Phagocytosis in the Brain: Homeostasis and Disease. Front. Immunol. 2019, 10, 790. [Google Scholar] [CrossRef] [Green Version]
- Elliott, M.R.; Koster, K.M.; Murphy, P.S. Efferocytosis Signaling in the Regulation of Macrophage Inflammatory Responses. J. Immunol. 2017, 198, 1387–1394. [Google Scholar] [CrossRef]
- Hickman, S.; Izzy, S.; Sen, P.; Morsett, L.; El Khoury, J. Microglia in neurodegeneration. Nat. Neurosci. 2018, 21, 1359–1369. [Google Scholar] [CrossRef]
- Healy, L.M.; Perron, G.; Won, S.Y.; Michell-Robinson, M.A.; Rezk, A.; Ludwin, S.K.; Moore, C.S.; Hall, J.A.; Bar-Or, A.; Antel, J.P. MerTK Is a Functional Regulator of Myelin Phagocytosis by Human Myeloid Cells. J. Immunol. 2016, 196, 3375–3384. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kierdorf, K.; Prinz, M. Microglia in steady state. J. Clin. Investig. 2017, 127, 3201–3209. [Google Scholar] [CrossRef] [Green Version]
- Saijo, K.; Glass, C.K. Microglial cell origin and phenotypes in health and disease. Nat. Rev. Immunol. 2011, 11, 775–787. [Google Scholar] [CrossRef] [PubMed]
- Barclay, A.N.; Wright, G.J.; Brooke, G.; Brown, M.H. CD200 and membrane protein interactions in the control of myeloid cells. Trends Immunol. 2002, 23, 285–290. [Google Scholar] [CrossRef]
- Hoek, R.M.; Ruuls, S.R.; Murphy, C.A.; Wright, G.J.; Goddard, R.; Zurawski, S.M.; Blom, B.; Homola, M.E.; Streit, W.J.; Brown, M.H.; et al. Down-regulation of the macrophage lineage through interaction with OX2 (CD200). Science 2000, 290, 1768–1771. [Google Scholar] [CrossRef] [PubMed]
- Biber, K.; Neumann, H.; Inoue, K.; Boddeke, H.W. Neuronal ‘On’ and ‘Off’ signals control microglia. Trends Neurosci. 2007, 30, 596–602. [Google Scholar] [CrossRef] [PubMed]
- Kierdorf, K.; Prinz, M. Factors regulating microglia activation. Front. Cell. Neurosci. 2013, 7, 44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zusso, M.; Methot, L.; Lo, R.; Greenhalgh, A.D.; David, S.; Stifani, S. Regulation of postnatal forebrain amoeboid microglial cell proliferation and development by the transcription factor Runx1. J. Neurosci. 2012, 32, 11285–11298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Minten, C.; Terry, R.; Deffrasnes, C.; King, N.J.; Campbell, I.L. IFN regulatory factor 8 is a key constitutive determinant of the morphological and molecular properties of microglia in the CNS. PLoS ONE 2012, 7, e49851. [Google Scholar] [CrossRef] [Green Version]
- Stence, N.; Waite, M.; Dailey, M.E. Dynamics of microglial activation: A confocal time-lapse analysis in hippocampal slices. Glia 2001, 33, 256–266. [Google Scholar] [CrossRef]
- Kettenmann, H.; Hanisch, U.K.; Noda, M.; Verkhratsky, A. Physiology of microglia. Physiol. Rev. 2011, 91, 461–553. [Google Scholar] [CrossRef]
- McKercher, S.R.; Torbett, B.E.; Anderson, K.L.; Henkel, G.W.; Vestal, D.J.; Baribault, H.; Klemsz, M.; Feeney, A.J.; Wu, G.E.; Paige, C.J.; et al. Targeted disruption of the PU.1 gene results in multiple hematopoietic abnormalities. EMBO J. 1996, 15, 5647–5658. [Google Scholar] [CrossRef]
- Beers, D.R.; Henkel, J.S.; Xiao, Q.; Zhao, W.; Wang, J.; Yen, A.A.; Siklos, L.; McKercher, S.R.; Appel, S.H. Wild-type microglia extend survival in PU.1 knockout mice with familial amyotrophic lateral sclerosis. Proc. Natl. Acad. Sci. USA 2006, 103, 16021–16026. [Google Scholar] [CrossRef] [Green Version]
- Bennett, M.L.; Bennett, F.C.; Liddelow, S.A.; Ajami, B.; Zamanian, J.L.; Fernhoff, N.B.; Mulinyawe, S.B.; Bohlen, C.J.; Adil, A.; Tucker, A. New tools for studying microglia in the mouse and human CNS. Proc. Natl. Acad. Sci. USA 2016, 113, E1738–E1746. [Google Scholar] [CrossRef] [Green Version]
- 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. Identification of a unique TGF-β–dependent molecular and functional signature in microglia. Nat. Neurosci. 2014, 17, 131. [Google Scholar] [CrossRef] [Green Version]
- Grabert, K.; Michoel, T.; Karavolos, M.H.; Clohisey, S.; Baillie, J.K.; Stevens, M.P.; Freeman, T.C.; Summers, K.M.; McColl, B.W. Microglial brain region-dependent diversity and selective regional sensitivities to aging. Nat. Neurosci. 2016, 19, 504–516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Masuda, T.; Sankowski, R.; Staszewski, O.; Bottcher, C.; Amann, L.; Sagar; Scheiwe, C.; Nessler, S.; Kunz, P.; van Loo, G.; et al. Spatial and temporal heterogeneity of mouse and human microglia at single-cell resolution. Nature 2019, 566, 388–392. [Google Scholar] [CrossRef] [PubMed]
- Kim, C.C.; Nakamura, M.C.; Hsieh, C.L. Brain trauma elicits non-canonical macrophage activation states. J. Neuroinflamm. 2016, 13, 117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ransohoff, R.M. A polarizing question: Do M1 and M2 microglia exist? Nat. Neurosci. 2016, 19, 987–991. [Google Scholar] [CrossRef]
- Joers, V.; Tansey, M.G.; Mulas, G.; Carta, A.R. Microglial phenotypes in Parkinson’s disease and animal models of the disease. Prog. Neurobiol. 2017, 155, 57–75. [Google Scholar] [CrossRef]
- Scott, A.; Khan, K.; Cook, J.; Duronio, V. What is “inflammation”? Are we ready to move beyond Celsus? Br. J. Sports Med. 2004, 38, 248–249. [Google Scholar] [CrossRef] [Green Version]
- Issazadeh, S.; Mustafa, M.; Ljungdahl, A.; Hojeberg, B.; Dagerlind, A.; Elde, R.; Olsson, T. Interferon gamma, interleukin 4 and transforming growth factor beta in experimental autoimmune encephalomyelitis in Lewis rats: Dynamics of cellular mRNA expression in the central nervous system and lymphoid cells. J. Neurosci. Res. 1995, 40, 579–590. [Google Scholar] [CrossRef]
- DiSabato, D.J.; Quan, N.; Godbout, J.P. Neuroinflammation: The devil is in the details. J. Neurochem. 2016, 139, 136–153. [Google Scholar] [CrossRef] [Green Version]
- Gehrmann, J.; Mies, G.; Bonnekoh, P.; Banati, R.; Iijima, T.; Kreutzberg, G.W.; Hossmann, K.A. Microglial reaction in the rat cerebral cortex induced by cortical spreading depression. Brain Pathol. 1993, 3, 11–17. [Google Scholar] [CrossRef]
- Kettenmann, H.; Banati, R.; Walz, W. Electrophysiological behavior of microglia. Glia 1993, 7, 93–101. [Google Scholar] [CrossRef] [PubMed]
- Kettenmann, H.; Hoppe, D.; Gottmann, K.; Banati, R.; Kreutzberg, G. Cultured microglial cells have a distinct pattern of membrane channels different from peritoneal macrophages. J. Neurosci. Res. 1990, 26, 278–287. [Google Scholar] [CrossRef] [PubMed]
- Kigerl, K.A.; de Rivero Vaccari, J.P.; Dietrich, W.D.; Popovich, P.G.; Keane, R.W. Pattern recognition receptors and central nervous system repair. Exp. Neurol. 2014, 258, 5–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Block, M.L.; Zecca, L.; Hong, J.-S. Microglia-mediated neurotoxicity: Uncovering the molecular mechanisms. Nat. Rev. Neurosci. 2007, 8, 57–69. [Google Scholar] [CrossRef] [PubMed]
- Walz, W.; Ilschner, S.; Ohlemeyer, C.; Banati, R.; Kettenmann, H. Extracellular ATP activates a cation conductance and a K conductance in cultured microglial cells from mouse brain. J. Neurosci. 1993, 13, 4403–4411. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Whittemore, E.R.; Korotzer, A.R.; Etebari, A.; Cotman, C.W. Carbachol increases intracellular free calcium in cultured rat microglia. Brain Res. 1993, 621, 59–64. [Google Scholar] [CrossRef]
- Priller, J.; Haas, C.A.; Reddington, M.; Kreutzberg, G.W. Calcitonin gene-related peptide and ATP induce immediate early gene expression in cultured rat microglial cells. Glia 1995, 15, 447–457. [Google Scholar] [CrossRef]
- Aloisi, F.; Ria, F.; Adorini, L. Regulation of T-cell responses by CNS antigen-presenting cells: Different roles for microglia and astrocytes. Immuno Today 2000, 21, 141–147. [Google Scholar] [CrossRef]
- Cserr, H.F.; Harling-Berg, C.J.; Knopf, P.M. Drainage of brain extracellular fluid into blood and deep cervical lymph and its immunological significance. Brain Pathol. 1992, 2, 269–276. [Google Scholar] [CrossRef]
- Kida, S.; Pantazis, A.; Weller, R.O. CSF drains directly from the subarachnoid space into nasal lymphatics in the rat. Anatomy, histology and immunological significance. Neuropathol. Appl. Neurobiol. 1993, 19, 480–488. [Google Scholar] [CrossRef]
- Louveau, A.; Smirnov, I.; Keyes, T.J.; Eccles, J.D.; Rouhani, S.J.; Peske, J.D.; Derecki, N.C.; Castle, D.; Mandell, J.W.; Lee, K.S.; et al. Structural and functional features of central nervous system lymphatic vessels. Nature 2015, 523, 337–341. [Google Scholar] [CrossRef] [PubMed]
- Raper, D.; Louveau, A.; Kipnis, J. How Do Meningeal Lymphatic Vessels Drain the CNS? Trends Neurosci. 2016, 39, 581–586. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Greenwood, J.; Heasman, S.J.; Alvarez, J.I.; Prat, A.; Lyck, R.; Engelhardt, B. Review: Leucocyte-endothelial cell crosstalk at the blood-brain barrier: A prerequisite for successful immune cell entry to the brain. Neuropathol. Appl. Neurobiol. 2011, 37, 24–39. [Google Scholar] [CrossRef] [PubMed]
- Weller, R.O. Pathology of Cerebrospinal Fluid and Interstitial Fluid of the CNS: Significance for Alzheimer Disease, Prion Disorders and Multiple Sclerosis. J. Neuropathol. Exp. Neurol. 1998, 57, 885–894. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hailer, N.P.; Bechmann, I.; Heizmann, S.; Nitsch, R. Adhesion molecule expression on phagocytic microglial cells following anterograde degeneration of perforant path axons. Hippocampus 1997, 7, 341–349. [Google Scholar] [CrossRef]
- Hu, X.; Li, P.; Guo, Y.; Wang, H.; Leak, R.K.; Chen, S.; Gao, Y.; Chen, J. Microglia/macrophage polarization dynamics reveal novel mechanism of injury expansion after focal cerebral ischemia. Stroke 2012, 43, 3063–3070. [Google Scholar] [CrossRef] [Green Version]
- Wang, G.; Zhang, J.; Hu, X.; Zhang, L.; Mao, L.; Jiang, X.; Liou, A.K.; Leak, R.K.; Gao, Y.; Chen, J. Microglia/macrophage polarization dynamics in white matter after traumatic brain injury. J. Cereb. Blood Flow. Metab. 2013, 33, 1864–1874. [Google Scholar] [CrossRef] [Green Version]
- Lalancette-Hebert, M.; Gowing, G.; Simard, A.; Weng, Y.C.; Kriz, J. Selective ablation of proliferating microglial cells exacerbates ischemic injury in the brain. J. Neurosci. 2007, 27, 2596–2605. [Google Scholar] [CrossRef] [Green Version]
- Holmin, S.; Mathiesen, T. Long-term intracerebral inflammatory response after experimental focal brain injury in rat. Neuroreport 1999, 10, 1889–1891. [Google Scholar] [CrossRef]
- Gentleman, S.M.; Leclercq, P.D.; Moyes, L.; Graham, D.I.; Smith, C.; Griffin, W.S.T.; Nicoll, J.A.R. Long-term intracerebral inflammatory response after traumatic brain injury. Forensic Sci. Int. 2004, 146, 97–104. [Google Scholar] [CrossRef]
- Alzheimer, A. Über eine eigenartige Erkrankung der Hirnrinde. Allg. Z Psych. Psych.-Gerich. Med. 1907, 64, 146–148. (In German) [Google Scholar]
- Fischer, O. Miliare Nekrosen mit drusigen Wucherungen der Neurofibrillen, eine regelmässige Veränderung der Hirnrinde bei seniler Demenz. Mon. Psychiatr. Neurol. 1907, 22, 361–372. [Google Scholar] [CrossRef] [Green Version]
- Fischer, O. Die presbyophrene Demenz, deren anatomische Grundlage und klinische Abgrenzung. Zschr. Ges Neur. Psychiat. 1910, 3, 371–471. [Google Scholar] [CrossRef] [Green Version]
- Eikelenboom, P.; Veerhuis, R.; Scheper, W.; Rozemuller, A.J.M.; van Gool, W.A.; Hoozemans, J.J.M. The significance of neuroinflammation in understanding Alzheimer’s disease. J. Neural. Transm. 2006, 113, 1685. [Google Scholar] [CrossRef]
- Wyss-Coray, T.; Mucke, L. Inflammation in neurodegenerative disease–a double-edged sword. Neuron 2002, 35, 419–432. [Google Scholar] [CrossRef] [Green Version]
- Franceschi, C.; Bonafe, M.; Valensin, S.; Olivieri, F.; De Luca, M.; Ottaviani, E.; De Benedictis, G. Inflamm-aging. An evolutionary perspective on immunosenescence. Ann. N. Y. Acad. Sci. 2000, 908, 244–254. [Google Scholar] [CrossRef]
- Kennedy, B.K.; Berger, S.L.; Brunet, A.; Campisi, J.; Cuervo, A.M.; Epel, E.S.; Franceschi, C.; Lithgow, G.J.; Morimoto, R.I.; Pessin, J.E.; et al. Geroscience: Linking aging to chronic disease. Cell 2014, 159, 709–713. [Google Scholar] [CrossRef] [Green Version]
- Franceschi, C.; Garagnani, P.; Parini, P.; Giuliani, C.; Santoro, A. Inflammaging: A new immune-metabolic viewpoint for age-related diseases. Nat. Rev. Endocrinol. 2018, 14, 576–590. [Google Scholar] [CrossRef]
- Tian, L.; Hui, C.W.; Bisht, K.; Tan, Y.; Sharma, K.; Chen, S.; Zhang, X.; Tremblay, M.-E. Microglia under psychosocial stressors along the aging trajectory: Consequences on neuronal circuits, behavior, and brain diseases. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2017, 79, 27–39. [Google Scholar] [CrossRef] [Green Version]
- Puntener, U.; Booth, S.G.; Perry, V.H.; Teeling, J.L. Long-term impact of systemic bacterial infection on the cerebral vasculature and microglia. J. Neuroinflammation 2012, 9, 146. [Google Scholar] [CrossRef] [Green Version]
- Ohmoto, Y.; Wood, M.J.; Charlton, H.M.; Kajiwara, K.; Perry, V.H.; Wood, K.J. Variation in the immune response to adenoviral vectors in the brain: Influence of mouse strain, environmental conditions and priming. Gene Ther. 1999, 6, 471–481. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Perry, V.H. Contribution of systemic inflammation to chronic neurodegeneration. Acta Neuropathol. 2010, 120, 277–286. [Google Scholar] [CrossRef] [PubMed]
- Cunningham, C. Microglia and neurodegeneration: The role of systemic inflammation. Glia 2013, 61, 71–90. [Google Scholar] [CrossRef] [PubMed]
- Olsen, L.K.; Cairns, A.G.; Ådén, J.; Moriarty, N.; Cabre, S.; Alamilla, V.R.; Almqvist, F.; Dowd, E.; McKernan, D.P. Viral mimetic priming enhances α-synuclein-induced degeneration: Implications for Parkinson’s disease. Brain Behav. Immun. 2019, 80, 525–535. [Google Scholar] [CrossRef] [PubMed]
- Sadasivan, S.; Sharp, B.; Schultz-Cherry, S.; Smeyne, R.J. Synergistic effects of influenza and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) can be eliminated by the use of influenza therapeutics: Experimental evidence for the multi-hit hypothesis. NPJ Parkinson’s Dis. 2017, 3, 18. [Google Scholar] [CrossRef] [PubMed]
- Frank-Cannon, T.C.; Alto, L.T.; McAlpine, F.E.; Tansey, M.G. Does neuroinflammation fan the flame in neurodegenerative diseases? Mol. Neurodegener. 2009, 4, 47. [Google Scholar] [CrossRef] [Green Version]
- Murphy, M.P.; LeVine, H., 3rd. Alzheimer’s disease and the amyloid-beta peptide. J. Alzheimer’s Dis. 2010, 19, 311–323. [Google Scholar] [CrossRef] [Green Version]
- Sasaki, A.; Yamaguchi, H.; Ogawa, A.; Sugihara, S.; Nakazato, Y. Microglial activation in early stages of amyloid β protein deposition. Acta Neuropathol. 1997, 94, 316–322. [Google Scholar] [CrossRef]
- Meda, L.; Cassatella, M.A.; Szendrei, G.I.; Otvos Jr, L.; Baron, P.; Villalba, M.; Ferrari, D.; Rossi, F. Activation of microglial cells by β-amyloid protein and interferon-γ. Nature 1995, 374, 647. [Google Scholar] [CrossRef]
- Xiang, Z.; Haroutunian, V.; Ho, L.; Purohit, D.; Pasinetti, G.M. Microglia activation in the brain as inflammatory biomarker of Alzheimer’s disease neuropathology and clinical dementia. Dis. Markers 2006, 22, 95–102. [Google Scholar] [CrossRef] [Green Version]
- Nopoulos, P.C. Huntington disease: A single-gene degenerative disorder of the striatum. Dialogues Clin. Neurosci. 2016, 18, 91–98. [Google Scholar] [PubMed]
- Graveland, G.A.; Williams, R.S.; DiFiglia, M. Evidence for degenerative and regenerative changes in neostriatal spiny neurons in Huntington’s disease. Science 1985, 227, 770–773. [Google Scholar] [CrossRef] [PubMed]
- Sapp, E.; Kegel, K.B.; Aronin, N.; Hashikawa, T.; Uchiyama, Y.; Tohyama, K.; Bhide, P.G.; Vonsattel, J.P.; DiFiglia, M. Early and progressive accumulation of reactive microglia in the Huntington disease brain. J. Neuropathol. Exp. Neurol. 2001, 60, 161–172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Singhrao, S.K.; Neal, J.W.; Morgan, B.P.; Gasque, P. Increased complement biosynthesis by microglia and complement activation on neurons in Huntington’s disease. Exp. Neurol. 1999, 159, 362–376. [Google Scholar] [CrossRef] [PubMed]
- Tai, Y.F.; Pavese, N.; Gerhard, A.; Tabrizi, S.J.; Barker, R.A.; Brooks, D.J.; Piccini, P. Microglial activation in presymptomatic Huntington’s disease gene carriers. Brain 2007, 130, 1759–1766. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Parkinson, J. An essay on the shaking palsy. 1817. J. Neuropsychiatry Clin. Neurosci. 2002, 14, 223–236. [Google Scholar] [CrossRef]
- Charcot, J.; Vulpian, A. Revue clinique: De la paralysie agitante, a ‘propos d’un cas tire’ de la clinique du Professeur Oppolzer. Gaz. Hebd. Med. Chir. 1861, 9, 54–59. [Google Scholar]
- Sveinbjornsdottir, S. The clinical symptoms of Parkinson’s disease. J. Neurochem. 2016, 139, 318–324. [Google Scholar] [CrossRef] [Green Version]
- Polymeropoulos, M.H.; Lavedan, C.; Leroy, E.; Ide, S.E.; Dehejia, A.; Dutra, A.; Pike, B.; Root, H.; Rubenstein, J.; Boyer, R.; et al. Mutation in the alpha-synuclein gene identified in families with Parkinson’s disease. Science 1997, 276, 2045–2047. [Google Scholar] [CrossRef] [Green Version]
- Zimprich, A.; Biskup, S.; Leitner, P.; Lichtner, P.; Farrer, M.; Lincoln, S.; Kachergus, J.; Hulihan, M.; Uitti, R.J.; Calne, D.B.; et al. Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology. Neuron 2004, 44, 601–607. [Google Scholar] [CrossRef] [Green Version]
- Tanner, C.M.; Kamel, F.; Ross, G.W.; Hoppin, J.A.; Goldman, S.M.; Korell, M.; Marras, C.; Bhudhikanok, G.S.; Kasten, M.; Chade, A.R.; et al. Rotenone, paraquat, and Parkinson’s disease. Environ. Health Perspect. 2011, 119, 866–872. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- de Lau, L.M.L.; Breteler, M.M.B. Epidemiology of Parkinson’s disease. Lancet Neurol. 2006, 5, 525–535. [Google Scholar] [CrossRef]
- Fearnley, J.M.; Lees, A.J. Ageing and Parkinson’s disease: Substantia nigra regional selectivity. Brain 1991, 114, 2283–2301. [Google Scholar] [CrossRef]
- Ross, G.W.; Petrovitch, H.; Abbott, R.D.; Nelson, J.; Markesbery, W.; Davis, D.; Hardman, J.; Launer, L.; Masaki, K.; Tanner, C.M.; et al. Parkinsonian signs and substantia nigra neuron density in decendents elders without PD. Ann. Neurol. 2004, 56, 532–539. [Google Scholar] [CrossRef] [PubMed]
- Lewy, F. Paralysis agitans, I. Pathologische Anatomie. In Handbuch der Neurologie; Springer: Berlin, Germany, 1912. [Google Scholar]
- Spillantini, M.G.; Schmidt, M.L.; Lee, V.M.; Trojanowski, J.Q.; Jakes, R.; Goedert, M. Alpha-synuclein in Lewy bodies. Nature 1997, 388, 839–840. [Google Scholar] [CrossRef] [PubMed]
- Hirsch, E.C.; Hunot, S. Neuroinflammation in Parkinson’s disease: a target for neuroprotection? Lancet Neurol. 2009, 8, 382–397. [Google Scholar] [CrossRef]
- Boka, G.; Anglade, P.; Wallach, D.; Javoy-Agid, F.; Agid, Y.; Hirsch, E.C. Immunocytochemical analysis of tumor necrosis factor and its receptors in Parkinson’s disease. Neurosci. Lett. 1994, 172, 151–154. [Google Scholar] [CrossRef]
- Mogi, M.; Harada, M.; Riederer, P.; Narabayashi, H.; Fujita, K.; Nagatsu, T. Tumor necrosis factor-α (TNF-α) increases both in the brain and in the cerebrospinal fluid from parkinsonian patients. Neurosci. Lett. 1994, 165, 208–210. [Google Scholar] [CrossRef]
- Hunot, S.; Dugas, N.; Faucheux, B.; Hartmann, A.; Tardieu, M.; Debre, P.; Agid, Y.; Dugas, B.; Hirsch, E.C. FcepsilonRII/CD23 is expressed in Parkinson’s disease and induces, in vitro, production of nitric oxide and tumor necrosis factor-alpha in glial cells. J. Neurosci. 1999, 19, 3440–3447. [Google Scholar] [CrossRef] [Green Version]
- Mogi, M.; Harada, M.; Kondo, T.; Riederer, P.; Inagaki, H.; Minami, M.; Nagatsu, T. Interleukin-1β, interleukin-6, epidermal growth factor and transforming growth factor-α are elevated in the brain from parkinsonian patients. Neurosci. Lett. 1994, 180, 147–150. [Google Scholar] [CrossRef]
- Mogi, M.; Kondo, T.; Mizuno, Y.; Nagatsu, T. p53 protein, interferon-γ, and NF-κB levels are elevated in the parkinsonian brain. Neurosci. Lett. 2007, 414, 94–97. [Google Scholar] [CrossRef] [PubMed]
- Knott, C.; Stern, G.; Wilkin, G.P. Inflammatory Regulators in Parkinson’s Disease: iNOS, Lipocortin-1, and Cyclooxygenases-1 and -2. Mol. Cell. Neurosci. 2000, 16, 724–739. [Google Scholar] [CrossRef] [PubMed]
- McGeer, P.; Itagaki, S.; Boyes, B.; McGeer, E. Reactive microglia are positive for HLA-DR in the substantia nigra of Parkinson’s and Alzheimer’s disease brains. Neurology 1988, 38, 1285. [Google Scholar] [CrossRef]
- Imamura, K.; Hishikawa, N.; Sawada, M.; Nagatsu, T.; Yoshida, M.; Hashizume, Y. Distribution of major histocompatibility complex class II-positive microglia and cytokine profile of Parkinson’s disease brains. Acta Neuropathol. 2003, 106, 518–526. [Google Scholar] [CrossRef]
- Banati, R.B.; Daniel, S.E.; Blunt, S.B. Glial pathology but absence of apoptotic nigral neurons in long-standing Parkinson’s disease. Mov. Disord. 1998, 13, 221–227. [Google Scholar] [CrossRef] [PubMed]
- Langston, J.W.; Forno, L.S.; Tetrud, J.; Reeves, A.G.; Kaplan, J.A.; Karluk, D. Evidence of active nerve cell degeneration in the substantia nigra of humans years after 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine exposure. Ann. Neurol. 1999, 46, 598–605. [Google Scholar] [CrossRef]
- Langston, J.W. The MPTP Story. J. Parkinson’s Dis. 2017, 7, S11–S19. [Google Scholar] [CrossRef] [Green Version]
- Hamza, T.H.; Zabetian, C.P.; Tenesa, A.; Laederach, A.; Montimurro, J.; Yearout, D.; Kay, D.M.; Doheny, K.F.; Paschall, J.; Pugh, E.; et al. Common genetic variation in the HLA region is associated with late-onset sporadic Parkinson’s disease. Nat. Genet. 2010, 42, 781–785. [Google Scholar] [CrossRef] [PubMed]
- Kannarkat, G.T.; Cook, D.A.; Lee, J.K.; Chang, J.; Chung, J.; Sandy, E.; Paul, K.C.; Ritz, B.; Bronstein, J.; Factor, S.A.; et al. Common genetic variant association with altered HLA expression, synergy with pyrethroid exposure, and risk for Parkinson’s disease: An observational and case–control study. NPJ Parkinson’s Dis. 2015, 1, 15002. [Google Scholar] [CrossRef]
- Wissemann, W.T.; Hill-Burns, E.M.; Zabetian, C.P.; Factor, S.A.; Patsopoulos, N.; Hoglund, B.; Holcomb, C.; Donahue, R.J.; Thomson, G.; Erlich, H.; et al. Association of Parkinson Disease with Structural and Regulatory Variants in the HLA Region. Am. J. Hum. Genet. 2013, 93, 984–993. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hill-Burns, E.M.; Factor, S.A.; Zabetian, C.P.; Thomson, G.; Payami, H. Evidence for more than one Parkinson’s disease-associated variant within the HLA region. PLoS ONE 2011, 6, e27109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ahmed, I.; Tamouza, R.; Delord, M.; Krishnamoorthy, R.; Tzourio, C.; Mulot, C.; Nacfer, M.; Lambert, J.C.; Beaune, P.; Laurent-Puig, P.; et al. Association between Parkinson’s disease and the HLA-DRB1 locus. Mov. Disord. 2012, 27, 1104–1110. [Google Scholar] [CrossRef] [PubMed]
- Singleton, A.B.; Farrer, M.; Johnson, J.; Singleton, A.; Hague, S.; Kachergus, J.; Hulihan, M.; Peuralinna, T.; Dutra, A.; Nussbaum, R.; et al. Alpha-Synuclein locus triplication causes Parkinson’s disease. Science 2003, 302, 841. [Google Scholar] [CrossRef] [Green Version]
- Cremades, N.; Chen, S.W.; Dobson, C.M. Structural Characteristics of alpha-Synuclein Oligomers. Int. Rev. Cell. Mol. Biol. 2017, 329, 79–143. [Google Scholar] [PubMed]
- Rojanathammanee, L.; Murphy, E.J.; Combs, C.K. Expression of mutant alpha-synuclein modulates microglial phenotype in vitro. J. Neuroinflammation 2011, 8, 44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Roodveldt, C.; Labrador-Garrido, A.; Gonzalez-Rey, E.; Fernandez-Montesinos, R.; Caro, M.; Lachaud, C.C.; Waudby, C.A.; Delgado, M.; Dobson, C.M.; Pozo, D. Glial innate immunity generated by non-aggregated alpha-synuclein in mouse: Differences between wild-type and Parkinson’s disease-linked mutants. PLoS ONE 2010, 5, e13481. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Austin, S.A.; Floden, A.M.; Murphy, E.J.; Combs, C.K. Alpha-synuclein expression modulates microglial activation phenotype. J. Neurosci. 2006, 26, 10558–10563. [Google Scholar] [CrossRef] [Green Version]
- Park, J.Y.; Paik, S.R.; Jou, I.; Park, S.M. Microglial phagocytosis is enhanced by monomeric alpha-synuclein, not aggregated alpha-synuclein: Implications for Parkinson’s disease. Glia 2008, 56, 1215–1223. [Google Scholar] [CrossRef]
- Bliederhaeuser, C.; Grozdanov, V.; Speidel, A.; Zondler, L.; Ruf, W.P.; Bayer, H.; Kiechle, M.; Feiler, M.S.; Freischmidt, A.; Brenner, D.; et al. Age-dependent defects of alpha-synuclein oligomer uptake in microglia and monocytes. Acta Neuropathol. 2016, 131, 379–391. [Google Scholar] [CrossRef]
- Zhang, W.; Wang, T.; Pei, Z.; Miller, D.S.; Wu, X.; Block, M.L.; Wilson, B.; Zhang, W.; Zhou, Y.; Hong, J.S.; et al. Aggregated alpha-synuclein activates microglia: A process leading to disease progression in Parkinson’s disease. FASEB J. 2005, 19, 533–542. [Google Scholar] [CrossRef]
- Klegeris, A.; Pelech, S.; Giasson, B.I.; Maguire, J.; Zhang, H.; McGeer, E.G.; McGeer, P.L. Alpha-synuclein activates stress signaling protein kinases in THP-1 cells and microglia. Neurobiol. Aging 2008, 29, 739–752. [Google Scholar] [CrossRef] [PubMed]
- Lee, S.B.; Park, S.M.; Ahn, K.J.; Chung, K.C.; Paik, S.R.; Kim, J. Identification of the amino acid sequence motif of alpha-synuclein responsible for macrophage activation. Biochem. Biophys. Res. Commun. 2009, 381, 39–43. [Google Scholar] [CrossRef] [PubMed]
- Couch, Y.; Alvarez-Erviti, L.; Sibson, N.R.; Wood, M.J.; Anthony, D.C. The acute inflammatory response to intranigral alpha-synuclein differs significantly from intranigral lipopolysaccharide and is exacerbated by peripheral inflammation. J. Neuroinflammation 2011, 8, 166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Su, X.; Federoff, H.J.; Maguire-Zeiss, K.A. Mutant alpha-synuclein overexpression mediates early proinflammatory activity. Neurotox Res. 2009, 16, 238–254. [Google Scholar] [CrossRef] [Green Version]
- Hoenen, C.; Gustin, A.; Birck, C.; Kirchmeyer, M.; Beaume, N.; Felten, P.; Grandbarbe, L.; Heuschling, P.; Heurtaux, T. Alpha-Synuclein Proteins Promote Pro-Inflammatory Cascades in Microglia: Stronger Effects of the A53T Mutant. PLoS ONE 2016, 11, e0162717. [Google Scholar] [CrossRef] [Green Version]
- Choi, I.; Kim, B.; Byun, J.W.; Baik, S.H.; Huh, Y.H.; Kim, J.H.; Mook-Jung, I.; Song, W.K.; Shin, J.H.; Seo, H.; et al. LRRK2 G2019S mutation attenuates microglial motility by inhibiting focal adhesion kinase. Nat. Commun. 2015, 6, 8255. [Google Scholar] [CrossRef] [Green Version]
- Ma, B.; Xu, L.; Pan, X.; Sun, L.; Ding, J.; Xie, C.; Koliatsos, V.E.; Cai, H. LRRK2 modulates microglial activity through regulation of chemokine (C–X3–C) receptor 1–mediated signalling pathways. Hum. Mol. Genet. 2016, 25, 3515–3523. [Google Scholar] [CrossRef] [Green Version]
- Walsh, S.; Finn, D.P.; Dowd, E. Time-course of nigrostriatal neurodegeneration and neuroinflammation in the 6-hydroxydopamine-induced axonal and terminal lesion models of Parkinson’s disease in the rat. Neuroscience 2011, 175, 251–261. [Google Scholar] [CrossRef]
- Sherer, T.B.; Betarbet, R.; Kim, J.H.; Greenamyre, J.T. Selective microglial activation in the rat rotenone model of Parkinson’s disease. Neurosci. Lett. 2003, 341, 87–90. [Google Scholar] [CrossRef]
- Concannon, R.M.; Okine, B.N.; Finn, D.P.; Dowd, E. Upregulation of the cannabinoid CB2 receptor in environmental and viral inflammation-driven rat models of Parkinson’s disease. Exp. Neurol. 2016, 283, 204–212. [Google Scholar] [CrossRef]
- Wu, D.C.; Jackson-Lewis, V.; Vila, M.; Tieu, K.; Teismann, P.; Vadseth, C.; Choi, D.K.; Ischiropoulos, H.; Przedborski, S. Blockade of microglial activation is neuroprotective in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model of Parkinson disease. J. Neurosci. 2002, 22, 1763–1771. [Google Scholar] [CrossRef]
- Smeyne, R.J.; Breckenridge, C.B.; Beck, M.; Jiao, Y.; Butt, M.T.; Wolf, J.C.; Zadory, D.; Minnema, D.J.; Sturgess, N.C.; Travis, K.Z.; et al. Assessment of the Effects of MPTP and Paraquat on Dopaminergic Neurons and Microglia in the Substantia Nigra Pars Compacta of C57BL/6 Mice. PLoS ONE 2016, 11, e0164094. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cho, B.P.; Song, D.Y.; Sugama, S.; Shin, D.H.; Shimizu, Y.; Kim, S.S.; Kim, Y.S.; Joh, T.H. Pathological dynamics of activated microglia following medial forebrain bundle transection. Glia 2006, 53, 92–102. [Google Scholar] [CrossRef] [PubMed]
- Concannon, R.M.; Okine, B.N.; Finn, D.P.; Dowd, E. Differential upregulation of the cannabinoid CB(2) receptor in neurotoxic and inflammation-driven rat models of Parkinson’s disease. Exp. Neurol. 2015, 269, 133–141. [Google Scholar] [CrossRef] [PubMed]
- Deleidi, M.; Hallett, P.J.; Koprich, J.B.; Chung, C.Y.; Isacson, O. The Toll-like receptor-3 agonist polyinosinic: Polycytidylic acid triggers nigrostriatal dopaminergic degeneration. J. Neurosci. 2010, 30, 16091–16101. [Google Scholar] [CrossRef] [Green Version]
- McCabe, K.; Concannon, R.M.; McKernan, D.P.; Dowd, E. Time-course of striatal Toll-like receptor expression in neurotoxic, environmental and inflammatory rat models of Parkinson’s disease. J. Neuroimmunol. 2017, 310, 103–106. [Google Scholar] [CrossRef]
- Mechoulam, R. The Pharmacohistory of Cannabis Sativa. In Cannabis as Therapeutic Agent; Mechoulam, R., Ed.; CRC Press: Boca Raton, FL, USA, 1986; pp. 1–19. [Google Scholar]
- Mechoulam, R.; Shvo, Y.; Hashish, I. The structure of cannabidiol. Tetrahedron 1963, 19, 2073–2078. [Google Scholar] [CrossRef]
- Mechoulam, R.; Gaoni, Y. A Total Synthesis of Dl-Delta-1-Tetrahydrocannabinol, the Active Constituent of Hashish. J. Am. Chem. Soc. 1965, 87, 3273–3275. [Google Scholar] [CrossRef]
- Matsuda, L.A.; Lolait, S.J.; Brownstein, M.J.; Young, A.C.; Bonner, T.I. Structure of a cannabinoid receptor and functional expression of the cloned cDNA. Nature 1990, 346, 561–564. [Google Scholar] [CrossRef]
- Hua, T.; Vemuri, K.; Nikas, S.P.; Laprairie, R.B.; Wu, Y.; Qu, L.; Pu, M.; Korde, A.; Jiang, S.; Ho, J.-H.; et al. Crystal structures of agonist-bound human cannabinoid receptor CB1. Nature 2017, 547, 468–471. [Google Scholar] [CrossRef]
- Shao, Z.; Yan, W.; Chapman, K.; Ramesh, K.; Ferrell, A.J.; Yin, J.; Wang, X.; Xu, Q.; Rosenbaum, D.M. Structure of an allosteric modulator bound to the CB1 cannabinoid receptor. Nat. Chem. Biol. 2019, 15, 1199–1205. [Google Scholar] [CrossRef] [PubMed]
- Herkenham, M.; Lynn, A.B.; Little, M.D.; Johnson, M.R.; Melvin, L.S.; de Costa, B.R.; Rice, K.C. Cannabinoid receptor localization in brain. Proc. Natl. Acad. Sci. USA 1990, 87, 1932–1936. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Glass, M.; Dragunow, M.; Faull, R.L. Cannabinoid receptors in the human brain: A detailed anatomical and quantitative autoradiographic study in the fetal, neonatal and adult human brain. Neuroscience 1997, 77, 299–318. [Google Scholar] [CrossRef]
- Cota, D.; Marsicano, G.; Tschop, M.; Grubler, Y.; Flachskamm, C.; Schubert, M.; Auer, D.; Yassouridis, A.; Thone-Reineke, C.; Ortmann, S.; et al. The endogenous cannabinoid system affects energy balance via central orexigenic drive and peripheral lipogenesis. J. Clin. Investig. 2003, 112, 423–431. [Google Scholar] [CrossRef] [PubMed]
- Osei-Hyiaman, D.; DePetrillo, M.; Pacher, P.; Liu, J.; Radaeva, S.; Batkai, S.; Harvey-White, J.; Mackie, K.; Offertaler, L.; Wang, L.; et al. Endocannabinoid activation at hepatic CB1 receptors stimulates fatty acid synthesis and contributes to diet-induced obesity. J. Clin. Investig. 2005, 115, 1298–1305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nakata, M.; Yada, T. Cannabinoids inhibit insulin secretion and cytosolic Ca2+ oscillation in islet beta-cells via CB1 receptors. Regul. Pept. 2008, 145, 49–53. [Google Scholar] [CrossRef]
- Cavuoto, P.; McAinch, A.J.; Hatzinikolas, G.; Janovská, A.; Game, P.; Wittert, G.A. The expression of receptors for endocannabinoids in human and rodent skeletal muscle. Biochem. Biophys. Res. Commun. 2007, 364, 105–110. [Google Scholar] [CrossRef]
- Munro, S.; Thomas, K.L.; Abu-Shaar, M. Molecular characterization of a peripheral receptor for cannabinoids. Nature 1993, 365, 61–65. [Google Scholar] [CrossRef]
- Li, X.; Hua, T.; Vemuri, K.; Ho, J.-H.; Wu, Y.; Wu, L.; Popov, P.; Benchama, O.; Zvonok, N.; Locke, K.A.; et al. Crystal Structure of the Human Cannabinoid Receptor CB2. Cell 2019, 176, 459–467. [Google Scholar] [CrossRef] [Green Version]
- Galiègue, S.; Mary, S.; Marchand, J.; Dussossoy, D.; Carrière, D.; Carayon, P.; Bouaboula, M.; Shire, D.; LE Fur, G.; Casellas, P. Expression of central and peripheral cannabinoid receptors in human immune tissues and leukocyte subpopulations. Eur. J. Biochem. 1995, 232, 54–61. [Google Scholar] [CrossRef]
- Carlisle, S.J.; Marciano-Cabral, F.; Staab, A.; Ludwick, C.; Cabral, G.A. Differential expression of the CB2 cannabinoid receptor by rodent macrophages and macrophage-like cells in relation to cell activation. Int. Immunopharmacol. 2002, 2, 69–82. [Google Scholar] [CrossRef]
- Maresz, K.; Carrier, E.J.; Ponomarev, E.D.; Hillard, C.J.; Dittel, B.N. Modulation of the cannabinoid CB2 receptor in microglial cells in response to inflammatory stimuli. J. Neurochem. 2005, 95, 437–445. [Google Scholar] [CrossRef] [PubMed]
- Atwood, B.K.; Mackie, K. CB2: A cannabinoid receptor with an identity crisis. Br. J. Pharmacol. 2010, 160, 467–479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brown, S.M.; Wager-Miller, J.; Mackie, K. Cloning and molecular characterization of the rat CB2 cannabinoid receptor. Biochim. Biophys. Acta 2002, 1576, 255–264. [Google Scholar] [CrossRef]
- Lutz, B. Molecular biology of cannabinoid receptors. Prostaglandins Leukot. Essent. Fatty Acids 2002, 66, 123–142. [Google Scholar] [CrossRef]
- Howlett, A.; Barth, F.; Bonner, T.; Cabral, G.; Casellas, P.; Devane, W.; Felder, C.; Herkenham, M.; Mackie, K.; Martin, B.; et al. International Union of Pharmacology. XXVII. Classification of Cannabinoid Receptors. Pharmacol. Rev. 2002, 54, 161–202. [Google Scholar] [CrossRef]
- Sugiura, T.; Kondo, S.; Kishimoto, S.; Miyashita, T.; Nakane, S.; Kodaka, T.; Suhara, Y.; Takayama, H.; Waku, K. Evidence that 2-arachidonoylglycerol but not N-palmitoylethanolamine or anandamide is the physiological ligand for the cannabinoid CB2 receptor. Comparison of the agonistic activities of various cannabinoid receptor ligands in HL-60 cells. J. Biol. Chem. 2000, 275, 605–612. [Google Scholar] [CrossRef] [Green Version]
- Viscomi, M.T.; Oddi, S.; Latini, L.; Pasquariello, N.; Florenzano, F.; Bernardi, G.; Molinari, M.; Maccarrone, M. Selective CB2 receptor agonism protects central neurons from remote axotomy-induced apoptosis through the PI3K/Akt pathway. J. Neurosci. 2009, 29, 4564–4570. [Google Scholar] [CrossRef] [Green Version]
- Hanus, L.; Abu-Lafi, S.; Fride, E.; Breuer, A.; Vogel, Z.; Shalev, D.E.; Kustanovich, I.; Mechoulam, R. 2-arachidonyl glyceryl ether, an endogenous agonist of the cannabinoid CB1 receptor. Proc. Natl. Acad. Sci. USA 2001, 98, 3662–3665. [Google Scholar] [CrossRef] [Green Version]
- Re, G.; Barbero, R.; Miolo, A.; Di Marzo, V. Palmitoylethanolamide, endocannabinoids and related cannabimimetic compounds in protection against tissue inflammation and pain: Potential use in companion animals. Vet. J. 2007, 173, 21–30. [Google Scholar] [CrossRef]
- Rodriguez de Fonseca, F.; Navarro, M.; Gomez, R.; Escuredo, L.; Nava, F.; Fu, J.; Murillo-Rodriguez, E.; Giuffrida, A.; LoVerme, J.; Gaetani, S.; et al. An anorexic lipid mediator regulated by feeding. Nature 2001, 414, 209–212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Freund, T.F.; Katona, I.; Piomelli, D. Role of endogenous cannabinoids in synaptic signaling. Physiol. Rev. 2003, 83, 1017–1066. [Google Scholar] [CrossRef] [PubMed]
- Chevaleyre, V.; Takahashi, K.A.; Castillo, P.E. Endocannabinoid-mediated synaptic plasticity in the CNS. Annu. Rev. Neurosci. 2006, 29, 37–76. [Google Scholar] [CrossRef] [PubMed]
- Kano, M.; Ohno-Shosaku, T.; Hashimotodani, Y.; Uchigashima, M.; Watanabe, M. Endocannabinoid-mediated control of synaptic transmission. Physiol. Rev. 2009, 89, 309–380. [Google Scholar] [CrossRef]
- Gonsiorek, W.; Lunn, C.; Fan, X.; Narula, S.; Lundell, D.; Hipkin, R.W. Endocannabinoid 2-arachidonyl glycerol is a full agonist through human type 2 cannabinoid receptor: Antagonism by anandamide. Mol. Pharmacol. 2000, 57, 1045–1050. [Google Scholar]
- Smart, D.; Gunthorpe, M.J.; Jerman, J.C.; Nasir, S.; Gray, J.; Muir, A.I.; Chambers, J.K.; Randall, A.D.; Davis, J.B. The endogenous lipid anandamide is a full agonist at the human vanilloid receptor (hVR1). Br. J. Pharmacol. 2000, 129, 227–230. [Google Scholar] [CrossRef] [Green Version]
- Ross, R.A. Anandamide and vanilloid TRPV1 receptors. Br. J. Pharmacol. 2003, 140, 790–801. [Google Scholar] [CrossRef] [Green Version]
- Andersson, D.A.; Adner, M.; Hogestatt, E.D.; Zygmunt, P.M. Mechanisms underlying tissue selectivity of anandamide and other vanilloid receptor agonists. Mol. Pharmacol. 2002, 62, 705–713. [Google Scholar] [CrossRef] [Green Version]
- Ryberg, E.; Larsson, N.; Sjogren, S.; Hjorth, S.; Hermansson, N.O.; Leonova, J.; Elebring, T.; Nilsson, K.; Drmota, T.; Greasley, P.J. The orphan receptor GPR55 is a novel cannabinoid receptor. Br. J. Pharmacol. 2007, 152, 1092–1101. [Google Scholar] [CrossRef]
- Pertwee, R.G.; Howlett, A.C.; Abood, M.E.; Alexander, S.P.; Di Marzo, V.; Elphick, M.R.; Greasley, P.J.; Hansen, H.S.; Kunos, G.; Mackie, K.; et al. International Union of Basic and Clinical Pharmacology. LXXIX. Cannabinoid receptors and their ligands: Beyond CB(1) and CB(2). Pharmacol. Rev. 2010, 62, 588–631. [Google Scholar] [CrossRef] [Green Version]
- Sun, Y.; Bennett, A. Cannabinoids: A new group of agonists of PPARs. PPAR Res. 2007, 2007, 23513. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Di Marzo, V.; Fontana, A.; Cadas, H.; Schinelli, S.; Cimino, G.; Schwartz, J.C.; Piomelli, D. Formation and inactivation of endogenous cannabinoid anandamide in central neurons. Nature 1994, 372, 686–691. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Okamoto, Y.; Morishita, J.; Tsuboi, K.; Tonai, T.; Ueda, N. Molecular characterization of a phospholipase D generating anandamide and its congeners. J. Biol. Chem. 2004, 279, 5298–5305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bisogno, T.; Howell, F.; Williams, G.; Minassi, A.; Cascio, M.G.; Ligresti, A.; Matias, I.; Schiano-Moriello, A.; Paul, P.; Williams, E.J.; et al. Cloning of the first sn1-DAG lipases points to the spatial and temporal regulation of endocannabinoid signaling in the brain. J. Cell Biol. 2003, 163, 463–468. [Google Scholar] [CrossRef] [PubMed]
- Blankman, J.L.; Simon, G.M.; Cravatt, B.F. A Comprehensive Profile of Brain Enzymes that Hydrolyze the Endocannabinoid 2-Arachidonoylglycerol. Chem. Biol. 2007, 14, 1347–1356. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Klein, T.W.; Newton, C.; Friedman, H. Cannabinoid receptors and immunity. Immuno. Today 1998, 19, 373–381. [Google Scholar] [CrossRef]
- Klein, T.W.; Newton, C.; Larsen, K.; Lu, L.; Perkins, I.; Nong, L.; Friedman, H. The cannabinoid system and immune modulation. J. Leukoc. Biol. 2003, 74, 486–496. [Google Scholar] [CrossRef] [Green Version]
- Tindall, B.; Cooper, D.; Donovan, B.; Barnes, T.; Philpot, C.; Gold, J.; Penny, R. The Sydney AIDS Project: development of acquired immunodeficiency syndrome in a group of HIV seropositive homosexual men. Austr. N. Z. J. Med. 1988, 18, 8–15. [Google Scholar] [CrossRef]
- Newell, G.R.; Mansell, P.W.A.; Wilson, M.B.; Lynch, H.K.; Spitz, M.R.; Hersh, E.M. Risk factor analysis among men referred for possible acquired immune deficiency syndrome. Prev. Med. 1985, 14, 81–91. [Google Scholar] [CrossRef]
- Caiaffa, W.T.; Vlahov, D.; Graham, N.M.; Astemborski, J.; Solomon, L.; Nelson, K.E.; Munoz, A. Drug smoking, Pneumocystis carinii pneumonia, and immunosuppression increase risk of bacterial pneumonia in human immunodeficiency virus-seropositive injection drug users. Am. J. Respir. Crit. Care Med. 1994, 150, 1493–1498. [Google Scholar] [CrossRef]
- Baldwin, G.C.; Tashkin, D.P.; Buckley, D.M.; Park, A.N.; Dubinett, S.M.; Roth, M.D. Marijuana and cocaine impair alveolar macrophage function and cytokine production. Am. J. Respir. 1997, 156, 1606–1613. [Google Scholar] [CrossRef] [PubMed]
- Benito, C.; Romero, J.P.; Tolon, R.M.; Clemente, D.; Docagne, F.; Hillard, C.J.; Guaza, C.; Romero, J. Cannabinoid CB1 and CB2 receptors and fatty acid amide hydrolase are specific markers of plaque cell subtypes in human multiple sclerosis. J. Neurosci. 2007, 27, 2396–2402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- De Lago, E.; Moreno-Martet, M.; Cabranes, A.; Ramos, J.A.; Fernández-Ruiz, J. Cannabinoids ameliorate disease progression in a model of multiple sclerosis in mice, acting preferentially through CB1 receptor-mediated anti-inflammatory effects. Neuropharmacology 2012, 62, 2299–2308. [Google Scholar] [CrossRef] [PubMed]
- Rossi, S.; Furlan, R.; Chiara, V.D.; Muzio, L.; Musella, A.; Motta, C.; Studer, V.; Cavasinni, F.; Bernardi, G.; Martino, G.; et al. Cannabinoid CB1 receptors regulate neuronal TNF-α effects in experimental autoimmune encephalomyelitis. Brain Behav. Immun. 2011, 25, 1242–1248. [Google Scholar] [CrossRef] [PubMed]
- Arevalo-Martin, A.; Vela, J.M.; Molina-Holgado, E.; Borrell, J.; Guaza, C. Therapeutic action of cannabinoids in a murine model of multiple sclerosis. J. Neurosci. 2003, 23, 2511–2516. [Google Scholar] [CrossRef] [PubMed]
- Maresz, K.; Pryce, G.; Ponomarev, E.D.; Marsicano, G.; Croxford, J.L.; Shriver, L.P.; Ledent, C.; Cheng, X.; Carrier, E.J.; Mann, M.K.; et al. Direct suppression of CNS autoimmune inflammation via the cannabinoid receptor CB1 on neurons and CB2 on autoreactive T cells. Nat. Med. 2007, 13, 492–497. [Google Scholar] [CrossRef]
- Mestre, L.; Inigo, P.M.; Mecha, M.; Correa, F.G.; Hernangomez-Herrero, M.; Loria, F.; Docagne, F.; Borrell, J.; Guaza, C. Anandamide inhibits Theiler’s virus induced VCAM-1 in brain endothelial cells and reduces leukocyte transmigration in a model of blood brain barrier by activation of CB(1) receptors. J. Neuroinflamm. 2011, 8, 102. [Google Scholar] [CrossRef] [Green Version]
- Werner, P.; Pitt, D.; Raine, C.S. Multiple sclerosis: Altered glutamate homeostasis in lesions correlates with oligodendrocyre and axonal damage. Ann. Neurol. 2001, 50, 169–180. [Google Scholar] [CrossRef]
- Srinivasan, R.; Sailasuta, N.; Hurd, R.; Nelson, S.; Pelletier, D. Evidence of elevated glutamate in multiple sclerosis using magnetic resonance spectroscopy at 3 T. Brain 2005, 128, 1016–1025. [Google Scholar] [CrossRef] [Green Version]
- Kostic, M.; Zivkovic, N.; Stojanovic, I. Multiple sclerosis and glutamate excitotoxicity. Rev. Neurosci. 2013, 24, 71–88. [Google Scholar] [CrossRef]
- Van der Stelt, M.; Veldhuis, W.B.; Bar, P.R.; Veldink, G.A.; Vliegenthart, J.F.; Nicolay, K. Neuroprotection by Delta9-tetrahydrocannabinol, the main active compound in marijuana, against ouabain-induced in vivo excitotoxicity. J. Neurosci. 2001, 21, 6475–6479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ashton, J.C.; Glass, M. The cannabinoid CB2 receptor as a target for inflammation-dependent neurodegeneration. Curr. Neuropharmacol. 2007, 5, 73–80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Matias, I.; Pochard, P.; Orlando, P.; Salzet, M.; Pestel, J.; Di Marzo, V. Presence and regulation of the endocannabinoid system in human dendritic cells. Eur. J. Biochem. 2002, 269, 3771–3778. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, S.F.; Newton, C.; Widen, R.; Friedman, H.; Klein, T.W. Differential expression of cannabinoid CB(2) receptor mRNA in mouse immune cell subpopulations and following B cell stimulation. Eur. J. Pharmacol. 2001, 423, 235–241. [Google Scholar] [CrossRef]
- Carayon, P.; Marchand, J.; Dussossoy, D.; Derocq, J.M.; Jbilo, O.; Bord, A.; Bouaboula, M.; Galiegue, S.; Mondiere, P.; Penarier, G.; et al. Modulation and functional involvement of CB2 peripheral cannabinoid receptors during B-cell differentiation. Blood 1998, 92, 3605–3615. [Google Scholar] [CrossRef]
- Schwarz, H.; Blanco, F.J.; Lotz, M. Anadamide, an endogenous cannabinoid receptor agonist inhibits lymphocyte proliferation and induces apoptosis. J. Neuroimmunol. 1994, 55, 107–115. [Google Scholar] [CrossRef]
- Zhu, W.; Friedman, H.; Klein, T.W. Delta9-tetrahydrocannabinol induces apoptosis in macrophages and lymphocytes: Involvement of Bcl-2 and caspase-1. J. Pharmacol. Exp. Ther. 1998, 286, 1103–1109. [Google Scholar]
- McKallip, R.J.; Lombard, C.; Martin, B.R.; Nagarkatti, M.; Nagarkatti, P.S. Delta(9)-tetrahydrocannabinol-induced apoptosis in the thymus and spleen as a mechanism of immunosuppression in vitro and in vivo. J. Pharmacol. Exp. Ther. 2002, 302, 451–465. [Google Scholar] [CrossRef]
- Lombard, C.; Nagarkatti, M.; Nagarkatti, P. CB2 cannabinoid receptor agonist, JWH-015, triggers apoptosis in immune cells: Potential role for CB2-selective ligands as immunosuppressive agents. Clin. Immunol. 2007, 122, 259–270. [Google Scholar] [CrossRef] [Green Version]
- Lombard, C.; Nagarkatti, M.; Nagarkatti, P.S. Targeting cannabinoid receptors to treat leukemia: Role of cross-talk between extrinsic and intrinsic pathways in Delta9-tetrahydrocannabinol (THC)-induced apoptosis of Jurkat cells. Leuk. Res. 2005, 29, 915–922. [Google Scholar] [CrossRef]
- McKallip, R.J.; Jia, W.; Schlomer, J.; Warren, J.W.; Nagarkatti, P.S.; Nagarkatti, M. Cannabidiol-induced apoptosis in human leukemia cells: A novel role of cannabidiol in the regulation of p22phox and Nox4 expression. Mol. Pharmacol. 2006, 70, 897–908. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- McKallip, R.J.; Lombard, C.; Fisher, M.; Martin, B.R.; Ryu, S.; Grant, S.; Nagarkatti, P.S.; Nagarkatti, M. Targeting CB2 cannabinoid receptors as a novel therapy to treat malignant lymphoblastic disease. Blood 2002, 100, 627–634. [Google Scholar] [CrossRef] [PubMed]
- Lee, M.; Kyu Hwan, Y.; Kaminski, N.E. Effects of putative cannabinoid receptor ligands, anandamide and 2- arachidonyl-glycerol, on immune function in B6C3F1 mouse splenocytes. J. Pharmacol. Exp. Ther. 1995, 275, 529–536. [Google Scholar] [PubMed]
- Derocq, J.M.; Segui, M.; Marchand, J.; Le Fur, G.; Casellas, P. Cannabinoids enhance human B-cell growth at low nanomolar concentrations. FEBS Lett. 1995, 369, 177–182. [Google Scholar] [CrossRef] [Green Version]
- Klein, T.W.; Newton, C.A.; Widen, R.; Friedman, H. The effect of delta-9-tetrahydrocannabinol and 11-hydroxy-delta-9-tetrahydrocannabinol on t-lymphocyte and b-lymphocyte mitogen responses. Immunopharmacol. Immunotoxicol. 1985, 7, 451–466. [Google Scholar] [CrossRef]
- Patrini, G.; Sacerdote, P.; Fuzio, D.; Manfredi, B.; Parolaro, D. Regulation of immune functions in rat splenocytes after acute and chronic in vivo treatment with CP-55,940, a synthetic cannabinoid compound. J. Neuroimmunol. 1997, 80, 143–148. [Google Scholar] [CrossRef]
- Sacerdote, P.; Massi, P.; Panerai, A.E.; Parolaro, D. In vivo and in vitro treatment with the synthetic cannabinoid CP55,940 decreases the in vitro migration of macrophages in the rat: Involvement of both CB1 and CB2 receptors. J. Neuroimmunol. 2000, 109, 155–163. [Google Scholar] [CrossRef]
- Shoemaker, J.L.; Ruckle, M.B.; Mayeux, P.R.; Prather, P.L. Agonist-directed trafficking of response by endocannabinoids acting at CB2 receptors. J. Pharmacol. Exp. Ther. 2005, 315, 828–838. [Google Scholar] [CrossRef] [Green Version]
- Joseph, J.; Niggemann, B.; Zaenker, K.S.; Entschladen, F. Anandamide is an endogenous inhibitor for the migration of tumor cells and T lymphocytes. Cancer Immunol. Immunother. 2004, 53, 723–728. [Google Scholar] [CrossRef]
- Ghosh, S.; Preet, A.; Groopman, J.E.; Ganju, R.K. Cannabinoid receptor CB2 modulates the CXCL12/CXCR4-mediated chemotaxis of T lymphocytes. Mol. Immunol. 2006, 43, 2169–2179. [Google Scholar] [CrossRef]
- Miller, A.M.; Stella, N. CB2 receptor-mediated migration of immune cells: It can go either way. Br. J. Pharmacol. 2008, 153, 299–308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhu, L.X.; Sharma, S.; Stolina, M.; Gardner, B.; Roth, M.D.; Tashkin, D.P.; Dubinett, S.M. Δ-9-tetrahydrocannabinol inhibits antitumor immunity by a CB2 receptor-mediated, cytokine-dependent pathway. J. Immunol. 2000, 165, 373–380. [Google Scholar] [CrossRef] [PubMed]
- Smith, S.R.; Terminelli, C.; Denhardt, G. Effects of Cannabinoid Receptor Agonist and Antagonist Ligands on Production of Inflammatory Cytokines and Anti-Inflammatory Interleukin-10 in Endotoxemic Mice. J. Pharmacol. Exp. Ther. 2000, 293, 136. [Google Scholar] [PubMed]
- Klein, T.W.; Newton, C.A.; Nakachi, N.; Friedman, H. Delta 9-tetrahydrocannabinol treatment suppresses immunity and early IFN-gamma, IL-12, and IL-12 receptor beta 2 responses to Legionella pneumophila infection. J. Immunol. 2000, 164, 6461–6466. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ouyang, Y.; Hwang, S.G.; Han, S.H.; Kaminski, N.E. Suppression of Interleukin-2 by the Putative Endogenous Cannabinoid 2-Arachidonyl-Glycerol Is Mediated through Down-regulation of the Nuclear Factor of Activated T Cells. Mol. Pharmacol. 1998, 53, 676. [Google Scholar] [CrossRef] [Green Version]
- Yuan, M.; Kiertscher, S.M.; Cheng, Q.; Zoumalan, R.; Tashkin, D.P.; Roth, M.D. Δ9-Tetrahydrocannabinol regulates Th1/Th2 cytokine balance in activated human T cells. J. Neuroimmunol. 2002, 133, 124–131. [Google Scholar] [CrossRef]
- Panitch, H.; Haley, A.; Hirsch, R.; Johnson, K. Exacerbations of multiple sclerosis in patients treated with gamma interferon. Lancet 1987, 329, 893–895. [Google Scholar] [CrossRef]
- Kusaba, M.; Honda, J.; Fukuda, T.; Oizumi, K. Analysis of type 1 and type 2 T cells in synovial fluid and peripheral blood of patients with rheumatoid arthritis. J. Rheumatol. 1998, 25, 1466–1471. [Google Scholar]
- Bo, X.; Broome, U.; Remberger, M.; Sumitran-Holgersson, S. Tumour necrosis factor alpha impairs function of liver derived T lymphocytes and natural killer cells in patients with primary sclerosing cholangitis. Gut 2001, 49, 131–141. [Google Scholar] [CrossRef] [Green Version]
- Mageed, R.A.; Adams, G.; Woodrow, D.; Podhajcer, O.L.; Chernajovsky, Y. Prevention of collagen-induced arthritis by gene delivery of soluble p75 tumour necrosis factor receptor. Gene Ther. 1998, 5, 1584–1592. [Google Scholar] [CrossRef] [Green Version]
- Malfait, A.M.; Gallily, R.; Sumariwalla, P.F.; Malik, A.S.; Andreakos, E.; Mechoulam, R.; Feldmann, M. The nonpsychoactive cannabis constituent cannabidiol is an oral anti-arthritic therapeutic in murine collagen-induced arthritis. Proc. Natl. Acad. Sci. USA 2000, 97, 9561–9566. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lu, Y.; Chen, B.; Song, J.-H.; Zhen, T.; Wang, B.-Y.; Li, X.; Liu, P.; Yang, X.; Zhang, Q.-L.; Xi, X.-D.; et al. Eriocalyxin B ameliorates experimental autoimmune encephalomyelitis by suppressing Th1 and Th17 cells. Proc. Natl. Acad. Sci. USA 2013, 110, 2258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nizri, E.; Irony-Tur-Sinai, M.; Lory, O.; Orr-Urtreger, A.; Lavi, E.; Brenner, T. Activation of the cholinergic anti-inflammatory system by nicotine attenuates neuroinflammation via suppression of Th1 and Th17 responses. J. Immunol. 2009, 183, 6681–6688. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Salzet, M.; Breton, C.; Bisogno, T.; Di Marzo, V. Comparative biology of the endocannabinoid system. Eur. J. Biochem. 2000, 267, 4917–4927. [Google Scholar] [CrossRef]
- Carrier, E.J.; Kearn, C.S.; Barkmeier, A.J.; Breese, N.M.; Yang, W.; Nithipatikom, K.; Pfister, S.L.; Campbell, W.B.; Hillard, C.J. Cultured rat microglial cells synthesize the endocannabinoid 2-arachidonylglycerol, which increases proliferation via a CB2 receptor-dependent mechanism. Mol. Pharmacol. 2004, 65, 999–1007. [Google Scholar] [CrossRef] [Green Version]
- Walter, L.; Franklin, A.; Witting, A.; Wade, C.; Xie, Y.; Kunos, G.; Mackie, K.; Stella, N. Nonpsychotropic cannabinoid receptors regulate microglial cell migration. J. Neurosci. 2003, 23, 1398–1405. [Google Scholar] [CrossRef] [Green Version]
- Witting, A.; Walter, L.; Wacker, J.; Moller, T.; Stella, N. P2X7 receptors control 2-arachidonoylglycerol production by microglial cells. Proc. Natl. Acad. Sci. USA 2004, 101, 3214–3219. [Google Scholar] [CrossRef] [Green Version]
- Stella, N. Endocannabinoid signaling in microglial cells. Neuropharmacology 2009, 56, 244–253. [Google Scholar] [CrossRef] [Green Version]
- Witting, A.; Chen, L.; Cudaback, E.; Straiker, A.; Walter, L.; Rickman, B.; Möller, T.; Brosnan, C.; Stella, N. Experimental autoimmune encephalomyelitis disrupts endocannabinoid-mediated neuroprotection. Proc. Natl. Acad. Sci. USA 2006, 103, 6362. [Google Scholar] [CrossRef] [Green Version]
- Mecha, M.; Feliú, A.; Carrillo-Salinas, F.J.; Rueda-Zubiaurre, A.; Ortega-Gutiérrez, S.; de Sola, R.G.; Guaza, C. Endocannabinoids drive the acquisition of an alternative phenotype in microglia. Brain Behav. Immun. 2015, 49, 233–245. [Google Scholar] [CrossRef]
- Muccioli, G.G.; Xu, C.; Odah, E.; Cudaback, E.; Cisneros, J.A.; Lambert, D.M.; López Rodríguez, M.L.; Bajjalieh, S.; Stella, N. Identification of a Novel Endocannabinoid-Hydrolyzing Enzyme Expressed by Microglial Cells. J. Neurosci. 2007, 27, 2883. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Marrs, W.R.; Blankman, J.L.; Horne, E.A.; Thomazeau, A.; Lin, Y.H.; Coy, J.; Bodor, A.L.; Muccioli, G.G.; Hu, S.S.-J.; Woodruff, G.; et al. The serine hydrolase ABHD6 controls the accumulation and efficacy of 2-AG at cannabinoid receptors. Nat. Neurosci. 2010, 13, 951–957. [Google Scholar] [CrossRef] [Green Version]
- Schatz, A.R.; Lee, M.; Condie, R.B.; Pulaski, J.T.; Kaminski, N.E. Cannabinoid receptors CB1 and CB2: A characterization of expression and adenylate cyclase modulation within the immune system. Toxicol. Appl. Pharmacol. 1997, 142, 278–287. [Google Scholar] [CrossRef]
- Griffin, G.; Wray, E.J.; Tao, Q.; McAllister, S.D.; Rorrer, W.K.; Aung, M.M.; Martin, B.R.; Abood, M.E. Evaluation of the cannabinoid CB2 receptor-selective antagonist, SR144528: Further evidence for cannabinoid CB2 receptor absence in the rat central nervous system. Eur. J. Pharmacol. 1999, 377, 117–125. [Google Scholar] [CrossRef]
- Becher, B.; Antel, J.P. Comparison of phenotypic and functional properties of immediately ex vivo and cultured human adult microglia. Glia 1996, 18, 1–10. [Google Scholar] [CrossRef]
- Klegeris, A.; Bissonnette, C.J.; McGeer, P.L. Reduction of human monocytic cell neurotoxicity and cytokine secretion by ligands of the cannabinoid-type CB2 receptor. Br. J. Pharmacol. 2003, 139, 775–786. [Google Scholar] [CrossRef] [Green Version]
- Rock, R.B.; Gekker, G.; Hu, S.; Sheng, W.S.; Cabral, G.A.; Martin, B.R.; Peterson, P.K. WIN55,212-2-Mediated Inhibition of HIV-1 Expression in Microglial Cells: Involvement of Cannabinoid Receptors. J. Neuroimmune Pharmacol. 2007, 2, 178–183. [Google Scholar] [CrossRef] [PubMed]
- Ashton, J.C.; Rahman, R.M.; Nair, S.M.; Sutherland, B.A.; Glass, M.; Appleton, I. Cerebral hypoxia-ischemia and middle cerebral artery occlusion induce expression of the cannabinoid CB2 receptor in the brain. Neurosci. Lett. 2007, 412, 114–117. [Google Scholar] [CrossRef] [PubMed]
- Benito, C.; Kim, W.K.; Chavarria, I.; Hillard, C.J.; Mackie, K.; Tolon, R.M.; Williams, K.; Romero, J. A glial endogenous cannabinoid system is upregulated in the brains of macaques with simian immunodeficiency virus-induced encephalitis. J. Neurosci. 2005, 25, 2530–2536. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Naguib, M.; Xu, J.J.; Diaz, P.; Brown, D.L.; Cogdell, D.; Bie, B.; Hu, J.; Craig, S.; Hittelman, W.N. Prevention of paclitaxel-induced neuropathy through activation of the central cannabinoid type 2 receptor system. Anesth. Analg. 2012, 114, 1104–1120. [Google Scholar] [CrossRef] [Green Version]
- Zarruk, J.G.; Fernandez-Lopez, D.; Garcia-Yebenes, I.; Garcia-Gutierrez, M.S.; Vivancos, J.; Nombela, F.; Torres, M.; Burguete, M.C.; Manzanares, J.; Lizasoain, I.; et al. Cannabinoid type 2 receptor activation downregulates stroke-induced classic and alternative brain macrophage/microglial activation concomitant to neuroprotection. Stroke 2012, 43, 211–219. [Google Scholar] [CrossRef] [PubMed]
- Zhang, J.; Hoffert, C.; Vu, H.K.; Groblewski, T.; Ahmad, S.; O’Donnell, D. Induction of CB2 receptor expression in the rat spinal cord of neuropathic but not inflammatory chronic pain models. Eur. J. Neurosci. 2003, 17, 2750–2754. [Google Scholar] [CrossRef] [PubMed]
- Stefano, G.B.; Liu, Y.; Goligorsky, M.S. Cannabinoid receptors are coupled to nitric oxide release in invertebrate immunocytes, microglia, and human monocytes. J. Biol. Chem. 1996, 271, 19238–19242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Waksman, Y.; Olson, J.M.; Carlisle, S.J.; Cabral, G.A. The central cannabinoid receptor (CB1) mediates inhibition of nitric oxide production by rat microglial cells. J. Pharmacol. Exp. Ther. 1999, 288, 1357–1366. [Google Scholar] [PubMed]
- Puffenbarger, R.A.; Boothe, A.C.; Cabral, G.A. Cannabinoids inhibit LPS-inducible cytokine mRNA expression in rat microglial cells. Glia 2000, 29, 58–69. [Google Scholar] [CrossRef]
- Facchinetti, F.; Del Giudice, E.; Furegato, S.; Passarotto, M.; Leon, A. Cannabinoids ablate release of TNFα in rat microglial cells stimulated with lypopolysaccharide. Glia 2003, 41, 161–168. [Google Scholar] [CrossRef]
- Romero-Sandoval, E.A.; Horvath, R.; Landry, R.P.; DeLeo, J.A. Cannabinoid receptor type 2 activation induces a microglial anti-inflammatory phenotype and reduces migration via MKP induction and ERK dephosphorylation. Mol. Pain. 2009, 5, 25. [Google Scholar] [CrossRef] [Green Version]
- Ramirez, B.G.; Blazquez, C.; Gomez del Pulgar, T.; Guzman, M.; de Ceballos, M.L. Prevention of Alzheimer’s disease pathology by cannabinoids: Neuroprotection mediated by blockade of microglial activation. J. Neurosci. 2005, 25, 1904–1913. [Google Scholar] [CrossRef] [Green Version]
- Correa, F.; Hernangomez, M.; Mestre, L.; Loria, F.; Spagnolo, A.; Docagne, F.; Di Marzo, V.; Guaza, C. Anandamide enhances IL-10 production in activated microglia by targeting CB(2) receptors: Roles of ERK1/2, JNK, and NF-kappaB. Glia 2010, 58, 135–147. [Google Scholar] [CrossRef]
- Reiner, A.; Heldt, S.A.; Presley, C.S.; Guley, N.H.; Elberger, A.J.; Deng, Y.; D’Surney, L.; Rogers, J.T.; Ferrell, J.; Bu, W.; et al. Motor, visual and emotional deficits in mice after closed-head mild traumatic brain injury are alleviated by the novel CB2 inverse agonist SMM-189. Int. J. Mol. Sci. 2014, 16, 758–787. [Google Scholar] [CrossRef]
- Offertaler, L.; Mo, F.M.; Batkai, S.; Liu, J.; Begg, M.; Razdan, R.K.; Martin, B.R.; Bukoski, R.D.; Kunos, G. Selective ligands and cellular effectors of a G protein-coupled endothelial cannabinoid receptor. Mol. Pharmacol. 2003, 63, 699–705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Horvath, R.J.; Nutile-McMenemy, N.; Alkaitis, M.S.; Deleo, J.A. Differential migration, LPS-induced cytokine, chemokine, and NO expression in immortalized BV-2 and HAPI cell lines and primary microglial cultures. J. Neurochem. 2008, 107, 557–569. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Benito, C.; Nunez, E.; Tolon, R.M.; Carrier, E.J.; Rabano, A.; Hillard, C.J.; Romero, J. Cannabinoid CB2 receptors and fatty acid amide hydrolase are selectively overexpressed in neuritic plaque-associated glia in Alzheimer’s disease brains. J. Neurosci. 2003, 23, 11136–11141. [Google Scholar] [CrossRef] [Green Version]
- Solas, M.; Francis, P.T.; Franco, R.; Ramirez, M.J. CB2 receptor and amyloid pathology in frontal cortex of Alzheimer’s disease patients. Neurobiol. Aging 2013, 34, 805–808. [Google Scholar] [CrossRef] [PubMed]
- Yiangou, Y.; Facer, P.; Durrenberger, P.; Chessell, I.P.; Naylor, A.; Bountra, C.; Banati, R.R.; Anand, P. COX-2, CB2 and P2X7-immunoreactivities are increased in activated microglial cells/macrophages of multiple sclerosis and amyotrophic lateral sclerosis spinal cord. BMC Neurol. 2006, 6, 12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nunez, E.; Benito, C.; Tolon, R.M.; Hillard, C.J.; Griffin, W.S.; Romero, J. Glial expression of cannabinoid CB(2) receptors and fatty acid amide hydrolase are beta amyloid-linked events in Down’s syndrome. Neuroscience 2008, 151, 104–110. [Google Scholar] [CrossRef]
- Bie, B.; Wu, J.; Foss, J.F.; Naguib, M. An overview of the cannabinoid type 2 receptor system and its therapeutic potential. Curr. Opin. Anaesthesiol. 2018, 31, 407–414. [Google Scholar] [CrossRef]
- Belayev, L.; Busto, R.; Zhao, W.; Ginsberg, M.D. HU-211, a novel noncompetitive N-methyl-D-aspartate antagonist, improves neurological deficit and reduces infarct volume after reversible focal cerebral ischemia in the rat. Stroke 1995, 26, 2313–2319. [Google Scholar] [CrossRef]
- Shohami, E.; Gallily, R.; Mechoulam, R.; Bass, R.; Ben-Hur, T. Cytokine production in the brain following closed head injury: Dexanabinol (HU-211) is a novel TNF-alpha inhibitor and an effective neuroprotectant. J. Neuroimmunol. 1997, 72, 169–177. [Google Scholar] [CrossRef]
- Zhang, M.; Adler, M.W.; Abood, M.E.; Ganea, D.; Jallo, J.; Tuma, R.F. CB2 receptor activation attenuates microcirculatory dysfunction during cerebral ischemic/reperfusion injury. Microvasc. Res. 2009, 78, 86–94. [Google Scholar] [CrossRef] [Green Version]
- Elliott, M.B.; Tuma, R.F.; Amenta, P.S.; Barbe, M.F.; Jallo, J.I. Acute effects of a selective cannabinoid-2 receptor agonist on neuroinflammation in a model of traumatic brain injury. J. Neurotrauma 2011, 28, 973–981. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Suzuki, N.; Suzuki, M.; Murakami, K.; Hamajo, K.; Tsukamoto, T.; Shimojo, M. Cerebroprotective effects of TAK-937, a cannabinoid receptor agonist, on ischemic brain damage in middle cerebral artery occluded rats and non-human primates. Brain Res. 2012, 1430, 93–100. [Google Scholar] [CrossRef]
- Maas, A.I.; Murray, G.; Henney, H., 3rd; Kassem, N.; Legrand, V.; Mangelus, M.; Muizelaar, J.P.; Stocchetti, N.; Knoller, N.; Pharmos, T.i. Efficacy and safety of dexanabinol in severe traumatic brain injury: Results of a phase III randomised, placebo-controlled, clinical trial. Lancet Neurol. 2006, 5, 38–45. [Google Scholar] [CrossRef]
- Shohami, E.; Novikov, M.; Bass, R. Long-term effect of HU-211, a novel non-competitive NMDA antagonist, on motor and memory functions after closed head injury in the rat. Brain Res. 1995, 674, 55–62. [Google Scholar] [CrossRef]
- Martin-Moreno, A.M.; Brera, B.; Spuch, C.; Carro, E.; Garcia-Garcia, L.; Delgado, M.; Pozo, M.A.; Innamorato, N.G.; Cuadrado, A.; de Ceballos, M.L. Prolonged oral cannabinoid administration prevents neuroinflammation, lowers beta-amyloid levels and improves cognitive performance in Tg APP 2576 mice. J. Neuroinflamm. 2012, 9, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lourbopoulos, A.; Grigoriadis, N.; Lagoudaki, R.; Touloumi, O.; Polyzoidou, E.; Mavromatis, I.; Tascos, N.; Breuer, A.; Ovadia, H.; Karussis, D. Administration of 2-arachidonoylglycerol ameliorates both acute and chronic experimental autoimmune encephalomyelitis. Brain Res. 2011, 1390, 126–141. [Google Scholar] [CrossRef]
- Ehrhart, J.; Obregon, D.; Mori, T.; Hou, H.; Sun, N.; Bai, Y.; Klein, T.; Fernandez, F.; Tan, J.; Shytle, R.D. Stimulation of cannabinoid receptor 2 (CB2) suppresses microglial activation. J. Neuroinflamm. 2005, 2, 29. [Google Scholar] [CrossRef] [Green Version]
- Tansey, M.G.; Goldberg, M.S. Neuroinflammation in Parkinson’s disease: Its role in neuronal death and implications for therapeutic intervention. Neurobiol. Dis. 2010, 37, 510–518. [Google Scholar] [CrossRef] [Green Version]
- Sanchez-Guajardo, V.; Barnum, C.J.; Tansey, M.G.; Romero-Ramos, M. Neuroimmunological processes in Parkinson’s disease and their relation to alpha-synuclein: microglia as the referee between neuronal processes and peripheral immunity. ASN Neuro 2013, 5, 113–139. [Google Scholar] [CrossRef] [Green Version]
- Garcia, M.C.; Cinquina, V.; Palomo-Garo, C.; Rabano, A.; Fernandez-Ruiz, J. Identification of CB(2) receptors in human nigral neurons that degenerate in Parkinson’s disease. Neurosci. Lett. 2015, 587, 1–4. [Google Scholar] [CrossRef]
- Gómez-Gálvez, Y.; Palomo-Garo, C.; Fernández-Ruiz, J.; García, C. Potential of the cannabinoid CB2 receptor as a pharmacological target against inflammation in Parkinson’s disease. Prog. Neuro-Psychopharmacol. 2016, 64, 200–208. [Google Scholar] [CrossRef] [PubMed]
- Herkenham, M.; Lynn, A.B.; Johnson, M.R.; Melvin, L.S.; de Costa, B.R.; Rice, K.C. Characterization and localization of cannabinoid receptors in rat brain: A quantitative in vitro autoradiographic study. J. Neurosci. 1991, 11, 563–583. [Google Scholar] [CrossRef] [PubMed]
- Mailleux, P.; Vanderhaeghen, J.J. Distribution of neuronal cannabinoid receptor in the adult rat brain: a comparative receptor binding radioautography and in situ hybridization histochemistry. Neuroscience 1992, 48, 655–668. [Google Scholar] [CrossRef]
- Hohmann, A.G.; Herkenham, M. Localization of cannabinoid CB(1) receptor mRNA in neuronal subpopulations of rat striatum: a double-label in situ hybridization study. Synapse 2000, 37, 71–80. [Google Scholar] [CrossRef]
- Hermann, H.; Marsicano, G.; Lutz, B. Coexpression of the cannabinoid receptor type 1 with dopamine and serotonin receptors in distinct neuronal subpopulations of the adult mouse forebrain. Neuroscience 2002, 109, 451–460. [Google Scholar] [CrossRef]
- Perez-Rial, S.; Garcia-Gutierrez, M.S.; Molina, J.A.; Perez-Nievas, B.G.; Ledent, C.; Leiva, C.; Leza, J.C.; Manzanares, J. Increased vulnerability to 6-hydroxydopamine lesion and reduced development of dyskinesias in mice lacking CB1 cannabinoid receptors. Neurobiol. Aging 2011, 32, 631–645. [Google Scholar] [CrossRef]
- Concannon, R.; Finn, D.P.; Dowd, E. Chapter 3-Cannabinoids in Parkinson’s disease. In Cannabinoids in Neurologic and Mental Disease; Fattore, L., Ed.; Academic Press: San Diego, CA, USA, 2015; pp. 35–59. [Google Scholar]
- Lastres-Becker, I.; Molina-Holgado, F.; Ramos, J.A.; Mechoulam, R.; Fernandez-Ruiz, J. Cannabinoids provide neuroprotection against 6-hydroxydopamine toxicity in vivo and in vitro: Relevance to Parkinson’s disease. Neurobiol. Dis. 2005, 19, 96–107. [Google Scholar] [CrossRef]
- Chung, E.S.; Bok, E.; Chung, Y.C.; Baik, H.H.; Jin, B.K. Cannabinoids prevent lipopolysaccharide-induced neurodegeneration in the rat substantia nigra in vivo through inhibition of microglial activation and NADPH oxidase. Brain Res. 2012, 1451, 110–116. [Google Scholar] [CrossRef]
- Chung, Y.C.; Bok, E.; Huh, S.H.; Park, J.Y.; Yoon, S.H.; Kim, S.R.; Kim, Y.S.; Maeng, S.; Park, S.H.; Jin, B.K. Cannabinoid receptor type 1 protects nigrostriatal dopaminergic neurons against MPTP neurotoxicity by inhibiting microglial activation. J. Immunol. 2011, 187, 6508–6517. [Google Scholar] [CrossRef] [Green Version]
- Benito, C.; Tolon, R.M.; Pazos, M.R.; Nunez, E.; Castillo, A.I.; Romero, J. Cannabinoid CB2 receptors in human brain inflammation. Br. J. Pharm. 2008, 153, 277–285. [Google Scholar] [CrossRef] [Green Version]
- Price, D.A.; Martinez, A.A.; Seillier, A.; Koek, W.; Acosta, Y.; Fernandez, E.; Strong, R.; Lutz, B.; Marsicano, G.; Roberts, J.L.; et al. WIN55,212-2, a cannabinoid receptor agonist, protects against nigrostriatal cell loss in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model of Parkinson’s disease. Eur. J. Neurosci. 2009, 29, 2177–2186. [Google Scholar] [CrossRef] [PubMed]
- Garcia, C.; Palomo-Garo, C.; Garcia-Arencibia, M.; Ramos, J.; Pertwee, R.; Fernandez-Ruiz, J. Symptom-relieving and neuroprotective effects of the phytocannabinoid Delta(9)-THCV in animal models of Parkinson’s disease. Br. J. Pharmacol. 2011, 163, 1495–1506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- García-Arencibia, M.; González, S.; de Lago, E.; Ramos, J.A.; Mechoulam, R.; Fernández-Ruiz, J. Evaluation of the neuroprotective effect of cannabinoids in a rat model of Parkinson’s disease: Importance of antioxidant and cannabinoid receptor-independent properties. Brain Res. 2007, 1134, 162–170. [Google Scholar] [CrossRef] [PubMed]
- Javed, H.; Azimullah, S.; Haque, M.E.; Ojha, S.K. Cannabinoid Type 2 (CB2) Receptors Activation Protects against Oxidative Stress and Neuroinflammation Associated Dopaminergic Neurodegeneration in Rotenone Model of Parkinson’s Disease. Front. Neurosci. 2016, 10, 321. [Google Scholar] [CrossRef] [Green Version]
- Ternianov, A.; Perez-Ortiz, J.M.; Solesio, M.E.; Garcia-Gutierrez, M.S.; Ortega-Alvaro, A.; Navarrete, F.; Leiva, C.; Galindo, M.F.; Manzanares, J. Overexpression of CB2 cannabinoid receptors results in neuroprotection against behavioral and neurochemical alterations induced by intracaudate administration of 6-hydroxydopamine. Neurobiol. Aging 2012, 33, 421.e1–421.e16. [Google Scholar] [CrossRef]
Species | Inflammagen | CB Treatment | Microglial Effects | Reference |
---|---|---|---|---|
CB2−/− mice (Deltagen, Jackson Lab)—C57BL/6 | IL-4 + IL-13 | ↓ Arg1 in KO microglia than WT microglia | [272] | |
IL-4 + IL-13 | ↓ Phagocytic activity in KO microglia measured by reduced engulfed fluorescent microspheres | |||
Rat (P0–P2)—Wistar | IL-4 + IL-13 | ↑ Arg1 mRNA and protein ↑ Acute (6 hr) in mRNA of CB2 and DAGLα ↓ Prolonged (24 hr) mRNA in MAGL, FAAH, and ↑ in CB1 and CB2, and NAPE-PLD | [272] | |
IL-4 + IL-13 | AM251 or AM630 | ↓ Cytokine-induced Arg1 mRNA and protein | ||
TGF-β | ↑ Acute (6 hr) in mRNA of CB1, CB2, NAPE-PLD, DAGLβ, and MAGL and ↓ FAAH Prolonged (24 hr) mRNA ↓ in NAPE-PLD, MAGL, and FAAH | |||
LPS | Acute (6 hr) ↓ in CB1, CB2, FAAH, NAPE-PLD, and MAGL with prolonged (24 hr) ↓ in FAAH and MAGL | |||
Rat (P1–P2)—Sprague Dawley | LPS | AEA | ↓ LPS-induced microglia cytokine mRNA of IL-1α and TNF | [287] |
LPS | SR144,528 | ↑ Microglia cytokine mRNA in dose-dependent manner (IL-1α, IL-1β, IL-6, TNF) | ||
Rat cortical (neonate)—Sprague Dawley | LPS | CP55,940 | ↓ LPS-induced production of TNFα | [288] |
LPS | AEA or 2-AG | ↓ LPS-induced production of TNFα | ||
Rat cortical (P2–P3)—Sprague Dawley | LPS | JWH015 | ↓ LPS-induced TNF protein expression ↓ LPS-stimulated microglia chemotaxis measured by # of cells that migrated toward the chemoattractant ADP | [289] |
LPS | JWH015 + AM630 | JWH015 effect on chemotaxis blocked by AM630, demonstrating CB2-specific effect on microglia migration | ||
Mouse forebrain (newborn)—Balb/c | LPS and IFNγ | Anandamide | ↑ Further the LPS/IFNγ-induced expression of IL-10 | [291] |
LPS and IFNγ | JWH133 | ↑ Further the LPS/IFNγ-induced expression of IL-10 that is ↓ with SR144528 (CB2 inverse agonist) and not by SR141716A (CB1 antagonist), suggesting CB2-mediated mechanism | ||
Rat cortical (neonate) | Fibrillar βA25–35 | HU-210, WIN55,212-2, or JWH133 | ↓ Fibrillar βA-induced TNF microglial release | [290] |
Human | LPS | SMM-189 | ↓ IFN-γ, IL-6, IL-12p70, and chemokines IL-8, MCP-1, CCL17 (TARC), macrophage derived-chemokine (MDC), and eotaxin-3 | [292] |
Species | Inflammagen | Cannabinoid Treatment | Treatment Timeline | Microglia/Inflammation Effects | Neuroprotective Effect | Reference |
---|---|---|---|---|---|---|
C57BL/6 mice | MPTP (4 × 20 mg/kg every 2 hrs) | HU-210 or WIN55,212-2 | 2 d before MPTP and again 12 hrs after MPTP and continue for 3 d (microglia analysis) and 7 d (neuron analysis) | ↓ MPTP-induced nigral Mac1 (CD11b) activation and production of O2−(ethidium accumulation) ↓ MPTP-induced 8-OHdG suggesting reduced protein oxidative damage ↓ MPTP-induced nigral TNF, IL-1β, and iNOS gene expression and TNF and IL-1β protein expression | ↑ MPTP-induced TH+ stereological nigral cell count ↑ MPTP-induced rotarod latency to fall and striatal dopamine content | [322] |
+AM251 (microglia/ neuron) or SR141716A (neuron) | 30 min prior to non-selective agonists | ↑ Agonist-induced nigral Mac1 (CD11b) activation and production of O2− (ethidium accumulation) ↑ Agonist-induced 8-OHdG suggesting oxidative damage ↑ Agonist-induced nigral TNF, IL-1β, and iNOS gene expression and TNF and IL-1β protein expression | ↓ Agonist-induced TH+ stereological nigral cell count (no difference with MPTP alone group) when treated with CB1 antagonists ↓ Agonist-induced rotarod latency to fall and striatal dopamine content (no difference with MPTP alone group) when treated with AM251 | |||
CB2−/− (C57BL/6 background) | Intrastriatal LPS | - | - | ↑ CD68-immunoreactivity in the nigra of KO mice compared to WT | Not evaluated | [313] |
C57BL/6 mice | Intrastriatal LPS | HU-308 | Daily injections for 2 weeks starting 16 hr after LPS | ↓ LPS-induced nigral CD68-immunoreactivity ↓ LPS-induced striatal iNOS gene expression | ↑ LPS-induced nigral TH-immunoreactivity | |
C57BL/6 P1 glia and cerebellar neural cultures | 6-OHDA | HU-210 directly to cultures | Neurons +/− conditioned media from glia culture 24 hrs after HU-210 | Not directly evaluated but neuroprotective effects from glia conditioned media suggest CB1 and CB2-mediated glial effects | ↑ cerebellar granule cell survival with direct HU-210 to neurons and greater protection when neurons treated with glia conditioned media treated with HU-210 | [320] |
Sprague Dawley rats | Intranigral LPS | HU-210 or WIN55,212-2 | ICV injections 1 hr prior to LPS | ↓ LPS-induced nigral CD11b activation and production of O2− (ethidium accumulation) ↓ TNF and IL-1β after WIN55,212-2 and ↓ IL-1β after HU-210 24 hrs after LPS as measured by ELISA ↓ p67phox and p47phox subunits in cytosol and membrane nigral fractions 12 hrs after LPS by western, suggesting reduced translocation of NADPH oxidase which was specific to CD11b+ cells | ↑ TH+ stereological nigral cell count | [321] |
C57BL/6 mice | MPTP (4 × 20 mg/kg every 2 hrs) | JWH015 (microglia) or WIN55,212-2 (microglia/ neuron) | Daily for 3d (microglia analysis) or 5 d (neuron analysis) starting 1 d after MPTP | ↓ MPTP-induced nigral Mac1 (CD11b) protein with WIN55,212-2 | ↑ MPTP-induced nigral TH+ stereological neuron counts (dose-dependent) ↑ MPTP-induced midbrain dopamine levels after WIN55,212-2 treatment | [324] |
WIN55,212-2 +JTE907 | 20 min before WIN55,212-2 | ↑ Agonist-induced Mac1 (CD11b) when administered alone or in conjunction with WIN55,212-2 agonist | Not evaluated | |||
Sprague Dawley rats | ICV 6-OHDA | Δ9-THCV or enriched CBD | Daily for 14 d starting 16 hrs after 6-OHDA | ↓ 6-OHDA-induced nigral OX-42-immunoreactivity with either Δ9-THCV or enriched CBD | ↑ 6-OHDA-induced nigral TH-immunoreactivity when treated with CBD but not Δ9-THCV | [325] |
C57BL/6 mice | Intrastriatal LPS | Δ9-THCV or HU-308 | Daily for 14 d starting 16 hrs after LPS | Not evaluated | ↑ LPS-induced nigral TH-immunoreactivity when treated with HU-308 or Δ9-THCV | |
Sprague Dawley rats | Unilateral MFB injection of 6-OHDA | CBD | Daily for 2 weeks, starting 16 hr after 6-OHDA | ↑ 6-OHDA induced striatal Cu,Zn-SOD gene expression suggesting a protection from endogenous oxidative stress | Not evaluated | [326] |
HU-308 | Daily for 2 weeks, starting 16 hr after 6-OHDA | Not evaluated | Did not alter striatal TH activity by HPLC or nigral TH mRNA levels compared to 6-OHDA+vehicle | |||
Wistar rats | Rotenone i.p. once daily for 4 weeks | βcaryophyllene (BCP) ± AM630 | Daily for 4 weeks and 30 min prior to rotenone | ↓ Rotenone-induced striatal Iba1+ activated microglia with BCP and blocked by AM630 ↓ Rotenone-induced striatal GFAP activated astrocytes with BCP and blocked by AM630 ↓ Rotenone-induced midbrain pro-inflammatory cytokines IL-1β, TNF, and IL-6 with BCP and blocked by AM630 ↓ Rotenone-induced striatal NFκB p65, COX-2, iNOS with BCP and blocked by AM630 | ↑ Rotenone-induced striatal and nigral TH-immunoreactivity with BCP and blocked by AM630 | [327] |
CB2xP mice (overexpression of mouse CB2) | Unilateral striatal 6-OHDA | - | 7 weeks | ↓ 6-OHDA-induced striatal GFAP expression vs. WT Overexpression of CB2 altered striatal Iba1 immunoreactivity, but not striatal levels of iNOS and COX2 vs. matched WT ↓ Striatal malonyldialdehyde (lipid peroxidation product) vs. WT at basal and 6-OHDA conditions ↓ 6-OHDA induced striatal ratio of oxidized GSSG:glutathione (oxidative stress marker) vs. WT | ↓ 6-OHDA-induced apomorphine rotations vs. WT ↑ 6-OHDA-induced time in open arms of elevated plus maze suggesting CB2 role in anxiety-like behavior vs. WT ↑ 6-OHDA-induced memory impairment in step-down inhibitory avoidance task vs. WT ↑ 6-OHDA-induced striatal and nigral TH immunostaining vs. WT | [328] |
© 2020 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
Kelly, R.; Joers, V.; Tansey, M.G.; McKernan, D.P.; Dowd, E. Microglial Phenotypes and Their Relationship to the Cannabinoid System: Therapeutic Implications for Parkinson’s Disease. Molecules 2020, 25, 453. https://doi.org/10.3390/molecules25030453
Kelly R, Joers V, Tansey MG, McKernan DP, Dowd E. Microglial Phenotypes and Their Relationship to the Cannabinoid System: Therapeutic Implications for Parkinson’s Disease. Molecules. 2020; 25(3):453. https://doi.org/10.3390/molecules25030453
Chicago/Turabian StyleKelly, Rachel, Valerie Joers, Malú G. Tansey, Declan P. McKernan, and Eilís Dowd. 2020. "Microglial Phenotypes and Their Relationship to the Cannabinoid System: Therapeutic Implications for Parkinson’s Disease" Molecules 25, no. 3: 453. https://doi.org/10.3390/molecules25030453
APA StyleKelly, R., Joers, V., Tansey, M. G., McKernan, D. P., & Dowd, E. (2020). Microglial Phenotypes and Their Relationship to the Cannabinoid System: Therapeutic Implications for Parkinson’s Disease. Molecules, 25(3), 453. https://doi.org/10.3390/molecules25030453