327
Views
0
CrossRef citations to date
0
Altmetric
Review

Astroglial 5-HT2B receptor in mood disorders

, , &
Pages 435-442 | Received 11 Sep 2017, Accepted 26 Mar 2018, Published online: 30 Mar 2018

References

  • Verkhratsky A, Butt AM. Glial physiology and pathophysiology. Chichester: Wiley-Blackwell; 2013.
  • Verkhratsky A, Parpura V. Physiology of astroglia: channels, receptors, transporters, ion signaling and gliotransmission. In: Colloquium series on neuroglia in biology and medicine: from physiology to disease (E-textbook series) Ed by A. Verkhratsky, V.Parpura. Morgan & Claypool Publishers; 2015. p. 172.
  • Pekny M, Pekna M, Messing A, et al. Astrocytes: a central element in neurological diseases. Acta Neuropathol. 2016;131:323–345.
  • Verkhratsky A, Zorec R, Parpura V. Stratification of astrocytes in healthy and diseased brain. Brain Pathol. 2017;27:629–644.
  • Zeidan-Chulia F, Salmina AB, Malinovskaya NA, et al. The glial perspective of autism spectrum disorders. Neurosci Biobehav Rev. 2014;38:160–172.
  • Verkhratsky A, Rodríguez JJ, Steardo L. Astrogliopathology: a central element of neuropsychiatric diseases? Neuroscientist. 2014;20:576–588.
  • Peng L, Verkhratsky A, Gu L, et al. Targeting astrocytes in major depression. Expert Rev Neurother. 2015;15:1299–1306.
  • Peng L, Li B, Verkhratsky A. Targeting astrocytes in bipolar disorder. Expert Rev Neurother. 2016;16:649–657.
  • Hertz L, Chen Y, Gibbs ME, et al. Astrocytic adrenoceptors: a major drug target in neurological and psychiatric disorders? Curr Drug Targets CNS Neurol Disord. 2014;3:239–267.
  • Saura J, Kettler R, Da Prada M, et al. Quantitative enzyme radioautography with 3H-Ro 41-1049 and 3H-Ro 19-6327 in vitro: localization and abundance of MAO-A and MAO-B in rat CNS, peripheral organs, and human brain. J Neurosci. 1992;12:1977–1999.
  • Fitzgerald LW, Kaplinsky L, Kimelberg HK. Serotonin metabolism by monoamine oxidase in rat primary astrocyte cultures. J Neurochem. 1990;55:2008–2014.
  • Inazu M, Takeda H, Matsumiya T. Functional expression of the norepinephrine transporter in cultured rat astrocytes. J Neurochem. 2003;84:136–144.
  • Pacholczyk T, Blakely RD, Amara SG. Expression cloning of a cocaine- and antidepressant-sensitive human noradrenaline transporter. Nature. 1991;350:350–354.
  • Karakaya S, Kipp M, Beyer C. Oestrogen regulates the expression and function of dopamine transporters in astrocytes of the nigrostriatal system. J Neuroendocrinol. 2007;19:682–690.
  • Malynn S, Campos-Torres A, Moynagh P, et al. The pro-inflammatory cytokine TNF-alpha regulates the activity and expression of the serotonin transporter (SERT) in astrocytes. Neurochem Res. 2013;38:694–704.
  • Kirischuk S, Parpura V, Verkhratsky A. Sodium dynamics: another key to astroglial excitability? Trends Neurosci. 2012;35:497–506.
  • Rose CR, Verkhratsky A. Principles of sodium homeostasis and sodium signalling in astroglia. Glia. 2016;64:1611–1627.
  • Hertz L, Lovatt D, Goldman SA, et al. Adrenoceptors in brain: cellular gene expression and effects on astrocytic metabolism and [Ca2+]i. Neurochem Int. 2010;57:411–420.
  • Ding F, O’Donnell J, As T, et al. α1-Adrenergic receptors mediate coordinated Ca2+ signaling of cortical astrocytes in awake, behaving mice. Cell Calcium. 2013;54:387–394.
  • Kirischuk S, Tuschick S, Verkhratsky A, et al. Calcium signalling in mouse Bergmann glial cells mediated by α1-adrenoreceptors and H1 histamine receptors. Eur J Neurosci. 1996;8:1198–1208.
  • Hertz L, Rothman DL, Li B, et al. Chronic SSRI stimulation of astrocytic 5-HT2B receptors change multiple gene expressions/editings and metabolism of glutamate, glucose and glycogen: a potential paradigm shift. Front Behav Neurosci. 2015;9:25.
  • Zhang S, Li B, Lovatt D, et al. 5-HT2B receptors are expressed on astrocytes from brain and in culture and are chronic target for all five conventional “serotonin-specific reuptake inhibitors”. Neuron Glia Biol. 2010;6:113–125.
  • Hertz L, Li B, Song D, et al. Astrocytes as a 5-HT2B-mediated SERT-independent SSRI target, slowly altering depression-associated genes and function. Curr Signal Transduct Ther. 2012;7:65–80.
  • Baez M, Kursar JD, Helton LA, et al. Molecular biology of serotonin receptors. Obes Res. 1995;Suppl 4:441S–447S.
  • Kursar JD, Nelson DL, Wainscott DB, et al. Molecular cloning, functional expression, and pharmacological characterization of a novel serotonin receptor (5-hydroxytryptamine2F) from rat stomach fundus. Mol Pharmacol. 1992;42:549–557.
  • Kursar JD, Nelson DL, Wainscott DB, et al. Molecular cloning, functional expression, and mRNA tissue distribution of the human 5-hydroxytryptamine2B receptor. Mol Pharmacol. 1994;46:227–234.
  • Choi DS, Maroteaux L. Immunohistochemical localisation of the serotonin 5-HT2B receptor in mouse gut, cardiovascular system, and brain. FEBS Lett. 1996;391:45–51.
  • Li B, Dong L, Wang B, et al. Cell type-specific gene expression and editing responses to chronic fluoxetine treatment in the in vivo mouse brain and their relevance for stress-induced anhedonia. Neurochem Res. 2012;37:2480–2495.
  • Roth BL, Willins DL, Kristiansen K, et al. 5-Hydroxytryptamine 2-family receptors (5-hydroxytryptamine2A, 5-hydroxytryptamine2B, 5-hydroxytryptamine2C): where structure meets function. Pharmacol Ther. 1998;79:231–257.
  • Sanden N, Thorlin T, Blomstrand F, et al. 5-hydroxytryptamine2B receptors stimulate Ca2+ increases in cultured astrocytes from three different brain regions. Neurochem Int. 2000;36:427–434.
  • Jerman JC, Brough SJ, Gager T, et al. Pharmacological characterisation of human 5-HT2 receptor subtypes. Eur J Pharmacol. 2001;414:23–30.
  • Hosli L, Hosli E. Receptors for dopamine and serotonin on astrocytes of cultured rat central nervous system. J Physiol (Paris). 1987;82:191–195.
  • Hertz L, Baldwin F, Schousboe A. Serotonin receptors on astrocytes in primary cultures: effects of methysergide and fluoxetine. Can J Physiol Pharmacol. 1979;57:223–226.
  • Magistretti PJ, Manthorpe M, Bloom FE, et al. Functional receptors for vasoactive intestinal polypeptide in cultured astroglia from neonatal rat brain. Regul Pept. 1983;6:71–80.
  • Hertz L, Tamir H. Some properties of an astrocytic protein fraction that binds serotonin. J Neurochem. 1981;37:1331–1334.
  • Carson MJ, Thomas EA, Danielson PE, et al. The 5HT5A serotonin receptor is expressed predominantly by astrocytes in which it inhibits cAMP accumulation: a mechanism for neuronal suppression of reactive astrocytes. Glia. 1996;17:317–326.
  • Maxishima M, Shiga T, Shutoh F, et al. Serotonin 2A receptor-like immunoreactivity is detected in astrocytes but not in oligodendrocytes of rat spinal cord. Brain Res. 2001;889:270–273.
  • Wu C, Singh SK, Dias P, et al. Activated astrocytes display increased 5-HT2a receptor expression in pathological states. Exp Neurol. 1999;158:529–533.
  • Cohen Z, Bouchelet I, Olivier A, et al. Multiple microvascular and astroglial 5-hydroxytryptamine receptor subtypes in human brain: molecular and pharmacologic characterization. J Cereb Blood Flow Metab. 1999;19:908–917.
  • Hirst WD, Cheung NY, Rattray M, et al. Cultured astrocytes express messenger RNA for multiple serotonin receptor subtypes, without functional coupling of 5-ht1 receptor subtypes to adenylyl cyclase. Brain Res Mol Brain Res. 1998;61:90–99.
  • Li B, Zhang S, Li M, et al. Serotonin increases ERK1/2 phosphorylation in astrocytes by stimulation of 5-HT2B and 5-HT2C receptors. Neurochem Int. 2010;57:432–439.
  • Schipke CG, Heuser I, Peters O. Antidepressants act on glial cells: SSRIs and serotonin elicit astrocyte calcium signaling in the mouse prefrontal cortex. J Psychiatr Res. 2011;45:242–248.
  • Bai Q, Song D, Gu L, et al. Bi-phasic regulation of glycogen content in astrocytes via Cav-1/PTEN/PI3K/AKT/GSK-3β pathway by fluoxetine. Psychopharmacology. 2017;234:1069–1077.
  • Li B, Zhang S, Zhang H, et al. Fluoxetine-mediated 5-HT2B receptor stimulation in astrocytes causes EGF receptor transactivation and ERK phosphorylation. Psychopharmacology. 2008;201:443–458.
  • Sowa G. Novel insights into the role of caveolin-2 in cell- and tissue-specific signaling and function. Biochem Res Int. 2011;2011:809259.
  • Smart EJ, Graf GA, McNiven MA, et al. Caveolins, liquid-ordered domains, and signal transduction. Mol Cell Biol. 1999;19:7289–7304.
  • Stary CM, Tsutsumi YM, Patel PM, et al. Caveolins: targeting pro-survival signaling in the heart and brain. Front Physiol. 2012;3:393.
  • Zschocke J, Manthey D, Bayatti N, et al. Estrogen receptor α-mediated silencing of caveolin gene expression in neuronal cells. J Biol Chem. 2002;277:38772–38780.
  • Shin T, Kim H, Jin JK, et al. Expression of caveolin-1, -2, and -3 in the spinal cords of Lewis rats with experimental autoimmune encephalomyelitis. J Neuroimmunol. 2005;165:11–20.
  • Robbins HL, Hague A. The PI3K/Akt pathway in tumors of endocrine tissues. Front Endocrinol (Lausanne). 2016;6:188.
  • Xia H, Khalil W, Kahm J, et al. Pathologic caveolin-1 regulation of PTEN in idiopathic pulmonary fibrosis. Am J Pathol. 2010;176:2626–2637.
  • Gould TD, Manji HK. Glycogen synthase kinase-3: a putative molecular target for lithium mimetic drugs. Neuropsychopharmacology. 2005;30:1223–1237.
  • Perez-Costas E, Gandy JC, Melendez-Ferro M, et al. Light and electron microscopy study of glycogen synthase kinase-3beta in the mouse brain. PLoS ONE. 2010;5:e8911.
  • Fang X, Yu SX, Lu Y, et al. Phosphorylation and inactivation of glycogen synthase kinase 3 by protein kinase A. Proc Natl Acad Sci U S A. 2000;97:11960–11965.
  • Karege F, Perroud N, Burkhardt S, et al. Protein levels of β-catenin and activation state of glycogen synthase kinase-3β in major depression. A study with postmortem prefrontal cortex. Affect Disord. 2012;136:185–188.
  • Zhang K, Song X, Xu Y, et al. Continuous GSK-3β overexpression in the hippocampal dentate gyrus induces prodepressant-like effects and increases sensitivity to chronic mild stress in mice. J Affect Disord. 2013;146:45–52.
  • Verkhratsky A, Rodriguez JJ, Steardo L. Astrogliopathology: a central element of neuropsychiatric diseases? Neuroscientist. 2014;20:576–588.
  • Rajkowska G, Stockmeier CA. Astrocyte pathology in major depressive disorder: insights from human postmortem brain tissue. Curr Drug Targets. 2013;14:1225–1236.
  • Jaggi AS, Bhatia N, Kumar N, et al. A review on animal models for screening potential anti-stress agents. Neurol Sci. 2011;32:993–1005.
  • Feng G, Mellor RH, Bernstein M, et al. Imaging neuronal subsets in transgenic mice expressing multiple spectral variants of GFP. Neuron. 2000;28:41–51.
  • Seki M, Nawa H, Morioka T, et al. Establishment of a novel enzyme-linked immunosorbent assay for Thy-1; quantitative assessment of neuronal degeneration. Neurosci Lett. 2002;329:185–188.
  • Dong L, Li B, Verkhratsky A, et al. Cell type-specific in vivo expression of genes encoding signalling molecules in the brain in response to chronic mild stress and chronic treatment with fluoxetine. Psychopharmacology. 2015;232:2827–2835.
  • Bass BL. RNA editing by adenosine deaminases that act on RNA. Annu Rev Biochem. 2002;71:817–846.
  • Peng L, Gu L, Li B, et al. Fluoxetine and all other SSRIs are 5-HT2B agonists – importance for their therapeutic effects. Curr Neuropharmacol. 2014;12:365–379.
  • Hornykiewicz O, Kish SJ. Biochemical pathophysiology of Parkinson’s disease. Adv Neurol. 1987;45:19–34.
  • Costa FH, Rosso AL, Maultasch H, et al. Depression in Parkinson’s disease: diagnosis and treatment. Arq Neuropsiquiatr. 2012;70:617–620.
  • Zhang X, Song D, Gu L, et al. Decrease of gene expression of astrocytic 5-HT2B receptors parallels development of depressive phenotype in a mouse model of Parkinson’s disease. Front Cell Neurosci. 2015;9:388.
  • Verkhratsky A, Nedergaard M. Physiology of astroglia. Physiol Rev. 2018;98:239–389.
  • Peng L, Parpura V, Verkhratsky A. Neuroglia as a central element of neurological diseases: an underappreciated target for therapeutic intervention. Curr Neuropharmacol. 2014;12:303–307.
  • Oberheim NA, Takano T, Han X, et al. Uniquely hominid features of adult human astrocytes. J Neurosci. 2009;29:3276–3287.
  • Sosunov AA, Wu X, Tsankova NM, et al. Phenotypic heterogeneity and plasticity of isocortical and hippocampal astrocytes in the human brain. J Neurosci. 2014;34:2285–2298.
  • Jones VC, Atkinson-Dell R, Verkhratsky A, et al. Aberrant iPSC-derived human astrocytes in Alzheimer’s disease. Cell Death Dis. 2017;8:e2696.
  • Hallmann AL, Arauzo-Bravo MJ, Mavrommatis L, et al. Astrocyte pathology in a human neural stem cell model of frontotemporal dementia caused by mutant TAU protein. Sci Rep. 2017;7:42991.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.