GET THE APP

Astrocyte Function in Alcohol Reward and Addiction
Journal of Alcoholism & Drug Dependence

Journal of Alcoholism & Drug Dependence
Open Access

ISSN: 2329-6488

+44 1223 790975

Editorial - (2015) Volume 3, Issue 4

Astrocyte Function in Alcohol Reward and Addiction

Louise Adermark*
Addiction Biology Unit, Sahlgrenska Academy, Institute of Neuroscience and Physiology, University of Gothenburg, Box 410, SE-405 30 Gothenburg, Sweden
*Corresponding Author: Louise Adermark, Addiction Biology Unit, Sahlgrenska Academy, Institute of Neuroscience and Physiology, University of Gothenburg, Box 410, SE-405 30 Gothenburg, Sweden, Tel: +46-31 786-3975 Email:

Abstract

Glial cells, particularly astrocytes, play essential roles in the regulation of neurotransmission, metabolism, and supply of energy substrates for synaptic transmission. One astrocyte can receive inputs from several hundreds of synapses, and synchronized neuronal activity correlates with astrocyte calcium signaling. Astrocyte pathology is a common feature of ethanol exposure in both humans and animal models, and brief alcohol intake is sufficient to cause long-lasting changes in astrocyte gene expression, activity and proliferation. Recent research also suggests that astrocytes shape the rewarding sensation of ethanol, and might be involved in modulating alcohol consumption. Considering the role of astrocytes in regulating glutamate homeostasis, a crucial component of alcohol abuse disorders, the astrocyte might be an important target for the development of new pharmacological treatments of alcoholism.

Keywords: Alcohol; Astrocytes; Astroglia; Glia; GFAP; Dopamine; Ethanol

Astrocytes

The astroglial cell, or astrocyte, is the most numerous cell type of the glial cell family. Astrocytes give structural and metabolic support to surrounding neurons and are pivotal for neuronal functioning and signal processing in the CNS [1,2] (Figure 1). The end feet of astrocytic processes encapsulate blood vessels and participate in building up the blood brain barrier [3,4]. The contact with blood vessels also enables the astrocytes to provide neurons with lactate under anaerobic conditions [1,5] (Figure 1). The astrocyte was first acknowledged as the primary cell type responsible for potassium buffering [6], and combined with the clearance of amino acids from the extracellular space, astrocytes warrants a high signal to noise ratio, and reduced receptor desensitization [7,8]. One single astrocyte can enwrap several neuronal somata and receive inputs from thousands of synapses, enabling them to sense and integrate synaptic activity [9,10]. Activation of astrocytes also appears to be a crucial component for the induction of synaptic plasticity mechanisms, and both long-term potentiation (LTP) and long-term depression (LTD) in the hippocampus may be regulated by astrocytes [11,12].

alcoholism-drug-dependence-integrate-neuronal-functioning

Figure 1: Astrocytes sense and integrate neuronal functioning.

Astrocytes enwrap both presynaptic and postsynaptic terminals, forming the tripartite synapse. Astrocytic end feet encapsulate blood vessels, enabling the astrocyte to give metabolic support to surrounding neurons. Astrocytic clearance of neuroactive substances is crucial for synaptic transmission. The excitatory amino acid glutamate (Glu) is rapidly metabolized to its non-excitable precursor glutamine (Gln), released back to the extracellular space, and taken up by neurons for synthesis of glutamate or GABA. See text for further details.

Astrocytic transporters

One of the most prominent functions of the astrocyte is the clearance of glutamate, and astrocytic glutamate transporters EAAT2 (GLT-1) and EAAT1 (GLAST) are the most abundant transporters for removal of glutamate in the brain [13]. Impaired astrocytic glutamate clearance appears to contribute to neuronal dysfunction and degeneration in Huntington’s disease, major depressive disorder and Alzheimer’s disease [14-17], and might also play a role in alcohol abuse [18]. Following reuptake, glutamate is converted by glutamine synthetase to its non-excitable precursor glutamine, which is released back to the extracellular space [19] (Figure 1). This astrocyte dependent glutamate-glutamine cycle is required to maintain active neurotransmission at excitatory terminals, but may also play a vital role for GABAergic neurotransmission [20,21]. Inhibitory transmission is further regulated by astrocytes through glycine transporters GlyT1 and GlyT2, and GABA transporters GAT-1 and GAT-3 [22,23], while adenosine is transported through the equilibrative nucleoside transporter 1 and 3 (ENT1, ENT3) [24,25].

Astrocytic calcium signaling

Even though astrocytes are not electrically excitable they express receptors for the majority of neurotransmitters and neuromodulators [26-31]. Upon activation, astrocytes generate various complex changes in intracellular Ca2+, which can initiate the release of gliotransmittors such as adenosine, D-serine and glutamate [32-35]. The main Ca2+- signaling pathway is mediated via G-protein receptor activated phospholipase C (PLC) and formation of inositol (1,4,5) trisphosphate (IP3) through hydrolysis of phosphatidylinositol 4,5-bisphosphate (PIP2), which induces Ca2+-release from internal stores. Photorelease of Ca2+ or IP3 within a single astrocyte is also sufficient to cause glutamaterelease and to modulate neuronal excitation [32,34,36-40]. In addition to Ca2+ release from internal stores, activation of G-coupled receptors can also initiate influx of Ca2+ through different types of voltageindependent Ca2+ channels [41,42] (Figure 2).

alcoholism-drug-dependence-Receptor-activated-calcium

Figure 2: Receptor activated calcium signaling in astrocytes.

Intracellular Ca2+-signaling is primarily mediated through IP3- gated release of Ca2+ from internal stores, which may be followed by Ca2+-influx through Ca2+ channels. Gap junction channels, which are dynamically regulated by neuronal activity and intracellular homeostasis, allow Ca2+ waves to spread through the astrocytic network. (CIF = Calcium Influx Factor; DOCC = Depletion-Operated Calcium Channel; SMOCC = Second Messenger-Operated Calcium Channel; ROCC = Receptor-Operated Calcium Channel; ER = Endoplasmic Reticulum).

Gap junction channels

Astrocytes are highly interconnected through gap junction channels, built up by the protein connexin [43]. These gap junction channels concede bidirectional diffusion of charged and uncharged small molecules allowing Ca2+ waves to spread through the astrocytic syncytium [44]. Functional gap junctions might also exist between astrocytes and oligodendrocytes and to some extent neurons [45-47]. Coupling at gap junctions is regulated by intracellular ion homeostasis and influenced by neuronal activity and released neuroactive substances such as growth hormones, glutamate, 5-HT, ATP, and potassium [48- 53] (Figure 2). Connexins also form plasma membrane channels, hemi channels, through which gliotransmitters can be released [54,55]. Connexin-43 hemi channels are necessary for fear memory consolidation and has been suggested as pharmacological targets for treatment of post-traumatic stress disorder, but dysregulations of hemi channel properties could also be critical during homeostatic imbalances [54,56]. Importantly, the connectivity of the astrocyte network, as well as individual cells, appears to be brain region selective, where astrocytes in the striatum are highly interconnected and primarily categorized as passive, while a larger proportion of astrocytes in the hippocampus are classified as complex [2,49,57].

The Astrocyte during Acute and Chronic Alcohol Exposure

Ethanol effects on signal transduction appear to vary considerable with development, differentiation and functional activity [58]. Many membrane proteins, such as ion channels, transporters, receptors, enzymes and G-proteins are affected by ethanol in ways that lead to long-lasting changes in activity and gene expression [59], and astrocytes appear to be important target cells. Brief ethanol-exposure is sufficient to alter astrocytic gene expression and to restrict proliferation [60,61], and may acutely impair astrocytic regulation of neurotransmission. Ethanol elevates intracellular calcium in a subset of astrocytes and reduces gap junction coupling in a brain region selective manner, which could selectively affect signal processing [62-64]. Considering that gap junction coupling equalizes [Na+]i, the decline in connectivity might also reduce the capacity for amino acid uptake and potassium buffering, as well as lead to failure in coordinating other physiological responses that involves sodium [65,66]. Diminished immunoreactive puncta, and reduced levels of the gap junction protein connexin 43, is also seen in post mortem studies of subjects with alcohol abuse disorder, supporting a role for altered connectivity or hemi channelbased communication in the pathophysiology of alcoholism [67].

Astrocytic cell swelling

Ethanol induces cell swelling in a subset of astrocytes, which could lead to changes in size and geometry of the extracellular space and thereby altered volume transmission [64,68-70]. Swelling of the end feet of astrocytic perivascular processes may also lead to altered cerebral blood flow [71]. Cell swelling is rapidly followed by regulatory volume decrease and the release of organic osmolytes, of which the amino acid taurine is of especial interest [72-75]. Taurine has previously been shown to increase accumbal dopamine levels, and appears to be a key component in ethanol induced dopamine release [76-78]. Considering the role of dopamine in mediating the rewarding sensation of drugs of abuse, it is thus possible that astrocytes are indirectly involved in ethanol-induced reinforcement [65,79,80]. Rapid swelling and regulatory volume decrease is facilitated by astrocytic water channels built up by the protein aquaporin-4 [81,82], and reduced aquaporin-4 expression has in fact also been linked to excessive alcohol consumption [83]. Interestingly, aquaporin-4 expression is depressed by dopamine, while genetic deletion of aquaporin-4 reduces accumbal dopamine [81]. Antagonists targeting dopamine D1 and D2 receptors has also been shown to increase astrocyte calcium excitability, synchrony and gap junction coupling [84], indicating that astrocytes may monitor extra synaptic dopamine levels.

Astrocytic protein expression and morphological transformations

Changes in the expression of the astrocyte specific cytoskeletal protein glial fibrillary acidic protein (GFAP) have been detected in several psychiatric conditions such as Parkinson’s disease, schizophrenia, depression and Alzheimer’s disease [17,85-89], but it is not clear if this is in response to the injury, or part of the cause of the disorder. Voluntary intake of alcohol, as well as forced exposure, increases both GFAP and GFAP mRNA expression [90-93], while withdrawal reduces the number of GFAP positive cells [94,95]. Alcohol also affects astrocyte morphology and induces rearrangements of actin filaments, which may further impair Ca2+ signaling [64,96]. Post mortem analysis of human alcoholics reports reduced GFAP expression and altered astrocyte morphology [97]. These prominent changes in astrocyte morphology are not exclusive to alcoholics with liver pathology, and may persist despite the cessation of alcohol consumption [98,99].

Astrocytic amino acid transporters and ethanol

Astrocytic amino acid transporters are crucial regulators of neurotransmission, and ethanol may cause glutamatergic dysfunction by affecting astrocytic proteins responsible for maintaining extracellular homeostasis [100]. Over all, chronic ethanol exposure causes long-lasting alterations in the expression of glutamatergic receptors and transporters, producing a hyperglutamatergic state [101-104]. Pharmacological modulation of astrocytic clearance of glutamate might thus indirectly mitigate behavioral responses caused by ethanol exposure. Supporting this theory, selective up regulation of the glutamate transporter GLT-1 in the nucleus accumbens shell reduces voluntary ethanol consumption, and impairs methamphetamine and morphine induced conditioned place preference (CPP) [105,106]. However, mice with homozygous GLAST deletion show a pronounced reduction of voluntary alcohol consumption, and inhibited ethanolinduced CPP [107]. In addition, blocking central astrocytic glutamate uptake selectively attenuates ethanol binge drinking behavior in mice [108]. The role of glutamate transporters might thus depend on the brain region targeted, the amount of alcohol consumed, and/or the transporter subtype manipulated. A differential role for astrocyte glutamate transporters is also supported by the selective correlation between GLT-1 and alcohol consumption [106]. Interestingly, changes in adenosine levels, caused by ethanol exposure, may also indirectly alter GLT-1 expression, further affecting alcohol consumption [25,83].

Astrocytes also express transporters for glycine, which has been implemented in the rewarding properties of ethanol through the potentiation of NMDA receptors and activation of glycine receptors [109-111]. Especially, glycine receptors could be important regulators of ethanol induced reinforcement as antagonists targeting glycine receptors both blocks the dopamine-elevating property of ethanol in vivo, as well as prevents ethanol-induced changes in striatal neurotransmission ex vivo [79,111,112]. In addition, inhibition of the glycine transporter GlyT1 persistently reduces ethanol intake and relapse-like alcohol drinking in rats [113-116], and might thus be a pharmacological target for the treatment of alcohol abuse disorders. However, even though it was recently shown that the GlyT1 transporter inhibitor Org25935 robustly reduces the number of days with heavy drinking, the effect was not significant as compared to placebo [117]. More research is thus required in order to fully elucidate the role of extra synaptic glycine levels in the pathophysiology of alcohol abuse and addiction.

Conclusion

Astrocytes sense and integrate neuronal functioning and activation of astrocytes coincides with synchronized synaptic firing and plasticity. Due to their pivotal role in monitoring extra synaptic transmission, the astrocyte appears to be an important target during both the acute and chronic phase of ethanol exposure and may play a key role in alcohol abuse disorders. Finding ways to modulate astrocyte function might thus lead to novel pharmacological interventions for treatment of alcohol abuse disorders. Especially, restoring ethanol-induced effects on amino acid transporters, and reinstating glutamate homeostasis, might be efficient methods for inhibiting drug seeking, and heavy drinking. However, additional studies are required in order to define ethanol-induced changes in astrocyte function, and to determine the role of astrocytes in the pathophysiology of relapse and alcohol addiction.

Acknowledgements

This work was supported by the Swedish Brain foundation, the Swedish research council, the Åhlén foundation, and Wilhelm and Martina Lundgren Foundation.

References

  1. Kacem K, Lacombe P, Seylaz J, Bonvento G (1998) Structural organization of the perivascular astrocyte end feet and their relationship with the endothelial glucose transporter: a confocal microscopy study. Glia 23: 1-10.
  2. Hansson E (1990) Regional heterogeneity among astrocytes in the central nervous system. Neurochem Int 16: 237-245.
  3. Ambrosi G, Virgintino D, Benagiano V, Maiorano E, Bertossi M, et al. (1995) Glial cells and blood-brain barrier in the human cerebral cortex. Ital J Anat Embryol 100 Suppl 1: 177-184.
  4. Brightman M (1991) Implication of astroglia in the blood-brain barrier. Ann N Y Acad Sci 633: 343-347.
  5. Magistretti PJ, Pellerin L, Rothman DL, Shulman RG (1999) Energy on demand. Science 283: 496-497.
  6. Hertz L (1965) Possible role of neuroglia: a potassium-mediated neuronal--neuroglial--neuronal impulse transmission system. Nature 206: 1091-1094.
  7. Halassa MM, Fellin T, Haydon PG (2007) The tripartite synapse: roles for gliotransmission in health and disease. Trends Mol Med 13: 54-63.
  8. Hansson E, Muyderman H, Leonova J, Allansson L, Sinclair J, et al. (2000) Astroglia and glutamate in physiology and pathology: Aspects on glutamate transport, glutamate-induced cell swelling and gap-junction communication. Neurochem Int 37: 317-329.
  9. Rossi D (2015) Astrocyte physiopathology: At the crossroads of intercellular networking, inflammation and cell death. Prog Neurobiol 130: 86-120.
  10. Halassa MM, Fellin T, Takano H, Dong JH, Haydon PG (2007) Synaptic islands defined by the territory of a single astrocyte. J Neurosci 27: 6473-6477.
  11. Han J, Kesner P, Metna-Laurent M, Duan T, Xu L, et al. (2012) Acute cannabinoids impair working memory through astroglial CB1 receptor modulation of hippocampal LTD. Cell 148: 1039-1050.
  12. Henneberger C, Papouin T, Oliet SH, Rusakov DA (2010) Long-term potentiation depends on release of D-serine from astrocytes. Nature 463: 232-236.
  13. Zhou Y, Danbolt NC (2013) GABA and Glutamate Transporters in Brain. Front Endocrinol (Lausanne) 4: 165.
  14. Faideau M, Kim J, Cormier K, Gilmore R, Welch M, et al. (2010) In vivo expression of polyglutamine-expanded huntingtin by mouse striatal astrocytes impairs glutamate transport: A correlation with Huntington's disease subjects. Hum Mol Genet 19: 3053-3067.
  15. Fuller S, Münch G, Steele M (2009) Activated astrocytes: a therapeutic target in Alzheimer's disease? Expert Rev Neurother 9: 1585-1594.
  16. Kim K, Lee SG, Kegelman TP, Su ZZ, Das SK, et al. (2011) Role of excitatory amino acid transporter-2 (EAAT2) and glutamate in neurodegeneration: Opportunities for developing novel therapeutics. J Cell Physiol 226: 2484-2493.
  17. Rajkowska G, Stockmeier CA (2013) Astrocyte pathology in major depressive disorder: insights from human postmortem brain tissue. Curr Drug Targets 14: 1225-1236.
  18. Kryger R, Wilce PA (2010) The effects of alcoholism on the human basolateral amygdala. Neuroscience 167: 361-371.
  19. Yudkoff M, Nissim I, Daikhin Y, Lin ZP, Nelson D, et al. (1993) Brain glutamate metabolism: Neuronal-astroglial relationships. Dev Neurosci 15: 343-350.
  20. Kaczor P, Rakus D, Mozrzymas JW (2015) Neuron-astrocyte interaction enhance GABAergic synaptic transmission in a manner dependent on key metabolic enzymes. Front Cell Neurosci 9: 120.
  21. Tani H, Dulla CG, Farzampour Z, Taylor-Weiner A, Huguenard JR, et al. (2014) A local glutamate-glutamine cycle sustains synaptic excitatory transmitter release. Neuron 81: 888-900.
  22. Aroeira RI, Sebastião AM, Valente CA (2014) GlyT1 and GlyT2 in brain astrocytes: expression, distribution and function. Brain Struct Funct 219: 817-830.
  23. Stephan J, Friauf E (2014) Functional analysis of the inhibitory neurotransmitter transporters GlyT1, GAT-1, and GAT-3 in astrocytes of the lateral superior olive. Glia 62: 1992-2003.
  24. Song D, Xu J, Bai Q, Cai L, Hertz L, et al. (2014) Role of the intracellular nucleoside transporter ENT3 in transmitter and high K+ stimulation of astrocytic ATP release investigated using siRNA against ENT3. ASN Neuro 6: 1759091414543439.
  25. Ruby CL, O'Connor KM, Ayers-Ringler J, Choi DS (2014) Adenosine and glutamate in neuroglial interaction: implications for circadian disorders and alcoholism. Adv Neurobiol 11: 103-119.
  26. Fraser DD, Duffy S, Angelides KJ, Perez-Velazquez JL, Kettenmann H, et al. (1995) GABAA/benzodiazepine receptors in acutely isolated hippocampal astrocytes. J Neurosci 15: 2720-2732.
  27. Gimpl G, Kirchhoff F, Lang RE, Kettenmann H (1993) Identification of neuropeptide Y receptors in cultured astrocytes from neonatal rat brain. J Neurosci Res 34: 198-205.
  28. Pastor A, Chvátal A, Syková E, Kettenmann H (1995) Glycine- and GABA-activated currents in identified glial cells of the developing rat spinal cord slice. Eur J Neurosci 7: 1188-1198.
  29. Steinhäuser C, Jabs R, Kettenmann H (1994) Properties of GABA and glutamate responses in identified glial cells of the mouse hippocampal slice. Hippocampus 4: 19-35.
  30. Hertz L, Lovatt D, Goldman SA, Nedergaard M (2010) Adrenoceptors in brain: cellular gene expression and effects on astrocytic metabolism and [Ca (2+)] i. Neurochem Int 57: 411-420.
  31. Bernstein M, Lyons SA, Möller T, Kettenmann H (1996) Receptor-mediated calcium signalling in glial cells from mouse corpus callosum slices. J Neurosci Res 46: 152-163.
  32. D'Ascenzo M, Fellin T, Terunuma M, Revilla-Sanchez R, Meaney DF, et al. (2007) mGluR5 stimulates gliotransmission in the nucleus accumbens. Proc Natl Acad Sci U S A 104: 1995-2000.
  33. Haydon PG, Carmignoto G (2006) Astrocyte control of synaptic transmission and neurovascular coupling. Physiol Rev 86: 1009-1031.
  34. Fellin T, D'Ascenzo M, Haydon PG (2007) Astrocytes control neuronal excitability in the nucleus accumbens. Scientific World Journal 7: 89-97.
  35. Araque A, Sanzgiri RP, Parpura V, Haydon PG (1998) Calcium elevation in astrocytes causes an NMDA receptor-dependent increase in the frequency of miniature synaptic currents in cultured hippocampal neurons. J Neurosci 18: 6822-6829.
  36. Nedergaard M (1994) Direct signaling from astrocytes to neurons in cultures of mammalian brain cells. Science 263: 1768-1771.
  37. Wang F, Smith NA, Xu Q, Goldman S, Peng W, et al. (2013) Photolysis of caged Ca2+ but not receptor-mediated Ca2+ signaling triggers astrocytic glutamate release. J Neurosci 33: 17404-17412.
  38. Hua X, Malarkey EB, Sunjara V, Rosenwald SE, Li WH, et al. (2004) (Ca2+)-dependent glutamate release involves two classes of endoplasmic reticulum (Ca2+) stores in astrocytes. J Neurosci Res 76: 86-97.
  39. Hansson E, Simonsson P, Alling C (1987) 5-Hydroxytryptamine stimulates the formation of inositol phosphate in astrocytes from different regions of the brain. Neuropharmacology 26: 1377-1382.
  40. Fiacco TA, McCarthy KD (2004) Intracellular astrocyte calcium waves in situ increase the frequency of spontaneous AMPA receptor currents in CA1 pyramidal neurons. J Neurosci 24: 722-732.
  41. Shigetomi E, Bushong EA, Haustein MD, Tong X, Jackson-Weaver O, et al. (2013) Imaging calcium microdomains within entire astrocyte territories and end feet with GCaMPs expressed using adeno-associated viruses. J Gen Physiol 141: 633-647.
  42. Verkhratsky A, Kettenmann H (1996) Calcium signaling in glial cells. Trends Neurosci 19: 346-352.
  43. Giaume C, Fromaget C, el Aoumari A, Cordier J, Glowinski J, et al. (1991) Gap junctions in cultured astrocytes: Single-channel currents and characterization of channel-forming protein. Neuron 6: 133-143.
  44. Blomstrand F, Aberg ND, Eriksson PS, Hansson E, Rönnbäck L (1999) Extent of intercellular calcium wave propagation is related to gap junction permeability and level of connexin-43 expression in astrocytes in primary cultures from four brain regions. Neuroscience 92: 255-265.
  45. Alvarez-Maubecin V, Garcia-Hernandez F, Williams JT, Van Bockstaele EJ (2000) Functional coupling between neurons and glia. J Neurosci 20: 4091-4098.
  46. Fróes MM, Correia AH, Garcia-Abreu J, Spray DC, Campos de Carvalho AC, et al. (1999) Gap-junctional coupling between neurons and astrocytes in primary central nervous system cultures. Proc Natl Acad Sci U S A 96: 7541-7546.
  47. Nagy JI, Rash JE (2000) Connexins and gap junctions of astrocytes and oligodendrocytes in the CNS. Brain Res Brain Res Rev 32: 29-44.
  48. Aberg ND, Carlsson B, Rosengren L, Oscarsson J, Isaksson OG, et al. (2000) Growth hormone increases connexin-43 expression in the cerebral cortex and hypothalamus. Endocrinology 141: 3879-3886.
  49. Adermark L, Lovinger DM (2008) Electrophysiological properties and gap junction coupling of striatal astrocytes. Neurochem Int 52: 1365-1372.
  50. Blomstrand F, Khatibi S, Muyderman H, Hansson E, Olsson T, et al. (1999) 5-Hydroxytryptamine and glutamate modulate velocity and extent of intercellular calcium signaling in hippocampal astroglial cells in primary cultures. Neuroscience 88: 1241-1253.
  51. Enkvist MO, McCarthy KD (1994) Astroglial gap junction communication is increased by treatment with either glutamate or high K+ concentration. J Neurochem 62: 489-495.
  52. Giaume C, McCarthy KD (1996) Control of gap-junctional communication in astrocytic networks. Trends Neurosci 19: 319-325.
  53. Rouach N, Glowinski J, Giaume C (2000) Activity-dependent neuronal control of gap-junctional communication in astrocytes. J Cell Biol 149: 1513-1526.
  54. Montero TD, Orellana JA (2015) Hemichannels: New pathways for gliotransmitter release. Neuroscience 286: 45-59.
  55. Orellana JA, Stehberg J (2014) Hemichannels: new roles in astroglial function. Front Physiol 5: 193.
  56. Stehberg J, Moraga-Amaro R, Salazar C, Becerra A, Echeverría C, et al. (2012) Release of gliotransmitters through astroglial connexin 43 hemi channels is necessary for fear memory consolidation in the basolateral amygdala. FASEB J 26: 3649-3657.
  57. Isokawa M, McKhann GM 2nd (2005) Electrophysiological and morphological characterization of dentate astrocytes in the hippocampus. J Neurobiol 65: 125-134.
  58. Lovinger DM (1997) Alcohols and neurotransmitter gated ion channels: past, present and future. Naunyn Schmiedebergs Arch Pharmacol 356: 267-282.
  59. Diamond I, Gordon AS (1997) Cellular and molecular neuroscience of alcoholism. Physiol Rev 77: 1-20.
  60. Kane CJ, Berry A, Boop FA, Davies DL (1996) Proliferation of astroglia from the adult human cerebrum is inhibited by ethanol in vitro. Brain Res 731: 39-44.
  61. Pignataro L, Varodayan FP, Tannenholz LE, Protiva P, Harrison NL (2013) Brief alcohol exposure alters transcription in astrocytes via the heat shock pathway. Brain Behav 3: 114-133.
  62. Adermark L, Lovinger DM (2006) Ethanol effects on electrophysiological properties of astrocytes in striatal brain slices. Neuropharmacology 51: 1099-1108.
  63. Adermark L, Olsson T, Hansson E (2004) Ethanol acutely decreases astroglial gap junction permeability in primary cultures from defined brain regions. Neurochem Int 45: 971-978.
  64. Allansson L, Khatibi S, Olsson T, Hansson E (2001) Acute ethanol exposure induces [Ca2+]i transients, cell swelling and transformation of actin cytoskeleton in astroglial primary cultures. J Neurochem 76: 472-479.
  65. Reichenbach A (1991) Glial K+ permeability and CNS K+ clearance by diffusion and spatial buffering. Ann N Y Acad Sci 633: 272-286.
  66. Rose CR, Ransom BR (1997) Gap junctions equalize intracellular Na+ concentration in astrocytes. Glia 20: 299-307.
  67. Miguel-Hidalgo JJ, Wilson BA, Hussain S, Meshram A, Rajkowska G, et al. (2014) Reduced connexin 43 immunolabeling in the orbitofrontal cortex in alcohol dependence and depression. J Psychiatr Res 55: 101-109.
  68. Adermark L, Jonsson S, Ericson M, Söderpalm B (2011) Intermittent ethanol consumption depresses endocannabinoid-signaling in the dorsolateral striatum of rat. Neuropharmacology 61: 1160-1165.
  69. Aschner M, Allen JW, Mutkus LA, Cao C (2001) Ethanol-induced swelling in neonatal rat primary astrocyte cultures. Brain Res 900: 219-226.
  70. Vargová L, Syková E (2014) Astrocytes and extracellular matrix in extra synaptic volume transmission. Philos Trans R Soc Lond B Biol Sci 369: 20130608.
  71. Dietrich WD, Ginsberg MD, Busto R, Watson BD, Yoshida S (1986) Vascular aspects and hemodynamic consequences of central nervous system injury. Cent Nerv Syst Trauma 3: 265-280.
  72. Allen JW, Mutkus LA, Aschner M (2002) Chronic ethanol produces increased taurine transport and efflux in cultured astrocytes. Neurotoxicology 23: 693-700.
  73. Lallemand F, Dahchour A, Ward RJ, De Witte P (2000) Does taurine play an osmolarity role during ethanol intoxication? Adv Exp Med Biol 483: 203-212.
  74. Quertemont E, Devitgh A, De Witte P (2003) Systemic osmotic manipulations modulate ethanol-induced taurine release: a brain micro dialysis study. Alcohol 29: 11-19.
  75. Vitarella D, DiRisio DJ, Kimelberg HK, Aschner M (1994) Potassium and taurine release are highly correlated with regulatory volume decrease in neonatal primary rat astrocyte cultures. J Neurochem 63: 1143-1149.
  76. Ericson M, Chau P, Adermark L, Söderpalm B (2013) Rising taurine and ethanol concentrations in nucleus accumbens interact to produce the dopamine-activating effects of alcohol. Adv Exp Med Biol 775: 215-223.
  77. Ericson M, Chau P, Clarke RB, Adermark L, Söderpalm B (2011) Rising taurine and ethanol concentrations in nucleus accumbens interact to produce dopamine release after ethanol administration. Addict Biol 16: 377-385.
  78. Ericson M, Molander A, Stomberg R, Söderpalm B (2006) Taurine elevates dopamine levels in the rat nucleus accumbens; antagonism by strychnine. Eur J Neurosci 23: 3225-3229.
  79. Adermark L, Clarke RB, Olsson T, Hansson E, Söderpalm B, et al. (2011) Implications for glycine receptors and astrocytes in ethanol-induced elevation of dopamine levels in the nucleus accumbens. Addict Biol 16: 43-54.
  80. Di Chiara G, Imperato A (1985) Ethanol preferentially stimulates dopamine release in the nucleus accumbens of freely moving rats. Eur J Pharmacol 115: 131-132.
  81. Küppers E, Gleiser C, Brito V, Wachter B, Pauly T, et al. (2008) AQP4 expression in striatal primary cultures is regulated by dopamine--implications for proliferation of astrocytes. Eur J Neurosci 28: 2173-2182.
  82. Risher WC, Andrew RD, Kirov SA (2009) Real-time passive volume responses of astrocytes to acute osmotic and ischemic stress in cortical slices and in vivo revealed by two-photon microscopy. Glia 57: 207-221.
  83. Lee MR, Ruby CL, Hinton DJ, Choi S, Adams CA, et al. (2013) Striatal adenosine signaling regulates EAAT2 and astrocytic AQP4 expression and alcohol drinking in mice. Neuropsychopharmacology 38: 437-445.
  84. Bosson A, Boisseau S, Buisson A, Savasta M, Albrieux M (2015) Disruption of dopaminergic transmission remodels tripartite synapse morphology and astrocytic calcium activity within substantia nigra pars reticulata. Glia 63: 673-683.
  85. Gittins RA, Harrison PJ (2011) A morphometric study of glia and neurons in the anterior cingulate cortex in mood disorder. J Affect Disord 133: 328-332.
  86. Gouw AA, Seewann A, Vrenken H, van der Flier WM, Rozemuller JM, et al. (2008) Heterogeneity of white matter hyper intensities in Alzheimer's disease: Post-mortem quantitative MRI and neuropathology. Brain 131: 3286-3298.
  87. Hercher C, Chopra V, Beasley CL (2014) Evidence for morphological alterations in prefrontal white matter glia in schizophrenia and bipolar disorder. J Psychiatry Neurosci 39: 376-385.
  88. Oh DH, Son H, Hwang S, Kim SH (2012) Neuropathological abnormalities of astrocytes, GABAergic neurons, and pyramidal neurons in the dorsolateral prefrontal cortices of patients with major depressive disorder. Eur Neuropsychopharmacol 22: 330-338.
  89. Tong J, Ang LC, Williams B, Furukawa Y, Fitzmaurice P, et al. (2015) Low levels of astroglial markers in Parkinson's disease: relationship to α-synuclein accumulation. Neurobiol Dis 82: 243-253.
  90. Dalçik H, Yardimoglu M, Filiz S, Gonca S, Dalçik C, et al. (2009) Chronic ethanol-induced glial fibrillary acidic protein (GFAP) immunoreactivity: An immunocytochemical observation in various regions of adult rat brain. Int J Neurosci 119: 1303-1318.
  91. Fletcher TL, Shain W (1993) Ethanol-induced changes in astrocyte gene expression during rat central nervous system development. Alcohol Clin Exp Res 17: 993-1001.
  92. Udomuksorn W, Mukem S, Kumarnsit E, Vongvatcharanon S, Vongvatcharanon U (2011) Effects of alcohol administration during adulthood on parvalbumin and glial fibrillary acidic protein immunoreactivity in the rat cerebral cortex. Acta Histochem 113: 283-289.
  93. Vongvatcharanon U, Mukem S, Udomuksorn W, Kumarsit E, Vongvatcharanon S (2010) Alcohol administration during adulthood induces alterations of parvalbumin and glial fibrillary acidic protein immunoreactivity in rat hippocampus and cingulate cortex. Acta Histochem 112: 392-401.
  94. Bull C, Syed WA, Minter SC, Bowers MS (2015) Differential response of glial fibrillary acidic protein-positive astrocytes in the rat prefrontal cortex following ethanol self-administration. Alcohol Clin Exp Res 39: 650-658.
  95. Wilkins LH Jr, Prendergast MA, Blanchard J, Holley RC, Chambers ER, et al. (2006) Potential value of changes in cell markers in organotypic hippocampal cultures associated with chronic EtOH exposure and withdrawal: comparison with NMDA-induced changes. Alcohol Clin Exp Res 30: 1768-1780.
  96. Delbro D, Westerlund A, Björklund U, Hansson E (2009) In inflammatory reactive astrocytes co-cultured with brain endothelial cells nicotine-evoked Ca(2+) transients are attenuated due to interleukin-1beta release and rearrangement of actin filaments. Neuroscience 159: 770-779.
  97. Lewohl JM, Wixey J, Harper CG, Dodd PR (2005) Expression of MBP, PLP, MAG, CNP, and GFAP in the Human Alcoholic Brain. Alcohol Clin Exp Res 29: 1698-1705.
  98. Cullen KM, Halliday GM (1994) Chronic alcoholics have substantial glial pathology in the forebrain and diencephalon. Alcohol Alcohol Suppl 2: 253-257.
  99. Skuja S, Groma V, Smane L (2012) Alcoholism and cellular vulnerability in different brain regions. Ultrastruct Pathol 36: 40-47.
  100. Scofield MD, Kalivas PW (2014) Astrocytic dysfunction and addiction: consequences of impaired glutamate homeostasis. Neuroscientist 20: 610-622.
  101. Othman T, Sinclair CJ, Haughey N, Geiger JD, Parkinson FE (2002) Ethanol alters glutamate but not adenosine uptake in rat astrocytes: evidence for protein kinase C involvement. Neurochem Res 27: 289-296.
  102. Smith TL (1997) Regulation of glutamate uptake in astrocytes continuously exposed to ethanol. Life Sci 61: 2499-2505.
  103. Gass JT, Olive MF (2008) Glutamatergic substrates of drug addiction and alcoholism. Biochem Pharmacol 75: 218-265.
  104. Holmes A, Spanagel R, Krystal JH (2013) Glutamatergic targets for new alcohol medications. Psychopharmacology (Berl) 229: 539-554.
  105. Fujio M, Nakagawa T, Sekiya Y, Ozawa T, Suzuki Y, et al. (2005) Gene transfer of GLT-1, a glutamate transporter, into the nucleus accumbens shell attenuates methamphetamine- and morphine-induced conditioned place preference in rats. Eur J Neurosci 22: 2744-2754.
  106. Alhaddad H, Kim NT, Aal-Aaboda M, Althobaiti YS, Leighton J, et al. (2014) Effects of MS-153 on chronic ethanol consumption and GLT1 modulation of glutamate levels in male alcohol-preferring rats. Front Behav Neurosci 8: 366.
  107. Karlsson RM, Adermark L, Molander A, Perreau-Lenz S, Singley E, et al. (2012) Reduced alcohol intake and reward associated with impaired endocannabinoid signaling in mice with a deletion of the glutamate transporter GLAST. Neuropharmacology 63: 181-189.
  108. Smith KL, John CS, Sypek EI, Ongür D, Cohen BM, et al. (2014) Exploring the role of central astrocytic glutamate uptake in ethanol reward in mice. Alcohol Clin Exp Res 38: 1307-1314.
  109. Kiefer F, Jahn H, Koester A, Montkowski A, Reinscheid RK, et al. (2003) Involvement of NMDA receptors in alcohol-mediated behavior: Mice with reduced affinity of the NMDA R1 glycine binding site display an attenuated sensitivity to ethanol. Biol Psychiatry 53: 345-351.
  110. Boyce-Rustay JM, Cunningham CL (2004) The role of NMDA receptor binding sites in ethanol place conditioning. Behav Neurosci 118: 822-834.
  111. Molander A, Söderpalm B (2005) Accumbal strychnine-sensitive glycine receptors: an access point for ethanol to the brain reward system. Alcohol Clin Exp Res 29: 27-37.
  112. Adermark L, Clarke RB, Söderpalm B, Ericson M (2011) Ethanol-induced modulation of synaptic output from the dorsolateral striatum in rat is regulated by cholinergic interneurons. Neurochem Int 58: 693-699.
  113. Lidö HH, Marston H, Ericson M, Söderpalm B (2012) The glycine reuptake inhibitor Org24598 and acamprosate reduce ethanol intake in the rat; tolerance development to acamprosate but not to Org24598. Addict Biol 17: 897-907.
  114. Lidö HH, Stomberg R, Fagerberg A, Ericson M, Söderpalm B (2009) The glycine reuptake inhibitor org 25935 interacts with basal and ethanol-induced dopamine release in rat nucleus accumbens. Alcohol Clin Exp Res 33: 1151-1157.
  115. Molander A, Lidö HH, Löf E, Ericson M, Söderpalm B (2007) The glycine reuptake inhibitor Org 25935 decreases ethanol intake and preference in male wistar rats. Alcohol Alcohol 42: 11-18.
  116. Vengeliene V, Leonardi-Essmann F, Sommer WH, Marston HM, Spanagel R (2010) Glycine transporter-1 blockade leads to persistently reduced relapse-like alcohol drinking in rats. Biol Psychiatry 68: 704-711.
  117. de Bejczy A, Nations KR, Szegedi A, Schoemaker J, Ruwe F, et al. (2014) Efficacy and safety of the glycine transporter-1 inhibitor org 25935 for the prevention of relapse in alcohol-dependent patients: a randomized, double-blind, placebo-controlled trial. Alcohol Clin Exp Res 38: 2427-2435.
Citation: Adermark L (2015) Astrocyte Function in Alcohol Reward and Addiction. J Alcohol Drug Depend 3:e123.

Copyright: © 2015 Adermark L. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top