Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2016

Open Access 01-12-2016 | Research

β-Catenin signaling positively regulates glutamate uptake and metabolism in astrocytes

Authors: Victoria Lutgen, Srinivas D. Narasipura, Amit Sharma, Stephanie Min, Lena Al-Harthi

Published in: Journal of Neuroinflammation | Issue 1/2016

Login to get access

Abstract

Background

Neurological disorders have been linked to abnormal excitatory neurotransmission. Perturbations in glutamate cycling can have profound impacts on normal activity, lead to excitotoxicity and neuroinflammation, and induce and/or exacerbate impairments in these diseases. Astrocytes play a key role in excitatory signaling as they both clear glutamate from the synaptic cleft and house enzymes responsible for glutamate conversion to glutamine. However, mechanisms responsible for the regulation of glutamate cycling, including the main astrocytic glutamate transporter excitatory amino acid transporter 2 (EAAT2 or GLT-1 in rodents) and glutamine synthetase (GS) which catalyzes the ATP-dependent reaction of glutamate and ammonia into glutamine, remain largely undefined.

Methods

Gain and loss of function for β-catenin in human progenitor-derived astrocyte (PDAs) was used to assess EAAT2 and GS levels by PCR, western blot, luciferase reporter assays, and chromatin immunoprecipitation (ChIP). Further, morpholinos were stereotaxically injected into C57BL/6 mice and western blots measured the protein levels of β-catenin, GLT-1, and GS.

Results

β-Catenin, a transcriptional co-activator and the central mediator of Wnt/β-catenin signaling pathway, positively regulates EAAT2 and GS at the transcriptional level in PDAs by partnering with T cell factor 1 (TCF-1) and TCF-3, respectively. This pathway is conserved in vivo as the knockdown of β-catenin in the prefrontal cortex results in reduced GLT-1 and GS expression.

Conclusions

These studies confirm that β-catenin regulates key proteins responsible for excitatory glutamate neurotransmission in vitro and in vivo and reveal the therapeutic potential of β-catenin modulation in treating diseases with abnormal glutamatergic neurotransmission and excitotoxicity.
Literature
1.
go back to reference Mehta A, Prabhakar M, Kumar P, Deshmukh R, Sharma PL. Excitotoxicity: bridge to various triggers in neurodegenerative disorders. Eur J Pharmacol. 2013;698(1-3):6–18.CrossRefPubMed Mehta A, Prabhakar M, Kumar P, Deshmukh R, Sharma PL. Excitotoxicity: bridge to various triggers in neurodegenerative disorders. Eur J Pharmacol. 2013;698(1-3):6–18.CrossRefPubMed
2.
go back to reference Lau A, Tymianski M. Glutamate receptors, neurotoxicity and neurodegeneration. Pflugers Arch. 2010;460(2):525–42.CrossRefPubMed Lau A, Tymianski M. Glutamate receptors, neurotoxicity and neurodegeneration. Pflugers Arch. 2010;460(2):525–42.CrossRefPubMed
3.
go back to reference Cisneros IE, Ghorpade A. HIV-1, methamphetamine and astrocyte glutamate regulation: combined excitotoxic implications for neuro-AIDS. Curr HIV Res. 2012;10(5):392–406.CrossRefPubMedPubMedCentral Cisneros IE, Ghorpade A. HIV-1, methamphetamine and astrocyte glutamate regulation: combined excitotoxic implications for neuro-AIDS. Curr HIV Res. 2012;10(5):392–406.CrossRefPubMedPubMedCentral
4.
go back to reference Potter MC, Figuera-Losada M, Rojas C, Slusher BS. Targeting the glutamatergic system for the treatment of HIV-associated neurocognitive disorders. J Neuroimmune Pharmacol. 2013;8(3):594–607.CrossRefPubMedPubMedCentral Potter MC, Figuera-Losada M, Rojas C, Slusher BS. Targeting the glutamatergic system for the treatment of HIV-associated neurocognitive disorders. J Neuroimmune Pharmacol. 2013;8(3):594–607.CrossRefPubMedPubMedCentral
5.
go back to reference Lauriat TL, McInnes LA. EAAT2 regulation and splicing: relevance to psychiatric and neurological disorders. Mol Psychiatry. 2007;12(12):1065–78.CrossRefPubMed Lauriat TL, McInnes LA. EAAT2 regulation and splicing: relevance to psychiatric and neurological disorders. Mol Psychiatry. 2007;12(12):1065–78.CrossRefPubMed
6.
go back to reference Kim K, Lee SG, Kegelman TP, Su ZZ, Das SK, Dash R, et al. Role of excitatory amino acid transporter-2 (EAAT2) and glutamate in neurodegeneration: opportunities for developing novel therapeutics. J Cell Physiol. 2011;226(10):2484–93.CrossRefPubMedPubMedCentral Kim K, Lee SG, Kegelman TP, Su ZZ, Das SK, Dash R, et al. Role of excitatory amino acid transporter-2 (EAAT2) and glutamate in neurodegeneration: opportunities for developing novel therapeutics. J Cell Physiol. 2011;226(10):2484–93.CrossRefPubMedPubMedCentral
7.
go back to reference Sitcheran R, Gupta P, Fisher PB, Baldwin AS. Positive and negative regulation of EAAT2 by NF-kappaB: a role for N-myc in TNFalpha-controlled repression. Embo J. 2005;24(3):510–20.CrossRefPubMedPubMedCentral Sitcheran R, Gupta P, Fisher PB, Baldwin AS. Positive and negative regulation of EAAT2 by NF-kappaB: a role for N-myc in TNFalpha-controlled repression. Embo J. 2005;24(3):510–20.CrossRefPubMedPubMedCentral
8.
go back to reference Su ZZ, Leszczyniecka M, Kang DC, Sarkar D, Chao W, Volsky DJ, et al. Insights into glutamate transport regulation in human astrocytes: cloning of the promoter for excitatory amino acid transporter 2 (EAAT2). Proc Natl Acad Sci U S A. 2003;100(4):1955–60.CrossRefPubMedPubMedCentral Su ZZ, Leszczyniecka M, Kang DC, Sarkar D, Chao W, Volsky DJ, et al. Insights into glutamate transport regulation in human astrocytes: cloning of the promoter for excitatory amino acid transporter 2 (EAAT2). Proc Natl Acad Sci U S A. 2003;100(4):1955–60.CrossRefPubMedPubMedCentral
9.
go back to reference Ghosh M, Yang Y, Rothstein JD, Robinson MB. Nuclear factor-kappaB contributes to neuron-dependent induction of glutamate transporter-1 expression in astrocytes. J Neurosci. 2011;31(25):9159–69.CrossRefPubMedPubMedCentral Ghosh M, Yang Y, Rothstein JD, Robinson MB. Nuclear factor-kappaB contributes to neuron-dependent induction of glutamate transporter-1 expression in astrocytes. J Neurosci. 2011;31(25):9159–69.CrossRefPubMedPubMedCentral
10.
go back to reference Schlag BD, Vondrasek JR, Munir M, Kalandadze A, Zelenaia OA, Rothstein JD, et al. Regulation of the glial Na+-dependent glutamate transporters by cyclic AMP analogs and neurons. Mol Pharmacol. 1998;53(3):355–69.PubMed Schlag BD, Vondrasek JR, Munir M, Kalandadze A, Zelenaia OA, Rothstein JD, et al. Regulation of the glial Na+-dependent glutamate transporters by cyclic AMP analogs and neurons. Mol Pharmacol. 1998;53(3):355–69.PubMed
11.
go back to reference Gegelashvili G, Schousboe A. High affinity glutamate transporters: regulation of expression and activity. Mol Pharmacol. 1997;52(1):6–15.PubMed Gegelashvili G, Schousboe A. High affinity glutamate transporters: regulation of expression and activity. Mol Pharmacol. 1997;52(1):6–15.PubMed
12.
go back to reference Swanson RA, Liu J, Miller JW, Rothstein JD, Farrell K, Stein BA, et al. Neuronal regulation of glutamate transporter subtype expression in astrocytes. J Neurosci. 1997;17(3):932–40.PubMed Swanson RA, Liu J, Miller JW, Rothstein JD, Farrell K, Stein BA, et al. Neuronal regulation of glutamate transporter subtype expression in astrocytes. J Neurosci. 1997;17(3):932–40.PubMed
13.
go back to reference Eid T, Tu N, Lee TS, Lai JC. Regulation of astrocyte glutamine synthetase in epilepsy. Neurochem Int. 2013;63(7):670–81.CrossRefPubMed Eid T, Tu N, Lee TS, Lai JC. Regulation of astrocyte glutamine synthetase in epilepsy. Neurochem Int. 2013;63(7):670–81.CrossRefPubMed
14.
go back to reference Henderson LJ, Sharma A, Monaco MC, Major EO, Al-Harthi L. Human immunodeficiency virus type 1 (HIV-1) transactivator of transcription through its intact core and cysteine-rich domains inhibits Wnt/beta-catenin signaling in astrocytes: relevance to HIV neuropathogenesis. J Neurosci. 2012;32(46):16306–13.CrossRefPubMedPubMedCentral Henderson LJ, Sharma A, Monaco MC, Major EO, Al-Harthi L. Human immunodeficiency virus type 1 (HIV-1) transactivator of transcription through its intact core and cysteine-rich domains inhibits Wnt/beta-catenin signaling in astrocytes: relevance to HIV neuropathogenesis. J Neurosci. 2012;32(46):16306–13.CrossRefPubMedPubMedCentral
15.
go back to reference Audard V, Cavard C, Richa H, Infante M, Couvelard A, Sauvanet A, et al. Impaired E-cadherin expression and glutamine synthetase overexpression in solid pseudopapillary neoplasm of the pancreas. Pancreas. 2008;36(1):80–3.CrossRefPubMed Audard V, Cavard C, Richa H, Infante M, Couvelard A, Sauvanet A, et al. Impaired E-cadherin expression and glutamine synthetase overexpression in solid pseudopapillary neoplasm of the pancreas. Pancreas. 2008;36(1):80–3.CrossRefPubMed
16.
go back to reference Chao CC, Hu S, Ehrlich L, Peterson PK. Interleukin-1 and tumor necrosis factor-alpha synergistically mediate neurotoxicity: involvement of nitric oxide and of N-methyl-D-aspartate receptors. Brain Behav Immun. 1995;9(4):355–65.CrossRefPubMed Chao CC, Hu S, Ehrlich L, Peterson PK. Interleukin-1 and tumor necrosis factor-alpha synergistically mediate neurotoxicity: involvement of nitric oxide and of N-methyl-D-aspartate receptors. Brain Behav Immun. 1995;9(4):355–65.CrossRefPubMed
17.
go back to reference Blutstein T, Devidze N, Choleris E, Jasnow AM, Pfaff DW, Mong JA. Oestradiol up-regulates glutamine synthetase mRNA and protein expression in the hypothalamus and hippocampus: implications for a role of hormonally responsive glia in amino acid neurotransmission. J Neuroendocrinol. 2006;18(9):692–702.CrossRefPubMed Blutstein T, Devidze N, Choleris E, Jasnow AM, Pfaff DW, Mong JA. Oestradiol up-regulates glutamine synthetase mRNA and protein expression in the hypothalamus and hippocampus: implications for a role of hormonally responsive glia in amino acid neurotransmission. J Neuroendocrinol. 2006;18(9):692–702.CrossRefPubMed
18.
go back to reference Kruithof-de Julio M, Labruyere WT, Ruijter JM, Vermeulen JL, Stanulovic V, Stallen JM, et al. The RL-ET-14 cell line mediates expression of glutamine synthetase through the upstream enhancer/promoter region. J Hepatol. 2005;43(1):126–31.CrossRefPubMed Kruithof-de Julio M, Labruyere WT, Ruijter JM, Vermeulen JL, Stanulovic V, Stallen JM, et al. The RL-ET-14 cell line mediates expression of glutamine synthetase through the upstream enhancer/promoter region. J Hepatol. 2005;43(1):126–31.CrossRefPubMed
19.
go back to reference Cadoret A, Ovejero C, Terris B, Souil E, Levy L, Lamers WH, et al. New targets of beta-catenin signaling in the liver are involved in the glutamine metabolism. Oncogene. 2002;21(54):8293–301.CrossRefPubMed Cadoret A, Ovejero C, Terris B, Souil E, Levy L, Lamers WH, et al. New targets of beta-catenin signaling in the liver are involved in the glutamine metabolism. Oncogene. 2002;21(54):8293–301.CrossRefPubMed
20.
go back to reference Narasipura SD, Henderson LJ, Fu SW, Chen L, Kashanchi F, Al-Harthi L. Role of beta-catenin and TCF/LEF family members in transcriptional activity of HIV in astrocytes. J Virol. 2012;86(4):1911–21.CrossRefPubMedPubMedCentral Narasipura SD, Henderson LJ, Fu SW, Chen L, Kashanchi F, Al-Harthi L. Role of beta-catenin and TCF/LEF family members in transcriptional activity of HIV in astrocytes. J Virol. 2012;86(4):1911–21.CrossRefPubMedPubMedCentral
21.
go back to reference Al-Harthi L. Wnt/beta-catenin and its diverse physiological cell signaling pathways in neurodegenerative and neuropsychiatric disorders. J Neuroimmune Pharmacol. 2012;7(4):725–30.CrossRefPubMedPubMedCentral Al-Harthi L. Wnt/beta-catenin and its diverse physiological cell signaling pathways in neurodegenerative and neuropsychiatric disorders. J Neuroimmune Pharmacol. 2012;7(4):725–30.CrossRefPubMedPubMedCentral
22.
go back to reference Allritz C, Bette S, Figiel M, Engele J. Comparative structural and functional analysis of the GLT-1/EAAT-2 promoter from man and rat. J Neurosci Res. 2010;88(6):1234–41.PubMed Allritz C, Bette S, Figiel M, Engele J. Comparative structural and functional analysis of the GLT-1/EAAT-2 promoter from man and rat. J Neurosci Res. 2010;88(6):1234–41.PubMed
23.
go back to reference Henderson LJ, Al-Harthi L. Role of beta-catenin/TCF-4 signaling in HIV replication and pathogenesis: insights to informing novel anti-HIV molecular therapeutics. J Neuroimmune Pharmacol. 2011;6(2):247–59.CrossRefPubMed Henderson LJ, Al-Harthi L. Role of beta-catenin/TCF-4 signaling in HIV replication and pathogenesis: insights to informing novel anti-HIV molecular therapeutics. J Neuroimmune Pharmacol. 2011;6(2):247–59.CrossRefPubMed
24.
go back to reference Maguschak KA, Ressler KJ. The dynamic role of beta-catenin in synaptic plasticity. Neuropharmacology. 2012;62(1):78–88.CrossRefPubMed Maguschak KA, Ressler KJ. The dynamic role of beta-catenin in synaptic plasticity. Neuropharmacology. 2012;62(1):78–88.CrossRefPubMed
25.
go back to reference Mills F, Bartlett TE, Dissing-Olesen L, Wisniewska MB, Kuznicki J, Macvicar BA, et al. Cognitive flexibility and long-term depression (LTD) are impaired following beta-catenin stabilization in vivo. Proc Natl Acad Sci U S A. 2014;111(23):8631–6.CrossRefPubMedPubMedCentral Mills F, Bartlett TE, Dissing-Olesen L, Wisniewska MB, Kuznicki J, Macvicar BA, et al. Cognitive flexibility and long-term depression (LTD) are impaired following beta-catenin stabilization in vivo. Proc Natl Acad Sci U S A. 2014;111(23):8631–6.CrossRefPubMedPubMedCentral
27.
go back to reference Orellana AM, Vasconcelos AR, Leite JA, de Sa LL, Andreotti DZ, Munhoz CD, et al. Age-related neuroinflammation and changes in AKT-GSK-3β and WNT/β-CATENIN signaling in rat hippocampus. Aging (Albany NY). 2015;7(12):1094–111.CrossRef Orellana AM, Vasconcelos AR, Leite JA, de Sa LL, Andreotti DZ, Munhoz CD, et al. Age-related neuroinflammation and changes in AKT-GSK-3β and WNT/β-CATENIN signaling in rat hippocampus. Aging (Albany NY). 2015;7(12):1094–111.CrossRef
28.
go back to reference Inestrosa NC, Montecinos-Oliva C, Fuenzalida M. Wnt signaling: role in Alzheimer disease and schizophrenia. J Neuroimmune Pharmacol. 2012;7(4):788–807.CrossRefPubMed Inestrosa NC, Montecinos-Oliva C, Fuenzalida M. Wnt signaling: role in Alzheimer disease and schizophrenia. J Neuroimmune Pharmacol. 2012;7(4):788–807.CrossRefPubMed
29.
go back to reference Berwick DC, Harvey K. The importance of Wnt signalling for neurodegeneration in Parkinson’s disease. Biochem Soc Trans. 2012;40(5):1123–8.CrossRefPubMed Berwick DC, Harvey K. The importance of Wnt signalling for neurodegeneration in Parkinson’s disease. Biochem Soc Trans. 2012;40(5):1123–8.CrossRefPubMed
30.
go back to reference De Ferrari GV, Moon RT. The ups and downs of Wnt signaling in prevalent neurological disorders. Oncogene. 2006;25(57):7545–53.CrossRefPubMed De Ferrari GV, Moon RT. The ups and downs of Wnt signaling in prevalent neurological disorders. Oncogene. 2006;25(57):7545–53.CrossRefPubMed
31.
go back to reference Singh S, Mishra A, Shukla S. ALCAR exerts neuroprotective and pro-neurogenic effects by inhibition of glial activation and oxidative stress via activation of the Wnt/beta-catenin signaling in Parkinsonian rats. Mol Neurobiol. 2016;53(7):4286–301. doi:10.1007/s12035-015-9361-5. Epub 2015 Jul 30. Singh S, Mishra A, Shukla S. ALCAR exerts neuroprotective and pro-neurogenic effects by inhibition of glial activation and oxidative stress via activation of the Wnt/beta-catenin signaling in Parkinsonian rats. Mol Neurobiol. 2016;53(7):4286–301. doi:10.​1007/​s12035-015-9361-5. Epub 2015 Jul 30.
32.
go back to reference L’Episcopo F, Tirolo C, Caniglia S, Testa N, Morale MC, Serapide MF, et al. Targeting Wnt signaling at the neuroimmune interface for dopaminergic neuroprotection/repair in Parkinson’s disease. J Mol Cell Biol. 2014;6(1):13–26.CrossRefPubMedPubMedCentral L’Episcopo F, Tirolo C, Caniglia S, Testa N, Morale MC, Serapide MF, et al. Targeting Wnt signaling at the neuroimmune interface for dopaminergic neuroprotection/repair in Parkinson’s disease. J Mol Cell Biol. 2014;6(1):13–26.CrossRefPubMedPubMedCentral
33.
go back to reference Ernst T, Jiang CS, Nakama H, Buchthal S, Chang L. Lower brain glutamate is associated with cognitive deficits in HIV patients: a new mechanism for HIV-associated neurocognitive disorder. J Magn Reson Imaging. 2011;32(5):1045–53.CrossRef Ernst T, Jiang CS, Nakama H, Buchthal S, Chang L. Lower brain glutamate is associated with cognitive deficits in HIV patients: a new mechanism for HIV-associated neurocognitive disorder. J Magn Reson Imaging. 2011;32(5):1045–53.CrossRef
34.
go back to reference Mohamed MA, Barker PB, Skolasky RL, Selnes OA, Moxley RT, Pomper MG, et al. Brain metabolism and cognitive impairment in HIV infection: a 3-T magnetic resonance spectroscopy study. Magn Reson Imaging. 2010;28(9):1251–7.CrossRefPubMedPubMedCentral Mohamed MA, Barker PB, Skolasky RL, Selnes OA, Moxley RT, Pomper MG, et al. Brain metabolism and cognitive impairment in HIV infection: a 3-T magnetic resonance spectroscopy study. Magn Reson Imaging. 2010;28(9):1251–7.CrossRefPubMedPubMedCentral
35.
go back to reference Sailasuta N, Shriner K, Ross B. Evidence of reduced glutamate in the frontal lobe of HIV-seropositive patients. NMR Biomed. 2009;22(3):326–31.CrossRefPubMed Sailasuta N, Shriner K, Ross B. Evidence of reduced glutamate in the frontal lobe of HIV-seropositive patients. NMR Biomed. 2009;22(3):326–31.CrossRefPubMed
36.
go back to reference Meisner F, Neuen-Jacob E, Sopper S, Schmidt M, Schlammes S, Scheller C, et al. Disruption of excitatory amino acid transporters in brains of SIV-infected rhesus macaques is associated with microglia activation. J Neurochem. 2008;104(1):202–9.PubMed Meisner F, Neuen-Jacob E, Sopper S, Schmidt M, Schlammes S, Scheller C, et al. Disruption of excitatory amino acid transporters in brains of SIV-infected rhesus macaques is associated with microglia activation. J Neurochem. 2008;104(1):202–9.PubMed
37.
go back to reference Koutsilieri E, Sopper S, Heinemann T, Scheller C, Lan J, Stahl-Hennig C, et al. Involvement of microglia in cerebrospinal fluid glutamate increase in SIV-infected rhesus monkeys (Macaca mulatta). AIDS Res Hum Retroviruses. 1999;15(5):471–7.CrossRefPubMed Koutsilieri E, Sopper S, Heinemann T, Scheller C, Lan J, Stahl-Hennig C, et al. Involvement of microglia in cerebrospinal fluid glutamate increase in SIV-infected rhesus monkeys (Macaca mulatta). AIDS Res Hum Retroviruses. 1999;15(5):471–7.CrossRefPubMed
38.
go back to reference Kustova Y, Ha JH, Espey MG, Sei Y, Morse D, Basile AS. The pattern of neurotransmitter alterations in LP-BM5 infected mice is consistent with glutamatergic hyperactivation. Brain Res. 1998;793(1-2):119–26.CrossRefPubMed Kustova Y, Ha JH, Espey MG, Sei Y, Morse D, Basile AS. The pattern of neurotransmitter alterations in LP-BM5 infected mice is consistent with glutamatergic hyperactivation. Brain Res. 1998;793(1-2):119–26.CrossRefPubMed
39.
go back to reference Plaitakis A, Caroscio JT. Abnormal glutamate metabolism in amyotrophic lateral sclerosis. Ann Neurol. 1987;22(5):575–9.CrossRefPubMed Plaitakis A, Caroscio JT. Abnormal glutamate metabolism in amyotrophic lateral sclerosis. Ann Neurol. 1987;22(5):575–9.CrossRefPubMed
40.
go back to reference Rothstein JD, Martin LJ, Kuncl RW. Decreased glutamate transport by the brain and spinal cord in amyotrophic lateral sclerosis. N Engl J Med. 1992;326(22):1464–8.CrossRefPubMed Rothstein JD, Martin LJ, Kuncl RW. Decreased glutamate transport by the brain and spinal cord in amyotrophic lateral sclerosis. N Engl J Med. 1992;326(22):1464–8.CrossRefPubMed
41.
go back to reference Moon RT, Brown JD, Torres M. WNTs modulate cell fate and behavior during vertebrate development. Trends Genet. 1997;13(4):157–62.CrossRefPubMed Moon RT, Brown JD, Torres M. WNTs modulate cell fate and behavior during vertebrate development. Trends Genet. 1997;13(4):157–62.CrossRefPubMed
42.
go back to reference Torp R, Lekieffre D, Levy LM, Haug FM, Danbolt NC, Meldrum BS, et al. Reduced postischemic expression of a glial glutamate transporter, GLT1, in the rat hippocampus. Exp Brain Res. 1995;103(1):51–8.CrossRefPubMed Torp R, Lekieffre D, Levy LM, Haug FM, Danbolt NC, Meldrum BS, et al. Reduced postischemic expression of a glial glutamate transporter, GLT1, in the rat hippocampus. Exp Brain Res. 1995;103(1):51–8.CrossRefPubMed
43.
go back to reference Martin LJ, Brambrink AM, Lehmann C, Portera-Cailliau C, Koehler R, Rothstein J, et al. Hypoxia-ischemia causes abnormalities in glutamate transporters and death of astroglia and neurons in newborn striatum. Ann Neurol. 1997;42(3):335–48.CrossRefPubMed Martin LJ, Brambrink AM, Lehmann C, Portera-Cailliau C, Koehler R, Rothstein J, et al. Hypoxia-ischemia causes abnormalities in glutamate transporters and death of astroglia and neurons in newborn striatum. Ann Neurol. 1997;42(3):335–48.CrossRefPubMed
44.
go back to reference Li W, Henderson LJ, Major EO, Al-Harthi L. IFN-gamma mediates enhancement of HIV replication in astrocytes by inducing an antagonist of the beta-catenin pathway (DKK1) in a STAT 3-dependent manner. J Immunol. 2011;186(12):6771–8.CrossRefPubMedPubMedCentral Li W, Henderson LJ, Major EO, Al-Harthi L. IFN-gamma mediates enhancement of HIV replication in astrocytes by inducing an antagonist of the beta-catenin pathway (DKK1) in a STAT 3-dependent manner. J Immunol. 2011;186(12):6771–8.CrossRefPubMedPubMedCentral
45.
go back to reference Gonsalves FC, Klein K, Carson BB, Katz S, Ekas LA, Evans S, et al. An RNAi-based chemical genetic screen identifies three small-molecule inhibitors of the Wnt/wingless signaling pathway. Proc Natl Acad Sci U S A. 2011;108(15):5954–63.CrossRefPubMedPubMedCentral Gonsalves FC, Klein K, Carson BB, Katz S, Ekas LA, Evans S, et al. An RNAi-based chemical genetic screen identifies three small-molecule inhibitors of the Wnt/wingless signaling pathway. Proc Natl Acad Sci U S A. 2011;108(15):5954–63.CrossRefPubMedPubMedCentral
Metadata
Title
β-Catenin signaling positively regulates glutamate uptake and metabolism in astrocytes
Authors
Victoria Lutgen
Srinivas D. Narasipura
Amit Sharma
Stephanie Min
Lena Al-Harthi
Publication date
01-12-2016
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2016
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-016-0691-7

Other articles of this Issue 1/2016

Journal of Neuroinflammation 1/2016 Go to the issue