Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2017

Open Access 01-12-2017 | Research

Inflammation-associated regulation of RGS in astrocytes and putative implication in neuropathic pain

Authors: Pierre J. Doyen, Maxime Vergouts, Amandine Pochet, Nathalie Desmet, Sabien van Neerven, Gary Brook, Emmanuel Hermans

Published in: Journal of Neuroinflammation | Issue 1/2017

Login to get access

Abstract

Background

Regulators of G-protein signaling (RGS) are major physiological modulators of G-protein-coupled receptors (GPCR) signaling. Several GPCRs expressed in both neurons and astrocytes participate in the central control of pain processing, and the reduced efficacy of analgesics in neuropathic pain conditions may rely on alterations in RGS function. The expression and the regulation of RGS in astrocytes is poorly documented, and we herein hypothesized that neuroinflammation which is commonly observed in neuropathic pain could influence RGS expression in astrocytes.

Methods

In a validated model of neuropathic pain, the spared nerve injury (SNI), the regulation of RGS2, RGS3, RGS4, and RGS7 messenger RNA (mRNA) was examined up to 3 weeks after the lesion. Changes in the expression of the same RGS were also studied in cultured astrocytes exposed to defined activation protocols or to inflammatory cytokines.

Results

We evidenced a differential regulation of these RGS in the lumbar spinal cord of animals undergoing SNI. In particular, RGS3 appeared upregulated at early stages after the lesion whereas expression of RGS2 and RGS4 was decreased at later stages. Decrease in RGS7 expression was already observed after 3 days and outlasted until 21 days after the lesion. In cultured astrocytes, we observed that changes in the culture conditions distinctly influenced the constitutive expression of these RGS. Also, brief exposures (4 to 8 h) to either interleukin-1β, interleukin-6, or tumor necrosis factor α caused rapid changes in the mRNA levels of the RGS, which however did not strictly recapitulate the regulations observed in the spinal cord of lesioned animals. Longer exposure (48 h) to inflammatory cytokines barely influenced RGS expression, confirming the rapid but transient regulation of these cell signaling modulators.

Conclusion

Changes in the environment of astrocytes mimicking the inflammation observed in the model of neuropathic pain can affect RGS expression. Considering the role of astrocytes in the onset and progression of neuropathic pain, we propose that the inflammation-mediated modulation of RGS in astrocytes constitutes an adaptive mechanism in a context of neuroinflammation and may participate in the regulation of nociception.
Literature
1.
go back to reference Haydon PG, Carmignoto G. Astrocyte control of synaptic transmission and neurovascular coupling. Physiol Rev. 2006;86:1009–31.CrossRefPubMed Haydon PG, Carmignoto G. Astrocyte control of synaptic transmission and neurovascular coupling. Physiol Rev. 2006;86:1009–31.CrossRefPubMed
2.
go back to reference Hansson E, Ronnback L. Glial neuronal signaling in the central nervous system. FASEB J. 2003;17:341–8.CrossRefPubMed Hansson E, Ronnback L. Glial neuronal signaling in the central nervous system. FASEB J. 2003;17:341–8.CrossRefPubMed
3.
go back to reference Bradley SJ, Challiss RA. G protein-coupled receptor signalling in astrocytes in health and disease: a focus on metabotropic glutamate receptors. Biochem Pharmacol. 2012;84:249–59.CrossRefPubMed Bradley SJ, Challiss RA. G protein-coupled receptor signalling in astrocytes in health and disease: a focus on metabotropic glutamate receptors. Biochem Pharmacol. 2012;84:249–59.CrossRefPubMed
4.
go back to reference Stogsdill JA, Eroglu C. The interplay between neurons and glia in synapse development and plasticity. Curr Opin Neurobiol. 2016;42:1–8.CrossRefPubMed Stogsdill JA, Eroglu C. The interplay between neurons and glia in synapse development and plasticity. Curr Opin Neurobiol. 2016;42:1–8.CrossRefPubMed
5.
6.
go back to reference Hansson E. Could chronic pain and spread of pain sensation be induced and maintained by glial activation? Acta Physiol (Oxf). 2006;187:321–7.CrossRefPubMed Hansson E. Could chronic pain and spread of pain sensation be induced and maintained by glial activation? Acta Physiol (Oxf). 2006;187:321–7.CrossRefPubMed
7.
go back to reference Old EA, Clark AK, Malcangio M. The role of glia in the spinal cord in neuropathic and inflammatory pain. Handb Exp Pharmacol. 2015;227:145–70.CrossRefPubMed Old EA, Clark AK, Malcangio M. The role of glia in the spinal cord in neuropathic and inflammatory pain. Handb Exp Pharmacol. 2015;227:145–70.CrossRefPubMed
9.
go back to reference Kronschlager MT, Drdla-Schutting R, Gassner M, Honsek SD, Teuchmann HL, Sandkuhler J. Gliogenic LTP spreads widely in nociceptive pathways. Science. 2016;354:1144–8.CrossRefPubMed Kronschlager MT, Drdla-Schutting R, Gassner M, Honsek SD, Teuchmann HL, Sandkuhler J. Gliogenic LTP spreads widely in nociceptive pathways. Science. 2016;354:1144–8.CrossRefPubMed
11.
go back to reference Tsuda M, Kohro Y, Yano T, Tsujikawa T, Kitano J, Tozaki-Saitoh H, et al. JAK-STAT3 pathway regulates spinal astrocyte proliferation and neuropathic pain maintenance in rats. Brain. 2011;134:1127–39.CrossRefPubMedPubMedCentral Tsuda M, Kohro Y, Yano T, Tsujikawa T, Kitano J, Tozaki-Saitoh H, et al. JAK-STAT3 pathway regulates spinal astrocyte proliferation and neuropathic pain maintenance in rats. Brain. 2011;134:1127–39.CrossRefPubMedPubMedCentral
12.
go back to reference Pan HL, ZZ W, Zhou HY, Chen SR, Zhang HM, Li DP. Modulation of pain transmission by G-protein-coupled receptors. Pharmacol Ther. 2008;117:141–61.CrossRefPubMed Pan HL, ZZ W, Zhou HY, Chen SR, Zhang HM, Li DP. Modulation of pain transmission by G-protein-coupled receptors. Pharmacol Ther. 2008;117:141–61.CrossRefPubMed
13.
go back to reference Ji RR, Strichartz G. Cell signaling and the genesis of neuropathic pain. Sci STKE. 2004;2004:reE14.PubMed Ji RR, Strichartz G. Cell signaling and the genesis of neuropathic pain. Sci STKE. 2004;2004:reE14.PubMed
14.
go back to reference Maione S, Marabese I, Leyva J, Palazzo E, de Novellis V, Rossi F. Characterisation of mGluRs which modulate nociception in the PAG of the mouse. Neuropharmacology. 1998;37:1475–83.CrossRefPubMed Maione S, Marabese I, Leyva J, Palazzo E, de Novellis V, Rossi F. Characterisation of mGluRs which modulate nociception in the PAG of the mouse. Neuropharmacology. 1998;37:1475–83.CrossRefPubMed
15.
go back to reference Luongo L, de Novellis V, Gatta L, Palazzo E, Vita D, Guida F, et al. Role of metabotropic glutamate receptor 1 in the basolateral amygdala-driven prefrontal cortical deactivation in inflammatory pain in the rat. Neuropharmacology. 2013;66:317–29.CrossRefPubMed Luongo L, de Novellis V, Gatta L, Palazzo E, Vita D, Guida F, et al. Role of metabotropic glutamate receptor 1 in the basolateral amygdala-driven prefrontal cortical deactivation in inflammatory pain in the rat. Neuropharmacology. 2013;66:317–29.CrossRefPubMed
16.
go back to reference Berger JV, Knaepen L, Janssen SP, Jaken RJ, Marcus MA, Joosten EA, et al. Cellular and molecular insights into neuropathy-induced pain hypersensitivity for mechanism-based treatment approaches. Brain Res Rev. 2011;67:282–310.CrossRefPubMed Berger JV, Knaepen L, Janssen SP, Jaken RJ, Marcus MA, Joosten EA, et al. Cellular and molecular insights into neuropathy-induced pain hypersensitivity for mechanism-based treatment approaches. Brain Res Rev. 2011;67:282–310.CrossRefPubMed
17.
go back to reference Bender E, Buist A, Jurzak M, Langlois X, Baggerman G, Verhasselt P, et al. Characterization of an orphan G protein-coupled receptor localized in the dorsal root ganglia reveals adenine as a signaling molecule. Proc Natl Acad Sci U S A. 2002;99:8573–8.CrossRefPubMedPubMedCentral Bender E, Buist A, Jurzak M, Langlois X, Baggerman G, Verhasselt P, et al. Characterization of an orphan G protein-coupled receptor localized in the dorsal root ganglia reveals adenine as a signaling molecule. Proc Natl Acad Sci U S A. 2002;99:8573–8.CrossRefPubMedPubMedCentral
18.
go back to reference Calixto JB, Medeiros R, Fernandes ES, Ferreira J, Cabrini DA, Campos MM. Kinin B1 receptors: key G-protein-coupled receptors and their role in inflammatory and painful processes. Br J Pharmacol. 2004;143:803–18.CrossRefPubMedPubMedCentral Calixto JB, Medeiros R, Fernandes ES, Ferreira J, Cabrini DA, Campos MM. Kinin B1 receptors: key G-protein-coupled receptors and their role in inflammatory and painful processes. Br J Pharmacol. 2004;143:803–18.CrossRefPubMedPubMedCentral
19.
go back to reference Geppetti P, Veldhuis NA, Lieu T, Bunnett NW. G protein-coupled receptors: dynamic machines for signaling pain and itch. Neuron. 2015;88:635–49.CrossRefPubMed Geppetti P, Veldhuis NA, Lieu T, Bunnett NW. G protein-coupled receptors: dynamic machines for signaling pain and itch. Neuron. 2015;88:635–49.CrossRefPubMed
21.
go back to reference De Vries L, Zheng B, Fischer T, Elenko E, Farquhar MG. The regulator of G protein signaling family. Annu Rev Pharmacol Toxicol. 2000;40:235–71.CrossRefPubMed De Vries L, Zheng B, Fischer T, Elenko E, Farquhar MG. The regulator of G protein signaling family. Annu Rev Pharmacol Toxicol. 2000;40:235–71.CrossRefPubMed
22.
go back to reference Abramow-Newerly M, Roy AA, Nunn C, Chidiac P. RGS proteins have a signalling complex: interactions between RGS proteins and GPCRs, effectors, and auxiliary proteins. Cell Signal. 2006;18:579–91.CrossRefPubMed Abramow-Newerly M, Roy AA, Nunn C, Chidiac P. RGS proteins have a signalling complex: interactions between RGS proteins and GPCRs, effectors, and auxiliary proteins. Cell Signal. 2006;18:579–91.CrossRefPubMed
23.
go back to reference Hollinger S, Hepler JR. Cellular regulation of RGS proteins: modulators and integrators of G protein signaling. Pharmacol Rev. 2002;54:527–59.CrossRefPubMed Hollinger S, Hepler JR. Cellular regulation of RGS proteins: modulators and integrators of G protein signaling. Pharmacol Rev. 2002;54:527–59.CrossRefPubMed
24.
go back to reference Larminie C, Murdock P, Walhin JP, Duckworth M, Blumer KJ, Scheideler MA, et al. Selective expression of regulators of G-protein signaling (RGS) in the human central nervous system. Brain Res Mol Brain Res. 2004;122:24–34.CrossRefPubMed Larminie C, Murdock P, Walhin JP, Duckworth M, Blumer KJ, Scheideler MA, et al. Selective expression of regulators of G-protein signaling (RGS) in the human central nervous system. Brain Res Mol Brain Res. 2004;122:24–34.CrossRefPubMed
25.
go back to reference Gold SJ, Ni YG, Dohlman HG, Nestler EJ. Regulators of G-protein signaling (RGS) proteins: region-specific expression of nine subtypes in rat brain. J Neurosci. 1997;17:8024–37.PubMed Gold SJ, Ni YG, Dohlman HG, Nestler EJ. Regulators of G-protein signaling (RGS) proteins: region-specific expression of nine subtypes in rat brain. J Neurosci. 1997;17:8024–37.PubMed
26.
go back to reference Bosier B, Doyen PJ, Brolet A, Muccioli GG, Ahmed E, Desmet N, et al. Inhibition of the regulator of G protein signalling RGS4 in the spinal cord decreases neuropathic hyperalgesia and restores cannabinoid CB1 receptor signalling. Br J Pharmacol. 2015;172:5333–46.CrossRefPubMedPubMedCentral Bosier B, Doyen PJ, Brolet A, Muccioli GG, Ahmed E, Desmet N, et al. Inhibition of the regulator of G protein signalling RGS4 in the spinal cord decreases neuropathic hyperalgesia and restores cannabinoid CB1 receptor signalling. Br J Pharmacol. 2015;172:5333–46.CrossRefPubMedPubMedCentral
27.
go back to reference Sutton LP, Ostrovskaya O, Dao M, Xie K, Orlandi C, Smith R, et al. Regulator of G-protein signaling 7 regulates reward behavior by controlling opioid signaling in the striatum. Biol Psychiatry. 2016;80:235–45.CrossRefPubMed Sutton LP, Ostrovskaya O, Dao M, Xie K, Orlandi C, Smith R, et al. Regulator of G-protein signaling 7 regulates reward behavior by controlling opioid signaling in the striatum. Biol Psychiatry. 2016;80:235–45.CrossRefPubMed
28.
go back to reference Salaga M, Storr M, Martemyanov KA, Fichna J. RGS proteins as targets in the treatment of intestinal inflammation and visceral pain: new insights and future perspectives. BioEssays. 2016;38:344–54.CrossRefPubMedPubMedCentral Salaga M, Storr M, Martemyanov KA, Fichna J. RGS proteins as targets in the treatment of intestinal inflammation and visceral pain: new insights and future perspectives. BioEssays. 2016;38:344–54.CrossRefPubMedPubMedCentral
29.
go back to reference Costigan M, Samad TA, Allchorne A, Lanoue C, Tate S, Woolf CJ. High basal expression and injury-induced down regulation of two regulator of G-protein signaling transcripts, RGS3 and RGS4 in primary sensory neurons. Mol Cell Neurosci. 2003;24:106–16.CrossRefPubMed Costigan M, Samad TA, Allchorne A, Lanoue C, Tate S, Woolf CJ. High basal expression and injury-induced down regulation of two regulator of G-protein signaling transcripts, RGS3 and RGS4 in primary sensory neurons. Mol Cell Neurosci. 2003;24:106–16.CrossRefPubMed
30.
go back to reference Taccola G, Doyen PJ, Damblon J, Dingu N, Ballarin B, Steyaert A, et al. A new model of nerve injury in the rat reveals a role of regulator of G protein signaling 4 in tactile hypersensitivity. Exp Neurol. 2016;286:1–11.CrossRefPubMed Taccola G, Doyen PJ, Damblon J, Dingu N, Ballarin B, Steyaert A, et al. A new model of nerve injury in the rat reveals a role of regulator of G protein signaling 4 in tactile hypersensitivity. Exp Neurol. 2016;286:1–11.CrossRefPubMed
31.
go back to reference Stratinaki M, Varidaki A, Mitsi V, Ghose S, Magida J, Dias C, et al. Regulator of G protein signaling 4 [corrected] is a crucial modulator of antidepressant drug action in depression and neuropathic pain models. Proc Natl Acad Sci U S A. 2013;110:8254–9.CrossRefPubMedPubMedCentral Stratinaki M, Varidaki A, Mitsi V, Ghose S, Magida J, Dias C, et al. Regulator of G protein signaling 4 [corrected] is a crucial modulator of antidepressant drug action in depression and neuropathic pain models. Proc Natl Acad Sci U S A. 2013;110:8254–9.CrossRefPubMedPubMedCentral
32.
go back to reference Han MH, Renthal W, Ring RH, Rahman Z, Psifogeorgou K, Howland D, et al. Brain region specific actions of regulator of G protein signaling 4 oppose morphine reward and dependence but promote analgesia. Biol Psychiatry. 2010;67:761–9.CrossRefPubMed Han MH, Renthal W, Ring RH, Rahman Z, Psifogeorgou K, Howland D, et al. Brain region specific actions of regulator of G protein signaling 4 oppose morphine reward and dependence but promote analgesia. Biol Psychiatry. 2010;67:761–9.CrossRefPubMed
33.
go back to reference Gui WS, Wei X, Mai CL, Murugan M, LJ W, Xin WJ, et al. Interleukin-1beta overproduction is a common cause for neuropathic pain, memory deficit, and depression following peripheral nerve injury in rodents. Mol Pain. 2016;12 Gui WS, Wei X, Mai CL, Murugan M, LJ W, Xin WJ, et al. Interleukin-1beta overproduction is a common cause for neuropathic pain, memory deficit, and depression following peripheral nerve injury in rodents. Mol Pain. 2016;12
34.
go back to reference Berger JV, Deumens R, Goursaud S, Schafer S, Lavand'homme P, Joosten EA, et al. Enhanced neuroinflammation and pain hypersensitivity after peripheral nerve injury in rats expressing mutated superoxide dismutase 1. J Neuroinflammation. 2011;8:33.CrossRefPubMedPubMedCentral Berger JV, Deumens R, Goursaud S, Schafer S, Lavand'homme P, Joosten EA, et al. Enhanced neuroinflammation and pain hypersensitivity after peripheral nerve injury in rats expressing mutated superoxide dismutase 1. J Neuroinflammation. 2011;8:33.CrossRefPubMedPubMedCentral
35.
go back to reference Decosterd I, Woolf CJ. Spared nerve injury: an animal model of persistent peripheral neuropathic pain. Pain. 2000;87:149–58.CrossRefPubMed Decosterd I, Woolf CJ. Spared nerve injury: an animal model of persistent peripheral neuropathic pain. Pain. 2000;87:149–58.CrossRefPubMed
36.
go back to reference Luis AL, Amado S, Geuna S, Rodrigues JM, Simoes MJ, Santos JD, et al. Long-term functional and morphological assessment of a standardized rat sciatic nerve crush injury with a non-serrated clamp. J Neurosci Methods. 2007;163:92–104.CrossRefPubMed Luis AL, Amado S, Geuna S, Rodrigues JM, Simoes MJ, Santos JD, et al. Long-term functional and morphological assessment of a standardized rat sciatic nerve crush injury with a non-serrated clamp. J Neurosci Methods. 2007;163:92–104.CrossRefPubMed
37.
go back to reference Chaplan SR, Bach FW, Pogrel JW, Chung JM, Yaksh TL. Quantitative assessment of tactile allodynia in the rat paw. J Neurosci Methods. 1994;53:55–63.CrossRefPubMed Chaplan SR, Bach FW, Pogrel JW, Chung JM, Yaksh TL. Quantitative assessment of tactile allodynia in the rat paw. J Neurosci Methods. 1994;53:55–63.CrossRefPubMed
38.
go back to reference Xu N, Tang XH, Pan W, Xie ZM, Zhang GF, Ji MH, et al. Spared Nerve Injury Increases the Expression of Microglia M1 Markers in the Prefrontal Cortex of Rats and Provokes Depression-Like Behaviors. Front Neurosci. 2017;11:209. Xu N, Tang XH, Pan W, Xie ZM, Zhang GF, Ji MH, et al. Spared Nerve Injury Increases the Expression of Microglia M1 Markers in the Prefrontal Cortex of Rats and Provokes Depression-Like Behaviors. Front Neurosci. 2017;11:209.
39.
go back to reference Tanga FY, Raghavendra V, DeLeo JA. Quantitative real-time RT-PCR assessment of spinal microglial and astrocytic activation markers in a rat model of neuropathic pain. Neurochem Int. 2004;45:397–407.CrossRefPubMed Tanga FY, Raghavendra V, DeLeo JA. Quantitative real-time RT-PCR assessment of spinal microglial and astrocytic activation markers in a rat model of neuropathic pain. Neurochem Int. 2004;45:397–407.CrossRefPubMed
40.
go back to reference Mitsi V, Terzi D, Purushothaman I, Manouras L, Gaspari S, Neve RL, et al. RGS9-2—controlled adaptations in the striatum determine the onset of action and efficacy of antidepressants in neuropathic pain states. Proc Natl Acad Sci U S A. 2015;112:E5088–97.CrossRefPubMedPubMedCentral Mitsi V, Terzi D, Purushothaman I, Manouras L, Gaspari S, Neve RL, et al. RGS9-2—controlled adaptations in the striatum determine the onset of action and efficacy of antidepressants in neuropathic pain states. Proc Natl Acad Sci U S A. 2015;112:E5088–97.CrossRefPubMedPubMedCentral
41.
go back to reference Dripps IJ, Wang Q, Neubig RR, Rice KC, Traynor JR, Jutkiewicz EM. The role of regulator of G protein signaling 4 in delta-opioid receptor-mediated behaviors. Psychopharmacology. 2017;234:29–39.CrossRefPubMed Dripps IJ, Wang Q, Neubig RR, Rice KC, Traynor JR, Jutkiewicz EM. The role of regulator of G protein signaling 4 in delta-opioid receptor-mediated behaviors. Psychopharmacology. 2017;234:29–39.CrossRefPubMed
42.
go back to reference Hausmann ON, Hu WH, Keren-Raifman T, Witherow DS, Wang Q, Levay K, et al. Spinal cord injury induces expression of RGS7 in microglia/macrophages in rats. Eur J Neurosci. 2002;15:602–12.CrossRefPubMed Hausmann ON, Hu WH, Keren-Raifman T, Witherow DS, Wang Q, Levay K, et al. Spinal cord injury induces expression of RGS7 in microglia/macrophages in rats. Eur J Neurosci. 2002;15:602–12.CrossRefPubMed
43.
go back to reference Garnier M, Zaratin PF, Ficalora G, Valente M, Fontanella L, Rhee MH, et al. Up-regulation of regulator of G protein signaling 4 expression in a model of neuropathic pain and insensitivity to morphine. J Pharmacol Exp Ther. 2003;304:1299–306.CrossRefPubMed Garnier M, Zaratin PF, Ficalora G, Valente M, Fontanella L, Rhee MH, et al. Up-regulation of regulator of G protein signaling 4 expression in a model of neuropathic pain and insensitivity to morphine. J Pharmacol Exp Ther. 2003;304:1299–306.CrossRefPubMed
44.
go back to reference Krumins AM, Barker SA, Huang C, Sunahara RK, Yu K, Wilkie TM, et al. Differentially regulated expression of endogenous RGS4 and RGS7. J Biol Chem. 2004;279:2593–9.CrossRefPubMed Krumins AM, Barker SA, Huang C, Sunahara RK, Yu K, Wilkie TM, et al. Differentially regulated expression of endogenous RGS4 and RGS7. J Biol Chem. 2004;279:2593–9.CrossRefPubMed
45.
go back to reference Jaggi AS, Jain V, Singh N. Animal models of neuropathic pain. Fundam Clin Pharmacol. 2011;25:1–28.CrossRefPubMed Jaggi AS, Jain V, Singh N. Animal models of neuropathic pain. Fundam Clin Pharmacol. 2011;25:1–28.CrossRefPubMed
46.
go back to reference Saugstad JA, Marino MJ, Folk JA, Hepler JR, Conn PJ. RGS4 inhibits signaling by group I metabotropic glutamate receptors. J Neurosci. 1998;18:905–13.PubMed Saugstad JA, Marino MJ, Folk JA, Hepler JR, Conn PJ. RGS4 inhibits signaling by group I metabotropic glutamate receptors. J Neurosci. 1998;18:905–13.PubMed
47.
go back to reference Kim CF, Moalem-Taylor G. Detailed characterization of neuro-immune responses following neuropathic injury in mice. Brain Res. 2011;1405:95–108.CrossRefPubMed Kim CF, Moalem-Taylor G. Detailed characterization of neuro-immune responses following neuropathic injury in mice. Brain Res. 2011;1405:95–108.CrossRefPubMed
48.
go back to reference Gattlen C, Clarke CB, Piller N, Kirschmann G, Pertin M, Decosterd I, et al. Spinal cord T-cell infiltration in the rat spared nerve injury model: a time course study. Int J Mol Sci. 2016;17:352.CrossRefPubMedPubMedCentral Gattlen C, Clarke CB, Piller N, Kirschmann G, Pertin M, Decosterd I, et al. Spinal cord T-cell infiltration in the rat spared nerve injury model: a time course study. Int J Mol Sci. 2016;17:352.CrossRefPubMedPubMedCentral
49.
go back to reference Costigan M, Moss A, Latremoliere A, Johnston C, Verma-Gandhu M, Herbert TA, et al. T-cell infiltration and signaling in the adult dorsal spinal cord is a major contributor to neuropathic pain-like hypersensitivity. J Neurosci. 2009;29:14415–22.CrossRefPubMedPubMedCentral Costigan M, Moss A, Latremoliere A, Johnston C, Verma-Gandhu M, Herbert TA, et al. T-cell infiltration and signaling in the adult dorsal spinal cord is a major contributor to neuropathic pain-like hypersensitivity. J Neurosci. 2009;29:14415–22.CrossRefPubMedPubMedCentral
50.
go back to reference Ji RR, Kawasaki Y, Zhuang ZY, Wen YR, Decosterd I. Possible role of spinal astrocytes in maintaining chronic pain sensitization: review of current evidence with focus on bFGF/JNK pathway. Neuron Glia Biol. 2006;2:259–69.CrossRefPubMedPubMedCentral Ji RR, Kawasaki Y, Zhuang ZY, Wen YR, Decosterd I. Possible role of spinal astrocytes in maintaining chronic pain sensitization: review of current evidence with focus on bFGF/JNK pathway. Neuron Glia Biol. 2006;2:259–69.CrossRefPubMedPubMedCentral
51.
go back to reference Eusemann TN, Willmroth F, Fiebich B, Biber K, van Calker D. Adenosine receptors differentially regulate the expression of regulators of G-protein signalling (RGS) 2, 3 and 4 in astrocyte-like cells. PLoS One. 2015;10:e0134934.CrossRefPubMedPubMedCentral Eusemann TN, Willmroth F, Fiebich B, Biber K, van Calker D. Adenosine receptors differentially regulate the expression of regulators of G-protein signalling (RGS) 2, 3 and 4 in astrocyte-like cells. PLoS One. 2015;10:e0134934.CrossRefPubMedPubMedCentral
52.
go back to reference Scholz J, Woolf CJ. The neuropathic pain triad: neurons, immune cells and glia. Nat Neurosci. 2007;10:1361–8.CrossRefPubMed Scholz J, Woolf CJ. The neuropathic pain triad: neurons, immune cells and glia. Nat Neurosci. 2007;10:1361–8.CrossRefPubMed
53.
go back to reference Fedoroff S, McAuley WA, Houle JD, Devon RM. Astrocyte cell lineage. V. Similarity of astrocytes that form in the presence of dBcAMP in cultures to reactive astrocytes in vivo. J Neurosci Res. 1984;12:14–27.CrossRefPubMed Fedoroff S, McAuley WA, Houle JD, Devon RM. Astrocyte cell lineage. V. Similarity of astrocytes that form in the presence of dBcAMP in cultures to reactive astrocytes in vivo. J Neurosci Res. 1984;12:14–27.CrossRefPubMed
54.
go back to reference Tsugane M, Nagai Y, Kimura Y, Oka J, Kimura H. Differentiated astrocytes acquire sensitivity to hydrogen sulfide that is diminished by the transformation into reactive astrocytes. Antioxid Redox Signal. 2007;9:257–69.CrossRefPubMed Tsugane M, Nagai Y, Kimura Y, Oka J, Kimura H. Differentiated astrocytes acquire sensitivity to hydrogen sulfide that is diminished by the transformation into reactive astrocytes. Antioxid Redox Signal. 2007;9:257–69.CrossRefPubMed
55.
go back to reference Vermeiren C, Najimi M, Vanhoutte N, Tilleux S, de Hemptinne I, Maloteaux JM, et al. Acute up-regulation of glutamate uptake mediated by mGluR5a in reactive astrocytes. J Neurochem. 2005;94:405–16.CrossRefPubMed Vermeiren C, Najimi M, Vanhoutte N, Tilleux S, de Hemptinne I, Maloteaux JM, et al. Acute up-regulation of glutamate uptake mediated by mGluR5a in reactive astrocytes. J Neurochem. 2005;94:405–16.CrossRefPubMed
57.
go back to reference Neitzel KL, Hepler JR. Cellular mechanisms that determine selective RGS protein regulation of G protein-coupled receptor signaling. Semin Cell Dev Biol. 2006;17:383–9.CrossRefPubMed Neitzel KL, Hepler JR. Cellular mechanisms that determine selective RGS protein regulation of G protein-coupled receptor signaling. Semin Cell Dev Biol. 2006;17:383–9.CrossRefPubMed
58.
go back to reference Xie GX, Palmer PP. How regulators of G protein signaling achieve selective regulation. J Mol Biol. 2007;366:349–65.CrossRefPubMed Xie GX, Palmer PP. How regulators of G protein signaling achieve selective regulation. J Mol Biol. 2007;366:349–65.CrossRefPubMed
59.
go back to reference Kawasaki Y, Zhang L, Cheng JK, Ji RR. Cytokine mechanisms of central sensitization: distinct and overlapping role of interleukin-1beta, interleukin-6, and tumor necrosis factor-alpha in regulating synaptic and neuronal activity in the superficial spinal cord. J Neurosci. 2008;28:5189–94.CrossRefPubMedPubMedCentral Kawasaki Y, Zhang L, Cheng JK, Ji RR. Cytokine mechanisms of central sensitization: distinct and overlapping role of interleukin-1beta, interleukin-6, and tumor necrosis factor-alpha in regulating synaptic and neuronal activity in the superficial spinal cord. J Neurosci. 2008;28:5189–94.CrossRefPubMedPubMedCentral
60.
go back to reference Papouin T, Dunphy J, Tolman M, Foley JC, Haydon PG. Astrocytic control of synaptic function. Phil Trans R Soc B. 2017;372:20160154. Papouin T, Dunphy J, Tolman M, Foley JC, Haydon PG. Astrocytic control of synaptic function. Phil Trans R Soc B. 2017;372:20160154.
61.
go back to reference Deumens R, Steyaert A, Forget P, Schubert M, Lavand'homme P, Hermans E, et al. Prevention of chronic postoperative pain: cellular, molecular, and clinical insights for mechanism-based treatment approaches. Prog Neurobiol. 2013;104:1–37.CrossRefPubMed Deumens R, Steyaert A, Forget P, Schubert M, Lavand'homme P, Hermans E, et al. Prevention of chronic postoperative pain: cellular, molecular, and clinical insights for mechanism-based treatment approaches. Prog Neurobiol. 2013;104:1–37.CrossRefPubMed
62.
go back to reference Colburn RW, Rickman AJ, DeLeo JA. The effect of site and type of nerve injury on spinal glial activation and neuropathic pain behavior. Exp Neurol. 1999;157:289–304.CrossRefPubMed Colburn RW, Rickman AJ, DeLeo JA. The effect of site and type of nerve injury on spinal glial activation and neuropathic pain behavior. Exp Neurol. 1999;157:289–304.CrossRefPubMed
Metadata
Title
Inflammation-associated regulation of RGS in astrocytes and putative implication in neuropathic pain
Authors
Pierre J. Doyen
Maxime Vergouts
Amandine Pochet
Nathalie Desmet
Sabien van Neerven
Gary Brook
Emmanuel Hermans
Publication date
01-12-2017
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2017
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-017-0971-x

Other articles of this Issue 1/2017

Journal of Neuroinflammation 1/2017 Go to the issue