Skip to main content
Top
Published in: Brain Structure and Function 3/2018

01-04-2018 | Original Article

Mapping GPR88-Venus illuminates a novel role for GPR88 in sensory processing

Authors: Aliza T. Ehrlich, Meriem Semache, Julie Bailly, Stefan Wojcik, Tanzil M. Arefin, Christine Colley, Christian Le Gouill, Florence Gross, Viktoriya Lukasheva, Mireille Hogue, Emmanuel Darcq, Laura-Adela Harsan, Michel Bouvier, Brigitte L. Kieffer

Published in: Brain Structure and Function | Issue 3/2018

Login to get access

Abstract

GPR88 is an orphan G-protein coupled receptor originally characterized as a striatal-enriched transcript and is a potential target for neuropsychiatric disorders. At present, gene knockout studies in the mouse have essentially focused on striatal-related functions and a comprehensive knowledge of GPR88 protein distribution and function in the brain is still lacking. Here, we first created Gpr88-Venus knock-in mice expressing a functional fluorescent receptor to fine-map GPR88 localization in the brain. The receptor protein was detected in neuronal soma, fibers and primary cilia depending on the brain region, and remarkably, whole-brain mapping revealed a yet unreported layer-4 cortical lamination pattern specifically in sensory processing areas. The unique GPR88 barrel pattern in L4 of the somatosensory cortex appeared 3 days after birth and persisted into adulthood, suggesting a potential function for GPR88 in sensory integration. We next examined Gpr88 knockout mice for cortical structure and behavioral responses in sensory tasks. Magnetic resonance imaging of live mice revealed abnormally high fractional anisotropy, predominant in somatosensory cortex and caudate putamen, indicating significant microstructural alterations in these GPR88-enriched areas. Further, behavioral analysis showed delayed responses in somatosensory-, visual- and olfactory-dependent tasks, demonstrating a role for GPR88 in the integration rather than perception of sensory stimuli. In conclusion, our data show for the first time a prominent role for GPR88 in multisensory processing. Because sensory integration is disrupted in many psychiatric diseases, our study definitely positions GPR88 as a target to treat mental disorders perhaps via activity on cortical sensory networks.
Appendix
Available only for authorised users
Literature
go back to reference Bi YP (NJ, US), Dzierba, Carolyn Diane (Middletown, CT, US), Bronson, Joanne J. (Durham, CT, US), Fink, Cynthia (Lebanon, NJ, US), Green, Michael (Easton, PA, US), Kimball, David (East Windsor, NJ, US), Macor, John E. (Gullford, CT, US), Kwon, Soojin, Zhang, Yulian, Zipp, Greg (2013) Modulators of G protein-coupled receptor 88. United States Patent Bi YP (NJ, US), Dzierba, Carolyn Diane (Middletown, CT, US), Bronson, Joanne J. (Durham, CT, US), Fink, Cynthia (Lebanon, NJ, US), Green, Michael (Easton, PA, US), Kimball, David (East Windsor, NJ, US), Macor, John E. (Gullford, CT, US), Kwon, Soojin, Zhang, Yulian, Zipp, Greg (2013) Modulators of G protein-coupled receptor 88. United States Patent
go back to reference Braff DL, Geyer MA, Light GA, Sprock J, Perry W, Cadenhead KS, Swerdlow NR (2001) Impact of prepulse characteristics on the detection of sensorimotor gating deficits in schizophrenia. Schizophr Res 49(1–2):171–178CrossRefPubMed Braff DL, Geyer MA, Light GA, Sprock J, Perry W, Cadenhead KS, Swerdlow NR (2001) Impact of prepulse characteristics on the detection of sensorimotor gating deficits in schizophrenia. Schizophr Res 49(1–2):171–178CrossRefPubMed
go back to reference Brown LL, Hand PJ, Divac I (1996) Representation of a single vibrissa in the rat neostriatum: peaks of energy metabolism reveal a distributed functional module. Neuroscience 75(3):717–728CrossRefPubMed Brown LL, Hand PJ, Divac I (1996) Representation of a single vibrissa in the rat neostriatum: peaks of energy metabolism reveal a distributed functional module. Neuroscience 75(3):717–728CrossRefPubMed
go back to reference Del Zompo M, Deleuze JF, Chillotti C, Cousin E, Niehaus D, Ebstein RP, Ardau R, Mace S, Warnich L, Mujahed M, Severino G, Dib C, Jordaan E, Murad I, Soubigou S, Koen L, Bannoura I, Rocher C, Laurent C, Derock M, Faucon Biguet N, Mallet J, Meloni R (2014) Association study in three different populations between the GPR88 gene and major psychoses. Mol Genet Genomic Med 2(2):152–159. https://doi.org/10.1002/mgg3.54 CrossRefPubMed Del Zompo M, Deleuze JF, Chillotti C, Cousin E, Niehaus D, Ebstein RP, Ardau R, Mace S, Warnich L, Mujahed M, Severino G, Dib C, Jordaan E, Murad I, Soubigou S, Koen L, Bannoura I, Rocher C, Laurent C, Derock M, Faucon Biguet N, Mallet J, Meloni R (2014) Association study in three different populations between the GPR88 gene and major psychoses. Mol Genet Genomic Med 2(2):152–159. https://​doi.​org/​10.​1002/​mgg3.​54 CrossRefPubMed
go back to reference Divac I, Rosvold HE, Szwarcbart MK (1967) Behavioral effects of selective ablation of the caudate nucleus. J Comp Physiol Psychol 63(2):184–190CrossRefPubMed Divac I, Rosvold HE, Szwarcbart MK (1967) Behavioral effects of selective ablation of the caudate nucleus. J Comp Physiol Psychol 63(2):184–190CrossRefPubMed
go back to reference Dzierba CD, Bi Y, Dasgupta B, Hartz RA, Ahuja V, Cianchetta G, Kumi G, Dong L, Aleem S, Fink C, Garcia Y, Green M, Han J, Kwon S, Qiao Y, Wang J, Zhang Y, Liu Y, Zipp G, Liang Z, Burford N, Ferrante M, Bertekap R, Lewis M, Cacace A, Grace J, Wilson A, Nouraldeen A, Westphal R, Kimball D, Carson K, Bronson JJ, Macor JE (2015) Design, synthesis, and evaluation of phenylglycinols and phenyl amines as agonists of GPR88. Bioorg Med Chem Lett 25(7):1448–1452. https://doi.org/10.1016/j.bmcl.2015.01.036 CrossRefPubMed Dzierba CD, Bi Y, Dasgupta B, Hartz RA, Ahuja V, Cianchetta G, Kumi G, Dong L, Aleem S, Fink C, Garcia Y, Green M, Han J, Kwon S, Qiao Y, Wang J, Zhang Y, Liu Y, Zipp G, Liang Z, Burford N, Ferrante M, Bertekap R, Lewis M, Cacace A, Grace J, Wilson A, Nouraldeen A, Westphal R, Kimball D, Carson K, Bronson JJ, Macor JE (2015) Design, synthesis, and evaluation of phenylglycinols and phenyl amines as agonists of GPR88. Bioorg Med Chem Lett 25(7):1448–1452. https://​doi.​org/​10.​1016/​j.​bmcl.​2015.​01.​036 CrossRefPubMed
go back to reference Erbs E, Faget L, Scherrer G, Matifas A, Filliol D, Vonesch JL, Koch M, Kessler P, Hentsch D, Birling MC, Koutsourakis M, Vasseur L, Veinante P, Kieffer BL, Massotte D (2015) A mu-delta opioid receptor brain atlas reveals neuronal co-occurrence in subcortical networks. Brain Struct Funct 220(2):677–702. https://doi.org/10.1007/s00429-014-0717-9 CrossRefPubMed Erbs E, Faget L, Scherrer G, Matifas A, Filliol D, Vonesch JL, Koch M, Kessler P, Hentsch D, Birling MC, Koutsourakis M, Vasseur L, Veinante P, Kieffer BL, Massotte D (2015) A mu-delta opioid receptor brain atlas reveals neuronal co-occurrence in subcortical networks. Brain Struct Funct 220(2):677–702. https://​doi.​org/​10.​1007/​s00429-014-0717-9 CrossRefPubMed
go back to reference Fishell G, van der Kooy D (1989) Pattern formation in the striatum: developmental changes in the distribution of striatonigral projections. Brain Res Dev Brain Res 45(2):239–255CrossRefPubMed Fishell G, van der Kooy D (1989) Pattern formation in the striatum: developmental changes in the distribution of striatonigral projections. Brain Res Dev Brain Res 45(2):239–255CrossRefPubMed
go back to reference Fox MW (1965) The visual cliff test for the study of visual depth perception in the mouse. Anim Behav 13(2):232–233CrossRefPubMed Fox MW (1965) The visual cliff test for the study of visual depth perception in the mouse. Anim Behav 13(2):232–233CrossRefPubMed
go back to reference Hodges A, Strand AD, Aragaki AK, Kuhn A, Sengstag T, Hughes G, Elliston LA, Hartog C, Goldstein DR, Thu D, Hollingsworth ZR, Collin F, Synek B, Holmans PA, Young AB, Wexler NS, Delorenzi M, Kooperberg C, Augood SJ, Faull RL, Olson JM, Jones L, Luthi-Carter R (2006) Regional and cellular gene expression changes in human Huntington’s disease brain. Hum Mol Genet 15(6):965–977. https://doi.org/10.1093/hmg/ddl013 CrossRefPubMed Hodges A, Strand AD, Aragaki AK, Kuhn A, Sengstag T, Hughes G, Elliston LA, Hartog C, Goldstein DR, Thu D, Hollingsworth ZR, Collin F, Synek B, Holmans PA, Young AB, Wexler NS, Delorenzi M, Kooperberg C, Augood SJ, Faull RL, Olson JM, Jones L, Luthi-Carter R (2006) Regional and cellular gene expression changes in human Huntington’s disease brain. Hum Mol Genet 15(6):965–977. https://​doi.​org/​10.​1093/​hmg/​ddl013 CrossRefPubMed
go back to reference Ingallinesi M, Le Bouil L, Biguet NF, Thi AD, Mannoury la Cour C, Millan MJ, Ravassard P, Mallet J, Meloni R (2015) Local inactivation of Gpr88 in the nucleus accumbens attenuates behavioral deficits elicited by the neonatal administration of phencyclidine in rats. Mol Psychiatry 20(8):951–958. https://doi.org/10.1038/mp.2014.92 CrossRefPubMed Ingallinesi M, Le Bouil L, Biguet NF, Thi AD, Mannoury la Cour C, Millan MJ, Ravassard P, Mallet J, Meloni R (2015) Local inactivation of Gpr88 in the nucleus accumbens attenuates behavioral deficits elicited by the neonatal administration of phencyclidine in rats. Mol Psychiatry 20(8):951–958. https://​doi.​org/​10.​1038/​mp.​2014.​92 CrossRefPubMed
go back to reference Logue SF, Grauer SM, Paulsen J, Graf R, Taylor N, Sung MA, Zhang L, Hughes Z, Pulito VL, Liu F, Rosenzweig-Lipson S, Brandon NJ, Marquis KL, Bates B, Pausch M (2009) The orphan GPCR, GPR88, modulates function of the striatal dopamine system: a possible therapeutic target for psychiatric disorders? Mol Cell Neurosci 42(4):438–447. https://doi.org/10.1016/j.mcn.2009.09.007 CrossRefPubMed Logue SF, Grauer SM, Paulsen J, Graf R, Taylor N, Sung MA, Zhang L, Hughes Z, Pulito VL, Liu F, Rosenzweig-Lipson S, Brandon NJ, Marquis KL, Bates B, Pausch M (2009) The orphan GPCR, GPR88, modulates function of the striatal dopamine system: a possible therapeutic target for psychiatric disorders? Mol Cell Neurosci 42(4):438–447. https://​doi.​org/​10.​1016/​j.​mcn.​2009.​09.​007 CrossRefPubMed
go back to reference Ludewig K, Geyer MA, Etzensberger M, Vollenweider FX (2002) Stability of the acoustic startle reflex, prepulse inhibition, and habituation in schizophrenia. Schizophr Res 55(1–2):129–137CrossRefPubMed Ludewig K, Geyer MA, Etzensberger M, Vollenweider FX (2002) Stability of the acoustic startle reflex, prepulse inhibition, and habituation in schizophrenia. Schizophr Res 55(1–2):129–137CrossRefPubMed
go back to reference Mai JK, Voss T, Paxinos G (2008) Atlas of the human brain, 3rd edn. Elsevier/Academic Press, Amsterdam Mai JK, Voss T, Paxinos G (2008) Atlas of the human brain, 3rd edn. Elsevier/Academic Press, Amsterdam
go back to reference Maier DL, Mani S, Donovan SL, Soppet D, Tessarollo L, McCasland JS, Meiri KF (1999) Disrupted cortical map and absence of cortical barrels in growth-associated protein (GAP)-43 knockout mice. Proc Natl Acad Sci USA 96(16):9397–9402CrossRefPubMedPubMedCentral Maier DL, Mani S, Donovan SL, Soppet D, Tessarollo L, McCasland JS, Meiri KF (1999) Disrupted cortical map and absence of cortical barrels in growth-associated protein (GAP)-43 knockout mice. Proc Natl Acad Sci USA 96(16):9397–9402CrossRefPubMedPubMedCentral
go back to reference Massart R, Mignon V, Stanic J, Munoz-Tello P, Becker JA, Kieffer BL, Darmon M, Sokoloff P, Diaz J (2016) Developmental and adult expression patterns of the G-protein-coupled receptor GPR88 in the rat: establishment of a dual nuclear-cytoplasmic localization. J Comp Neurol 524(14):2776–2802. https://doi.org/10.1002/cne.23991 CrossRefPubMed Massart R, Mignon V, Stanic J, Munoz-Tello P, Becker JA, Kieffer BL, Darmon M, Sokoloff P, Diaz J (2016) Developmental and adult expression patterns of the G-protein-coupled receptor GPR88 in the rat: establishment of a dual nuclear-cytoplasmic localization. J Comp Neurol 524(14):2776–2802. https://​doi.​org/​10.​1002/​cne.​23991 CrossRefPubMed
go back to reference Perry W, Minassian A, Feifel D, Braff DL (2001) Sensorimotor gating deficits in bipolar disorder patients with acute psychotic mania. Biol Psychiatry 50(6):418–424CrossRefPubMed Perry W, Minassian A, Feifel D, Braff DL (2001) Sensorimotor gating deficits in bipolar disorder patients with acute psychotic mania. Biol Psychiatry 50(6):418–424CrossRefPubMed
go back to reference Schallert T, Whishaw IQ (1984) Bilateral cutaneous stimulation of the somatosensory system in hemidecorticate rats. Behav Neurosci 98(3):518–540CrossRefPubMed Schallert T, Whishaw IQ (1984) Bilateral cutaneous stimulation of the somatosensory system in hemidecorticate rats. Behav Neurosci 98(3):518–540CrossRefPubMed
go back to reference Song DD, Harlan RE (1994) Genesis and migration patterns of neurons forming the patch and matrix compartments of the rat striatum. Brain Res Dev Brain Res 83(2):233–245CrossRefPubMed Song DD, Harlan RE (1994) Genesis and migration patterns of neurons forming the patch and matrix compartments of the rat striatum. Brain Res Dev Brain Res 83(2):233–245CrossRefPubMed
go back to reference Swerdlow NR, Lipska BK, Weinberger DR, Braff DL, Jaskiw GE, Geyer MA (1995) Increased sensitivity to the sensorimotor gating-disruptive effects of apomorphine after lesions of medial prefrontal cortex or ventral hippocampus in adult rats. Psychopharmacology 122(1):27–34CrossRefPubMed Swerdlow NR, Lipska BK, Weinberger DR, Braff DL, Jaskiw GE, Geyer MA (1995) Increased sensitivity to the sensorimotor gating-disruptive effects of apomorphine after lesions of medial prefrontal cortex or ventral hippocampus in adult rats. Psychopharmacology 122(1):27–34CrossRefPubMed
go back to reference Van der Loos H, Woolsey TA (1973) Somatosensory cortex: structural alterations following early injury to sense organs. Science 179(4071):395–398CrossRefPubMed Van der Loos H, Woolsey TA (1973) Somatosensory cortex: structural alterations following early injury to sense organs. Science 179(4071):395–398CrossRefPubMed
go back to reference Wilson JS, Hull CD, Buchwald NA (1983) Intracellular studies of the convergence of sensory input on caudate neurons of cat. Brain Res 270(2):197–208CrossRefPubMed Wilson JS, Hull CD, Buchwald NA (1983) Intracellular studies of the convergence of sensory input on caudate neurons of cat. Brain Res 270(2):197–208CrossRefPubMed
Metadata
Title
Mapping GPR88-Venus illuminates a novel role for GPR88 in sensory processing
Authors
Aliza T. Ehrlich
Meriem Semache
Julie Bailly
Stefan Wojcik
Tanzil M. Arefin
Christine Colley
Christian Le Gouill
Florence Gross
Viktoriya Lukasheva
Mireille Hogue
Emmanuel Darcq
Laura-Adela Harsan
Michel Bouvier
Brigitte L. Kieffer
Publication date
01-04-2018
Publisher
Springer Berlin Heidelberg
Published in
Brain Structure and Function / Issue 3/2018
Print ISSN: 1863-2653
Electronic ISSN: 1863-2661
DOI
https://doi.org/10.1007/s00429-017-1547-3

Other articles of this Issue 3/2018

Brain Structure and Function 3/2018 Go to the issue