Hostname: page-component-7c8c6479df-ph5wq Total loading time: 0 Render date: 2024-03-29T09:12:10.436Z Has data issue: false hasContentIssue false

Developmental regulation of group I metabotropic glutamate receptors in the premature brain and their protective role in a rodent model of periventricular leukomalacia

Published online by Cambridge University Press:  14 December 2011

Lauren L. Jantzie
Affiliation:
Department of Neurology, Children's Hospital Boston and Harvard Medical School, Boston, MA, USA
Delia M. Talos
Affiliation:
Department of Neurology, Children's Hospital Boston and Harvard Medical School, Boston, MA, USA
Debra B. Selip
Affiliation:
Department of Neurology, Children's Hospital Boston and Harvard Medical School, Boston, MA, USA
Li An
Affiliation:
Department of Neurology, Children's Hospital Boston and Harvard Medical School, Boston, MA, USA
Michele C. Jackson
Affiliation:
Department of Neurology, Children's Hospital Boston and Harvard Medical School, Boston, MA, USA
Rebecca D. Folkerth
Affiliation:
Department of Pathology, Children's Hospital Boston and Harvard Medical School, Boston, MA, USA
Wenbin Deng
Affiliation:
Department of Cell Biology and Human Anatomy, Institute for Pediatric Regenerative Medicine, School of Medicine, University of California, Sacramento, CA, USA
Frances E. Jensen*
Affiliation:
Department of Neurology, Children's Hospital Boston and Harvard Medical School, Boston, MA, USA
*
Correspondence should be addressed to: Frances E. Jensen, Department of Neurology, Children's Hospital Boston, CLS 14073, 300 Longwood Avenue, Boston, MA 02115, USA phone: 617-919-2445 fax: 617-919-2769 email: frances.jensen@childrens.harvard.edu

Abstract

Cerebral white matter injury in premature infants, known as periventricular leukomalacia (PVL), is common after hypoxia–ischemia (HI). While ionotropic glutamate receptors (iGluRs) can mediate immature white matter injury, we have previously shown that excitotoxic injury to premyelinating oligodendrocytes (preOLs) in vitro can be attenuated by group I metabotropic glutamate receptor (mGluR) agonists. Thus, we evaluated mGluR expression in developing white matter in rat and human brain, and tested the protective efficacy of a central nervous system (CNS)-penetrating mGluR agonist on injury to developing oligodendrocytes (OLs) in vivo. Group I mGluRs (mGluR1 and mGluR5) were strongly expressed on OLs in neonatal rodent cerebral white matter throughout normal development, with highest expression early in development on preOLs. Specifically at P6, mGluR1 and mGLuR5 were most highly expressed on GalC-positive OLs compared to neurons, axons, astrocytes and microglia. Systemic administration of (1S,3R) 1-aminocyclopentane-trans-1,3,-dicarboxylic acid (ACPD) significantly attenuated the loss of myelin basic protein in the white matter following HI in P6 rats. Assessment of postmortem human tissue showed both mGluR1 and mGluR5 localized on immature OLs in white matter throughout development, with mGluR5 highest in the preterm period. These data indicate group I mGluRs are highly expressed on OLs during the peak period of vulnerability to HI and modulation of mGluRs is protective in a rodent model of PVL. Group I mGluRs may represent important therapeutic targets for protection from HI-mediated white matter injury.

Type
Research Article
Copyright
Copyright © Cambridge University Press 2011

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

REFERENCES

Alexander, G.M. and Godwin, D.W. (2006) Metabotropic glutamate receptors as a strategic target for the treatment of epilepsy. Epilepsy Research 71, 122.CrossRefGoogle ScholarPubMed
Allin, M., Walshe, M., Fern, A., Nosarti, C., Cuddy, M., Rifkin, L. et al. (2008) Cognitive maturation in preterm and term born adolescents. Journal of Neurology, Neurosurgery, and Psychiatry 79, 381386.CrossRefGoogle ScholarPubMed
Back, S.A., Luo, N.L., Borenstein, N.S., Levine, J.M., Volpe, J.J. and Kinney, H.C. (2001) Late oligodendrocyte progenitors coincide with the developmental window of vulnerability for human perinatal white matter injury. Journal of Neuroscience 21, 13021312.CrossRefGoogle ScholarPubMed
Back, S.A., Riddle, A. and McClure, M.M. (2007) Maturation-dependent vulnerability of perinatal white matter in premature birth. Stroke 38, 724730.CrossRefGoogle ScholarPubMed
Bao, W.L., Williams, A.J., Faden, A.I. and Tortella, F.C. (2001) Selective mGluR5 receptor antagonist or agonist provides neuroprotection in a rat model of focal cerebral ischemia. Brain Research 922, 173179.CrossRefGoogle ScholarPubMed
Bayless, S. and Stevenson, J. (2007) Executive functions in school-age children born very prematurely. Early Human Development 83, 247254.Google Scholar
Bear, M.F. (2005) Therapeutic implications of the mGluR theory of fragile X mental retardation. Genes, Brain and Behaviour 4, 393398.CrossRefGoogle ScholarPubMed
Benveniste, H. (1991) The excitotoxin hypothesis in relation to cerebral ischemia. Cerebrovascular & Brain Metabolism Reviews 3, 213245.Google ScholarPubMed
Biber, K., Laurie, D.J., Berthele, A., Sommer, B., Tolle, T.R., Gebicke-Harter, P.J. et al. (1999) Expression and signaling of group I metabotropic glutamate receptors in astrocytes and microglia. Journal of Neurochemistry 72, 16711680.Google Scholar
Boer, K., Encha-Razavi, F., Sinico, M. and Aronica, E. (2010) Differential distribution of group I metabotropic glutamate receptors in developing human cortex. Brain Research 1324, 2433.CrossRefGoogle Scholar
Butt, A.M. (2006) Neurotransmitter-mediated calcium signaling in oligodendrocyte physiology and pathology. Glia 54, 666675.CrossRefGoogle ScholarPubMed
Byrnes, K.R., Loane, D.J. and Faden, A.I. (2009a) Metabotropic glutamate receptors as targets for multipotential treatment of neurological disorders. Neurotherapeutics 6, 94107.Google Scholar
Byrnes, K.R., Stoica, B., Loane, D.J., Riccio, A., Davis, M.I. and Faden, A.I. (2009b) Metabotropic glutamate receptor 5 activation inhibits microglial associated inflammation and neurotoxicity. Glia 57, 550560.CrossRefGoogle ScholarPubMed
Carroll, F.I. (2008) Antagonists at metabotropic glutamate receptor subtype 5: structure activity relationships and therapeutic potential for addiction. Annals of the New York Academy of Sciences 1141, 221232.CrossRefGoogle ScholarPubMed
Catania, M.V., D'Antoni, S., Bonaccorso, C.M., Aronica, E., Bear, M.F. and Nicoletti, F. (2007) Group I metabotropic glutamate receptors: a role in neurodevelopmental disorders? Molecular Neurobiology 35, 298307.Google Scholar
Dean, J.M., Moravec, M.D., Grafe, M., Abend, N., Ren, J., Gong, X. et al. (2011) Strain-specific differences in perinatal rodent oligodendrocytes lineage progression and its correlation with human. Developmental Neuroscience, doi: 10.1159/000327242. Epub ahead of Print, August 24, 2011.CrossRefGoogle ScholarPubMed
Deng, W., Wang, H., Rosenberg, P.A., Volpe, J.J. and Jensen, F.E. (2004) Role of metabotropic glutamate receptors in oligodendrocyte excitotoxicity and oxidative stress. Proceedings of the National Academy of Sciences of the U.S.A. 101, 77517756.CrossRefGoogle ScholarPubMed
D'Antoni, S., Berretta, A., Bonaccorso, C.M., Bruno, V., Aronica, E., Nicoletti, F. et al. (2008) Metabotropic glutamate receptors in glial cells. Neurochemistry Research 33, 24362443.CrossRefGoogle ScholarPubMed
Di Giorgi Gerevini, V.D., Caruso, A., Cappuccio, I., Ricci Vitiani, L., Romeo, S., Della Rocca, C. et al. (2004) The mGlu5 metabotropic glutamate receptor is expressed in zones of active neurogenesis of the embryonic and postnatal brain. Brain Research. Developmental Brain Research 150, 1722.Google Scholar
Drouin-Ouellet, J., Bownell, A.-L., Saint-Pierre, M., Fasano, C., Emond, V., Trudeau, L.-E. et al. (2011) Neuroinflammation is associated with changes in glial mGluR5 expression and the development of neonatal excitotoxic lesions. Glia 59, 188199.CrossRefGoogle ScholarPubMed
Farso, M.C., O'Shea, R.D. and Beart, P.M. (2009) Evidence group I mGluR drugs modulate the activation profile of lipopolysaccharide-exposed microglia in culture. Neurochemical Research 34, 17211728.CrossRefGoogle Scholar
Fern, R. and Moller, T. (2000) Rapid ischemic cell death in immature oligodendrocytes: a fatal glutamate release feedback loop. Journal of Neuroscience 20, 3442.Google Scholar
Ferraguti, F. and Shigemoto, R. (2006) Metabotropic glutamate receptors. Cell and Tissue Research 326, 483504.CrossRefGoogle ScholarPubMed
Ferriero, D. (2004) Neonatal brain injury. New England Journal of Medicine 351, 19851995.Google Scholar
Folkerth, R.D., Haynes, R.L., Borenstein, N.S., Belliveau, R.A., Trachtenberg, F., Rosenberg, P.A. et al. (2004) Developmental lag in superoxide dismutases relative to other anti-oxidant enzymes in premyelinated human telencephalic white matter. Journal of Neuropathology and Experimental Neurology 63, 990999.Google Scholar
Follett, P.L., Deng, W., Dai, W., Talos, D.M., Massillon, L.J., Rosenberg, P.A. et al. (2004) Glutamate receptor-mediated oligodendrocyte toxicity in periventricular leukomalacia: a protective role for topiramate. Journal of Neuroscience 24, 44124420.CrossRefGoogle ScholarPubMed
Follett, P.L., Rosenberg, P.A., Volpe, J.J. and Jensen, F.E. (2000) NBQX attenuates excitotoxic injury in developing white matter. Journal of Neuroscience 20, 92359241.CrossRefGoogle ScholarPubMed
Gonsette, R.E. (2008) Oxidative stress and excitotoxicity: a therapeutic issue in multiple sclerosis? Multiple Sclerosis 14, 2234.Google Scholar
Haynes, R.L., Folkerth, R.D., Keefe, R.J., Sung, I., Swzeda, L.I., Rosenberg, P.A. et al. (2003) Nitrosative and oxidative injury to premyelinating oligodendrocytes in periventricular leukomalacia. Journal of Neuropathology and Experimental Neurology 62, 441450.CrossRefGoogle ScholarPubMed
Hodge, C.W., Miles, M.F., Sharko, A.C., Stevenson, R.A., Hillmann, J.R., Lepoutre, V. et al. (2006) The mGluR antagonist MPEP selectively inhibits the onset and maintenance of ethanol self-admnistration in C57BL/6J mice. Psychopharmacology 183, 429438.CrossRefGoogle ScholarPubMed
Jantzie, L.L., Cheung, P.Y. and Todd, K.G. (2005) Doxycycline reduces cleaved caspase-3 and microglial activation in an animal model of neonatal hypoxia–ischemia. Journal of Cerebral Blood Flow Metabolism 25, 314324.CrossRefGoogle Scholar
Jantzie, L.L. and Todd, K.G. (2010) Doxycycline inhibits proinflammatory cytokines but not acute cerebral cytogenesis after hypoxia–ischemia in neonatal rats. Journal of Psychiatry and Neuroscience 35, 2032.CrossRefGoogle Scholar
Jensen, F. (2006) Developmental factors regulating susceptibility to perinatal brain injury and seizures. Current Opinion in Pediatrics 18, 628633.CrossRefGoogle ScholarPubMed
Karadottir, R. and Attwell, D. (2007) Neurotransmitter receptors in the life and death of oligodendrocytes. Neuroscience 145, 14261438.Google Scholar
Kelland, E.E. and Toms, N.J. (2001) Group I metabotropic glutamate receptors limit AMPA receptor-mediated oligodendrocyte progenitor cell death. European Journal of Pharmacology 424, R3R4.Google Scholar
Kinney, H.C. and Back, S.A. (1998) Human oligodendroglial development: relationship to periventricular leukomalacia. Seminars in Pediatric Neurology 5, 180189.CrossRefGoogle ScholarPubMed
Kobaly, K., Schluchter, M., Minich, N., Friedman, H., Taylor, H.G., Wilson-Costello, D. et al. (2008) Outcomes of extremely low birth weight (<1 kg) and extremely low gestational age (<28 weeks) infants with bronchopulmonary dysplasia: effects of practice changes in 2000 to 2003. Pediatrics 121, 7381.Google Scholar
Larroque, B., Ancel, P.Y., Marret, S., Marchand, L., Andre, M., Arnaud, C. et al. (2008) Neurodevelopmental disabilities and special care of 5-year-old children born before 33 weeks of gestation (the EPIPAGE study): a longitudinal cohort study. Lancet 371, 813820.Google Scholar
Lechpammer, M., Manning, S.M., Samonte, F., Nelligan, J., Sabo, E., Talos, D.M. et al. (2008) Minocycline treatment following hypoxic/ischaemic injury attenuates white matter injury in a rodent model of periventricular leukcomalacia. Neuropathology and Applied Neurobiology 34, 379393.Google Scholar
Liu, C., Lin, N., Wu, B. and Qiu, Y. (2009) Neuroprotective effect of memantine combined with topiramate in hypoxic-ischemic brain injury. Brain Research 1282, 173182.Google Scholar
Lojkova, D. and Mares, P. (2005) Anticonvulsant action of an antagonist of metabotropic glutamate receptor mGluR5 MPEP in immature rats. Neuropharmacology 49, 219229.Google Scholar
Lujan, R., Roberts, J.D., Shigemoto, R., Ohishi, H. and Somogyi, P. (1997) Differential plasma membrane distribution of metabotropic glutamate receptors mGluR1α, mGluR2, mGluR5, relative to neurotransmitter release sites. Journal of Chemical Neuroanatomy 13, 219241.Google Scholar
Luscher, C. and Huber, K.M. (2010) Group 1 mGluR-dependent synaptic long-term depression: mechanisms and implications for circuitry and disease. Neuron 65, 445459.CrossRefGoogle ScholarPubMed
Luyt, K., Varadi, A., Durant, C.F. and Molnar, E. (2006) Oligodendroglial metabotropic glutamate receptors are developmentally regulated and involved in the prevention of apoptosis. Journal of Neurochemistry 99, 641656.CrossRefGoogle ScholarPubMed
Luyt, K., Varadi, A. and Molnar, E. (2003) Functional metabotropic glutamate receptors are expressed in oligodendrocyte progenitor cells. Journal of Neurochemistry 84, 14521464.CrossRefGoogle ScholarPubMed
Maiese, K., Chong, Z.Z. and Li, F. (2005) Driving cellular plasticity and survival through the signal transduction pathways of metabotropic glutamate receptors. Current Neurovascular Research 2, 425446.CrossRefGoogle ScholarPubMed
Manning, S.M., Talos, D.M., Zhou, C., Selip, D.B., Park, H.K., Park, C.J. et al. (2008) NMDA receptor blockade with memantine attenuates white matter injury in a rat model of periventricular leukomalacia. Journal of Neuroscience 28, 66706678.CrossRefGoogle Scholar
Martin, J.A., Kung, H.C., Mathews, T.J., Hoyert, D.L., Strobino, D.M., Guyer, B. et al. (2008) Annual summary of vital statistics: 2006. Pediatrics 121, 788801.CrossRefGoogle ScholarPubMed
McDonald, J.W. and Johnston, M.V. (1990) Physiological and pathological roles of excitatory amino acids during central nervous system development. Brain Research. Brain Research Reviews 15, 4170.Google Scholar
McDonald, J.W., Fix, A.S., Tizzano, J.P. and Schoepp, D.D. (1993) Seizures and brain injury in neonatal rats induced by 1S,3R-ACPD, a metabotropic glutamate receptor agonist. Journal of Neuroscience 13, 44454455.CrossRefGoogle ScholarPubMed
Olive, M.F. (2009) Metabotropic glutamate receptor ligands as potential therapeutics for addiction. Current Drug Abuse Reviews 2, 8398.Google Scholar
Rakhade, S.N., Zhou, C., Aujla, P.K., Fishman, R., Sucher, N.J. and Jensen, F.E. (2008) Early alterations of AMPA receptors mediate synaptic potentiation induced by neonatal seizures. Journal of Neuroscience 28, 79797990.Google Scholar
Ribeiro, F.M., Paquet, M., Cregan, S.P. and Ferguson, S.S. (2010) Group I metabotropic glutamate receptor signaling and its implication in neurological disease. CNS & Neurological Disorders – Drug Targets 9, 574595.CrossRefGoogle Scholar
Romano, C., Sesma, M.A., McDonald, C.T., O'Malley, K., Van Den Pol, A.N. and Olney, J.W. (1995) Distribution of metabotropic glutamate receptor mGluR immunoreactivity in rat brain. Journal of Comparative Neurology 355, 455469.CrossRefGoogle Scholar
Silbereis, J.C., Huang, E.J., Back, S.A. and Rowitch, D.H. (2010) Towards improved animal models of neonatal white matter injury associated with cerebral palsy. Disease Models & Mechanisms 3, 678688.Google Scholar
Snyder, E.M., Philpot, B.D., Huber, K.M., Dong, X., Fallon, J.R. and Bear, M.F. (2001) Internalization of ionotropic glutamate receptors in response to mGluR activation. Nature Neuroscience 4, 10791085.Google Scholar
Talos, D., Fishman, R., Park, H., Folkerth, R., Follett, P., Volpe, J.J. et al. (2006a) Developmental regulation of alpha-amino-3-hydroxy-5-methyl-4-isoxazole-proprionic acid receptor subunit expression in forebrain and relationship to regional susceptibility to hypoxic/ischemic injury. I. Rodent cerebral white matter and cortex. Journal of Comparative Neurology 497, 4261.Google Scholar
Talos, D., Follett, P., Folkerth, R., Fishman, R., Trachtenberg, F., Volpe, J.J. et al. (2006b) Developmental regulation of alpha-amino-3-hydroxy-5-methyl-4-isoxazole-proprionic acid receptor subunit expression in forebrain and relationship to regional susceptibility to hypoxic/ischemic injury. II. Human cerebral white matter and cortex. Journal of Comparative Neurology 497, 6177.Google Scholar
Taylor, D.L., Pirianov, G., Holland, S., McGinnity, C.J., Norman, A.L., Reali, C. et al. (2010) Attenuation of proliferation in oligodendroctye precursor cells by activated microglia. Journal of Neuroscience Research 88, 16321644.Google Scholar
Ure, J., Baudry, M. and Perassolo, M. (2006) Metabotropic glutamate receptors and epilepsy. Journal of the Neurological Sciences 247, 19.Google Scholar
Vannucci, R.C. (2000) Hypoxic-ischemic encephalopathy. American Journal of Perinatology 17, 113119.CrossRefGoogle ScholarPubMed
Volpe, J.J. (2009) Brain injury in premature infants: a complex amalgam of destructive and developmental disturbances. Lancet Neurology 8, 110124.CrossRefGoogle ScholarPubMed
Wake, H., Lee, P.R. and Fields, R.D. (2011) Control of local protein synthesis and initial events in myelination by action potentials. Science 333, 16471651.Google Scholar
Zhong, J., Gerber, G., Kojic, L. and Randic, M. (2000) Dual modulation of excitatory synaptic transmission by agonists at group I metabotropic glutamate receptors in the rat spinal dorsal horn. Brain Research 887, 359377.CrossRefGoogle Scholar
Supplementary material: File

Jensen Supplementary Figure Legends

Jensen Supplementary Figure Legends

Download Jensen Supplementary Figure Legends(File)
File 20 KB
Supplementary material: PDF

Jensen Supplementary Figures

Jensen Supplementary Figures

Download Jensen Supplementary Figures(PDF)
PDF 211.6 KB