319
Views
13
CrossRef citations to date
0
Altmetric
REVIEW

Blockade of N-acetylaspartylglutamate peptidases: a novel protective strategy for brain injuries and neurological disorders

, &
Pages 867-873 | Received 06 Jan 2014, Accepted 31 Jan 2014, Published online: 13 Feb 2014

References

  • Conn PJ. Physiological roles and therapeutic potential of metabotropic glutamate receptors. Ann NY Acad Sci 2003; 1003:12–21.
  • Whelan J. NAALADase inhibitors: a novel approach to glutamate regulation. Drug Discov Today 2000;5:171–2.
  • Muir K, Grosset D. Neuroprotection for acute stroke: making clinical trials work. Stroke 1999;30:180–2.
  • Waxman EA, Lynch DR. N-methyl-D-aspartate receptor subtypes: multiple roles in excitotoxicity and neurological disease. Neuroscientist 2005;11:37–49.
  • Faden AI, Demediuk P, Panter SS, The role of excitatory amino acids and NMDA receptors in traumatic brain injury. Science 1989;244:798–800.
  • Katayama Y, Becker DP, Tamura T, Massive increases in extracellular potassium and the indiscriminate release of glutamate following concussive brain injury. J Neurosurg 1990;73:889–900.
  • Carlton SM, Hargett GL, Coggeshall RE. Localization of metabotropic glutamate receptors 2/3 on primary afferent axons in the rat. Neuroscience 2001;105:957–69.
  • Neugebauer V. Metabotropic glutamate receptors—important modulators of nociception and pain behavior. Pain 2002; 98:1–8.
  • Berent-Spillson A, Robinson AM, Golovoy D, Protection against glucose-induced neuronal death by NAAG and GCP II inhibition is regulated by mGluR3. J Neurochem 2004;89:90–9.
  • Tomiyama M, Furusawa K, Kamijo M, Upregulation of mRNAs coding for AMPA and NMDA receptor subunits and metabotropic glutamate receptors in the dorsal horn of the spinal cord in a rat model of diabetes mellitus. Brain Res Mol Brain Res 2005;136:275–81.
  • Rothstein JD. Of mice and men: reconciling preclinical ALS mouse studies and human clinical trials. Ann Neurol 2003;53:423–6.
  • Strong MJ. Amyotrophic lateral sclerosis: contemporary concepts in etiopathogenesis and pharmacotherapy. Expert Opin Investig Drugs 2004;13:1593–614.
  • Tsai G, Coyle JT. Glutamatergic mechanisms in schizophrenia. Annu Rev Pharmacol Toxicol 2002;42:165–79.
  • Javitt DC, Coyle JT. Decoding schizophrenia. Sci Am 2004;290:48–55.
  • Bullock R, Lyeth BG, Muizelaar JP. Current status of neuroprotection trials for traumatic brain injury: lessons from animal models and clinical studies. Neurosurgery 1999;45:207–17.
  • Narayan RK, Michel ME, Ansell B, Clinical trials in head injury. J. Neurotrauma 2002;19:503–57.
  • Neale JH, Bzdega T, Wroblewska B. N-Acetylaspartyl glutamate: the most abundant peptide neurotransmitter in the mammalian central nervous system. J Neurochem 2000; 75:443–52.
  • Neale JH, Olszewski RT, Gehl LM, The neurotransmitter N-acetylaspartylglutamate in models of pain, ALS, diabetic neuropathy, CNS injury and schizophrenia. Trends Pharmacol Sci 2005;26:477–84.
  • Zhou J, Neale JH, Pomper MG, NAAG peptidase inhibitors and their potential for diagnosis and therapy. Nat Rev Drug Discov 2005;4:1015–26.
  • Bařinka C, Rojas C, Slusher B, Glutamate carboxypeptidase II in diagnosis and treatment of neurologic disorders and prostate cancer. Curr Med Chem 2012;19:856–70.
  • Durand D, Carniglia L, Caruso C, mGlu3 receptor and astrocytes: partners in neuroprotection. Neuropharmacology. 2013;66:1–11.
  • Coyle JT. The nagging question of the function of N-acetylaspartylglutamate. Neurobiol Dis 1997;4:231–8.
  • Neale JH, Olszewski RT, Zuo D, Advances in understanding the peptide neurotransmitter NAAG and appearance of a new member of the NAAG neuropeptide family. J Neurochem, 2011;118:490–8.
  • Wroblewska B, Wroblewski JT, Saab OH, N-acetylaspartylglutamate inhibits forskolin-stimulated cyclic AMP levels via a metabotropic glutamate receptor in cultured cerebellar granule cells. J Neurochem 1993;61:943–8.
  • Wroblewska B, Wroblewski JT, Pshenichkin S, N-acetylaspartylglutamate selectively activates mGluR3 receptors in transfected cells, J Neurochem 1997;69:174–81.
  • Wroblewska B, Santi MR, Neale JH. N-acetylaspartylglutamate activates cyclic AMP-coupled metabotropic glutamate receptors in cerebellar astrocytes. Glia 1998;24:172–9.
  • Cartmell J, Schoepp DD. Regulation of neurotransmitter release by metabotropic glutamate receptors. J Neurochem 2000;75:889–907.
  • Bruno V, Battaglia G, Copani A, Metabotropic glutamate receptor subtypes as targets for neuroprotective drugs. J Cereb Blood Flow Metab 2001;21:1013–33.
  • Schweitzer C, Kratzeisen C, Adam G, Characterization of -LY354740 binding to rat mGlu2 and mGlu3 receptors expressed in CHO cells using Semliki Forest virus vectors. Neuropharmacology 2000;39:1700–6.
  • Bischofberger J, Schild D. Glutamate and N-acetylaspartylglutamate block HVA calcium currents in frog olfactory bulb interneurons via an mGluR2/3-like receptor. J Neurophysiol 1996;76:2089–92.
  • Zhao J, Ramadan E, Cappiello M, et al. NAAG inhibits KCl-induced [(3)H]-GABA release via mGluR3, cAMP, PKA and L-type calcium conductance. Eur J Neurosci 2001;13:340–6.
  • Xi ZX, Baker DA, Shen H, Group II metabotropic glutamate receptors modulate extracellular glutamate in the nucleus accumbens. J Pharmacol Exp Therap 2002;300:162–71
  • Garrido Sanabria ER, Wozniak KM, Slusher BS, GCP II (NAALADase) inhibition suppresses mossy fiber-CA3 synaptic neurotransmission by a presynaptic mechanism. J Neurophysiol 2004;91:182–93.
  • Luthi-Carter R, Berger UV, Barczak AK, Isolation and expression of a rat brain cDNA encoding glutamate carboxypeptidase II. Proc Natl Acad Sci USA 1998;95:3215–20.
  • Bzdega T, Crowe SL, Ramadan ER, The cloning and characterization of a second brain enzyme with NAAG peptidase activity. J Neurochem 2004;89:627–35.
  • Hlouchová K, Barinka C, Klusák V, Biochemical characterization of human glutamate carboxypeptidase III. J Neurochem 2007;101:682–96.
  • Bacich DJ, Ramadan E, O'Keefe DS, Deletion of the glutamate carboxypeptidase II gene in mice reveals a second enzyme activity that hydrolyzes N-acetylaspartylglutamate. J Neurochem 2002;83:20–9.
  • Jackson PF, Cole DC, Slusher BS, Design, synthesis, and biological activity of a potent inhibitor of the neuropeptidase N-acetylated-α-linked acidic dipeptidase. J Med Chem 1996;39:619–22.
  • Majer P, Jackson PF, Delahanty G, Synthesis and biological evaluation of thiol-based inhibitors of glutamate carboxypeptidase II: discovery of an orally active GCP II inhibitor. J Med Chem 2003;46:1989–96.
  • Kozikowski AP, Nan F, Conti P, Design of remarkably simple, yet potent urea-based inhibitors of glutamate carboxypeptidase II (NAALADase). J Med Chem 2001;44:298–301.
  • Tsukamoto T, Wozniak KM, Slusher BS. Progress in the discovery and development of glutamate carboxypeptidase II inhibitors. Drug Discov Today 2007;12:767–76.
  • Slusher BS, Vornov JJ, Thomas AG, Selective inhibition of NAALADase, which converts NAAG to glutamate, reduces ischemic brain injury. Nat Med 1999;5:1396–402.
  • Jackson PF, Slusher BS. Design of NAALADase inhibitors: a novel neuroprotective strategy. Curr Med Chem 2001;8:949–57.
  • Tsukamoto T, Flanary JM, Rojas C, et al. Phosphonate and phosphinate analogues of N-acylated-glutamylglutamate: potent inhibitors of glutamate carboxypeptidase II. Bioorg Med Chem Lett 2002;12:2189–92.
  • Oliver AJ, Wiest O, Helquist P, Conformational and SAR analysis of NAALADase and PSMA inhibitors. Bioorg Med Chem 2003;11:4455–61.
  • Maung J, Mallari JP, Girtsman TA, Probing for a hydrophobic a binding register in prostate-speci. c membrane antigen with phenylalkylphosphonamidates. Bioorg Med Chem 2004;12:4969–79.
  • van der Post JP, de Visser SJ, de Kam ML, The central nervous system effects, pharmacokinetics and safety of the NAALADase-inhibitor GPI 5693. Br J Clin Pharmacol 2005;60:128–36.
  • Grella B, Adams J, Berry JF, The discovery and structure–activity relationships of indole-based inhibitors of glutamate carboxypeptidase II. Bioorg Med Chem Lett, 2010;20:7222–25.
  • Nan F, Bzdega T, Pshenichkin S, Dual function glutamate-related ligands: discovery of a novel, potent inhibitor of glutamate carboxypeptidase II possessing mGluR3 agonist activity. J Med Chem 2000;43:772–4.
  • Jackson PF, Tays KL, Maclin KM, Design and pharmacological activity of phosphinic acid based NAALADase inhibitors. J Med Chem 2001;44:4170–5.
  • Kozikowski AP, Zhang J, Nan F, Synthesis of urea-based inhibitors as active site probes of glutamate carboxypeptidase II: efficacy as analgesic agents. J Med Chem 2004;47:1729–38.
  • Barinka C, Byun Y, Dusich CL, Interactions between human glutamate carboxypeptidase II and urea-based inhibitors: structural characterization. J Med Chem 2008;51:7737–43.
  • Choy CJ, Fulton MD, Davis AL, Rationally designed sulfamides as glutamate carboxypeptidase II inhibitors. Chem Biol Drug Des. 2013;82:612–9.
  • Plechanovová A, Byun Y, Alquicer G, Novel substrate-based inhibitors of human glutamate carboxypeptidase II with enhanced lipophilicity. J Med Chem 2011;54:7535–46.
  • Barinka C, Rovenská M, Mlcochová P, Structural insight into the pharmacophore pocket of human glutamate carboxypeptidase II. J Med Chem 2007;250:3267–73.(83)
  • Hlouchova K, Barinka C, Konvalinka J, et al. Structural insight into the evolutionary and pharmacologic homology of glutamate carboxypeptidases II and III. FEBS J. 2009;276:4448–62.
  • Wang H, Byun Y, Barinka C, Bioisosterism of urea-based GCPII inhibitors: synthesis and structure–activity relationship studies. Bioorg Med Chem Lett 2010;20:392–7.
  • Lu XM, Tang Z, Liu W, , N-acetylaspartylglutamate protects against transient focal cerebral ischemia in rats. Eur J Pharmacol 2000;408:233–9.
  • Williams AJ, Lu XM, Slusher B, Electroencephalogram analysis and neuroprotective profile of the N-acetylated-alpha-linked acidic dipeptidase inhibitor, GPI5232, in normal and brain-injured rats. J Pharmacol Exp Ther 2001;299:48–57.
  • Bacich DJ, Wozniak KM, Lu XC, Mice lacking glutamate carboxypeptidase II are protected from peripheral neuropathy and ischemic brain injury. J Neurochem 2005;95:314–23.
  • Cai Z, Lin S, Rhodes PG. Neuroprotective effects of N-acetylaspartylglutamate in a neonatal rat model of hypoxia-ischemia. Eur J Pharmacol 2002;437:139–45.
  • Zhong C, Zhao X, Sarva J, NAAG peptidase inhibitor reduces acute neuronal degeneration and astrocyte damage following lateral fluid percussion TBI in rats. J. Neurotrauma 2005;22:266–76.
  • Zhong C, Zhao X, Van LC, NAAG peptidase inhibitor increases dialysate NAAG and reduces glutamate, aspartate and GABA levels in the dorsal hippocampus following fluid percussion injury in the rat. J Neurochem 2006;97:1015–25.
  • Yamamoto T, Nozaki-Taguchi N, Sakashita T. Spinal N-acetylated-α-linked acidic dipeptidase (NAALADase) inhibition attenuates mechanical allodynia induced by paw carrageenan injection in the rat. Brain Res 2001;909:138–44.
  • Yamamoto T, Nozaki-Taguchi N, Sakashita Y, Inhibition of spinal N-acetylated-α-linked acidic dipeptidase produces an antinociceptive effect in the rat formalin test. Neurosci 2001;102:473–9.
  • Chen SR, Wozniak KM, Slusher BS, Effect of 2-(phosphonomethyl)-pentanedioic acid on allodynia and afferent ectopic discharges in a rat model of neuropathic pain. J Pharmacol Exp Ther 2002;300:662–7.
  • Carpenter KJ, Sen S, Matthews EA, Effects of GCP-II inhibition on responses of dorsal horn neurones after inflammation and neuropathy: an electrophysiological study in the rat. Neuropeptides 2003;37:298–306.
  • Yamamoto T, Hirasawa S, Wroblewska B, Antinociceptive effects of N-acetylaspartylglutamate (NAAG) peptidase inhibitors ZJ 11, ZJ 17 and ZJ 43 in the rat formalin test and in the rat neuropathic pain model. Eur J Neurosci 2004;20: 483–94.
  • Zhang W., Slusher B., Murakawa Y., et al. GCPII (NAALADase) inhibition prevents long-term diabetic neuropathy in type 1 diabetic BB/Wor rats. J Neurol Sci 2002;194:21–8.
  • Bruno V, Wroblewska B, Wroblewski JT, Neuroprotective activity of N-acetylaspartylglutamate in cultured cortical cells. Neuroscience 1998;85:751–7.
  • Thomas AG, Liu W, Olkowski JL, , Neuroprotection mediated by glutamate carboxypeptidase II (NAALADase) inhibition requires TGF-beta. Eur J Pharmacol 2001;430: 33–40.
  • Cleveland DW, Rothstein JD. From Charcot to Lou Gehrig: deciphering selective motor neuron death in ALS. Nature Rev Neurosci 2001;2:806–19.
  • Ghadge GD, Slusher BS, Bodner A, Glutamate carboxypeptidase II inhibition protects motor neurons from death in familial amyotrophic lateral sclerosis models. Proc Natl Acad Sci USA 2003;100:9554–9.
  • Javitt DC, Zukin SR. Recent advances in the phencyclidine model of schizophrenia. Am J Psychiatry 1991;148:1301–8.
  • Krystal JH, Anand A, Moghaddam B. Effects of NMDA receptor antagonists: implications for the pathophysiology of schizophrenia. Arch Gen Psychiatry 2002;59:663–4.
  • Moghaddam B, Adams BW. Reversal of phencyclidine effects by a group II metabotropic glutamate receptor agonist in rats. Science 1998;281:1349–52.
  • Cartmell J, Monn JA, Schoepp DD. The metabotropic glutamate 2/3 receptor agonists LY354740 and LY379268 selectively attenuate phencyclidine versus d-amphetamine motor behaviors in rats. J Pharmacol Exp Ther 1999;291:161–70.
  • Olszewski R, Buhkari N, Zhou J, NAAG peptidase inhibition reduces locomotor activity and some stereotypes in the PCP model of schizophrenia via group II mGluR. J Neurochem 2004;89:876–85.
  • Olszewski RT, Bzdega T, Neale JH. mGluR3 and not mGluR2 receptors mediate the efficacy of NAAG peptidase inhibitor in validated model of schizophrenia. Schizophr Res 2012;136:160–1.
  • Ferraguti F, Corti C, Valerio E, Activated astrocytes in areas of kainate-induced neuronal injury upregulate the expression of the metabotropic glutamate receptors 2/3 and 5. Exp Brain Res 2001;137:1–11.
  • Aronica E, Gorter JA, Ijlst-Keizers H, Expression and functional role of mGluR3 and mGluR5 in human astrocytes and glioma cells: opposite regulation of glutamate transporter proteins. Eur J Neurosci 2003;17:2106–18.
  • Thomas AG, Liu W, Olkowski JL, Neuroprotection mediated by glutamate carboxypeptidase II (NAALADase) inhibition requires TGF-beta. Eur J Pharmacol 2001;430:33–40.
  • Byrnes KR, Loane DJ, Faden AI. Metabotropic glutamate receptors as targets for multipotential treatment of neurological disorders. Neurotherapeutics 2009;6:94–107.
  • Urazaev AK, Buttram JG Jr, Deen JP, Mechanisms for clearance of released N-acetylaspartylglutamate in crayfish nerve fibers: implications for axon-glia signaling. Neuroscience 2001;107:697–703.
  • Tsukamoto T, Wozniak KM, Slusher BS. Progress in the discovery and development of glutamate carboxypeptidase II inhibitors. Drug Discov Today 2007;12:767–76
  • Baslow MH. The astrocyte surface NAAG receptor and NAAG peptidase signaling complex as a therapeutic target. Drug News Perspect 2008;21:251–7.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.