Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

NAAG peptidase inhibitors and their potential for diagnosis and therapy

Key Points

  • Glutamate (Glu) is the most abundant excitatory transmitter in the central nervous system. However, excessive glutamatergic transmission can damage or kill neurons, and has therefore been implicated in a variety of neurological disorders.

  • N-Acetyl-L-aspartyl-L-glutamate (NAAG), one of the three most prevalent neurotransmitters, acts as an agonist at group II metabotropic glutamate receptors with preference for metabotropic glutamate (mGlu3) receptors on neurons and glia. Two extracellular enzymes, glutamate carboxypeptidase II and III (GCPII and III), hydrolyse NAAG to N-acetylaspartate (NAA) and glutamate following its release into the synaptic cleft.

  • Inhibition of these NAAG peptidases (NPs) is thought to provide neuroprotection by increasing the intrasynaptic concentration of NAAG. NAAG decreases the release of glutamate by activation of presynaptic group II mGlu receptors and stimulates release of trophic factors from glia. Those actions of NAAG may provide neuroprotection in clinical conditions in which glutamate mediates and mGlu3 receptor activation reduces pathology.

  • Importantly, NP inhibitors do not seem to affect normal glutamate function. NP inhibition enhances a natural ongoing regulatory process rather than chronically activating or inhibiting receptors in a manner that is unrelated to ongoing chemical neurotransmission. NP represents an intriguing target for drug discovery aimed at unmet medical needs.

  • Additionally, human GCPII has also been identified as prostate-specific membrane antigen (PSMA), a cell surface protein expressed in elevated levels by prostate cancer. Its X-ray crystal structure was recently reported.

  • Studies using small-molecule-based NP inhibitors have confirmed their beneficial effects in animal models relevant to neurodegenerative diseases as well as cancer.

  • NP inhibitors therefore have significant potential for use as both diagnostic and therapeutic agents. Specific applications include neuropathic and inflammatory pain, traumatic brain injury, ischemic stroke, schizophrenia, diabetic neuropathy, amyotrophic lateral sclerosis, drug addiction, as well as prostate cancer.

Abstract

Modulation of N-acetyl-L-aspartyl-L-glutamate peptidase activity with small-molecule inhibitors holds promise for a wide variety of diseases that involve glutamatergic transmission, and has implications for the diagnosis and therapy of cancer. This new class of compounds, of which at least one has entered clinical trials and proven to be well tolerated, has demonstrated efficacy in experimental models of pain, schizophrenia, amyotrophic lateral sclerosis, traumatic brain injury and, when appropriately functionalized, can image prostate cancer. Further investigation of these promising drug candidates will be needed to bring them to the marketplace. The recent publication of the X-ray crystal structure for the enzymatic target of these compounds should facilitate the development of other new agents with enhanced activity that could improve both the diagnosis and treatment of neurological disorders.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Metabotropic glutamate (mGlu) receptor subtypes.
Figure 2: Mechanism of NAAG action on presynaptic endings.
Figure 3: Structure model of the GCPII extracellular domain.
Figure 4: Crystal structural of prostate-specific mebrane antigen: ribbon diagrams of side and top view.
Figure 5: Selected small-molecule-based NAAG peptidase inhibitors.
Figure 6: Complex model between GCPII and PBDA.
Figure 7: Chemical structures of two radiolabelled urea-based potent NAAG peptidase inhibitors.
Figure 8: Small animal imaging of [11C]DCMC and [125I]DCIT in experimental prostate cancer.

Similar content being viewed by others

References

  1. Research and Markets. Neuroprotection — Drugs, Markets and Companies [online], <http://www.researchandmarkets.com/reportinfo.asp?cat_id=0&report_id=39073&q=neuroprotection&p=1> (2005).

  2. Conn, P. J. Physiological roles and therapeutic potential of metabotropic glutamate receptors. Ann. NY Acad. Sci. 1003, 12–21 (2003).

    CAS  PubMed  Google Scholar 

  3. Whelan, J. NAALADase inhibitors: a novel approach to glutamate regulation. Drug Discov. Today 5, 171–172 (2000).

    CAS  PubMed  Google Scholar 

  4. Neale, J. H., Bzdega, T. & Wroblewska, B. N-Acetylaspartylglutamate: the most abundant peptide neurotransmitter in the mammalian central nervous system. J. Neurochem. 75, 443–452 (2000). An excellent review that extensively describes the roles of NAAG.

    CAS  PubMed  Google Scholar 

  5. Coyle, J. T. The nagging question of the function of N-acetylaspartylglutamate. Neurobiol. Dis. 4, 231–238 (1997).

    CAS  PubMed  Google Scholar 

  6. Wroblewska, B., Wroblewski, J. T., Pshenichkin, S., Surin, A., Sullivan, S. E. & Neale, J. H. N-Acetylaspartylglutamate selectively activates mGluR3 receptors in transfected cells. J. Neurochem. 69, 174–182 (1997).

    CAS  PubMed  Google Scholar 

  7. Wroblewska, B., Santi, M. R. & Neale, J. H. N-Acetylaspartylglutamate activates cyclic-AMP coupled metabotropic glutamate receptors in cerebellar astrocytes. Glia 24, 172–180 (1998).

    CAS  PubMed  Google Scholar 

  8. Wroblewska, B., Wroblewski, J. T., Saab, O. & Neale, J. H. N-Acetylaspartylglutamate inhibits forskolin-stimulated cyclic AMP levels via a metabotropic glutamate receptor in cultured cerebellar granule cells. J. Neurochem. 61, 943–948 (1993).

    CAS  PubMed  Google Scholar 

  9. Wroblewska, B., Wegorzewska, I. N., Bzdega, T., Olszewski, R. T. & Neale, J. H. Differential negative coupling of type 3 metabotropic glutamate receptor to cGMP levels in neurons and astrocytes. J. Neurochem. (in the press).

  10. Cartmell, J. & Schoepp, D. D. Regulation of neurotransmitter release by metabotropic glutamate receptors. J. Neurochem. 75, 889–907 (2000). Outstanding review that addresses the neurochemical evidence for mGluR-mediated regulation of neurotransmitters.

    CAS  PubMed  Google Scholar 

  11. Varney, M. A. & Gereau, R. W. Metabotropic glutamate receptor involvement in models of acute and persistent pain: Prospects for the development of novel analgesics. Curr. Drug Targets CNS Neurol. Disord. 1, 283–296 (2002).

    CAS  PubMed  Google Scholar 

  12. Riveros, N. & Orrego, F. A study of possible excitatory effects of N-acetylaspartylglutamate in different in vivo and in vitro brain preparations. Brain Res. 299, 393–395 (1984).

    CAS  PubMed  Google Scholar 

  13. Robinson, M. B., Blakely, R. D., Couto, R. & Coyle, J. T. Hydrolysis of the brain dipeptide N-acetyl-L-aspartyl-L-glutamate. J. Biol. Chem. 262, 14498–14506 (1987).

    CAS  PubMed  Google Scholar 

  14. Carter, R. E., Feldman, A. R. & Coyle, J. T. Prostate-specific membrane antigen is a hydrolase with substrate and pharmacologic characteristics of a neuropeptidase. Proc. Natl Acad. Sci. USA 93, 749–753 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Bzdega, T. et al. Molecular cloning of a peptidase against N-acetylaspartylglutamate (NAAG) from a rat hippocampal cDNA library. J. Neurochem. 69, 2270–2277 (1997).

    CAS  PubMed  Google Scholar 

  16. Bzdega, T. et al. The cloning and characterization of a second brain enzyme with NAAG peptidase activity. J. Neurochem. 89, 627–635 (2004).

    CAS  PubMed  Google Scholar 

  17. O'Keefe, D. S. et al. Mapping, genomic organization and promoter analysis of the human prostate-specific membrane antigen gene. Biochim. Biophys. Acta 1443, 113–127 (1998).

    CAS  PubMed  Google Scholar 

  18. Bacich, D. J., Pinto, J. T., Tong, W. P. & Heston, W. D. Cloning, expression, genomic localization, and enzymatic activities of the mouse homolog of prostate-specific membrane antigen/NAALADase/folate hydrolase. Mamm. Genome 12, 117–123 (2001).

    CAS  PubMed  Google Scholar 

  19. Zhao, J. et al. NAAG inhibits [3H]-GABA release from cortical neurons via the type 3 metabotropic glutamate receptor. Eur. J. Neurosci. 13, 340–346 (2001).

    CAS  PubMed  Google Scholar 

  20. Sanabria, E. R., Wozniak, K. M., Slusher, B. S. & Keller, A. GCP II (NAALADase) Inhibition suppresses mossy fiber-CA3 synaptic neurotransmission by a presynaptic mechanism. J. Neurophysiol. 91, 182–193 (2004).

    PubMed  Google Scholar 

  21. Bruno, V., Wroblewska, B., Wroblewski, J. T., Fiore & L. & Nicoletti, F. Neuroprotective activity of N-acetylaspartylglutamate in cultured cortical cells. Neurosci. 85, 751–757 (1998).

    CAS  Google Scholar 

  22. Slusher, B. S. et al. Selective inhibition of NAALADase, which converts NAAG to glutamate, reduces ischemic brain injury. Nature Med. 5, 1396–1402 (1999).

    CAS  PubMed  Google Scholar 

  23. Thomas, A. G. et al. Neuroprotection mediated by glutamate carboxypeptidase II (NAALADase) inhibition requires TGF-β. Eur. J. Pharmacol. 430, 33–40 (2001).

    CAS  PubMed  Google Scholar 

  24. Jackson, P. F. et al. Design, synthesis, and biological activity of a potent inhibitor of neuropeptidase N-acetylated-α-linked acidic dipeptidase. J. Med. Chem. 39, 619–622 (1996).

    CAS  PubMed  Google Scholar 

  25. Yourick, D. L., Koenig, M. L., Durden,. & Long, J. B. N-acetylaspartylglutamate and β-NAAG protect against injury induced by NMDA and hypoxia in primary spinal cord cultures. Brain Res. 991, 56–64 (2003).

    CAS  PubMed  Google Scholar 

  26. Parsons, C. G., Danysz, W. & Quack, G. Glutamate in CNS disorders as a target for drug development: an update. Drug News Perspect. 11, 523–569 (1998). Excellent review that describes the role of glutamate in CNS disorders.

    CAS  PubMed  Google Scholar 

  27. van der Post, J. P. et al. The central nervous system effects, pharmacokinetics and safety of the NAALADase-inhibitor GPI 5693. Br. J. Clin. Pharmacol. 60, 128–136 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Speno, H. S. et al. Site-directed mutagenesis of predicted active site residues in glutamate carboxypeptidase II. Mol. Pharmacol. 55, 179–185 (1999). An important paper describing the enzyme structure and function of GCPII.

    CAS  PubMed  Google Scholar 

  29. Tsai, G. et al. Early embryonic death of glutamate carboxypeptidase II (NAALADase) homozygous mutants. Synapse 50, 285–292 (2003).

    CAS  PubMed  Google Scholar 

  30. Maras, B. et al. Aminopeptidase from Streptomyces griseus: primary structure and comparison with other zinc containing aminopeptidases. Eur. J. Biochem. 236, 843–846 (1996).

    CAS  PubMed  Google Scholar 

  31. van Heeke, G., Denslow, S., Watkins, J., Wilson, K. & Wagner, F. Cloning and nucleotide sequence of the Vibrio proteolyticus aminopeptidase gene. Biochim. Biophys. Acta 1131, 337–340 (1992).

    CAS  PubMed  Google Scholar 

  32. Heston, W. D. Significance of prostate-specific membrane antigen (PSMA). A neurocarboxypeptidase and membrane folate hydrolase. Urologe A. 35, 400–407 (1996).

    CAS  PubMed  Google Scholar 

  33. Rawlings, N. D. & Barrett, A. J. Structure of membrane glutamate carboxypeptidase. Biochim. Biophys. Acta 1339, 247–252 (1997).

    CAS  PubMed  Google Scholar 

  34. Mahadevan, D. & Saldanha, J. W. The extracellular regions of PSMA and the transferring receptor contain an aminopeptidase domain: implications for drug design. Protein Sci. 8, 2546–2549 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Rong, S. B. et al. Molecular modeling of the interactions of glutamate carboxypeptidase II with its potent NAAG-based inhibitors. J. Med. Chem. 45, 4140–4152 (2002). An outstanding paper describing the molecular modelling of NAAG peptidase inhibitors.

    CAS  PubMed  Google Scholar 

  36. Davis, M. I., Bennett, M. J., Thomas, L. M. & Bjorkman, P. J. Crystal structure of prostate-specific membrane antigen, a tumor marker and peptidase. Proc. Natl Acad. Sci. USA 102, 5981–5986 (2005). An outstanding paper that describes the crystal structure of PSMA (GCPII) peptidase.

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Subasinghe, N. et al. Synthesis of acyclic and dehydroaspartic acid analogues of Ac-Asp-Glu-OH and their inhibition of rat brain N-acetylated-linked acidic dipeptidase (NAALA dipeptidase). J. Med. Chem. 33, 2734–2744 (1990).

    CAS  PubMed  Google Scholar 

  38. Serval, V., Galli, T., Cheramy, A., Glowinski, J. & Lavielle, S. In vitro and in vivo inhibition of N-acetyl-L-aspartyl-L-glutamate catabolism by N-acylated-L-glutamate analogs. J. Pharmacol. Exp. Ther. 260, 1093–1100 (1992).

    CAS  PubMed  Google Scholar 

  39. Jackson, P. F. & Slusher, B. S. Design of NAALADase inhibitors: a novel neuroprotective strategy. Curr. Med. Chem. 8, 949–957 (2001). An important and early review describing NAAG peptidase inhibitors of phosphinic acid derivatives.

    CAS  PubMed  Google Scholar 

  40. Bennett, B. & Holz, R. C. Inhibition of the aminopeptidase from Aeromonas Proteolytica by L-leucinephosphonic acid, a transition state analogue of peptide hydrolysis. J. Am. Chem. Soc. 120, 12139–12140 (1998).

    CAS  Google Scholar 

  41. Jackson, P. F. et al. Design and pharmacological activity of phosphinic acid based NAALADase inhibitors. J. Med. Chem. 44, 4170–4175 (2001).

    CAS  PubMed  Google Scholar 

  42. Tsukamoto, T. et al. Phosphonate and phosphinate analogues of N-acylated-glutamylglutamate: Potent inhibitors of glutamate carboxypeptidase II. Bioorg. Med. Chem. Lett. 12, 2189–2192 (2002).

    CAS  PubMed  Google Scholar 

  43. Tang, H. et al. Prostate targeting ligands based on N-acetylated α-linked acidic dipeptidase. Biochem. Biophys. Res. Commun. 307, 8–14 (2003).

    CAS  PubMed  Google Scholar 

  44. Oliver, A. J., Wiest, O., Helquist, P., Miller, M. J. & Tenniswood, M. Conformational and SAR analysis of NAALADase and PSMA inhibitors. Bioorg. Med. Chem. 11, 4455–4461 (2003).

    CAS  PubMed  Google Scholar 

  45. Maung, J. et al. Probing for a hydrophobic a binding register in prostate-speci. c membrane antigen with phenylalkylphosphonamidates. Bioorg. Med. Chem. 12, 4969–4979 (2004).

    CAS  PubMed  Google Scholar 

  46. Tsukamoto, T. et al. Enantiospecificity of glutamate carboxypeptidase II inhibition. J. Med. Chem. 48, 2319–2324 (2005).

    CAS  PubMed  Google Scholar 

  47. Vitharana, D. et al. Synthesis and biological evaluation of (R)- and (S)-2-(phosphonomethyl)pentanedioic acids as inhibitors of glutamate carboxypeptidase II. Tetrahedron: Asymm. 13, 1609–1614 (2002).

    CAS  Google Scholar 

  48. Majer, P. et al. Synthesis and biological evaluation of thiol-based inhibitors of glutamate carboxypeptidase II: Discovery of an orally active GCP II inhibitor. J. Med. Chem. 46, 1989–1996 (2003).

    CAS  PubMed  Google Scholar 

  49. Tsukamoto, T., Grella, B. & Majer, P. Indoles as NAALADase inhibitors. US patent 2005/0080128A1. April 14, 2005.

  50. Stoermer, D. et al. Synthesis and biological evaluation of hydroxamate-based Inhibitors of glutamate Carboxypeptidase II. Bioorg. Med. Chem. Lett. 13, 2097–2100 (2003).

    CAS  PubMed  Google Scholar 

  51. Ding, P. et al. Syntheses of conformationally constricted molecules as potential NAALADase/PSMA inhibitors. Org. Lett. 6, 1805–1808 (2004).

    CAS  PubMed  Google Scholar 

  52. Maison, W., Grohs, D. C. & Prenzel, A. H. G. P. Efficient synthesis of structurally diverse diazabicycloalkanes: scaffolds for modular dipeptide mimetics with tunable backbone conformations. Eur. J. Org. Chem. 17, 1527–1543 (2004).

    Google Scholar 

  53. Grohs, D. C. & Maison, W. Synthesis of modular dipeptide mimetics on the basis of diazabicycloalkanes and derivatives thereof with sulphur containing side chains. Amino Acids 29, 131–138 (2005).

    CAS  PubMed  Google Scholar 

  54. Nan, F. et al. Dual function glutamate-related ligands: Discovery of a novel, potent inhibitor of glutamate carboxypeptidase II possessing mGluR3 agonist activity. J. Med. Chem. 43, 772–774 (2000).

    CAS  PubMed  Google Scholar 

  55. Kozikowski, A. P. et al. Design of remarkably simple, yet potent urea-based inhibitors of glutamate carboxypeptidase II (NAALADase). J. Med. Chem. 44, 298–301 (2001).

    CAS  PubMed  Google Scholar 

  56. Kozikowski, A. P. et al. Synthesis of urea-based inhibitors as active site probes of glutamate carboxypeptidase II: efficacy as analgesic agents. J. Med. Chem. 47, 1729–1738 (2004).

    CAS  PubMed  Google Scholar 

  57. 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. 909, 138–144 (2001).

    CAS  PubMed  Google Scholar 

  58. Yamamoto, T., Nozaki-Taguchi, N., Sakashita, Y. & Inagaki, T. Inhibition of spinal N-acetylated-α-linked acidic dipeptidase produces an antinociceptive effect in the rat formalin test. Neurosci. 102, 473–479 (2001).

    CAS  Google Scholar 

  59. Chen, S. R., Wozniak, K. M., Slusher, B. S. & Pan, H. L. Effect of 2-(phosphonomethyl)-pentanedioic acid on allodynia and afferent ectopic discharges in a rat model of neuropathic pain. J. Pharmacol. Exp. Ther. 300, 662–667 (2002).

    CAS  PubMed  Google Scholar 

  60. Carpenter, K. J. et al. Effects of GCP-II inhibition on responses of dorsal horn neurones after inflammation and neuropathy: an electrophysiological study in the rat. Neuropeptides 37, 298–306 (2003).

    CAS  PubMed  Google Scholar 

  61. Yamamoto, T. et al. 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. 20, 483–494 (2004).

    PubMed  Google Scholar 

  62. Losi, G., Vicini, S. & Neale, J. NAAG fails to antagonize synaptic and extrasynaptic NMDA receptors in cerebellar granule neurons. Neuropharmacology 46, 490–496 (2004).

    CAS  PubMed  Google Scholar 

  63. Bergeron, R., Coyle, J. T., Tsai, G. & Greene, R. W. NAAG reduces NMDA receptor current in CA1 hippocampal pyramidal neurons of acute slices and dissociated neurons. Neuropsychopharmacology 30, 7–16 (2005).

    CAS  PubMed  Google Scholar 

  64. Narayan, R. K. &, Michael, M. E. The clinical trials in head injury study group. Clinical trials in head injury. J. Neurotrauma 19, 503–557 (2002).

    PubMed  Google Scholar 

  65. Murray, C. J. L. & Lopez, A. D. Global mortality, disability and the contribution of the risk factors: global burden of disease study. Lancet 349, 1436–1442 (1997).

    CAS  PubMed  Google Scholar 

  66. Tolias, C. M. & Bullock, M. R. Critical appraisal of neuroprotection trials in head injury: what have we learned? J. Am. Soc. Exp. NeuroTherapeutics 1, 71–79 (2004). An outstanding review that describes the recent development of drug discovery in head injury.

    Google Scholar 

  67. Faden, A. I., Demediuk, P., Panter, S. S. & Vink, R. The role of excitatory amino acids and NMDA receptors in traumatic brain injury. Science 244, 798–800 (1989).

    CAS  PubMed  Google Scholar 

  68. Katayama, Y., Becker, D. P., Tamura, T. & Hovda, D. A. Massive increases in extracellular potassium and the indiscriminate release of glutamate following concussive brain injury. J. Neurosurg. 73, 889–900 (1990).

    CAS  PubMed  Google Scholar 

  69. Globus, M. Y., Alonso, O., Dietrich, W. D., Busto, R. & Ginsberg, M. D. Glutamate release and free radical production following brain injury. J. Neurochem. 65, 1704–1711 (1995).

    CAS  PubMed  Google Scholar 

  70. Bullock, R., Lyeth, B. G. & Muizelaar, J. P. Current status of neuroprotection trials for traumatic brain injury: lessons from animal models and clinical studies. Neurosurgery 45, 207–217 (1999).

    CAS  PubMed  Google Scholar 

  71. Zhong, C. et al. NAAG peptidase inhibitor reduces acute neuronal degeneration and astrocyte damage following lateral fluid percussion TBI in rats. J. Neurotrauma 22, 266–276 (2005).

    PubMed  Google Scholar 

  72. Xiong, Z. et al. Neuroprotection in ischemia: Blocking calcium-permeable acid-sensing ion channels. Cell 118, 687–698 (2004).

    CAS  PubMed  Google Scholar 

  73. Martin, L. et al. Neurodegeneration in excitotoxicity, global cerebral ischemia, and target deprivation: a perspective on the contributions of apoptosis and necrosis. Brain Res. Bull. 46, 281–309 (1998).

    CAS  PubMed  Google Scholar 

  74. Muir, K. & Grosset, D. Neuroprotection for acute stroke: making clinical trials work. Stroke 30, 180–182 (1999).

    CAS  PubMed  Google Scholar 

  75. Williams, A. J., Lu, X. M., Slusher, B. & Tortella, F. C. Electroencephalogram analysis and neuroprotective profile of the N-acetylated-α-linked acidic dipeptidase inhibitor, GPI5232, in normal and brain-injured rats. J. Pharmacol. Exp. Ther. 299, 48–57 (2001).

    CAS  PubMed  Google Scholar 

  76. Cai, Z., Lin, S. & Rhodes, P. G. Neuroprotective effects of N-acetylaspartylglutamate in a neonatal rat model of hypoxia-ischemia. Eur. J. Pharmacol. 437, 139–45 (2002).

    CAS  PubMed  Google Scholar 

  77. Carlsson, A. The current status of the dopamine hypothesis of schizophrenia. Neuropsychopharmacology 1, 179–186 (1988).

    CAS  PubMed  Google Scholar 

  78. Tsai, G. et al. Abnormal excitatory neurotransmitter metabolism in schizophrenic brains. Arch. Gen. Psychiatry 52, 829–836 (1995).

    CAS  PubMed  Google Scholar 

  79. Flores, C. & Coyle, J. T. Regulation of glutamate carboxypeptidase II function in corticolimbic regions of rat brain by phencyclidine, haloperidol, and clozapine. Neuropsychopharmacology 28, 1227–1234 (2003).

    CAS  PubMed  Google Scholar 

  80. Tsai, G. & Coyle, J. T. Glutamatergic mechanisms in schizophrenia. Annu. Rev. Pharmacol. Toxicol. 42, 165–179 (2002).

    CAS  PubMed  Google Scholar 

  81. Marek, G. J. Glutamate and schizophrenia: pathophysiology and therapeutics. Curr. Med. Chem.-Central Nervous System Agents 2, 29–44 (2002). Excellent review that describes the involvement of glutamatergic neurotransmission in schizophrenia as well as the relevant therapeutics.

    CAS  Google Scholar 

  82. McCarley, R. W. et al. Cognitive dysfunction in schizophrenia: unifying basic research and clinical aspects. Eur. Arch. Psychiatry Clin. Neurosci. 249, (Suppl. 4), 69–82 (1999).

    PubMed  PubMed Central  Google Scholar 

  83. Johnstone, M., Evans, V., Baigel, S. Sernyl (Cl-395) in clinical anaesthesia. J. Anaesth. 31, 433–439 (1959).

    CAS  Google Scholar 

  84. Luby, E. D., Cohen, B. D., Rosenbaum, F., Gottlieb, J. & Kelley, R. Study of a new schizophrenomimetic drug, Sernyl. Arch. Neurol. Psychiatry 81, 363–369 (1959).

    CAS  Google Scholar 

  85. Javitt, D. C. & Zukin, S. R. Recent advances in the phencyclidine model of schizophrenia. Am. J. Psychiatry 148, 1301–1308 (1991).

    CAS  PubMed  Google Scholar 

  86. Krystal, J. H. et al. NMDA agonists and antagonists as probes of glutamatergic dysfunction and pharmacotherapies in neuropsychiatric disorders. Harv. Rev. Psychiatry 7, 125–143 (1999).

    CAS  PubMed  Google Scholar 

  87. Moghaddam, B. & Adams, B. W. Reversal of phencyclidine effects by a group II metabotropic glutamate receptor agonist in rats. Science 281, 1349–1352 (1998).

    CAS  PubMed  Google Scholar 

  88. Cartmell, J., Monn, J. A. & Schoepp, D. D. The metabotropic glutamate 2/3 receptor agonists LY354740 and LY379268 selectively attenuate phencyclidine versus d-amphetamine motor behaviors in rats. J. Pharmacol. Exp. Ther. 291, 161–170 (1999).

    CAS  PubMed  Google Scholar 

  89. Olszewski, R. T. et al. NAAG peptidase inhibition reduces locomotor activity and some stereotypes in the PCP model of schizophrenia via group II mGluR. J. Neurochem. 89, 876–885 (2004).

    CAS  PubMed  Google Scholar 

  90. Sima, A. A. F. Diabetic neuropathy: pathogenetic background, current and future therapies. Expert Rev. Neurotherapeutics 1, 225–238 (2001).

    CAS  Google Scholar 

  91. Berent-Spillson, A. et al. Protection against glucose-induced neuronal death by NAAG and GCP II inhibition is regulated by mGluR3. J. Neurochem. 89, 90–99 (2004).

    CAS  PubMed  Google Scholar 

  92. Rosson, G. D., Dellon, A. L. Vascular risk factors and diabetic neuropathy. N. Engl. J. Med. 352, 1925–1927 (2005).

    CAS  PubMed  Google Scholar 

  93. Cameron, N. E. & Cotter, M. A. in Chronic Complications in Diabetes. Oxidative Stress and Abnormal Lipid Metabolism in Diabetic Complication. Frontiers in Animal Diabetes Research vol. 1 (ed. Sima, A. A. F.) 97–130 (Harwood, Amsterdam, 2000).

    Google Scholar 

  94. Coppey, L. J., Davidson, E. P., Dunlap, J. A., Lund, D. D. & Yorek, M. A. Slowing of motor nerve conduction velocity in streptozotocin-induced diabetic rats is preceded by impaired vasodilatation in arterioles that overlie the sciatic nerve. Int. J. Exp. Diabetes Res. 1, 131–143 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Srinivasan, S., Stevens, M. & Wiley, J. W. Diabetic peripheral neuropathy: evidence for apoptosis and associated mitochondrial dysfunction. Diabetes 49, 1932–1938 (2000).

    CAS  PubMed  Google Scholar 

  96. Tomiyama, M. et al. 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. 136, 275–281 (2005).

    CAS  PubMed  Google Scholar 

  97. Zhang, W. et al. GCPII (NAALADase) inhibition prevents long-term diabetic neuropathy in type 1 diabetic BB/Wor rats. J. Neurological Sci. 194, 21–28 (2002).

    CAS  Google Scholar 

  98. National Institute of Neurological Disorders and Stroke. Neuroscience news [online] <http://www.ninds.nih.gov/index.htm> (2005).

  99. Cleveland, D. W. & Rothstein, J. D. From Charcot to Lou Gehrig: deciphering selective motor neuron death in ALS. Nature Rev. Neurosci. 2, 806–819 (2001).

    CAS  Google Scholar 

  100. Cluskey, S. & Ramsden, D. B. Mechanisms of neurodegeneration in amyotrophic lateral sclerosis. Mol. Pathol. 54, 386–392 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Bruneteau, G., Demeret, S. & Meininger, V. Physiopathology of ALS: therapeutic approach. Rev. Neurol. (Paris) 160, 235–241 (2004).

    CAS  Google Scholar 

  102. McGeer, E. G., & McGeer, P. L. Pharmacologic approaches to the treatment of amyotrophic lateral sclerosis. BioDrugs 19, 31–37 (2005).

    CAS  PubMed  Google Scholar 

  103. Kwak, S. & Kawahara, Y. Deficient RNA editing of GluR2 and neuronal death in amyotropic lateral sclerosis. J. Mol. Med. 83, 110–120 (2005).

    CAS  PubMed  Google Scholar 

  104. Rembach, A. et al. Antisense peptide nucleic acid targeting GluR3 delays disease onset and progression in the SOD1 G93A mouse model of familial ALS. J. Neurosci. Res. 77, 573–582 (2004).

    CAS  PubMed  Google Scholar 

  105. Vanoni, C. et al. Increased internalisation and degradation of GLT-1 glial glutamate transporter in a cell model for familial amyotrophic lateral sclerosis (ALS). J. Cell Sci. 117, 5417–5426 (2004).

    CAS  PubMed  Google Scholar 

  106. Iwasaki, Y., Ikeda, K. & Kinoshita, M. Molecular and cellular mechanism of glutamate receptors in relation to amyotrophic lateral sclerosis. Curr. Drug Targets 1, 511–518 (2002).

    CAS  Google Scholar 

  107. Ghadge, G. D. et al. Glutamate carboxypeptidase II inhibition protects motor neurons from death in familial amyotrophic lateral sclerosis models. Proc. Natl Acad. Sci. USA 100, 9554–9559 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Zhou, J. et al. Biaryl analogues of conformationally constrained tricyclic tropanes as potent and selective norepinephrine reuptake inhibitors: Synthesis and evaluation of their uptake inhibition at monoamine transporter sites. J. Med. Chem. 46, 1997–2007 (2003).

    CAS  PubMed  Google Scholar 

  109. Kalivas, P. W. Glutamate systems in cocaine addiction. Curr. Opin. Pharmacol. 4, 23–29 (2004).

    CAS  PubMed  Google Scholar 

  110. Kalivas, P. W. et al. Glutamate transmission and addiction to cocaine. Ann. N. Y. Acad. Sci. 1003, 169–175 (2003).

    CAS  PubMed  Google Scholar 

  111. Baker, D. A. et al. Neuroadaptations in cystine-glutamate exchange underlie cocaine relapse. Nature Neurosci. 6, 743–749 (2003).

    CAS  PubMed  Google Scholar 

  112. Sutton, M. A. et al. Extinction-induced upregulation in AMPA receptors reduces cocaine seeking behaviour. Nature 421, 70–75 (2003).

    CAS  PubMed  Google Scholar 

  113. Robbe, D., Bockaert, J. & Manzoni, O. J. Metabotropic glutamate receptor 2/3-dependent long-term depression in the nucleus accumbens is blocked in morphine withdrawn mice. Eur. J. Neurosci. 16, 2231–2235 (2002).

    PubMed  Google Scholar 

  114. Vekovischeva, O. Y. et al. Morphine-induced dependence and sensitization are altered in mice deficient in AMPA-type glutamate receptor-A subunits. J. Neurosci. 21, 4451–4459 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Watkins, S. S., Koob, G. F. & Markou, A. Neural mechanisms underlying nicotine addiction: acute positive reinforcement and withdrawal. Nicotine Tob. Res. 2, 19–37 (2000).

    CAS  PubMed  Google Scholar 

  116. De Witte, P. Imbalance between neuroexcitatory and neuroinhibitory amino acids causes craving for ethanol. Addict. Behav. 29, 1325–1339 (2004).

    PubMed  Google Scholar 

  117. Spanagel, R. The role of the glutamatergic system in alcohol addiction. Fortschr. Neurol. Psychiatr. 71, (Suppl 1), S33–S35 (2003).

    PubMed  Google Scholar 

  118. Tessari, M., Pilla, M., Andreoli, M., Hutcheson, D. M. & Heidbreder, C. A. Antagonism at metabotropic glutamate 5 receptors inhibits nicotine- and cocaine-taking behaviours and prevents nicotine-triggered relapse to nicotine-seeking. Eur. J. Pharmacol. 499, 121–133 (2004).

    CAS  PubMed  Google Scholar 

  119. Kenny, P. J. & Markou, A. The ups and downs of addiction: role of metabotropic glutamate receptors. Trends Pharmacol. Sci. 25, 265–272 (2004).

    CAS  PubMed  Google Scholar 

  120. Lovinger, D. M., Partridge, J. G. & Tang, K. C. Plastic control of striatal glutamatergic transmission by ensemble actions of several neurotransmitters and targets for drugs of abuse. Ann. N. Y. Acad. Sci. 1003, 226–240 (2003).

    CAS  PubMed  Google Scholar 

  121. Tzschentke, T. M. & Schmidt, W. J. Glutamatergic mechanisms in addiction. Mol. Psychiatr. 8, 373–382 (2003).

    CAS  Google Scholar 

  122. Heidbreder, C. Recent advances in the pharmacotherapeutic management of drug dependence and addiction. Curr. Psychiatry Rev. 1, 45–67 (2005). Outstanding review that summarizes the most promising therapeutic strategies in the field of drug abuse.

    CAS  Google Scholar 

  123. Slusher, B. S., Thomas, A., Paul, M., Schad, C. A. & Ashby, Jr., C. R. Expression and acquisition of the conditioned place preference response to cocaine in rats is blocked by selective inhibitors of the enzyme N-acetylated-α-linked-acidic dipeptidase (NAALADase). Synapse 41, 22–28 (2001).

    CAS  PubMed  Google Scholar 

  124. Witkin, J. M. et al. NAALADase (GCP II) inhibition prevents cocaine-kindled seizures. Neuropharmacology 43, 348–356 (2002).

    CAS  PubMed  Google Scholar 

  125. Popik, P., Kozela, E., Wrobel, M., Wozniak, K. M. & Slusher, B. S. Morphine tolerance and reward but not expression of morphine dependence are inhibited by the selective glutamate carboxypeptidase II (GCP II, NAALADase) inhibitor, 2-PMPA. Neuropsychopharmacology 28, 457–467 (2003).

    CAS  PubMed  Google Scholar 

  126. Mckinzie, D. L., Li, T. -K., Mcbride, W. J. & Slusher, B. S. NAALADase inhibition reduces alcohol consumption in the alcohol-preferring (P) line of rats. Addict. Biol. 5, 411–416 (2000).

    CAS  PubMed  Google Scholar 

  127. Senkowska, A. & Ossowska, K. Role of metabotropic glutamate receptors in animal models of Parkinson's disease. Pol. J. Pharmacol. 55, 935–950 (2003).

    CAS  PubMed  Google Scholar 

  128. Kieburtz, K. Antiglutamate therapies in Huntington's disease. J. Neural Transm. Suppl. 55, 97–102 (1999).

    CAS  PubMed  Google Scholar 

  129. Hynd, M. R., Scott, H. L. & Dodd, P. R. Glutamate-mediated excitotoxicity and neurodegeneration in Alzheimer's disease. Neurochem. Int. 45, 583–595 (2004).

    CAS  PubMed  Google Scholar 

  130. Lee Q. et al. Aberrant expression of metabotropic glutamate receptor 2 in the vulnerable neurons of Alzheimer's disease. Acta. Neuropathol. (Berl). 107, 365–371 (2004).

    CAS  Google Scholar 

  131. Jaarsma, D., Veenma-van der Duin, L. & Korf, J. N-Acetylaspartate and N-acetylaspartylglutamate levels in Alzheimer's disease post-mortem brain tissue. J. Neurol. Sci. 127, 230–233 (1994).

    CAS  PubMed  Google Scholar 

  132. Passani, L. A., Vonsattel, J. P. & Coyle, J. T. Distribution of N-acetylaspartylglutamate immunoreactivity in human brain and its alteration in neurodegenerative disease. Brain Res. 772, 9–22 (1997).

    CAS  PubMed  Google Scholar 

  133. Passani, L. A., Vonsattel, J. P., Carter, R. E. & Coyle, J. T. N-Acetylaspartylglutamate, N-acetylaspartate, and N-acetylated α-linked acidic dipeptidase in human brain and their alterations in Huntington and Alzheimer's diseases. Mol. Chem. Neuropathol. 31, 97–118 (1997).

    CAS  PubMed  Google Scholar 

  134. Jemal, A., Thomas, A., Murray, T. & Thun, M. Cancer statistics. CA Cancer J. Clin. 52, 23–47 (2002).

    PubMed  Google Scholar 

  135. Leach, F. Targeting prostate-specific membrane antigen in cancer therapy: can molecular medicine be brought to the surface? Cancer Biol. Ther. 3, 559–560 (2004).

    PubMed  Google Scholar 

  136. Chang, S. S. et al. Prostate-specific membrane antigen is produced in tumor-associated neovasculature. Clin. Cancer Res. 5, 2674–2681 (1999).

    CAS  PubMed  Google Scholar 

  137. Chang, S. S. et al. Five different anti-prostate-specific membrane antigen (PSMA) antibodies confirm PSMA expression in tumor-associated neovasculature. Cancer Res. 59, 3192–3198 (1999).

    CAS  PubMed  Google Scholar 

  138. Chang, S. S. Monoclonal antibodies and prostate specific membrane antigen. Curr. Opin. Investig. Drugs 5, 611–615 (2004).

    CAS  PubMed  Google Scholar 

  139. Ghosh, A. & Heston, W. D. Tumor target prostate specific membrane antigen (PSMA) and its regulation in prostate cancer. J. Cell Biochem. 91, 528–539 (2004).

    CAS  PubMed  Google Scholar 

  140. Gong, M. C. et al. Overview of evolving strategies incorporating prostate-specific membrane antigen as target for therapy. Mol. Urol. 4, 217–222 (2000).

    CAS  PubMed  Google Scholar 

  141. Chang, S. S. & Heston, W. D. The clinical role of prostate-specific membrane antigen (PSMA). Urol. Oncol. 7, 7–12 (2002).

    CAS  PubMed  Google Scholar 

  142. She, Y. et al. 2-MPPA, a selective inhibitor of PSMA, delays prostate cancer growth and attenuates taxol-induced neuropathy in mice. Am. Soc. Clin. Oncol. Annu. Mtg Alexandria, Virginia May 13–17 (2005).

  143. Milowsky, M. I. et al. Phase I trial of yttrium-90-labeled anti-prostate-specific membrane antigen monoclonal antibody J591 for androgen-independent prostate cancer. J. Clin. Oncol. 22, 2522–2531 (2004).

    CAS  PubMed  Google Scholar 

  144. Nargund, V. et al. Imaging with radiolabelled monoclonal antibody (MUJ591) to prostate-specific membrane antigen in staging of clinically localized prostatic carcinoma: comparison with clinical, surgical and histological staging. BJU Int. 95, 1232–1236 (2005).

    PubMed  Google Scholar 

  145. Pomper, M. G. et al. 11C-MCG: Synthesis, uptake selectivity, and primate PET of a probe for glutamate carboxypeptidase II (NAALADase). Mol. Imaging 1, 96–101 (2002).

    CAS  PubMed  Google Scholar 

  146. Foss, C. A. et al. Radiolabeled small-molecule ligands for prostate-specific membrane antigen: In vivo imaging in experimental models of prostate cancer. Clin. Cancer Res. 11, 4022–4028 (2005). A key paper that describes NAAG peptidase inhibitors as promising PET/SPECT imaging ligands for prostate cancer.

    CAS  PubMed  Google Scholar 

  147. Tasch, J., Gong, M., Sadelain, M. & Heston, W. D. A unique folate hydrolase, prostate-specific membrane antigen (PSMA): a target for immunotherapy? Crit. Rev. Immunol. 21, 249–261 (2001).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank W. Tueckmantel of Acenta Discovery, Inc. for helpful discussions. This work was supported by the National Institutes of Health (NIH), including a National Institute of Mental Health grant, National Institute of Neurological Disorders and Stroke grants and a National Cancer Institute grant.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jia Zhou.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

Related links

DATABASES

Entrez Gene

GCPII

GCPIII

mGlu2

mGlu3

SOD1

OMIM

Alzheimer's disease

amyotrophic lateral sclerosis

epilepsy

Huntington's disease

multiple sclerosis

Parkinson's disease

prostate cancer

schizophrenia

stroke

FURTHER INFORMATION

ALS Association

American Stroke Association

Glossary

NEUROPROTECTIVE

An agent that reduces nerve cell death, particularly that resulting from excess glutamate release.

ISCHAEMIA

Loss of blood flow to a tissue; in this review we primarily consider loss of blood flow to the nervous system.

IONOTROPIC

Receptors that are also ion channels.

METABOTROPIC

Receptors that are not ion channels but rather are coupled to second-messenger cascades.

GLUTAMATERGIC

Neurons using glutamate as one of their neurotransmitters.

HYPERALGESIA

An increase in pain perception above the normal response to a stimulus.

PSYCHOMIMETIC

An agent that induces a psychotic-like state.

NEURITE

An extension of a nerve cell that is used to communicate information back to the body of the cell.

SUPEROXIDE DISMUTASE

An enzyme that mediates the conversion of toxic superoxide radicals to peroxide and then to oxygen and water.

PROSTASCINT

A monoclonal antibody to the prostate-specific membrane antigen labeled with indium-111 for the detection of prostate cancer.

POSITRON-EMISSION TOMOGRAPHY

A noninvasive, molecular imaging technique of high sensitivity that detects species labelled with positron-emitting radionuclides in vivo.

DRUGGABILITY

The suitability of a lead candidate that has the requisite physico-chemical/absorption, distribution, metabolism and excretion properties for development as a drug candidate.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Zhou, J., Neale, J., Pomper, M. et al. NAAG peptidase inhibitors and their potential for diagnosis and therapy. Nat Rev Drug Discov 4, 1015–1026 (2005). https://doi.org/10.1038/nrd1903

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrd1903

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing