Glutamate carboxypeptidase II

Jump to navigation Jump to search
VALUE_ERROR (nil)
Identifiers
Aliases
External IDsGeneCards: [1]
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

n/a

n/a

RefSeq (protein)

n/a

n/a

Location (UCSC)n/an/a
PubMed searchn/an/a
Wikidata
View/Edit Human
glutamate carboxypeptidase II
File:Gcp2-naag.svg
Reaction Scheme of NAAG Degradation by GCPII: GCPII + NAAG → GCPII-NAAG complex → Glutamate + NAA
Identifiers
EC number3.4.17.21
CAS number111070-04-3
Databases
IntEnzIntEnz view
BRENDABRENDA entry
ExPASyNiceZyme view
KEGGKEGG entry
MetaCycmetabolic pathway
PRIAMprofile
PDB structuresRCSB PDB PDBe PDBsum
Gene OntologyAmiGO / QuickGO

Glutamate carboxypeptidase II (GCPII), also known as N-acetyl-L-aspartyl-L-glutamate peptidase I (NAALADase I), NAAG peptidase, or prostate-specific membrane antigen (PSMA) is an enzyme that in humans is encoded by the FOLH1 (folate hydrolase 1) gene.[1] Human GCPII contains 750 amino acids and weighs approximately 84 kDa.[2]

GCPII is a zinc metalloenzyme that resides in membranes. Most of the enzyme resides in the extracellular space. GCPII is a class II membrane glycoprotein. It catalyzes the hydrolysis of N-acetylaspartylglutamate (NAAG) to glutamate and N-acetylaspartate (NAA) according to the reaction scheme to the right.[3][4]

Neuroscientists primarily use the term NAALADase in their studies, while those studying folate metabolism use folate hydrolase, and those studying prostate cancer or oncology, PSMA. All of which refer to the same protein glutamate carboxypeptidase II.

Discovery

GCPII is mainly expressed in four tissues of the body, including prostate epithelium, the proximal tubules of the kidney, the jejunal brush border of the small intestine and ganglia of the nervous system.[2][5][6]

Indeed, the initial cloning of the cDNA encoding the gene expressing PSMA was accomplished with RNA from a prostate tumor cell line, LNCaP.[7] PSMA shares homology with the transferrin receptor and undergoes endocytosis but the ligand for inducing internalization has not been identified.[8] It was found that PSMA was the same as the membrane protein in the small intestine responsible for removal of gamma-linked glutamates from polygammaglutamate folate. This enables the freeing of folic acid, which then can be transported into the body for use as a vitamin. This resulted in the cloned genomic designation of PSMA as FOLH1 for folate hydrolase.[9]

PSMA(FOLH1)+ folate polygammaglutamate(n 1-7)---> PSMA (FOLH1) + folate(poly)gammaglutamate(n-1) + glutamate continuing until releasing folate.

Structure

The three domains of the extracellular portion of GCPII—the protease, apical and C-terminal domains—collaborate in substrate recognition.[4] The protease domain is a central seven-stranded mixed β-sheet. The β-sheet is flanked by 10 α-helices. The apical domain is located between the first and the second strands of the central β-sheet of the protease domain. The apical domain creates a pocket that facilitates substrate binding. The C-terminal domain is an Up-Down-Up-Down four-helix bundle.

The central pocket is approximately 2 nanometers in depth and opens from the extracellular space to the active site.[4] This active site contains two zinc ions. During inhibition, each acts as a ligand to an oxygen in 2-PMPA or phosphate. There is also one calcium ion coordinated in GCPII, far from the active site. It has been proposed that calcium holds together the protease and apical domains.[4] In addition, human GCPII has ten sites of potential glycosylation, and many of these sites (including some far from the catalytic domain) affect the ability of GCPII to hydrolyze NAAG.[2]

The FOLH1 gene has multiple potential start sites and splice forms, giving rise to differences in membrane protein structure, localization, and carboxypeptidase activity based on the parent tissue.[2][10]

Enzyme kinetics

The hydrolysis of NAAG by GCPII obeys Michaelis-Menten kinetics calculated the binding constant (Km) for NAAG as approximately 130 nM and the turnover constant (kcat) as approximately 4 s−1.[4] The apparent second-order rate constant is approximately 3 x 107 (M·s)−1.

Role in cancer

Human PSMA is highly expressed in the prostate, roughly a hundred times greater than in most other tissues. In some prostate cancers, PSMA is the second-most upregulated gene product, with an 8- to 12-fold increase over levels in noncancerous prostate cells.[11] Because of this high expression, PSMA is being developed as potential biomarker for therapy and imaging of some cancers.[12] In human prostate cancer, the higher expressing tumors are associated with quicker time to progression and a greater percentage of patients suffering relapse.[13][14] In vitro studies using prostate and breast cancer cell lines with decreased PSMA levels showed a significant decrease in the proliferation, migration, invasion, adhesion and survival of the cells.[15]

PSMA is the target of several nuclear medicine imaging agents for prostate cancer. Capromabpentide (marketed as PROSTASCINT) is bound to indium-111 for detection by a gamma camera.[16] Second-generation antibodies and low-molecular-weight ligands for imaging and therapy are also being developed.[12][17][18][19][20][21][22] PSMA expression can also be imaged with a gallium-68 PSMA scan for positron emission tomography.[23][24] In addition to nuclear medicine imaging, PSMA can also be used experimentally to target treatment. Lutetium-177 is a beta emitter, bound to PSMA to deliver treat prostate tumours.[25]

In addition to the expression in the human prostate and prostate cancer, PSMA is also found to be highly expressed in tumor neovasculature but not corresponding normal vasculature of all types of solid tumors as kidney, breast, colon, etc.[26] In terms of imaging, no non-tumor site such as normal kidney, small intestine, or CNS has been imaged using second-generation antibody imaging agents, while sites even in bone are being detected with better sensitivity than with technetium scans, and the tumors expressing PSMA in their neovasculature are also being imaged.[18] Low-molecular-weight ligands exhibit different binding with imaging seen in the kidney of the mouse, however mouse has much higher levels of PSMA in kidney and brain than the human. In the mouse, it was only the normal kidney and prostate tumors that were imaged, and not other tissues, not even CNS suggesting the imprortance of the blood–brain barrier.[17] In kidney, it is a subset of tubules that contain PSMA. Thus, imaging studies will have to be performed in humans with the low-molecular-weight ligands to define their potential for imaging and targeting. Still, in terms of potential toxicities, knockout animals were normal on most tests, which reduces somewhat concerns about toxicity in targeting PSMA.[27] In the CNS, PSMA is present only in a sub-set of glial cells, again suggesting that toxin targeting would likely have minimal toxicity to the host even if the blood–brain barrier were not intact.[28]

Neurotransmitter degradation

For those studying neural based diseases, NAAG is one of the three most prevalent neurotransmitters found in the central nervous system[29] and when it catylizes the reaction to produce glutamate it is also producing another neurotransmitter.[4] Glutamate is a common and abundant excitatory neurotransmitter in the central nervous system; however, if there is too much glutamate transmission, this can kill or at least damage neurons and has been implicated in many neurological diseases and disorders[29] therefore the balance that NAAG peptidase contributes to is quite important.

Potential therapeutic applications

Function in the brain

GCPII has been shown to both indirectly and directly increase the concentration of glutamate in the extracellular space.[29] GCPII directly cleaves NAAG into NAA and glutamate.[3][4] NAAG has been shown, in high concentration, to indirectly inhibit the release of neutrotransmitters, such as GABA and glutamate. It does this through interaction with and activation of presynaptic group II mGluRs.[29] Thus, in the presence of NAAG peptidase, the concentration of NAAG is kept in check, and glutamate and GABA, among other neurotransmitters, are not inhibited.

Researchers have been able to show that effective and selective GCPII inhibitors are able to decrease the brain's levels of glutamate and even provide protection from apoptosis or degradation of brain neurons in many animal models of stroke, amyotrophic lateral sclerosis, and neuropathic pain.[4] This inhibition of these NAAG peptidases, sometimes referred to as NPs, are thought to provide this protection from apoptosis or degradation of brain neurons by elevating the concentrations of NAAG within the synapse of neurons.[29] NAAG then reduces the release of glutamate while stimulating the release of some trophic factors from the glia cells in the central nervous system, resulting in the protection from apoptosis or degradation of brain neurons.[29] It is important to note, however, that these NP inhibitors do not seem to have any effect on normal glutamate function.[29] The NP inhibition is able to improve the naturally occurring regulation instead of activating or inhibiting receptors that would disrupt this process.[29] Research has also shown that small-molecule-based NP inhibitors are beneficial in animal models that are relevant to neurodegenerative diseases.[29] Some specific applications of this research include neuropathic and inflammatory pain, traumatic brain injury, ischemic stroke, schizophrenia, diabetic neuropathy, amyotrophic lateral sclerosis, as well as drug addiction.[29] Previous research has found that drugs that are able to reduce glutamate transmission can relieve the neuropathic pain, although the resultant side-effects have limited a great deal of their clinical applications.[30] Therefore, it appears that, since GCPII is exclusively recruited for the purpose of providing a glutamate source in hyperglutamatergic and excitotoxic conditions, this could be an alternative to avert these side-effects.[30] More research findings have shown that the hydrolysis of NAAG is disrupted in schizophrenia, and they have shown that specific anatomical regions of the brain may even show discrete abnormalities in the GCP II synthesis, so NPs may also be therapeutic for patients suffering with schizophrenia.[31] One major hurdle with using many of the potent GCPII inhibitors that have been prepared to date are typically highly polar compounds, which causes problems because they do not then penetrate the blood–brain barrier easily.[32]

Potential uses of NAAG peptidase inhibitors

Glutamate is the “primary excitatory neurotransmitter in the human nervous system”,[29] participating in a multitude of brain functions. Over-stimulation and -activation of glutamate receptors as well as “disturbances in the cellular mechanisms that protect against the adverse consequences of physiological glutamate receptor activation”[32] have been known to cause neuron damage and death, which have been associated with multiple neurological diseases.[29]

Due to the range of glutamate function and presence, it has been difficult to create glutamatergic drugs that do not negatively affect other necessary functions and cause unwanted side-effects.[33] NAAG peptidase inhibition has offered the possibility for specific drug targeting.

Specific inhibitors

Since its promise for possible neurological disease therapy and specific drug targeting, NAAG peptidase inhibitors have been widely created and studied. A few small molecule examples are those that follow:[29]

2-PMPA and analogues
Thiol and indole thiol derivatives
Hydroxamate derivatives
Conformationally constricted dipeptide mimetics
PBDA- and urea-based inhibitors.
File:CommonInhibitors.gif
Structures of Two Common Inhibitors: 2-PMPA and 2-MPPA

Other potential therapeutic applications

Neuropathic and inflammatory pain

Pain cause by injury to CNS or PNS has been associated with increase glutamate concentration. NAAG inhibition reduced glutamate presence and could, thus, diminish pain.[29] (Neale JH et al., 2005). Nagel et al.[33] used the inhibitor 2-PMPA to show the analgesic effect of NAAG peptidase inhibitions. This study followed one by Chen et al.,[34] which showed similar results.[33]

Head injury

Severe head injury (SHI) and traumatic brain injury (TBI) are widespread and have a tremendous impact. “They are the leading cause of death in children and young adults (<25 years) and account for a quarter of all deaths in the five to 15 years age group”.[35] Following initial impact, glutamate levels rise and cause excitotoxic damage in a process that has been well characterized.[29] With its ability to reduce glutamate levels, NAAG inhibition has shown promise in preventing neurological damage associated with SHI and TBI.

Stroke

According to the National Stroke Association,[36] stroke is the third-leading cause of death and the leading cause of adult disability. It is thought that glutamate levels cause underlying ischemic damage during a stroke, and, thus, NAAG inhibition might be able to diminish this damage.[29]

Schizophrenia

Schizophrenia is a mental disorder that affects 1% of people throughout the world.[37] It can be modeled by PCP in laboratory animals, and it has been shown that mGluR agonists have reduced the effects of the drug. NAAG is, such, an mGluR agonist. Thus, inhibition of the enzyme that reduces NAAG concentration, NAAG peptidase, could provide a practical treatment for reduction of schizophrenic symptoms.[29]

Diabetic neuropathy

Diabetes can lead to damaged nerves, causing loss of sensation, pain, or, if autonomic nerves are associated, damage to the circulatory, reproductive, or digestive systems, among others. Over 60% of diabetic patients are said to have some form of neuropathy,[29] however the severity ranges dramatically. Neuropathy not only directly causes harm and damage but also can indirectly lead to such problems as diabetic ulcerations, which in turn can lead to amputations. In fact, over half of all lower limb amputations in the United States are of patients with diabetes.[38]

Through the use of the NAAG peptidase inhibitor 2-PMPA, NAAG cleavage was inhibited and, with it, programmed DRG neuronal cell death in the presence of high glucose levels.[39] The researchers have proposed that the cause of this is NAAG's agonistic activity at mGluR3. In addition, NAAG also “prevented glucose-induced inhibition of neurite growth” (Berent- Spillson, et al. 2004). Overall, this makes GCPIII inhibition a clear model target for combating diabetic neuropathy.

Drug addiction

Schizophrenia, as previously described, is normally modeled in the laboratory through a PCP animal model. As GCPIII inhibition was shown to possibly limit schizophrenic behavior in this model,[29] this suggests that GCPIII inhibition, thus, reduces the effect of PCP. In addition, the reward action of many drugs (cocaine, PCP, alcohol, nicotine, etc.) have been shown with increasing evidence to be related to glutamate levels, on which NAAG and GCPIII can have some regulatory effect.[29]

In summary, the findings of multiple drug studies to conclude that:[29]

NAAG/NP system might be involved in neuronal mechanisms regulating cue-induced cocaine craving, the development of cocaine seizure kindling, and management of opioid addiction and alcohol consumptive behaviour. Therefore, NP inhibitors could provide a novel therapy for such conditions.

Other diseases and disorders

NAAG inhibition has also been studied as a treatment against prostate cancer, ALS, and other neurodegenerative diseases such as Parkinson's disease and Huntington's disease.[29]

References

  1. O'Keefe DS, Su SL, Bacich DJ, Horiguchi Y, Luo Y, Powell CT, Zandvliet D, Russell PJ, Molloy PL, Nowak NJ, Shows TB, Mullins C, Vonder Haar RA, Fair WR, Heston WD (November 1998). "Mapping, genomic organization and promoter analysis of the human prostate-specific membrane antigen gene". Biochimica et Biophysica Acta. 1443 (1–2): 113–27. doi:10.1016/s0167-4781(98)00200-0. PMID 9838072.
  2. 2.0 2.1 2.2 2.3 Barinka C, Sácha P, Sklenár J, Man P, Bezouska K, Slusher BS, Konvalinka J (June 2004). "Identification of the N-glycosylation sites on glutamate carboxypeptidase II necessary for proteolytic activity". Protein Science. 13 (6): 1627–35. doi:10.1110/ps.04622104. PMC 2279971. PMID 15152093.
  3. 3.0 3.1 Rojas C, Frazier ST, Flanary J, Slusher BS (November 2002). "Kinetics and inhibition of glutamate carboxypeptidase II using a microplate assay". Analytical Biochemistry. 310 (1): 50–4. doi:10.1016/S0003-2697(02)00286-5. PMID 12413472.
  4. 4.0 4.1 4.2 4.3 4.4 4.5 4.6 4.7 Mesters JR, Barinka C, Li W, Tsukamoto T, Majer P, Slusher BS, Konvalinka J, Hilgenfeld R (March 2006). "Structure of glutamate carboxypeptidase II, a drug target in neuronal damage and prostate cancer". The EMBO Journal. 25 (6): 1375–84. doi:10.1038/sj.emboj.7600969. PMC 1422165. PMID 16467855.
  5. Sácha P, Zámecník J, Barinka C, Hlouchová K, Vícha A, Mlcochová P, Hilgert I, Eckschlager T, Konvalinka J (February 2007). "Expression of glutamate carboxypeptidase II in human brain". Neuroscience. 144 (4): 1361–72. doi:10.1016/j.neuroscience.2006.10.022. PMID 17150306.
  6. Mhawech-Fauceglia P, Zhang S, Terracciano L, Sauter G, Chadhuri A, Herrmann FR, Penetrante R (March 2007). "Prostate-specific membrane antigen (PSMA) protein expression in normal and neoplastic tissues and its sensitivity and specificity in prostate adenocarcinoma: an immunohistochemical study using mutiple tumour tissue microarray technique". Histopathology. 50 (4): 472–83. doi:10.1111/j.1365-2559.2007.02635.x. PMID 17448023.
  7. Israeli RS, Powell CT, Fair WR, Heston WD (January 1993). "Molecular cloning of a complementary DNA encoding a prostate-specific membrane antigen". Cancer Research. 53 (2): 227–30. PMID 8417812.
  8. Goodman OB, Barwe SP, Ritter B, McPherson PS, Vasko AJ, Keen JH, Nanus DM, Bander NH, Rajasekaran AK (November 2007). "Interaction of prostate specific membrane antigen with clathrin and the adaptor protein complex-2". International Journal of Oncology. 31 (5): 1199–203. doi:10.3892/ijo.31.5.1199. PMID 17912448.
  9. Pinto JT, Suffoletto BP, Berzin TM, Qiao CH, Lin S, Tong WP, May F, Mukherjee B, Heston WD (September 1996). "Prostate-specific membrane antigen: a novel folate hydrolase in human prostatic carcinoma cells". Clinical Cancer Research. 2 (9): 1445–51. PMID 9816319.
  10. O'Keefe DS, Bacich DJ, Heston WD (2001). "Prostate Specific Membraen Antigen". In Simons JW, Chung LW, Isaacs WB. Prostate cancer: biology, genetics and the new therapeutics. Totowa, NJ: Humana Press. pp. 307–326. ISBN 978-0-89603-868-4.
  11. O'Keefe DS, Bacich DJ, Heston WD (February 2004). "Comparative analysis of prostate-specific membrane antigen (PSMA) versus a prostate-specific membrane antigen-like gene". The Prostate. 58 (2): 200–10. doi:10.1002/pros.10319. PMID 14716746.
  12. 12.0 12.1 Wang X, Yin L, Rao P, Stein R, Harsch KM, Lee Z, Heston WD (October 2007). "Targeted treatment of prostate cancer". Journal of Cellular Biochemistry. 102 (3): 571–9. doi:10.1002/jcb.21491. PMID 17685433.
  13. Perner S, Hofer MD, Kim R, Shah RB, Li H, Möller P, Hautmann RE, Gschwend JE, Kuefer R, Rubin MA (May 2007). "Prostate-specific membrane antigen expression as a predictor of prostate cancer progression". Human Pathology. 38 (5): 696–701. doi:10.1016/j.humpath.2006.11.012. PMID 17320151.
  14. Ross JS, Sheehan CE, Fisher HA, Kaufman RP, Kaur P, Gray K, Webb I, Gray GS, Mosher R, Kallakury BV (December 2003). "Correlation of primary tumor prostate-specific membrane antigen expression with disease recurrence in prostate cancer". Clinical Cancer Research. 9 (17): 6357–62. PMID 14695135.
  15. Zhang Y, Guo Z, Du T, Chen J, Wang W, Xu K, Lin T, Huang H (June 2013). "Prostate specific membrane antigen (PSMA): a novel modulator of p38 for proliferation, migration, and survival in prostate cancer cells". The Prostate. 73 (8): 835–41. doi:10.1002/pros.22627. PMID 23255296.
  16. Virgolini I, Decristoforo C, Haug A, Fanti S, Uprimny C (March 2018). "Current status of theranostics in prostate cancer". European Journal of Nuclear Medicine and Molecular Imaging. 45 (3): 471–495. doi:10.1007/s00259-017-3882-2. PMC 5787224. PMID 29282518.
  17. 17.0 17.1 Foss CA, Mease RC, Fan H, Wang Y, Ravert HT, Dannals RF, Olszewski RT, Heston WD, Kozikowski AP, Pomper MG (June 2005). "Radiolabeled small-molecule ligands for prostate-specific membrane antigen: in vivo imaging in experimental models of prostate cancer". Clinical Cancer Research. 11 (11): 4022–8. doi:10.1158/1078-0432.CCR-04-2690. PMID 15930336.
  18. 18.0 18.1 Milowsky MI, Nanus DM, Kostakoglu L, Sheehan CE, Vallabhajosula S, Goldsmith SJ, Ross JS, Bander NH (February 2007). "Vascular targeted therapy with anti-prostate-specific membrane antigen monoclonal antibody J591 in advanced solid tumors". Journal of Clinical Oncology. 25 (5): 540–7. doi:10.1200/JCO.2006.07.8097. PMID 17290063.
  19. Kularatne SA, Zhou Z, Yang J, Post CB, Low PS (May 2009). "Design, synthesis, and preclinical evaluation of prostate-specific membrane antigen targeted (99m)Tc-radioimaging agents". Molecular Pharmaceutics. 6 (3): 790–800. doi:10.1021/mp9000712. PMID 19361232.
  20. Kularatne SA, Venkatesh C, Santhapuram HK, Wang K, Vaitilingam B, Henne WA, Low PS (November 2010). "Synthesis and biological analysis of prostate-specific membrane antigen-targeted anticancer prodrugs". Journal of Medicinal Chemistry. 53 (21): 7767–77. doi:10.1021/jm100729b. PMID 20936874.
  21. Thomas M, Kularatne SA, Qi L, Kleindl P, Leamon CP, Hansen MJ, Low PS (September 2009). "Ligand-targeted delivery of small interfering RNAs to malignant cells and tissues". Annals of the New York Academy of Sciences. 1175: 32–9. doi:10.1111/j.1749-6632.2009.04977.x. PMID 19796075.
  22. He W, Kularatne SA, Kalli KR, Prendergast FG, Amato RJ, Klee GG, Hartmann LC, Low PS (October 2008). "Quantitation of circulating tumor cells in blood samples from ovarian and prostate cancer patients using tumor-specific fluorescent ligands". International Journal of Cancer. 123 (8): 1968–73. doi:10.1002/ijc.23717. PMC 2778289. PMID 18661519.
  23. Maurer T, Eiber M, Schwaiger M, Gschwend JE (April 2016). "Current use of PSMA-PET in prostate cancer management". Nature Reviews. Urology. 13 (4): 226–35. doi:10.1038/nrurol.2016.26. PMID 26902337.
  24. Fendler WP, Eiber M, Beheshti M, Bomanji J, Ceci F, Cho S, Giesel F, Haberkorn U, Hope TA, Kopka K, Krause BJ, Mottaghy FM, Schöder H, Sunderland J, Wan S, Wester HJ, Fanti S, Herrmann K (June 2017). "68Ga-PSMA PET/CT: Joint EANM and SNMMI procedure guideline for prostate cancer imaging: version 1.0" (PDF). European Journal of Nuclear Medicine and Molecular Imaging. 44 (6): 1014–1024. doi:10.1007/s00259-017-3670-z. PMID 28283702.
  25. Emmett L, Willowson K, Violet J, Shin J, Blanksby A, Lee J (March 2017). "Lutetium 177 PSMA radionuclide therapy for men with prostate cancer: a review of the current literature and discussion of practical aspects of therapy". Journal of Medical Radiation Sciences. 64 (1): 52–60. doi:10.1002/jmrs.227. PMC 5355374. PMID 28303694.
  26. Chang SS, Reuter VE, Heston WD, Bander NH, Grauer LS, Gaudin PB (July 1999). "Five different anti-prostate-specific membrane antigen (PSMA) antibodies confirm PSMA expression in tumor-associated neovasculature". Cancer Research. 59 (13): 3192–8. PMID 10397265.
  27. Bacich DJ, Wozniak KM, Lu XC, O'Keefe DS, Callizot N, Heston WD, Slusher BS (October 2005). "Mice lacking glutamate carboxypeptidase II are protected from peripheral neuropathy and ischemic brain injury". Journal of Neurochemistry. 95 (2): 314–23. doi:10.1111/j.1471-4159.2005.03361.x. PMID 16190866.
  28. Sácha P, Zámecník J, Barinka C, Hlouchová K, Vícha A, Mlcochová P, Hilgert I, Eckschlager T, Konvalinka J (February 2007). "Expression of glutamate carboxypeptidase II in human brain". Neuroscience. 144 (4): 1361–72. doi:10.1016/j.neuroscience.2006.10.022. PMID 17150306.
  29. 29.00 29.01 29.02 29.03 29.04 29.05 29.06 29.07 29.08 29.09 29.10 29.11 29.12 29.13 29.14 29.15 29.16 29.17 29.18 29.19 29.20 29.21 Zhou J, Neale JH, Pomper MG, Kozikowski AP (December 2005). "NAAG peptidase inhibitors and their potential for diagnosis and therapy". Nature Reviews. Drug Discovery. 4 (12): 1015–26. doi:10.1038/nrd1903. PMID 16341066.
  30. 30.0 30.1 Zhang W, Murakawa Y, Wozniak KM, Slusher B, Sima AA (September 2006). "The preventive and therapeutic effects of GCPII (NAALADase) inhibition on painful and sensory diabetic neuropathy". Journal of the Neurological Sciences. 247 (2): 217–23. doi:10.1016/j.jns.2006.05.052. PMID 16780883.
  31. Ghose S, Weickert CS, Colvin SM, Coyle JT, Herman MM, Hyde TM, Kleinman JE (January 2004). "Glutamate carboxypeptidase II gene expression in the human frontal and temporal lobe in schizophrenia". Neuropsychopharmacology. 29 (1): 117–25. doi:10.1038/sj.npp.1300304. PMID 14560319.
  32. 32.0 32.1 Kozikowski AP, Nan F, Conti P, Zhang J, Ramadan E, Bzdega T, Wroblewska B, Neale JH, Pshenichkin S, Wroblewski JT (February 2001). "Design of remarkably simple, yet potent urea-based inhibitors of glutamate carboxypeptidase II (NAALADase)". Journal of Medicinal Chemistry. 44 (3): 298–301. doi:10.1021/jm000406m. PMID 11462970.
  33. 33.0 33.1 33.2 Nagel J, Belozertseva I, Greco S, Kashkin V, Malyshkin A, Jirgensons A, Shekunova E, Eilbacher B, Bespalov A, Danysz W (December 2006). "Effects of NAAG peptidase inhibitor 2-PMPA in model chronic pain - relation to brain concentration". Neuropharmacology. 51 (7–8): 1163–71. doi:10.1016/j.neuropharm.2006.07.018. PMID 16926034.
  34. Chen SR, Wozniak KM, Slusher BS, Pan HL (February 2002). "Effect of 2-(phosphono-methyl)-pentanedioic acid on allodynia and afferent ectopic discharges in a rat model of neuropathic pain". The Journal of Pharmacology and Experimental Therapeutics. 300 (2): 662–7. doi:10.1124/jpet.300.2.662. PMID 11805230.
  35. Tolias C, Wasserberg J (2002). "Critical decision making in severe head injury management". Trauma. 4 (4): 211–221. doi:10.1191/1460408602ta246oa.
  36. "What is Stroke". National Stroke Association. Retrieved 2009-01-15.
  37. "Schizophrenia". National Mental Health Information Center. Retrieved 2009-01-15.
  38. "Diabetic Neuropathies: The Nerve Damage of Diabetes". National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health. 2009. Retrieved 2009-01-15.
  39. Berent-Spillson A, Robinson AM, Golovoy D, Slusher B, Rojas C, Russell JW (April 2004). "Protection against glucose-induced neuronal death by NAAG and GCP II inhibition is regulated by mGluR3". Journal of Neurochemistry. 89 (1): 90–9. doi:10.1111/j.1471-4159.2003.02321.x. PMID 15030392.

External links