Expression of NR1 and apoptosis levels in the hippocampal cells of mice treated with MK‑801

  • Authors:
    • Juan Ding
    • Hui‑Hui Zhou
    • Quan‑Rui Ma
    • Zhong‑Yi He
    • Jiang‑Bo Ma
    • Yin‑Ming Liu
    • Yi‑Wei Zhang
    • Yu‑Qing He
    • Juan Liu
  • View Affiliations

  • Published online on: September 29, 2017     https://doi.org/10.3892/mmr.2017.7674
  • Pages: 8359-8364
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The aim of the present study was to investigate the characteristics of N‑methyl‑D‑aspartate receptor R1 (NR1) expression and apoptosis in the nerve cells of the hippocampus in schizophrenia‑like mice. C57BL/6 mice were randomly allocated to the following groups: i) Blank group; ii) MK‑801 group; iii) MK‑801+NMDA group, according to body weight. The NMDAR antagonist, MK‑801 (0.6 mg/kg/d) was intraperitoneally injected daily for 14 days to induce a schizophrenia‑like phenotype mouse model, and the effect of the NMDA injection via the lateral ventricle was observed. The results demonstrated that the number of NR1 positive cells in the MK‑801 group increased in the CA1 and DG regions, indicating that NMDA may reverse this change. The level of damage decreased in the MK‑801 treated group when compared with the blank group in the CA3 region. The protein expression of NR1 increased however, at the mRNA expression level, NR1 was lower in the MK‑801 treated group when compared to the blank group; NMDA also reversed this change. In addition, early and total apoptosis detected in the hippocampal nerve cells was significantly increased in the MK‑801 group when compared with the blank group, which was reversible following treatment with NMDA. These results indicated that NMDA may regulate the expression of NR1 and suppress apoptosis in hippocampal nerve cells in schizophrenia‑like mice. Thus, NR1 may be a promising therapeutic target for the treatment of schizophrenia.

Introduction

Schizophrenia is a severe psychiatric disorder with a lifetime prevalence of around 1% (1). Cognitive impairments are the core symptoms of the disease which is characterized by disturbances in sensory information processing, attention, working memory and executive functions deficits (2) which are all hippocampus-dependent functions (3,4). Schizophrenia imposes a heavy financial burden and many potential safety hazards to families of sufferers and society as a whole.

Recent studies focusing on the pathogenesis and treatment of schizophrenia have made significant progress in understanding the biology underpinning the disease. The glutamatergic hypothesis of schizophrenia states that dysfunction of the glutamatergic system induces imbalance between the glutamatergic and dopaminergic systems in the central nervous system (CNS) and induces some symptoms of schizophrenia (5). N-methyl-D-aspartate receptors (NMDARs) may be a key factor in the pathogenesis of schizophrenia (68). NMDAR is one of the ionotropic glutamatergic receptors (5), is a tetrameric structure of seven subunits including at least one obligatory subunit, N-methyl-D-aspartate receptor R1 (NR1; it includes eight functional splice variants), and varying numbers of a family of NR2 (NR2A-D) or NR3 (NR3A-B) subunits (9,10). The properties of NMDARs are both complicated and diverse owing to their complex subunit compositions. The characteristics of NMDAR include high Ca2+ permeability and Mg2+ suppression closely associated with neurogenesis, neuronal survival, synaptic plasticity and the formation of learning and memory abilities (2,4,1113). Studies have demonstrated that schizophrenia patients show abnormal synaptic plasticity, LTP and cognitive dysfunction (11,1315). The hippocampus is involved with cognitive functions, particularly in learning and memory and so was chosen as the area of particular interest for the current study. NR1 subunits are essential components of NMDARs. NR1 is very important for understanding the distribution and functions of NMDARs (8), however, the expression and regulation of NR1 in the hippocampus of schizophrenia-like mice are not known.

To investigate these issues, our study examined the expression of the NR1 subunit in the granule cell layer CA1, CA3 and DG region, and discussed regulation of the NR1 subunit expression by NMDA. We also characterized the expression of the NR1 subunit mRNA and protein, and levels of apoptosis in the hippocampal nerve cells of schizophrenia-like mice. In conclusion, we demonstrated that NMDA can regulate the expression of NR1 and suppress apoptosis in the hippocampal nerve cells of schizophrenia-like mice.

Materials and methods

Materials

MK-801 (3 mg/ml in 0.9% saline) and NMDA (1.5 µg/ml in 0.9% saline) were purchased from Sigma-Aldrich (Merck KGaA, Darmstadt, Germany). The primary antibody of NR1 was purchased from Abcam (Cambridge, MA, USA), and β-actin antibody purchased from Nanjing KeyGen Biotech Co., Ltd. (Nanjing, China). The secondary antibody of goat anti-rabbit fluorescein isothiocyanate (FITC) was purchased from CWBIO (Beijing, China), and the HRP-conjugated goat anti-rabbit antibody purchased from ZSGB-BIO (Beijing, China). Annexin V-FITC/PI kit was purchased from Yeasen (USA). TRIzol reagent was purchased from Life Technologies (Thermo Fisher Scientific, Inc., Waltham, MA, USA). All PCR primers were synthesized by Sangon Biotech Co., Ltd. (Shanghai, China). RevertAid First Strand cDNA Synthesis kit and Maxima SYBR Green qPCR Master Mix were purchased from Thermo Fisher Scientific, Inc.

Animals

Male C57BL/6 mice (23–25 grams, at 3-month old) were used in this study and provided by the Experimental Animal Center of the Ningxia Medical University. Mice were housed in groups of five in cages with free access to food and tap water for 7 days. The animal room temperature was maintained at 23±2°C, with a 12/12 dark/light cycle (lights on 7:00-19:00) and 50% humidity. All animals were handled in accordance with the standards established by the institutional animal care and use committee of Ningxia Medical University.

Experimental groups and models

Male C57BL/6 mice were divided into 3 groups (n=12 in each group): A blank group, a MK-801 treated group and a MK-801+NMDA treated group. In the MK-801+NMDA treated group, mice were treated intra-peritoneally with MK-801 at dose of 0.6 mg/kg at the same time every day for 14 days (16). After 14 days, mice were anesthetized by intra-peritoneal injection of chloral hydrate and mounted on a stereotaxic frame for intra-cerebroventricular injection (mm from bregma: A −0.5, L −1.0, V-2.5) of NMDA (25 ng/µl, 3 ul) once each mouse (17). Brains were harvested 3 days after the operation (n=12). In the MK-801 group (schizophrenia-like group), saline was used instead of NMDA and all other operations were the same as intra-cerebroventricular injection groups (n=12). In the blank group, an equal volume of 0.9% saline was injected instead of MK-801 and NMDA (n=12).

Immunofluorescence (IF) staining

Mice (n=3 in each group) were transcardially perfused with ice-cold 0.01 M PBS (pH 7.4, 150 ml/mouse) and 4% paraformaldehyde solution (200 ml/mouse). Brains were harvested, and post-fixed for 24 h in 4% paraformaldehyde, followed by immersion in 20%, 30% sucrose solution overnight. Brains were washed once for 10 min with ice cold 0.01 M PBS, embedded in 1% gelose solution and 30 µm-thick coronal sections cut and used for immunofluorescent staining of NR1 in the hippocampal granule cell layer. Sections were permeabilized and blocked with blocking solution (7% BSA and 0.3% Triton X-100 in 0.01 M PBS) for 1 h at room temperature, and incubated overnight at 4°C with rabbit anti-NR1 (1:800). Sections were washed three times for 30 min with 0.01 M PBS followed by second antibody incubation with goat anti-rabbit fluorescein isothiocyanate (FITC, 1:200) for 2 h at room temperature. Sections were washed three times for 30 min with 0.01 M PBS. Finally, the number of NR1 positive cells in granule cell layer of hippocampus was measured using confocal laser scanning microscope.

Western blotting

Mice (n=3 in each group) were decapitated and hippocampi were harvested. The hippocampi were put into glass homogenates with ice-cold buffer containing protease inhibitor for 45 min. The homogenates were centrifuged at 12,000 × g for 20 min and the supernatant was collected (stored at −80°C). The protein levels were detected using the BCA method. Total protein in each group was separated (NR1, β-actin) by 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and electrophoretically transferred onto polyvinylidene difluoride (PVDF) membranes. PVDF membranes were immersed in blocking buffer (5% fat-free milk in PBST) for 1 h at room temperature and incubated overnight at 4°C with the primary rabbit antibodies respectively (NR1, 1:500; β-actin, 1:3,000). The membranes were then washed three times for 30 min using PBST and incubated HRP-conjugated goat anti-rabbit antibody (1:5,000) for 2 h at room temperature and washed three times for 30 min in PBST. Immuno-reactive proteins were visualized using enhanced chemiluminescence (ECL) detection and the signals were quantified by densitometry using a western blotting detection system.

Real-time PCR

Total RNA from the hippocampal tissue (n=3 in each group) was extracted using TRIzol reagent. One microgram of the purified total RNA was reverse transcribed using a RevertAid First Strand cDNA Synthesis kit. The mRNA level of NR1 was determined using the Maxima SYBR Green qPCR Master Mix. All PCR primers were synthesized by Sangon Biotech (Shanghai, China). The forward and reverse primer sequences for NR1 were 5′-CTTCCTCCAGCCACTACCC-3′ and 5′-AGAAAGCACCCCTGAAGCAC-3′, respectively; for β-actin, forward and reverse primer sequence were 5′-CCTAAGGCCAACCGTGAAAAG−3′and 5′-ACCAGAGGCATACAGGGACAAC-3′, respectively. β-actin was used as an internal reference to standardize each gene. Each sample was investigated in triplicate. The relative amount of mRNA was measured using the comparative threshold (Ct) method by normalizing target cDNA Ct values to that of β-actin, and the fold expression changes were calculated according to the 2−ΔΔCt method (18,19).

Flow cytometry analysis

Analysis of apoptosis was performed by flow cytometry of the hippocampi (n=3 in each group) isolated from each mouse. Samples were digested with 0.25% trypsin at 37°C for 20 min and cell suspension were then filtered through a cell strainer (20) and the number of apoptotics cells was determined by flow cytometry assay using Annexin V-FITC/PI kit (21) and analyzed using the FACS express v2.0 software.

Statistical analysis. Data are presented as the mean ± standard deviation. Statistical analysis was carried out in SPSS11.5 (SPSS, Inc., Chicago, IL, USA) using one-way ANOVA. P<0.05 was considered as statistically significant.

Results

Expression of NR1 subunits in the CA1, CA3 and DG of the hippocampus

The distribution patterns of NR1 positive cells (white arrows) in the CA1, CA3 and dentate gyrus (DG) of the hippocampus are shown in Fig. 1A. The number of NR1 positive cells is shown in Fig. 1B. The results showed that the number of NR1 positive cells in the MK-801 group increased in the CA1 (P>0.05) and DG (P<0.05) regions and this change was reversible by NMDA. The MK-801 group showed a decrease in NR1 compared with the Blank group in the CA3 region (P<0.05).

Expression of NR1 subunit protein levels in the hippocampus

Fig. 2 shows the expression of NR1 subunit protein levels in the hippocampus of experimental mice. The data showed the expression of NR1 subunit increased in the MK-801 treated group (P<0.05) compared with to the Blank group and again this trend was reversible by NMDA.

Expression of NR1 subunit mRNA levels in hippocampus

The mRNA levels of the NR1 subunit normalized to β-actin in hippocampus of mice are shown in Fig. 3. The data show that the mRNA expression of NR1 subunit decreased in the MK-801 treated group (P<0.05) compared to the Blank group. This change was reversed by NMDA although no statistical significance was detected (P>0.05).

Apoptosis of hippocampal nerve cells

From Fig. 4 it can be seen that the early and total levels of apoptosis in the hippocampal nerve cells significantly increased (P<0.05) in the MK-801 group compared to the Blank group. In contrast, these changes were reversible by NMDA (P<0.05). The normal live cells (NLC) are representative of the survival rate of hippocampus nerve cells. The number of NLC decreased in the MK-801 group compared to the blank group (P<0.05) and again these changes were reversible with NMDA.

Discussion

The prevalence of schizophrenia increased to 1.0% between 1990 and 2010 all over the world (22). The underlying pathophysiological mechanisms of the disease are complex and remain to be fully elucidated. NMDAR is a major glutamate receptor subtype that is known to play a key role in learning and memory. NMDARs include NR1, NR2 (2A-2D), NR3 (3A-3B). NR1 subunits are the essential and obligatory components of NMDARs. NR1 subunits play an important role in determining the properties of NMDARs. Recently, NMDAR has received great attention on schizophrenia research. Dysfunction of NMDAR in the hippocampus is very important for the formation of schizophrenia (8).

We adopted an MK-801 administrated mouse model to investigate NR1 expression pattern and hippocampal neuron survival deficit in schizophrenia. MK-801 is a NMDAR antagonist per se. We worried changes released in this model may not be schizophrenia-related, but just a classical receptor-ligand feedback response of physiological process. Actually, no animal model can accurately reproduce all aspects of schizophrenia but mouse models can mimic several important aspects of the disease. MK-801 has been used for inducing a schizophrenia-like phenotype in rodents (23,24) which leads to different degrees of cognitive impairment (2527). Some studies have shown hypofunction of the NMDA receptor by chronic treatment with MK-801 (28) or cell death in the hippocampus following NMDAR hypofunction (29). NMDAR subunits are known to play an important role in determining neurotoxic (30), functional and psychiatric effects in the hippocampus (31,32).

In this study, the results showed that the number of NR1 positive cells significantly increased in the hippocampal granule cell layer of the DG and CA1 regions, but decreased in the CA3 region of schizophrenia-like mice induced by MK-801. NMDA was shown to reverse these changes. The NR1 total protein levels significantly increased, whilste the mRNA levels were reduced in the hippocampus of schizophrenia-like mice induced by MK-801. NMDA could also reverse these changes. Other studies of NMDAR expression in schizophrenia have shown variable changes at the transcript and protein expression levels in different areas of the brain. For example, study have shown that the protein expression of NR1 in the anterior cingulate cortex increased (33) or was found to be unchanged in hippocampus (34). However, another study found that NR1 protein expression was reduced in the prefrontal cortex, hippocampus and hippocampal DG region (35). Furthermore, at the mRNA level, expression has been shown to be decreased in the hippocampus (36,37), and the expression of the NR1 protein and mRNA levels were contradictory in the cortex of schizophrenia mice (33).

Another study examined NR1 expression in postmortem samples from patients with schizophrenia and comparison subjects. The data showed that at the transcriptional level, NR1 levels were lower in the thalamus of schizophrenia patients compared to control subjects (38). These studies support our experimental results to a certain extent. The current study has demonstrated that transcriptional changes of the NMDAR subunit expression in cortical areas appear to be associated with specific regions (33). These data may be explained by three supporting hypotheses. Firstly, the regional specificity expression of NR1 protein levels comes from the mRNA and the differential expressions in the CA1, CA3 and DG region contribute to the function of different areas (4). Secondly, it is possible that the peak of expression is not the same point at the protein and mRNA levels. Finally, NR1 may be involved in regulation of other complex components of glutamatergic pathways that are associated with schizophrenia (33).

The levels of apoptosis in the hippocampal nerve cells increased in schizophrenia-like mice induced by MK-801 which could be reversed by NMDA. Studies have shown that hyperfunction of glutamate receptors such as NMDA receptors increases intracellular Ca2+ levels. The continual influx of Ca2+ through the open NMDA receptors results in mitochondrial stress which attempts to sequester and buffer Ca2+. As mitochondrial membrane potential decreases due to the overflow of Ca2+ and mitochondria reverse their ATP synthase in an attempt to restore Ca2+ homeostasis. Eventually, the excess Ca2+ uptake causes loss of mitochondrial membrane potential, mitochondrial swelling, opening of the mitochondrial permeability transition pore (39,40), outer membrane rupture and loss of Ca2+, and apoptogenic factors into the cytoplasm. This process ultimately results in neuronal cell death (38). Our experimental data show that the apoptosis of hippocampal nerve cells significantly increased in schizophrenia-like mice induced by MK-801 and these changes could be reversed by NMDA.

We demonstrated that NR1 significantly contributes to the acquisition and apoptosis of hippocampal nerve cells in schizophrenia-like mice. NMDA can effectively regulate the expression of NR1 and levels of apoptosis in hippocampal nerve cells in schizophrenia-like mice, whereas the exactly underprinning molecular mechanisms need to be further clarified.

In summary, we have demonstrated that the expression of NR1 markedly increased in the hippocampus of schizophrenia-like mice, but showed differential trends in the CA1, CA3 and DG regions. NMDAR hyperfunction induced apoptosis in hippocampal nerve cells. NMDA contributes to positive function in schizophrenia, and may be offer an important molecular pathway for therapeutic intervention. NR1 might hold much potential as target in the therapy of schizophrenia.

Acknowledgements

This work was supported by the National Natural Science Foundation of China (nos. 81160169, 81460214, 31460255, 31660270) and the Natural Science Foundation of Ningxia (NZ14058) and West China Top Class Discipline Project in Basic Medical Sciences, Ningxia Medical University.

References

1 

de Candia TR, Lee SH, Yang J, Browning BL, Gejman PV, Levinson DF, Mowry BJ, Hewitt JK, Goddard ME, O'Donovan MC, et al: Additive genetic variation in schizophrenia risk is shared by populations of African and European descent. Am J Hum Genet. 93:463–470. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Sánchez-Blázquez P, Rodríguez-Muñoz M and Garzón J: The cannabinoid receptor 1 associates with NMDA receptors to produce glutamatergic hypofunction: Implications in psychosis and schizophrenia. Front Pharmacol. 4:1692014. View Article : Google Scholar : PubMed/NCBI

3 

Nakashiba T, Cushman JD, Pelkey KA, Renaudineau S, Buhl DL, McHugh TJ, Barrera V Rodriguez, Chittajallu R, Iwamoto KS, McBain CJ, et al: Young dentate granule cells mediate pattern separation, whereas old granule cells facilitate pattern completion. Cell. 149:188–201. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Tannenholz L, Jimenez JC and Kheirbek MA: Local and regional heterogeneity underlying hippocampal modulation of cognition and mood. Front Behav Neurosci. 8:1472014. View Article : Google Scholar : PubMed/NCBI

5 

Gaspar PA, Bustamante ML, Silva H and Aboitiz F: Molecular mechanisms underlying glutamatergic dysfunction in schizophrenia: Therapeutic implications. J Neurochem. 111:891–900. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Howes OD and Murray RM: Schizophrenia: An integrated sociodevelopmental-cognitive model. Lancet. 383:1677–1687. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Penzes P, Cahill ME, Jones KA, VanLeeuwen JE and Woolfrey KM: Dendritic spine pathology in neuropsychiatric disorders. Nat Neurosci. 14:285–293. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Ju P and Cui D: The involvement of N-methyl-d-aspartate receptor (NMDAR) subunit NR1 in the pathophysiology of schizophrenia. Acta Biochim Biophys Sin (Shanghai). 48:209–219. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Ghasemi M, Phillips C, Trillo L, De Miguel Z, Das D and Salehi A: The role of NMDA receptors in the pathophysiology and treatment of mood disorders. Neurosci Biobehav Rev. 47:336–358. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Henson MA, Roberts AC, Pérez-Otaño I and Philpot BD: Influence of the NR3A subunit on NMDA receptor functions. Prog Neurobiol. 91:23–37. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Hardingham GE and Bading H: Synaptic versus extrasynaptic NMDA receptor signalling: Implications for neurodegenerative disorders. Nat Rev Neurosci. 11:682–696. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Lau CG and Zukin RS: NMDA receptor trafficking in synaptic plasticity and neuropsychiatric disorders. Nat Rev Neurosci. 8:413–426. 2007. View Article : Google Scholar : PubMed/NCBI

13 

Volianskis A, France G, Jensen MS, Bortolotto ZA, Jane DE and Collingridge GL: Long-term potentiation and the role of N-methyl-D-aspartate receptors. Brain Res. 1621:5–16. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Bliss TV, Collingridge GL and Morris RG: Synaptic plasticity in health and disease: Introduction and overview. Philos Trans R Soc Lond B Biol Sci. 369:201301292013. View Article : Google Scholar : PubMed/NCBI

15 

Soriano FX, Papadia S, Hofmann F, Hardingham NR, Bading H and Hardingham GE: Preconditioning doses of NMDA promote neuroprotection by enhancing neuronal excitability. J Neurosci. 26:4509–4518. 2006. View Article : Google Scholar : PubMed/NCBI

16 

Jones KS, Corbin JG and Huntsman MM: Neonatal NMDA receptor blockade disrupts spike timing and glutamatergic synapses in fast spiking interneurons in a NMDA receptor hypofunction model of schizophrenia. PLoS One. 9:e1093032014. View Article : Google Scholar : PubMed/NCBI

17 

Ahmad AS, Saleem S, Ahmad M and Doré S: Prostaglandin EP1 receptor contributes to excitotoxicity and focal ischemic brain damage. Toxicol Sci. 89:265–270. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Zapatero-Solana E, García-Giménez JL, Guerrero-Aspizua S, García M, Toll A, Baselga E, Durán-Moreno M, Markovic J, García-Verdugo JM, Conti CJ, et al: Oxidative stress and mitochondrial dysfunction in Kindler syndrome. Orphanet J Rare Dis. 9:2112014. View Article : Google Scholar : PubMed/NCBI

19 

Jiao Y, Liu C, Cui FM, Xu JY, Tong J, Qi XF, Wang LL and Zhu W: Long intergenic non-coding RNA induced by X-ray irradiation regulates DNA damage response signaling in the human bronchial epithelial BEAS-2B cell line. Oncol Lett. 9:169–176. 2015.PubMed/NCBI

20 

Zhang Z, Zhao C, Liu B, Liang D, Qin X, Li X, Zhang R, Li C, Wang H, Sun D and Cao F: Inositol pyrophosphates mediate the effects of aging on bone marrow mesenchymal stem cells by inhibiting Akt signaling. Stem Cell Res Ther. 5:332014. View Article : Google Scholar : PubMed/NCBI

21 

Pervaiz N and Hoffman-Goetz L: Immune cell inflammatory cytokine responses differ between central and systemic compartments in response to acute exercise in mice. Exerc Immunol Rev. 18:142–157. 2012.PubMed/NCBI

22 

Whiteford HA, Degenhardt L, Rehm J, Baxter AJ, Ferrari AJ, Erskine HE, Charlson FJ, Norman RE, Flaxman AD, Johns N, et al: Global burden of disease attributable to mental and substance use disorders: Findings from the global burden of disease study 2010. Lancet. 382:1575–1586. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Yu J, Qi D, Xing M, Li R, Jiang K, Peng Y and Cui D: MK-801 induces schizophrenic behaviors through downregulating Wnt signaling pathways in male mice. Brain Res. 1385:281–292. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Kim TW, Kang HS, Park JK, Lee SJ, Baek SB and Kim CJ: Voluntary wheel running ameliorates symptoms of MK-801-induced schizophrenia in mice. Mol Med Rep. 10:2924–2930. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Knott AB and Bossy-Wetzel E: Nitric oxide in health and disease of the nervous system. Antioxid Redox Signal. 11:541–554. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Mandillo S, Rinaldi A, Oliverio A and Mele A: Repeated administration of phencyclidine, amphetamine and MK-801 selectively impairs spatial learning in mice: A possible model of psychotomimetic drug-induced cognitive deficits. Behav Pharmacol. 14:533–544. 2003. View Article : Google Scholar : PubMed/NCBI

27 

Mohn AR, Gainetdinov RR, Caron MG and Koller BH: Mice with reduced NMDA receptor expression display behaviors related to schizophrenia. Cell. 98:427–436. 1999. View Article : Google Scholar : PubMed/NCBI

28 

Rujescu D, Bender A, Keck M, Hartmann AM, Ohl F, Raeder H, Giegling I, Genius J, McCarley RW, Möller HJ and Grunze H: A pharmacological model for psychosis based on N-methyl-D-aspartate receptor hypofunction: Molecular, cellular, functional and behavioral abnormalities. Biol Psychiatry. 59:721–729. 2006. View Article : Google Scholar : PubMed/NCBI

29 

Watanabe Y, Müller MK, von Engelhardt J, Sprengel R, Seeburg PH and Monyer H: Age-dependent degeneration of mature dentate gyrus granule cells following NMDA receptor ablation. Front Mol Neurosci. 8:872016. View Article : Google Scholar : PubMed/NCBI

30 

Anastasio NC, Xia Y, O'Connor ZR and Johnson KM: Differential role of N-methyl-D-aspartate receptor subunits 2A and 2B in mediating phencyclidine-induced perinatal neuronal apoptosis and behavioral deficits. Neuroscience. 163:1181–1191. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Kocsis B: Differential role of NR2A and NR2B subunits in N-methyl-D-aspartate receptor antagonist-induced aberrant cortical gamma oscillations. Biol Psychiatry. 71:987–995. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Inta D, Vogt MA, Luoni A, Filipović D, Lima-Ojeda JM, Pfeiffer N, Gasparini F, Riva MA and Gass P: Significant increase in anxiety during aging in mGlu5 receptor knockout mice. Behav Brain Res. 241:27–31. 2013. View Article : Google Scholar : PubMed/NCBI

33 

Kristiansen LV, Huerta I, Beneyto M and Meador-Woodruff JH: NMDA receptors and schizophrenia. Curr Opin Pharmacol. 7:48–55. 2007. View Article : Google Scholar : PubMed/NCBI

34 

Toro C and Deakin JF: NMDA receptor subunit NRI and postsynaptic protein PSD-95 in hippocampus and orbitofrontal cortex in schizophrenia and mood disorder. Schizophr Res. 80:323–330. 2005. View Article : Google Scholar : PubMed/NCBI

35 

Park JK, Lee SJ and Kim TW: Treadmill exercise enhances NMDA receptor expression in schizophrenia mice. J Exerc Rehabil. 10:15–21. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Law AJ and Deakin JF: Asymmetrical reductions of hippocampal NMDAR1 glutamate receptor mRNA in the psychoses. Neuroreport. 12:2971–2974. 2001. View Article : Google Scholar : PubMed/NCBI

37 

Gao XM, Sakai K, Roberts RC, Conley RR, Dean B and Tamminga CA: Ionotropic glutamate receptors and expression of N-methyl-D-aspartate receptor subunits in subregions of human hippocampus: Effects of schizophrenia. Am J Psychiatry. 157:1141–1149. 2000. View Article : Google Scholar : PubMed/NCBI

38 

Ibrahim HM, Hogg AJ Jr, Healy DJ, Haroutunian V, Davis KL and Meador-Woodruff JH: Ionotropic glutamate receptor binding and subunit mRNA expression in thalamic nuclei in schizophrenia. Am J Psychiatry. 157:1811–1823. 2000. View Article : Google Scholar : PubMed/NCBI

39 

Gono T, Kawaguchi Y, Kaneko H, Nishimura K, Hanaoka M, Kataoka S, Okamoto Y, Katsumata Y and Yamanaka H: Anti-NR2A antibody as a predictor for neuropsychiatric systemic lupus erythematosus. Rheumatology (Oxford). 50:1578–1585. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Faust TW, Chang EH, Kowal C, Berlin R, Gazaryan IG, Bertini E, Zhang J, Sanchez-Guerrero J, Fragoso-Loyo HE, Volpe BT, et al: Neurotoxic lupus autoantibodies alter brain function through two distinct mechanisms. Proc Natl Acad Sci USA. 107:pp. 18569–18574. 2010; View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2017
Volume 16 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ding J, Zhou HH, Ma QR, He ZY, Ma JB, Liu YM, Zhang YW, He YQ and Liu J: Expression of NR1 and apoptosis levels in the hippocampal cells of mice treated with MK‑801. Mol Med Rep 16: 8359-8364, 2017
APA
Ding, J., Zhou, H., Ma, Q., He, Z., Ma, J., Liu, Y. ... Liu, J. (2017). Expression of NR1 and apoptosis levels in the hippocampal cells of mice treated with MK‑801. Molecular Medicine Reports, 16, 8359-8364. https://doi.org/10.3892/mmr.2017.7674
MLA
Ding, J., Zhou, H., Ma, Q., He, Z., Ma, J., Liu, Y., Zhang, Y., He, Y., Liu, J."Expression of NR1 and apoptosis levels in the hippocampal cells of mice treated with MK‑801". Molecular Medicine Reports 16.6 (2017): 8359-8364.
Chicago
Ding, J., Zhou, H., Ma, Q., He, Z., Ma, J., Liu, Y., Zhang, Y., He, Y., Liu, J."Expression of NR1 and apoptosis levels in the hippocampal cells of mice treated with MK‑801". Molecular Medicine Reports 16, no. 6 (2017): 8359-8364. https://doi.org/10.3892/mmr.2017.7674