NMDA receptor blockade at rest triggers rapid behavioural antidepressant responses

Anita E Autry, Megumi Adachi, Elena Nosyreva, Elisa S Na, Maarten F Los, Peng-fei Cheng, Ege T Kavalali, Lisa M Monteggia, Anita E Autry, Megumi Adachi, Elena Nosyreva, Elisa S Na, Maarten F Los, Peng-fei Cheng, Ege T Kavalali, Lisa M Monteggia

Abstract

Clinical studies consistently demonstrate that a single sub-psychomimetic dose of ketamine, an ionotropic glutamatergic NMDAR (N-methyl-D-aspartate receptor) antagonist, produces fast-acting antidepressant responses in patients suffering from major depressive disorder, although the underlying mechanism is unclear. Depressed patients report the alleviation of major depressive disorder symptoms within two hours of a single, low-dose intravenous infusion of ketamine, with effects lasting up to two weeks, unlike traditional antidepressants (serotonin re-uptake inhibitors), which take weeks to reach efficacy. This delay is a major drawback to current therapies for major depressive disorder and faster-acting antidepressants are needed, particularly for suicide-risk patients. The ability of ketamine to produce rapidly acting, long-lasting antidepressant responses in depressed patients provides a unique opportunity to investigate underlying cellular mechanisms. Here we show that ketamine and other NMDAR antagonists produce fast-acting behavioural antidepressant-like effects in mouse models, and that these effects depend on the rapid synthesis of brain-derived neurotrophic factor. We find that the ketamine-mediated blockade of NMDAR at rest deactivates eukaryotic elongation factor 2 (eEF2) kinase (also called CaMKIII), resulting in reduced eEF2 phosphorylation and de-suppression of translation of brain-derived neurotrophic factor. Furthermore, we find that inhibitors of eEF2 kinase induce fast-acting behavioural antidepressant-like effects. Our findings indicate that the regulation of protein synthesis by spontaneous neurotransmission may serve as a viable therapeutic target for the development of fast-acting antidepressants.

©2011 Macmillan Publishers Limited. All rights reserved

Figures

Figure 1. Time-course of NMDAR antagonist-mediated antidepressant-like…
Figure 1. Time-course of NMDAR antagonist-mediated antidepressant-like behavioural effects
Mean immobility±SEM of C57BL/6 mice in FST following acute treatment of ketamine, CPP or MK801. Independent groups of mice were used at each time point and drug treatment to avoid behavioural habituation. ANOVA analysis F3,27 =30.31, P<.0001 for treatment groups, F3,27 =19.06, P<.0001 for duration of response, and F9,81=9.32, P<.0001 for treatment-duration interaction; therefore, we examined treatment effects by time-point. a, Ketamine (3.0 mg/kg) significantly reduced immobility, suggestive of an AD-like response, at 30-min., 3-hrs, 24-hrs, and 1-week compared to vehicle treatment (*P<0.05). b, CPP (0.5 mg/kg) significantly reduced immobility at 30-min., 3-hrs, and 24-hrs (*P<0.05) compared to vehicle treatment. c, MK-801 (0.1 mg/kg) produced significant decreases in immobility at 30-min and 3-hrs compared to vehicle treatment (*P<0.05), (n=10/group/time-point). Here and in all figures error bars represent Standard Error of the Mean (SEM).
Figure 2. BDNF translation in antidepressant effects…
Figure 2. BDNF translation in antidepressant effects of NMDAR antagonists
a, Immobility in FST following acute ketamine (3.0 mg/kg). At 30-min, ANOVA F1,35=17.13, P=0.0002 for drug, F1,35=7.57, P=0.0093 for genotype-drug interaction, multiple comparisons with t-test (*P<0.05). At 24-hr, in a separate cohort, ANOVA F1,29=3.77, P=0.0619 for treatment, multiple comparisons (*P<0.05) (n=7-12/group). b, Densitometric analysis of BDNF (normalized-GAPDH) in hippocampus following ketamine (3.0 mg/kg) or MK801 (0.1 mg/kg). 30-min., ANOVA F2,12=6.77, P=0 .0108 for treatment, Bonferroni post-hoc test *P<0.05. 24-hrs, no significance (n=5-6/group). c, Anisomycin and actinomycinD protocol. d, Immobility at 30-min., ANOVA F1,34=11.83, P=0.0016 for treatment and F1, 34 = 10.91, P =0.0023 for treatment-inhibitor interaction, multiple comparisons (*P<0.05)(n=8-10/group). e, Immobility at 24 hrs, ANOVA F1, 31=9.34, P =0.0046 for treatment, multiple comparisons (*P<0.05)(n=8-10/group). f, Immobility of WTs given vehicle or NMDA (75 mg/kg), tested 30-minutes later in FST. g, Immobility of WTs given NBQX (10 mg/kg) or PTX (1.0 mg/kg), tested 30-minutes later in FST. h, Immobility of WTs given vehicle, ketamine (3.0 mg/kg), ketamine+NBQX (10 mg/kg), tested 30-minutes later in FST. ANOVA F2,26=8.226, P<0.0019, Bonferroni post-hoc analysis shows ketamine effect reversed by NBQX, *P<0.05. i, Densitometric analysis of p-mTOR (normalized-mTOR) in hippocampus 30-minutes after vehicle or ketamine.
Figure 3. Ketamine blocks NMDAR spontaneous activity,…
Figure 3. Ketamine blocks NMDAR spontaneous activity, reduces the level of eEF2 phosphorylation, and strengthens synaptic responses
a, Representative western blots from hippocampal primary cultures. b, (left) Densitometric analysis of peEF2 (normalized-total eEF2). Data expressed as mean percentage±SEM. TTX alone does not alter peEF2 while AP5 or ketamine, with or without TTX, significantly decreases peEF2 as assessed by t-test analysis (*P<0.05). (right) Application of 1, 5 and 50 μM of ketamine produces dose-dependent decreases in peEF2 assessed by t-test analysis (*P<0.05). c, Representative traces of NMDAR spontaneous activity after application of 1, 5 and 50 μM. d, Quantification of charge transfer (10 sec) reveals significant effects (*P<0.05) for all ketamine concentrations compared to control (n=6-16) assessed by t-test analysis (*P<0.05). e, Field potential slopes are plotted as a function of time. Representative field potential traces, (average 2-min) are shown during baseline (1) and at 45-min (2). The asterisk refers to significantly different field potentials values (*P<0.05). For statistical analysis we used two-way repeated ANOVA with Bonferroni post-hoc analysis. The drug-time interaction was significant (F143,1430=6.723 P<0.001).
Figure 4. Rapid antidepressant-like behaviour mediated by…
Figure 4. Rapid antidepressant-like behaviour mediated by decreased p-eEF2 and increased BDNF translation
a, Images of CA1 pyramidal and stratum radiatum layers after acute vehicle, ketamine, or MK-801; scale bar=100 μm (red: peEF2, blue: DAPI). b, Stratum radiatum magnification; scale bar=20 μm. c, ImageJ analysis of average fluorescence intensity. ANOVA cell layer F2,23=13.13, P=0.0002 for treatment, dendrites F2,23=14.06, P=0.0001 for treatment (n=4/group). d, Densitometric analysis of peEF2 (normalized-total eEF2) in hippocampus after NMDAR antagonist. ANOVA F2,23=3.183, P=0.03 for treatment (n=8/group). e-h, Densitometric analysis. e, g, Significant increases in hippocampal BDNF protein (normalized-GAPDH) with rottlerin (5.0 mg/kg) versus vehicle (t-test *P<0.05), and NH125 (5.0 mg/kg) versus vehicle (*P<0.05). f, h, Significant decreases in peEF2 (normalized-total eEF2) versus vehicle (*P<0.05) and NH125 versus vehicle (t-test *P<0.05). i, Immobility in FST of WTs given acute rottlerin (5.0 mg/kg) or NH125 (5.0 mg/kg). ANOVA F3, 44= 8.13, P=0.0002 for treatment, Bonferroni post-hoc analysis shows significance with rottlerin or NH125 versus vehicle (*P<0.05), but not ERK inhibitor SL327 (10mg/kg). j, Immobility of BDNF KO or littermate CTLs given acute rottlerin (5.0 mg/kg), tested 30-minutes later in FST. ANOVA F1, 19=5.77, P=0.0267 for treatment, Bonferroni post-hoc analysis for rottlerin versus vehicle CTLs (*P<0.05) (n=5-7/group).

References

    1. Zarate CA, Jr, et al. A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression. Arch Gen Psychiatry. 2006;63(8):856–864.
    1. Berman RM, et al. Antidepressant effects of ketamine in depressed patients. Biol Psychiatry. 2000;47(4):351–354.
    1. Price RB, Nock MK, Charney DS, Mathew SJ. Effects of intravenous ketamine on explicit and implicit measures of suicidality in treatment-resistant depression. Biol Psychiatry. 2009;66(5):522–526.
    1. Maeng S, et al. Cellular mechanisms underlying the antidepressant effects of ketamine: role of alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptors. Biol Psychiatry. 2008;63(4):349–352.
    1. Detke MJ, Johnson J, Lucki I. Acute and chronic antidepressant drug treatment in the rat forced swimming test model of depression. Exp Clin Psychopharmacol. 1997;5(2):107–112.
    1. Sinner B, Graf BM. Ketamine. Handb Exp Pharmacol. 2008;(182):313–333.
    1. Schwartz PH, Wasterlain CG. Cardiac arrest and resuscitation alters the pharmacokinetics of MK-801 in the rat. Res Commun Chem Pathol Pharmacol. 1991;73(2):181–195.
    1. Kristensen JD, Hartvig P, Karlsten R, Gordh T, Halldin M. CSF and plasma pharmacokinetics of the NMDA receptor antagonist CPP after intrathecal, extradural and i.v. administration in anaesthetized pigs. Br J Anaesth. 1995;74(2):193–200.
    1. Chen B, Dowlatshahi D, MacQueen GM, Wang JF, Young LT. Increased hippocampal BDNF immunoreactivity in subjects treated with antidepressant medication. Biol Psychiatry. 2001;50(4):260–265.
    1. Monteggia LM, et al. Essential role of brain-derived neurotrophic factor in adult hippocampal function. Proc Natl Acad Sci U S A. 2004;101(29):10827–10832.
    1. Berton O, et al. Essential role of BDNF in the mesolimbic dopamine pathway in social defeat stress. Science. 2006;311(5762):864–868.
    1. Adachi M, Barrot M, Autry AE, Theobald D, Monteggia LM. Selective loss of brain-derived neurotrophic factor in the dentate gyrus attenuates antidepressant efficacy. Biol Psychiatry. 2008;63(7):642–649.
    1. Shirayama Y, Chen AC, Nakagawa S, Russell DS, Duman RS. Brain-derived neurotrophic factor produces antidepressant effects in behavioral models of depression. J Neurosci. 2002;22(8):3251–3261.
    1. Hoshaw BA, Malberg JE, Lucki I. Central administration of IGF-I and BDNF leads to long-lasting antidepressant-like effects. Brain Res. 2005;1037(1-2):204–208.
    1. Akbarian S, et al. Brain-derived neurotrophic factor is essential for opiate-induced plasticity of noradrenergic neurons. J Neurosci. 2002;22(10):4153–4162.
    1. Lattal KM, Abel T. Different requirements for protein synthesis in acquisition and extinction of spatial preferences and context-evoked fear. J Neurosci. 2001;21(15):5773–5780.
    1. Capasso A, Di Giannuario A, Loizzo A, Pieretti S, Sorrentino L. Actinomycin D blocks the reducing effect of dexamethasone on amphetamine and cocaine hypermotility in mice. Gen Pharmacol. 1996;27(4):707–712.
    1. Sutton MA, Taylor AM, Ito HT, Pham A, Schuman EM. Postsynaptic decoding of neural activity: eEF2 as a biochemical sensor coupling miniature synaptic transmission to local protein synthesis. Neuron. 2007;55(4):648–661.
    1. Poleszak E, et al. NMDA/glutamate mechanism of antidepressant-like action of magnesium in forced swim test in mice. Pharmacol Biochem Behav. 2007;88(2):158–164.
    1. Fernandez F, et al. Pharmacotherapy for cognitive impairment in a mouse model of Down syndrome. Nat Neurosci. 2007;10(4):411–413.
    1. Li N, et al. mTOR-dependent synapse formation underlies the rapid antidepressant effects of NMDA antagonists. Science. 329(5994):959–964.
    1. Cleary C, et al. Antidepressive-like effects of rapamycin in animal models: Implications for mTOR inhibition as a new target for treatment of affective disorders. Brain Res Bull. 2008;76(5):469–473.
    1. Atasoy D, et al. Spontaneous and evoked glutamate release activates two populations of NMDA receptors with limited overlap. J Neurosci. 2008;28(40):10151–10166.
    1. Espinosa F, Kavalali ET. NMDA receptor activation by spontaneous glutamatergic neurotransmission. J Neurophysiol. 2009;101(5):2290–2296.
    1. Tanaka J, et al. Protein synthesis and neurotrophin-dependent structural plasticity of single dendritic spines. Science. 2008;319(5870):1683–1687.
    1. Jakawich SK, et al. Local presynaptic activity gates homeostatic changes in presynaptic function driven by dendritic BDNF synthesis. Neuron. 68(6):1143–1158.
    1. Lindskog M, et al. Postsynaptic GluA1 enables acute retrograde enhancement of presynaptic function to coordinate adaptation to synaptic inactivity. Proc Natl Acad Sci U S A
    1. Sutton MA, Schuman EM. Partitioning the synaptic landscape: distinct microdomains for spontaneous and spike-triggered neurotransmission. Sci Signal. 2009;2(65):pe19.
    1. Kavalali ET, et al. Spontaneous neurotransmission: an independent pathway for neuronal signaling? Physiology (Bethesda) 26(1):45–53.
    1. Duman CH, Schlesinger L, Kodama M, Russell DS, Duman RS. A role for MAP kinase signalling in behavioral models of depression and antidepressant treatment. Biol Psychiatry. 2007;61:661–70.
    1. Porsolt RD, Le Pichon M, Jalfre M. Depression: A new animal model sensitive to antidepressant treatments. Nature. 1977;266:730–732.
    1. Park S, et al. Elongation factor 2 and fragile X mental retardation protein control the dynamic translation of Arc/Arg3.1 essential for mGluR-LTD. Neuron. 2008;59:70–83.
    1. Kavalali ET, Klingauf J, Tsien RW. Activity-dependent regulation of synaptic clustering in a hippocampal culture system. Proc Natl Acad Sci U S A. 1999;96:12893–12900.

Source: PubMed

3
Sottoscrivi