Altered expression of synapse and glutamate related genes in post-mortem hippocampus of depressed subjects

Vanja Duric, Mounira Banasr, Craig A Stockmeier, Arthur A Simen, Samuel S Newton, James C Overholser, George J Jurjus, Lesa Dieter, Ronald S Duman, Vanja Duric, Mounira Banasr, Craig A Stockmeier, Arthur A Simen, Samuel S Newton, James C Overholser, George J Jurjus, Lesa Dieter, Ronald S Duman

Abstract

Major depressive disorder (MDD) has been linked to changes in function and activity of the hippocampus, one of the central limbic regions involved in regulation of emotions and mood. The exact cellular and molecular mechanisms underlying hippocampal plasticity in response to stress are yet to be fully characterized. In this study, we examined the genetic profile of micro-dissected subfields of post-mortem hippocampus from subjects diagnosed with MDD and comparison subjects matched for sex, race and age. Gene expression profiles of the dentate gyrus and CA1 were assessed by 48K human HEEBO whole genome microarrays and a subgroup of identified genes was confirmed by real-time polymerase chain reaction (qPCR). Pathway analysis revealed altered expression of several gene families, including cytoskeletal proteins involved in rearrangement of neuronal processes. Based on this and evidence of hippocampal neuronal atrophy in MDD, we focused on the expression of cytoskeletal, synaptic and glutamate receptor genes. Our findings demonstrate significant dysregulation of synaptic function/structure related genes SNAP25, DLG2 (SAP93), and MAP1A, and 2-amino-3-(5-methyl-3-oxo-1,2-oxazol-4-yl)propanoic acid receptor subunit genes GLUR1 and GLUR3. Several of these human target genes were similarly dysregulated in a rat model of chronic unpredictable stress and the effects reversed by antidepressant treatment. Together, these studies provide new evidence that disruption of synaptic and glutamatergic signalling pathways contribute to the pathophysiology underlying MDD and provide interesting targets for novel therapeutic interventions.

Figures

Fig. 1
Fig. 1
Expression levels of synapse-related genes are altered in major depressive disorder (MDD). Microarray findings were validated by real-time polymerase chain reaction on subset of samples from the same cohort. mRNA levels of selected genes involved in synaptic functioning (a) SNAP25, (b) DLG2 (SAP93), (c) MAP1A, and (d) MAP1B) are shown. Data are expressed as mean fold change ±S.E.M.(n=6) ; * p≤0.05 compared to the healthy controls (paired Student’s t test). DG, dentate gyrus.
Fig. 2
Fig. 2
Expression levels of glutamate α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors are altered in major depressive disorder (MDD). Microarray findings were validated by real-time polymerase chain reaction on a subset of samples from the same cohort. mRNA levels of (a) GLUR1 and (b) GLUR3 are shown. Data are expressed as mean fold change±S.E.M. (n=6) ; * p≤0.05 compared to the healthy controls (paired Student’s t test). DG, dentate gyrus.
Fig. 3
Fig. 3
Expression levels of serotonin receptors are altered in major depressive disorder (MDD). Microarray findings were validated by real-time polymerase chain reaction on a subset of samples from the same cohort. mRNA levels of (a) HTR2C, (b) HTR4 and (c) HTR7 are shown. Data are expressed as mean fold change ±S.E.M.(n=5–6) ; * p≤0.05 compared to the healthy controls (paired Student’s t test). DG, dentate gyrus.
Fig. 4
Fig. 4
Effects of chronic unpredictable stress (CUS) and antidepressant treatment on expression of selected human target genes in the rat hippocampus. (a) Rats were exposed to CUS or control housing conditions ; saline (vehicle) or fluoxetine (Flx) were administered for the last 21 d of stress. CUS had no effect on the locomotor activity (LA), but evoked increased helpless behaviour in active avoidance test (AAT) and reduced sucrose intake in sucrose preference test (SPT) ; both of these behavioural effects of CUS were reversed by chronic Flx treatment (data not shown). Representative autoradiographs and quantitative analysis of (b) Snap25, (c) GluR1, (d) GluR3 and (e) Htr4 mRNA levels by in situ hybridization on coronal sections of dorsal rat hippocampus (scale bar, 1.0 mm). Results are expressed as mean±s.e.m. percent change over non-stressed control group (n=4or 5) ; * p<0.05 compared to the non-stressed control group, #p<0.05 compared to CUS group, $p<0.07 compared to CUS group (two-way analysis of variance and Fisher’s PLSD post-hoc analysis).

References

    1. APA . Diagnostic and Statistical Manual of Mental Health Disorders. 4th edn American Psychiatric Association; Washington DC: 1994.
    1. Banasr M, Chowdhury GM, Terwilliger R, Newton SS, et al. Glial pathology in an animal model of depression : reversal of stress-induced cellular, metabolic and behavioral deficits by the glutamate-modulating drug riluzole. Molecular Psychiatry. 2010;15:501–511.
    1. Banasr M, Duman RS. Regulation of neurogenesis and gliogenesis by stress and antidepressant treatment. CNS and Neurological Disorders Drug Targets. 2007;6:311–320.
    1. Banasr M, Duman RS. Glial loss in the prefrontal cortex is sufficient to induce depressive-like behaviors. Biological Psychiatry. 2008;64:863–870.
    1. Banasr M, Valentine GW, Li XY, Gourley SL, et al. Chronic unpredictable stress decreases cell proliferation in the cerebral cortex of the adult rat. Biological Psychiatry. 2007;62:496–504.
    1. Bear MF, Malenka RC. Synaptic plasticity : LTP and LTD. Current Opinion in Neurobiology. 1994;4:389–399.
    1. Beneyto M, Meador-Woodruff JH. Lamina-specific abnormalities of AMPA receptor trafficking and signaling molecule transcripts in the prefrontal cortex in schizophrenia. Synapse. 2006;60:585–598.
    1. Benowitz LI, Perrone-Bizzozero NI. The relationship of GAP-43 to the development and plasticity of synaptic connections. Annals of the New York Academy of Sciences. 1991;627:58–74.
    1. Bernard R, Kerman IA, Thompson RC, Jones EG, et al. Altered expression of glutamate signaling, growth factor, and glia genes in the locus coeruleus of patients with major depression. Molecular Psychiatry. 2010;16:634–646.
    1. Bruneau EG, Akaaboune M. Running to stand still : ionotropic receptor dynamics at central and peripheral synapses. Molecular Neurobiology. 2006;34:137–151.
    1. Coppen AJ, Doogan DP. Serotonin and its place in the pathogenesis of depression. Journal of Clinical Psychiatry. 1988;49(Suppl.):4–11.
    1. Cui X, Hwang JT, Qiu J, Blades NJ, et al. Improved statistical tests for differential gene expression by shrinking variance components estimates. Biostatistics. 2005;6:59–75.
    1. Duman RS. Structural alterations in depression: cellular mechanisms underlying pathology and treatment of mood disorders. CNS Spectrums. 2002;7:140–142. 144–147.
    1. Duman RS. Role of neurotrophic factors in the etiology and treatment of mood disorders. Neuromolecular Medicine. 2004;5:11–25.
    1. Duman RS. Neuronal damage and protection in the pathophysiology and treatment of psychiatric illness : stress and depression. Dialogues in Clinical Neuroscience. 2009;11:239–255.
    1. Duman RS, Heninger GR, Nestler EJ. A molecular and cellular theory of depression. Archives of General Psychiatry. 1997;54:597–606.
    1. Duric V, Banasr M, Licznerski P, Schmidt HD, et al. A negative regulator of MAP kinase causes depressive behavior. Nature Medicine. 2010;16:1328–1332.
    1. Errico M, Crozier RA, Plummer MR, Cowen DS. 5-HT(7) receptors activate the mitogen activated protein kinase extracellular signal related kinase in cultured rat hippocampal neurons. Neuroscience. 2001;102:361–367.
    1. Farley S, Apazoglou K, Witkin JM, Giros B, et al. Antidepressant-like effects of an AMPA receptor potentiator under a chronic mild stress paradigm. International Journal of Neuropsychopharmacology. 2010;13:1207–1218.
    1. Fava M, Davidson KG. Definition and epidemiology of treatment-resistant depression. Psychiatric Clinics of North America. 1996;19:179–200.
    1. Feyissa AM, Chandran A, Stockmeier CA, Karolewicz B. Reduced levels of NR2A and NR2B subunits of NMDA receptor and PSD-95 in the prefrontal cortex in major depression. Progress in Neuro-psychopharmacology and Biological Psychiatry. 2009;33:70–75.
    1. First MB, Donovan S, Frances A. Nosology of chronic mood disorders. Psychiatric Clinics of North America. 1996;19:29–39.
    1. Graeff FG, Guimaraes FS, De Andrade TG, Deakin JF. Role of 5-HT in stress, anxiety, and depression. Pharmacology, Biochemistry, and Behavior. 1996;54:129–141.
    1. Greenberg PE, Kessler RC, Birnbaum HG, Leong SA, et al. The economic burden of depression in the United States : how did it change between 1990 and 2000? Journal of Clinical Psychiatry. 2003;64:1465–1475.
    1. Hashimoto K, Sawa A, Iyo M. Increased levels of glutamate in brains from patients with mood disorders. Biological Psychiatry. 2007;62:1310–1316.
    1. Heisler LK, Zhou L, Bajwa P, Hsu J, et al. Serotonin 5-HT(2C) receptors regulate anxiety-like behavior. Genes, Brain, and Behavior. 2007;6:491–496.
    1. Hsiung SC, Adlersberg M, Arango V, Mann JJ, et al. Attenuated 5-HT1A receptor signaling in brains of suicide victims : involvement of adenylyl cyclase, phosphati-dylinositol 3-kinase, Akt and mitogen-activated protein kinase. Journal of Neurochemistry. 2003;87:182–194.
    1. Jones BJ, Blackburn TP. The medical benefit of 5-HT research. Pharmacology, Biochemistry, and Behavior. 2002;71:555–568.
    1. Kessler RC, Berglund P, Demler O, Jin R, et al. The epidemiology of major depressive disorder : results from the National Comorbidity Survey Replication (NCS-R) Journal of the American Medical Association. 2003;289:3095–3105.
    1. Kim JY, Duan X, Liu CY, Jang MH, et al. DISC1 regulates new neuron development in the adult brain via modulation of AKT-mTOR signaling through KIAA1212. Neuron. 2009;63:761–773.
    1. Knable MB, Barci BM, Webster MJ, Meador-Woodruff J, et al. Molecular abnormalities of the hippocampus in severe psychiatric illness : postmortem findings from the Stanley Neuropathology Consortium. Molecular Psychiatry. 2004;9:609–620.
    1. Koo JW, Duman RS. Evidence for IL-1 receptor blockade as a therapeutic strategy for the treatment of depression. Current Opinion in Investigational Drugs. 2009;10:664–671.
    1. Lajoie-Mazenc I, Tovar D, Penary M, Lortal B, et al. MAP1A light chain-2 interacts with GTP-RhoB to control epidermal growth factor (EGF)-dependent EGF receptor signaling. Journal of Biological Chemistry. 2008;283:4155–4164.
    1. Law AJ, Deakin JF. Asymmetrical reductions of hippocampal NMDAR1 glutamate receptor mRNA in the psychoses. Neuroreport. 2001;12:2971–2974.
    1. Li N, Liu RJ, Dwyer JM, Banasr M, et al. Glutamate N-methyl-d-aspartate receptor antagonists rapidly reverse behavioral and synaptic deficits caused by chronic stress exposure. Biological Psychiatry. 2011;69:754–761.
    1. Little A. Treatment-resistant depression. American Family Physician. 2009;80:167–172.
    1. Lopez JF, Vazquez DM, Chalmers DT, Watson SJ. Regulation of 5-HT receptors and the hypothalamic-pituitary-adrenal axis. Implications for the neurobiology of suicide. Annals of the New York Academy of Sciences. 1997;836:106–134.
    1. Lopez-Figueroa AL, Norton CS, Lopez-Figueroa MO, Armellini-Dodel D, et al. Serotonin 5-HT1A, 5-HT1B, and 5-HT2A receptor mRNA expression in subjects with major depression, bipolar disorder, and schizophrenia. Biological Psychiatry. 2004;55:225–233.
    1. Lucas G, Rymar VV, Du J, Mnie-Filali O, et al. Serotonin(4) (5-HT(4)) receptor agonists are putative antidepressants with a rapid onset of action. Neuron. 2007;55:712–725.
    1. McCullumsmith RE, Kristiansen LV, Beneyto M, Scarr E, et al. Decreased NR1, NR2A, and SAP102 transcript expression in the hippocampus in bipolar disorder. Brain Research. 2007;1127:108–118.
    1. Magarinos AM, Deslandes A, McEwen BS. Effects of antidepressants and benzodiazepine treatments on the dendritic structure of CA3 pyramidal neurons after chronic stress. European Journal of Pharmacology. 1999;371:113–122.
    1. Malenka RC. Synaptic plasticity and AMPA receptor trafficking. Annals of the New York Academy of Sciences. 2003;1003:1–11.
    1. Manji HK, Quiroz JA, Sporn J, Payne JL, et al. Enhancing neuronal plasticity and cellular resilience to develop novel, improved therapeutics for difficult-to-treat depression. Biological Psychiatry. 2003;53:707–742.
    1. Meador-Woodruff JH, Hogg AJ, II, Smith RE. Striatal ionotropic glutamate receptor expression in schizophrenia, bipolar disorder, and major depressive disorder. Brain Research Bulletin. 2001;55:631–640.
    1. Middlemiss DN, Price GW, Watson JM. Serotonergic targets in depression. Current Opinion in Pharmacology. 2002;2:18–22.
    1. Millan MJ. Serotonin 5-HT2C receptors as a target for the treatment of depressive and anxious states : focus on novel therapeutic strategies. Therapie. 2005;60:441–460.
    1. Millan MJ. Multi-target strategies for the improved treatment of depressive states : conceptual foundations and neuronal substrates, drug discovery and therapeutic application. Pharmacology and Therapeutics. 2006;110:135–370.
    1. Mintun MA, Sheline YI, Moerlein SM, Vlassenko AG, et al. Decreased hippocampal 5-HT2A receptor binding in major depressive disorder : in vivo measurement with [18F]altanserin positron emission tomography. Biological Psychiatry. 2004;55:217–224.
    1. Morris JA, Kandpal G, Ma L, Austin CP. DISC1 (Disrupted-In-Schizophrenia 1) is a centrosome-associated protein that interacts with MAP1A, MIPT3, ATF4/5 and NUDEL : regulation and loss of interaction with mutation. Human Molecular Genetics. 2003;12:1591–1608.
    1. Murua VS, Gomez RA, Andrea ME, Molina VA. Shuttle-box deficits induced by chronic variable stress : reversal by imipramine administration. Pharmacology, Biochemistry, and Behavior. 1991;38:125–130.
    1. Naylor P, Stewart CA, Wright SR, Pearson RC, et al. Repeated ECS induces GluR1 mRNA but not NMDAR1AG mRNA in the rat hippocampus. Brain Research. 1996;35:349–353.
    1. Neumeister A, Wood S, Bonne O, Nugent AC, et al. Reduced hippocampal volume in unmedicated, remitted patients with major depression vs. control subjects. Biological Psychiatry. 2005;57:935–937.
    1. Newton SS, Collier EF, Hunsberger J, Adams D, et al. Gene profile of electroconvulsive seizures : induction of neurotrophic and angiogenic factors. Journal of Neuroscience. 2003;23:10841–10851.
    1. Newton SS, Dow A, Terwilliger R, Duman R. A simplified method for combined immunohistochemistry and in-situ hybridization in fresh-frozen, cryocut mouse brain sections. Brain Research Protocols. 2002;9:214–219.
    1. Nudmamud-Thanoi S, Reynolds GP. The NR1 subunit of the glutamate/NMDA receptor in the superior temporal cortex in schizophrenia and affective disorders. Neuroscience Letters. 2004;372:173–177.
    1. Ossowska G, Nowa G, Kata R, Klenk-Majewska B, et al. Brain monoamine receptors in a chronic unpredictable stress model in rats. Journal of Neural Transmission. 2001;108:311–319.
    1. Pandey GN, Dwivedi Y, Rizavi HS, Ren X, et al. Higher expression of serotonin 5-HT(2A) receptors in the postmortem brains of teenage suicide victims. American Journal of Psychiatry. 2002;159:419–429.
    1. Pittenger C, Duman RS. Stress, depression, and neuroplasticity : a convergence of mechanisms. Neuropsychopharmacology. 2008;33:88–109.
    1. Rapp S, Baader M, Hu M, Jennen-Steinmetz C, et al. Differential regulation of synaptic vesicle proteins by antidepressant drugs. Pharmacogenomics Journal. 2004;4:110–113.
    1. Rosel P, Arranz B, Urretavizcaya M, Oros M, et al. Altered 5-HT2A and 5-HT4 postsynaptic receptors and their intracellular signalling systems IP3 and cAMP in brains from depressed violent suicide victims. Neuropsychobiology. 2004;49:189–195.
    1. Rosenzweig-Lipson S, Dunlop J, Marquis KL. 5-HT2C receptor agonists as an innovative approach for psychiatric disorders. Drug News and Perspectives. 2007a;20:565–571.
    1. Rosenzweig-Lipson S, Sabb A, Stack G, Mitchell P, et al. Antidepressant-like effects of the novel, selective, 5-HT2C receptor agonist WAY-163909 in rodents. Psychopharmacology. 2007b;192:159–170.
    1. Schmidt HD, Duman RS. The role of neurotrophic factors in adult hippocampal neurogenesis, antidepressant treatments and animal models of depressive-like behavior. Behavioural Pharmacology. 2007;18:391–418.
    1. Sheline YI, Gado MH, Kraemer HC. Untreated depression and hippocampal volume loss. American Journal of Psychiatry. 2003;160:1516–1518.
    1. Sheline YI, Wang PW, Gado MH, Csernansky JG, et al. Hippocampal atrophy in recurrent major depression. Proceedings of the National Academy of Sciences USA. 1996;93:3908–3913.
    1. Sheng M, Lee SH. AMPA receptor trafficking and the control of synaptic transmission. Cell. 2001;105:825–828.
    1. Simon GE. Social and economic burden of mood disorders. Biological Psychiatry. 2003;54:208–215.
    1. Stockmeier CA. Involvement of serotonin in depression : evidence from postmortem and imaging studies of serotonin receptors and the serotonin transporter. Journal of Psychiatric Research. 2003;37:357–373.
    1. Stockmeier CA, Mahajan GJ, Konick LC, Overholser JC, et al. Cellular changes in the postmortem hippocampus in major depression. Biological Psychiatry. 2004;56:640–650.
    1. Stockmeier CA, Shapiro LA, Dilley GE, Kolli TN, et al. Increase in serotonin-1A autoreceptors in the midbrain of suicide victims with major depression-postmortem evidence for decreased serotonin activity. Journal of Neuroscience. 1998;18:7394–7401.
    1. Storey JD, Tibshirani R. Statistical significance for genomewide studies. Proceedings of the National Academy of Sciences USA. 2003;100:9440–9445.
    1. Svenningsson P, Tzavara ET, Witkin JM, Fienberg AA, et al. Involvement of striatal and extrastriatal DARPP-32 in biochemical and behavioral effects of fluoxetine (Prozac) Proceedings of the National Academy of Sciences USA. 2002;99:3182–3187.
    1. Thompson PM, Egbufoama S, Vawter MP. SNAP-25 reduction in the hippocampus of patients with schizophrenia. Progress in Neuro-psychopharmacology and Biological Psychiatry. 2003;27:411–417.
    1. Tucker RP. The roles of microtubule-associated proteins in brain morphogenesis : a review. Brain Research Review. 1990;15:101–120.
    1. Vaidya VA, Terwilliger RM, Duman RS. Role of 5-HT2A receptors in the stress-induced down-regulation of brain-derived neurotrophic factor expression in rat hippocampus. Neuroscience Letters. 1999;262:1–4.
    1. Vawter MP, Thatcher L, Usen N, Hyde TM, et al. Reduction of synapsin in the hippocampus of patients with bipolar disorder and schizophrenia. Molecular Psychiatry. 2002;7:571–578.
    1. Wang M, Yang Y, Dong Z, Cao J, et al. NR2B-containing N-methyl-d-aspartate subtype glutamate receptors regulate the acute stress effect on hippocampal long-term potentiation/long-term depression in vivo. Neuroreport. 2006;17:1343–1346.
    1. Watanabe Y, Gould E, McEwen BS. Stress induces atrophy of apical dendrites of hippocampal CA3 pyramidal neurons. Brain Research. 1992;588:341–345.
    1. Wenthold RJ, Petralia RS, Blahos J, II, Niedzielski AS. Evidence for multiple AMPA receptor complexes in hippocampal CA1/CA2 neurons. Journal of Neuroscience. 1996;16:1982–1989.
    1. Willner P. Chronic mild stress (CMS) revisited : consistency and behavioural-neurobiological concordance in the effects of CMS. Neuropsychobiology. 2005;52:90–110.
    1. Wong ML, Smith MA, Licinio J, Doi SQ, et al. Differential effects of kindled and electrically induced seizures on a glutamate receptor (GluR1) gene expression. Epilepsy Research. 1993;14:221–227.
    1. Yamada M, Takahashi K, Tsunoda M, Nishioka G, et al. Differential expression of VAMP2/synaptobrevin2 after antidepressant and electroconvulsive treatment in rat frontal cortex. Pharmacogenomics Journal. 2002;2:377–382.

Source: PubMed

3
Subscribe