GPR56/ADGRG1 is associated with response to antidepressant treatment

Raoul Belzeaux, Victor Gorgievski, Laura M Fiori, Juan Pablo Lopez, Julien Grenier, Rixing Lin, Corina Nagy, El Chérif Ibrahim, Eduardo Gascon, Philippe Courtet, Stéphane Richard-Devantoy, Marcelo Berlim, Eduardo Chachamovich, Jean-François Théroux, Sylvie Dumas, Bruno Giros, Susan Rotzinger, Claudio N Soares, Jane A Foster, Naguib Mechawar, Gregory G Tall, Eleni T Tzavara, Sidney H Kennedy, Gustavo Turecki, Raoul Belzeaux, Victor Gorgievski, Laura M Fiori, Juan Pablo Lopez, Julien Grenier, Rixing Lin, Corina Nagy, El Chérif Ibrahim, Eduardo Gascon, Philippe Courtet, Stéphane Richard-Devantoy, Marcelo Berlim, Eduardo Chachamovich, Jean-François Théroux, Sylvie Dumas, Bruno Giros, Susan Rotzinger, Claudio N Soares, Jane A Foster, Naguib Mechawar, Gregory G Tall, Eleni T Tzavara, Sidney H Kennedy, Gustavo Turecki

Abstract

It remains unclear why many patients with depression do not respond to antidepressant treatment. In three cohorts of individuals with depression and treated with serotonin-norepinephrine reuptake inhibitor (N = 424) we show that responders, but not non-responders, display an increase of GPR56 mRNA in the blood. In a small group of subjects we also show that GPR56 is downregulated in the PFC of individuals with depression that died by suicide. In mice, we show that chronic stress-induced Gpr56 downregulation in the blood and prefrontal cortex (PFC), which is accompanied by depression-like behavior, and can be reversed by antidepressant treatment. Gpr56 knockdown in mouse PFC is associated with depressive-like behaviors, executive dysfunction and poor response to antidepressant treatment. GPR56 peptide agonists have antidepressant-like effects and upregulated AKT/GSK3/EIF4 pathways. Our findings uncover a potential role of GPR56 in antidepressant response.

Trial registration: ClinicalTrials.gov NCT00635219 NCT00599911 NCT01140906 NCT02209142.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1. GPR56 mRNA is related to…
Fig. 1. GPR56 mRNA is related to antidepressant response.
a In the discovery cohort, 237 patients in a major depressive episode were randomized to double-blind treatment with either duloxetine (n = 112) or placebo (n = 125), for up to 8 weeks. Using two-class paired significant analysis of microarray (SAM) with correction for multiple testing (FDR < 1%) in patients who responded to duloxetine, GPR56 mRNA is the most significantly upregulated mRNA in whole blood after duloxetine treatment, based on fold change and q-value (FC = 1.19, q-value < 0.01). General linear model (GLM) demonstrated a time x treatment interaction, F(1,199)=8.468, p = 0.004, that confirms the specificity of GPR56 mRNA increase in responders to duloxetine. b In the first replication cohort (Montréal), patients were treated with citalopram in an open-label trial. GPR56 mRNA in whole blood demonstrated an increase only in responders. GLM demonstrated a time x group interaction, F(1,61)=4.27, p = 0.043 (not adjusted for multiple testing). c In the second replication cohort (Marseille), psychiatrically healthy subjects and patients with depression were included in a naturalistic design. In patients who responded and achieved remission after 30 weeks of treatment (responders-remitters, n = 20), GPR56 mRNA is not different at inclusion and 2 weeks, however was then overexpressed at 8 weeks in comparison to others (n = 44) (two-sided t-test t = 2.085, p = 0.049) and remained stably overexpressed over a 30-week follow-up (n = 18 responders-remitters in comparison to others n = 31) two-sided t-test t = 3.076, p = 0.005); Linear Mixed model (F(1,230.199) = 14.79, p = 0.0001). Bars represent mean. Error bars represent standard error of the mean. **p < 0.01, *p < 0.05. Source data are provided as a Source Data file.
Fig. 2. Unpredictable chronic mild stress (UCMS)…
Fig. 2. Unpredictable chronic mild stress (UCMS) and antidepressant response dysregulate Gpr56 mRNA in blood and CNS in mice.
aGpr56 expression was analysed by qRT-PCR in blood and several brain regions, including the dorsal and ventral hippocampal areas (HD and HV, respectively), the prefrontal cortex (PFC) and the Nucleus Accumbens (NAcc) in non-stressed or stressed mice (exposed to UCMS) and receiving vehicle or fluoxetine. Mice treated by fluoxetine were classified as “responders” or “non-responders” according to behavioral tests. b In whole blood, a one-way ANOVA showed between group differences for Gpr56 expression (F = 6,150, p = 0.001). Blood Gpr56 mRNA expression was decreased in mice subjected to UCMS, while reversal of depressive-like behaviors with fluoxetine was paralleled by normalization of blood Gpr56 mRNA expression in responder mice (post hoc analysis p < 0.01). c In brain, a two-way ANOVA between group and brain regions showed a significant interaction between brain region and mice group (F(9,151) = 3.112; p = 0.0018). Post hoc analysis demonstrated a specific PFC effect, a decrease of Gpr56 in PFC between stressed and non-stressed mice, with a reversal effect of antidepressant only in responder mice. Sample numbers vary between tissues due to removal of poor quality RNA samples from the analyses. Bars represent mean. Error bars represent standard error of the mean. **p < 0.01. Graph represents Box and Whiskers Min to Max. Source data are provided as a Source Data file.
Fig. 3. Gpr56 regulates depressive-like behaviors.
Fig. 3. Gpr56 regulates depressive-like behaviors.
a Mice were injected bilaterally in the pre-frontal cortex with control (CTL) or lentivirus-Gpr56 constructs, as well as with CTL or lentivirus-sh-Gpr56 (inhibitor) constructs. b Overexpression of Gpr56 (n = 15) was associated with lower immobility time in the TST in comparison to control animals (n = 12), two-sided t = 3.07, p = 0.005. c Downregulation of Gpr56 (n = 8) produced increased immobility time, i.e. depressive-like behavior in the tail suspension test (TST) in comparison to control animals (n = 11), two-sided t = 2.203, p = 0.048. d Fluoxetine decreased immobility in control animals (n = 5), while this effect was strongly attenuated in sh-Gpr56-virus infused animals (n = 6); two-way ANOVA for repeated measures; treatment × group interaction F(1,9) = 6.80, p = 0.028; main effects for group F (1,9) = 25.4, p = 0.001 (as compared to control) and for treatment F(1,9) = 6.80 p = 0.001 (as compared to vehicle). e GPR56 agonist P7 has antidepressant-like effects. GPR56 agonist P7 peptide infused bilaterally with escalating doses in PFC decreases immobility time and demonstrates antidepressant-like effects, ANOVA F(1,4) = 4.88 p = 0.008. In comparison to vehicle (n = 13), both 1 mM (n = 7) and 2 mM (n = 4) doses demonstrate a significant decrease of immobility time (p < 0.01 and p = 0.01 respectively, two-sided post hoc test). Bars represent mean. Error bars represent standard error of the mean. **p < 0.01, *p < 0.05. Source data are provided as a Source Data file.
Fig. 4. GPR56 expression is altered in…
Fig. 4. GPR56 expression is altered in the prefrontal cortex from post-mortem brain tissue of individuals with depression and is related to pre-frontal cortex functioning in patients.
aGPR56 expression was measured by qRT-PCR in post-mortem brain tissue (BA44). Expression was lower in individuals with depression (MDD, n = 49) in comparison to psychiatrically healthy controls (n = 26). FC = 0.56, two-sided U = 385, Z = −2.81, p = 0.005. Graph represents Box and Whiskers Min to Max. b Changes in Stroop interference score, a neuropsychological test that involves pre-frontal cortex function, were correlated with changes in GPR56 expression in whole blood following antidepressant treatment (n = 12 from discovery cohort, Pearson coefficient of correlation = −0.71, two-sided p = 0.009). Reduction of interference score was associated with an improvement of Stroop interference test, i.e., an improvement in pre-frontal cortex functioning. Source data are provided as a Source Data file.

References

    1. Waraich P, Goldner EM, Somers JM, Hsu L. Prevalence and incidence studies of mood disorders: a systematic review of the literature. Can. J. Psychiatry. 2004;49:124–138. doi: 10.1177/070674370404900208.
    1. WHO. Depression and Other Common Mental Disorders: Global Health Estimates (2017).
    1. Kennedy SH, et al. Canadian Network for Mood and Anxiety Treatments (CANMAT) 2016 Clinical Guidelines for the Management of Adults with Major Depressive Disorder: Section 3. Pharmacological Treatments. Can. J. Psychiatry. 2016;61:540–560. doi: 10.1177/0706743716659417.
    1. Rush AJ, et al. Acute and longer-term outcomes in depressed outpatients requiring one or several treatment steps: a STAR*D report. Am. J. Psychiatry. 2006;163:1905–1917. doi: 10.1176/ajp.2006.163.11.1905.
    1. Apazoglou K, et al. Antidepressive effects of targeting ELK-1 signal transduction. Nat. Med. 2018;24:591–597. doi: 10.1038/s41591-018-0011-0.
    1. McIntosh AL, Gormley S, Tozzi L, Frodl T, Harkin A. Recent advances in translational magnetic resonance imaging in animal models of stress and depression. Front. Cell Neurosci. 2017;11:150. doi: 10.3389/fncel.2017.00150.
    1. Suzuki G, et al. Stress and electroconvulsive seizure differentially alter GPR56 expression in the adult rat brain. Brain Res. 2007;1183:21–31. doi: 10.1016/j.brainres.2007.09.020.
    1. Svenningsson P, et al. Involvement of striatal and extrastriatal DARPP-32 in biochemical and behavioral effects of fluoxetine (Prozac) Proc. Natl Acad. Sci. USA. 2002;99:3182–3187. doi: 10.1073/pnas.052712799.
    1. Wagner S, et al. Early improvement of executive test performance during antidepressant treatment predicts treatment outcome in patients with major depressive disorder. PLoS ONE. 2018;13:e0194574. doi: 10.1371/journal.pone.0194574.
    1. El Khoury MA, Gorgievski V, Moutsimilli L, Giros B, Tzavara ET. Interactions between the cannabinoid and dopaminergic systems: evidence from animal studies. Prog. Neuropsychopharmacol. Biol. Psychiatry. 2012;38:36–50. doi: 10.1016/j.pnpbp.2011.12.005.
    1. Stoveken HM, Hajduczok AG, Xu L, Tall GG. Adhesion G protein-coupled receptors are activated by exposure of a cryptic tethered agonist. Proc. Natl Acad. Sci. USA. 2015;112:6194–6199. doi: 10.1073/pnas.1421785112.
    1. Stoveken HM, Larsen SD, Smrcka AV, Tall GG. Gedunin- and Khivorin-derivatives are small-molecule partial agonists for adhesion G protein-coupled receptors GPR56/ADGRG1 and GPR114/ADGRG5. Mol. Pharm. 2018;93:477–488. doi: 10.1124/mol.117.111476.
    1. Gupta M, et al. TSPAN5, ERICH3 and selective serotonin reuptake inhibitors in major depressive disorder: pharmacometabolomics-informed pharmacogenomics. Mol. Psychiatry. 2016;21:1717–1725. doi: 10.1038/mp.2016.6.
    1. Qi XR, et al. Abnormal retinoid and TrkB signaling in the prefrontal cortex in mood disorders. Cereb. Cortex. 2015;25:75–83. doi: 10.1093/cercor/bht203.
    1. Subramanian A, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA. 2005;102:15545–15550. doi: 10.1073/pnas.0506580102.
    1. Zanos P, Gould TD. Mechanisms of ketamine action as an antidepressant. Mol. Psychiatry. 2018;23:801–811. doi: 10.1038/mp.2017.255.
    1. Beaulieu JM. A role for Akt and glycogen synthase kinase-3 as integrators of dopamine and serotonin neurotransmission in mental health. J. Psychiatry Neurosci. 2012;37:7–16. doi: 10.1503/jpn.110011.
    1. Gould TD, Manji HK. Glycogen synthase kinase-3: a putative molecular target for lithium mimetic drugs. Neuropsychopharmacology. 2005;30:1223–1237. doi: 10.1038/sj.npp.1300731.
    1. Aguilar-Valles A, et al. Translational control of depression-like behavior via phosphorylation of eukaryotic translation initiation factor 4E. Nat. Commun. 2018;9:2459. doi: 10.1038/s41467-018-04883-5.
    1. Bae BI, et al. Evolutionarily dynamic alternative splicing of GPR56 regulates regional cerebral cortical patterning. Science. 2014;343:764–768. doi: 10.1126/science.1244392.
    1. Bai Y, Du L, Shen L, Zhang Y, Zhang L. GPR56 is highly expressed in neural stem cells but downregulated during differentiation. Neuroreport. 2009;20:918–922. doi: 10.1097/WNR.0b013e32832c92d7.
    1. Giera S, et al. The adhesion G protein-coupled receptor GPR56 is a cell-autonomous regulator of oligodendrocyte development. Nat. Commun. 2015;6:6121. doi: 10.1038/ncomms7121.
    1. Giera, S. et al. Microglial transglutaminase-2 drives myelination and myelin repair via GPR56/ADGRG1 in oligodendrocyte precursor cells. Elife7, e33385 (2018).
    1. Peng YM, et al. Specific expression of GPR56 by human cytotoxic lymphocytes. J. Leukoc. Biol. 2011;90:735–740. doi: 10.1189/jlb.0211092.
    1. Hamann J, et al. International Union of Basic and Clinical Pharmacology. XCIV. Adhesion G protein-coupled receptors. Pharm. Rev. 2015;67:338–367. doi: 10.1124/pr.114.009647.
    1. Della Chiesa M, et al. GPR56 as a novel marker identifying the CD56dull CD16+ NK cell subset both in blood stream and in inflamed peripheral tissues. Int. Immunol. 2010;22:91–100. doi: 10.1093/intimm/dxp116.
    1. Luo R, et al. G protein-coupled receptor 56 and collagen III, a receptor-ligand pair, regulates cortical development and lamination. Proc. Natl Acad. Sci. USA. 2011;108:12925–12930. doi: 10.1073/pnas.1104821108.
    1. Chiang NY, et al. Heparin interacts with the adhesion GPCR GPR56, reduces receptor shedding, and promotes cell adhesion and motility. J. Cell Sci. 2016;129:2156–2169. doi: 10.1242/jcs.174458.
    1. Habib N, et al. Massively parallel single-nucleus RNA-seq with DroNc-seq. Nat. Methods. 2017;14:955–958. doi: 10.1038/nmeth.4407.
    1. Mamdani F, et al. Gene expression biomarkers of response to citalopram treatment in major depressive disorder. Transl. Psychiatry. 2011;1:e13. doi: 10.1038/tp.2011.12.
    1. Consoloni JL, et al. Serotonin transporter gene expression predicts the worsening of suicidal ideation and suicide attempts along a long-term follow-up of a major depressive episode. Eur. Neuropsychopharmacol. 2018;28:401–414. doi: 10.1016/j.euroneuro.2017.12.015.
    1. Belzeaux R, et al. Responder and nonresponder patients exhibit different peripheral transcriptional signatures during major depressive episode. Transl. Psychiatry. 2012;2:e185. doi: 10.1038/tp.2012.112.
    1. Farley S, Apazoglou K, Witkin JM, Giros B, Tzavara ET. Antidepressant-like effects of an AMPA receptor potentiator under a chronic mild stress paradigm. Int J. Neuropsychopharmacol. 2010;13:1207–1218. doi: 10.1017/S1461145709991076.
    1. Dournes C, Beeske S, Belzung C, Griebel G. Deep brain stimulation in treatment-resistant depression in mice: comparison with the CRF1 antagonist, SSR125543. Prog. Neuropsychopharmacol. Biol. Psychiatry. 2013;40:213–220. doi: 10.1016/j.pnpbp.2012.07.019.
    1. Crozatier C, et al. Calcineurin (protein phosphatase 2B) is involved in the mechanisms of action of antidepressants. Neuroscience. 2007;144:1470–1476. doi: 10.1016/j.neuroscience.2006.11.030.
    1. Birrell JM, Brown VJ. Medial frontal cortex mediates perceptual attentional set shifting in the rat. J. Neurosci. 2000;20:4320–4324. doi: 10.1523/JNEUROSCI.20-11-04320.2000.
    1. Paxinos, G. & Franklin, K. B. J. The Mouse Brain in Stereotaxic Coordinates, 2nd edn. (Academic Press, 2001).
    1. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15:550. doi: 10.1186/s13059-014-0550-8.
    1. Ritchie ME, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43:e47. doi: 10.1093/nar/gkv007.
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. Ser. B (Methodol.) 1995;57:289–300.
    1. Merico D, Isserlin R, Stueker O, Emili A, Bader GD. Enrichment map: a network-based method for gene-set enrichment visualization and interpretation. PLoS ONE. 2010;5:e13984. doi: 10.1371/journal.pone.0013984.
    1. Tusher VG, Tibshirani R, Chu G. Significance analysis of microarrays applied to the ionizing radiation response. Proc. Natl Acad. Sci. USA. 2001;98:5116–5121. doi: 10.1073/pnas.091062498.

Source: PubMed

3
Prenumerera