Brain region-specific alterations of RNA editing in PDE8A mRNA in suicide decedents

Fabrice Chimienti, Laurent Cavarec, Laurent Vincent, Nicolas Salvetat, Victoria Arango, Mark D Underwood, J John Mann, Jean-François Pujol, Dinah Weissmann, Fabrice Chimienti, Laurent Cavarec, Laurent Vincent, Nicolas Salvetat, Victoria Arango, Mark D Underwood, J John Mann, Jean-François Pujol, Dinah Weissmann

Abstract

Phosphodiesterases (PDE) are key modulators of signal transduction and are involved in inflammatory cell activation, memory and cognition. There is a two-fold decrease in the expression of phosphodiesterase 8A (PDE8A) in the temporal cortex of major depressive disorder (MDD) patients. Here, we studied PDE8A mRNA-editing profile in two architectonically distinct neocortical regions in a clinically well-characterized cohort of age- and sex-matched non-psychiatric drug-free controls and depressed suicide decedents. By using capillary electrophoresis single-stranded conformational polymorphism (CE-SSCP), a previously validated technique to identify A-to-I RNA modifications, we report the full editing profile of PDE8A in the brain, including identification of two novel editing sites. Editing of PDE8A mRNA displayed clear regional difference when comparing dorsolateral prefrontal cortex (BA9) and anterior cingulate cortex (BA24). Furthermore, we report significant intra-regional differences between non-psychiatric control individuals and depressed suicide decedents, which could discriminate the two populations. Taken together, our results (i) highlight the importance of immune/inflammatory markers in major depressive disorder and suicide and (ii) establish a direct relationship between A-to-I RNA modifications of peripheral markers and A-to-I RNA editing-related modifications in brain. This work provides the first immune response-related brain marker for suicide and could pave the way for the identification of a blood-based biomarker that predicts suicidal behavior.

Conflict of interest statement

J.J.M. receives royalties from the Research Foundation of Mental Hygiene for commercial use of the C-SSRS. The remaining authors declare that they have no conflict of interest.

Figures

Fig. 1. Editing sites in the intron…
Fig. 1. Editing sites in the intron 9 of PDE8A pre-mRNA from human brain.
Putative PDE8A intron 9 mRNA secondary structure as predicted by Vienna RNA Websuite. The mRNA positions where significant editing events have been detected are highlighted with arrows. Editing Sites A to G have been previously described in T cells. Newly identified sites in the brain are highlighted in red. Most of the editing sites are in close proximity to each other. All bases are annotated on chromosome 15 according to the last GRCh38 genebuild
Fig. 2. Relative isoform proportion of PDE8A…
Fig. 2. Relative isoform proportion of PDE8A mRNA in (a) Brodmann area 24 (BA24) and (b) Brodmann area 9 (BA9) measured by CE-SSCP on samples of the control group.
Histograms represent relative isoform proportion (%) of the 30 detected PDE8A isoform (mean ± s.e.m.; n = 8). Only isoforms representing more than 0.5% of relative proportion were included in the analysis
Fig. 3. Relative proportion of PDE8A mRNA…
Fig. 3. Relative proportion of PDE8A mRNA isoforms in two discrete brain regions of control (A) and suicide (B) subjects.
a Comparison of the relative isoform proportion of PDE8A in BA24 and BA9 within the control group. The PDE8A isoforms are depicted in order of significance and abundance in BA24. b Comparison of the relative isoform proportion of PDE8Ain BA24 and BA9 in the suicide group. In this representation, a negative percentage from the median indicates that the relative proportion of the isoform is higher in BA9 compared with BA24. Filled bars in (a) and (b) indicate the most significant differences of the isoform proportion between the two brain structures in both control and suicide groups. Criteria for the selection are (1) a p-value (FDR) ≤ 0.05 using the one-sample Wilcoxon signed rank test (where null hypothesis H0: median variation = 0) and (2) a median variation ≥ ± 20%. The symbol * indicates a p-value (FDR) ≤ 0.05, ** indicate a p-value (FDR) ≤ 0.01 and *** indicate a p-value (FDR) ≤ 0.001
Fig. 4. Brain regional specificity of PDE8A…
Fig. 4. Brain regional specificity of PDE8A mRNA editing changes in suicide with major depression.
a Comparison of the relative isoform proportion of PDE8A in control and suicide group within BA24 area. The PDE8A isoforms are depicted in order of significance and abundancy in suicide group. A negative value indicates relative decrease of the isoform whereas an increase in the relative proportion is indicated by a positive value. b Comparison of the relative isoform proportion of PDE8A in control and suicides within the BA9 area. Filled colored bars in (a) and (b) indicate most significant differences between the control and suicide groups. Criteria for the selection are (1) a p-value (FDR) ≤ 0,05 using the one-sample Wilcoxon signed rank test (where null hypothesis H0: median variation = 0) and (2) a median variation ≥ ± 20%. The symbol * indicate a p-value (FDR) ≤ 0.05, ** indicate a p-value (FDR) ≤ 0,01 and *** indicates a p-value (FDR) ≤ 0.001
Fig. 5. Suicide-induced alterations of the relative…
Fig. 5. Suicide-induced alterations of the relative proportion of PDE8A isoforms in BA24.
ac Comparison of the relative proportion of ABC, BCEG and ABDE isoforms in control (n = 8) and suicide groups (n = 8). Boxplot represents the distribution of the relative proportion of PDE8A mRNA in BA24 of both groups. d Boxplot representation of mROC combination of relative proportions (%) of ABC + BCEG + ABDE PDE8A isoforms in BA24 of both groups. e Table showing the most significant changes of the relative proportion of PDE8A mRNA isoforms in BA24 of both groups. The symbol * indicates, for Wilcoxon rank-sum test, a p-value ≤ 0.05, ** indicates a p-value ≤ 0.01 and *** indicate a p-value ≤ 0.001
Fig. 6. Suicide-induced alterations of the relative…
Fig. 6. Suicide-induced alterations of the relative proportion of PDE8A isoforms in BA9.
ac Comparison of the relative proportion of BD, BCEG and ABF isoforms in control (n = 8) and suicide groups (n = 8). Boxplot represents the distribution of the relative proportion of PDE8A mRNA in BA9 of both groups. d Boxplot representation of mROC combination of relative proportions (%) of BD + BCEG + ABF PDE8A isoforms in BA9 of both groups. e Table showing the most significant changes of the relative proportion of PDE8A mRNA isoforms in BA9 of both groups. The symbol * indicates, for Wilcoxon rank-sum test, a p-value ≤ 0.05, ** indicate a p-value ≤ 0.01 and *** indicate a p-value ≤ 0.001

References

    1. WHO. Preventing Suicide: a Global Imperative. 2014. .
    1. Arango V, Huang YY, Underwood MD, Mann JJ. Genetics of the serotonergic system in suicidal behavior. J. Psychiatr. Res. 2003;37:375–386. doi: 10.1016/S0022-3956(03)00048-7.
    1. Bani-Fatemi A, Howe AS, De Luca V. Epigenetic studies of suicidal behavior. Neurocase. 2015;21:134–143. doi: 10.1080/13554794.2013.826679.
    1. Labonte B, et al. Genome-wide methylation changes in the brains of suicide completers. Am. J. Psychiatry. 2013;170:511–520. doi: 10.1176/appi.ajp.2012.12050627.
    1. Dracheva S, et al. Increased serotonin 2C receptor mRNA editing: a possible risk factor for suicide. Mol. Psychiatry. 2008;13:1001–1010. doi: 10.1038/sj.mp.4002081.
    1. Weissmann D, et al. Region-specific alterations of A-to-I RNA editing of serotonin 2c receptor in the cortex of suicides with major depression. Transl. Psychiatry. 2016;6:e878. doi: 10.1038/tp.2016.121.
    1. Karanovic J, et al. Joint effect of ADARB1 gene, HTR2C gene and stressful life events on suicide attempt risk in patients with major psychiatric disorders. World J. Biol. Psychiatry. 2015;16:261–271. doi: 10.3109/15622975.2014.1000374.
    1. Walkley CR, Li JB. Rewriting the transcriptome: adenosine-to-inosine RNA editing by ADARs. Genome Biol. 2017;18:205. doi: 10.1186/s13059-017-1347-3.
    1. Eisenberg E., Levanon E. Y. A-to-I RNA editing—immune protector and transcriptome diversifier. Nat. Rev. Genet.19, 473-490 (2018).
    1. Jacobs MM, Fogg RL, Emeson RB, Stanwood GD. ADAR1 and ADAR2 expression and editing activity during forebrain development. Dev. Neurosci. 2009;31:223–237. doi: 10.1159/000210185.
    1. Picardi E, et al. Profiling RNA editing in human tissues: towards the inosinome Atlas. Sci. Rep. 2015;5:14941. doi: 10.1038/srep14941.
    1. Silberberg G, Lundin D, Navon R, Ohman M. Deregulation of the A-to-I RNA editing mechanism in psychiatric disorders. Human. Mol. Genet. 2012;21:311–321. doi: 10.1093/hmg/ddr461.
    1. Simmons M, Meador-Woodruff JH, Sodhi MS. Increased cortical expression of an RNA editing enzyme occurs in major depressive suicide victims. Neuroreport. 2010;21:993–997.
    1. Stamm S, Gruber SB, Rabchevsky AG, Emeson RB. The activity of the serotonin receptor 2C is regulated by alternative splicing. Human. Genet. 2017;136:1079–1091. doi: 10.1007/s00439-017-1826-3.
    1. Filip M, Bader M. Overview on 5-HT receptors and their role in physiology and pathology of the central nervous system. Pharmacol. Rep. 2009;61:761–777. doi: 10.1016/S1734-1140(09)70132-X.
    1. Giulietti M, et al. How much do we know about the coupling of G-proteins to serotonin receptors? Mol. brain. 2014;7:49. doi: 10.1186/s13041-014-0049-y.
    1. Chagraoui A, Thibaut F, Skiba M, Thuillez C, Bourin M. 5-HT2C receptors in psychiatric disorders: a review. Progress. Neuro-Psychopharmacol. Biol. Psychiatry. 2016;66:120–135. doi: 10.1016/j.pnpbp.2015.12.006.
    1. Conti M, Beavo J. Biochemistry and physiology of cyclic nucleotide phosphodiesterases: essential components in cyclic nucleotide signaling. Annu. Rev. Biochem. 2007;76:481–511. doi: 10.1146/annurev.biochem.76.060305.150444.
    1. Bender AT, Beavo JA. Cyclic nucleotide phosphodiesterases: molecular regulation to clinical use. Pharmacol. Rev. 2006;58:488–520. doi: 10.1124/pr.58.3.5.
    1. Cieslik EC, et al. Is there “one” DLPFC in cognitive action control? Evidence for heterogeneity from co-activation-based parcellation. Cereb. Cortex. 2013;23:2677–2689. doi: 10.1093/cercor/bhs256.
    1. Arango V, Underwood MD, Gubbi AV, Mann JJ. Localized alterations in pre and postsynaptic serotonin binding sites in the ventrolateral prefrontal cortex of suicide victims. Brain Res. 1995;688:121–133. doi: 10.1016/0006-8993(95)00523-S.
    1. Boldrini M, et al. Antidepressants increase neural progenitor cells in the human hippocampus. Neuropsychopharmacology. 2009;34:2376–2389. doi: 10.1038/npp.2009.75.
    1. Harrison PJ, et al. The relative importance of premortem acidosis and postmortem interval for human brain gene expression studies: selective mRNA vulnerability and comparison with their encoded proteins. Neurosci. Lett. 1995;200:151–154. doi: 10.1016/0304-3940(95)12102-A.
    1. Kelly TM, Mann JJ. Validity of DSM-III-R diagnosis by psychological autopsy: a comparison with clinician ante-mortem diagnosis. Acta Psychiatr. Scand. 1996;94:337–343. doi: 10.1111/j.1600-0447.1996.tb09869.x.
    1. Mann JJ, et al. A serotonin transporter gene promoter polymorphism (5-HTTLPR) and prefrontal cortical binding in major depression and suicide. Arch. General. Psychiatry. 2000;57:729–738. doi: 10.1001/archpsyc.57.8.729.
    1. Mayberg HS. Defining the neural circuitry of depression: toward a new nosology with therapeutic implications. Biol. Psychiatry. 2007;61:729–730. doi: 10.1016/j.biopsych.2007.01.013.
    1. Cavarec L, et al. In vitro screening for drug-induced depression and/or suicidal adverse effects: a new toxicogenomic assay based on CE-SSCP analysis of HTR2C mRNA editing in SH-SY5Y cells. Neurotox. Res. 2013;23:49–62. doi: 10.1007/s12640-012-9324-9.
    1. R. Core Team. R: A Language and Environment for Statistical Computing. (R Foundation for Statistical Computing, Vienna, Austria, 2014).
    1. Kramar A, Faraggi D, Fortune A, Reiser B. mROC: a computer program for combining tumour markers in predicting disease states. Comput. Methods Prog. Biomed. 2001;66:199–207. doi: 10.1016/S0169-2607(00)00129-2.
    1. Gruber AR, Lorenz R, Bernhart SH, Neubock R, Hofacker IL. The Vienna RNA websuite. Nucleic Acids Res. 2008;36:W70–74. doi: 10.1093/nar/gkn188.
    1. Ding Y, Chan CY, Lawrence CE. RNA secondary structure prediction by centroids in a Boltzmann weighted ensemble. RNA. 2005;11:1157–1166. doi: 10.1261/rna.2500605.
    1. Orlowski RJ, et al. Altered editing in cyclic nucleotide phosphodiesterase 8A1 gene transcripts of systemic lupus erythematosus T lymphocytes. Immunology. 2008;125:408–419. doi: 10.1111/j.1365-2567.2008.02850.x.
    1. Lehmann KA, Bass BL. Double-stranded RNA adenosine deaminases ADAR1 and ADAR2 have overlapping specificities. Biochemistry. 2000;39:12875–12884. doi: 10.1021/bi001383g.
    1. Daniel C, Widmark A, Rigardt D, Ohman M. Editing inducer elements increases A-to-I editing efficiency in the mammalian transcriptome. Genome Biol. 2017;18:195. doi: 10.1186/s13059-017-1324-x.
    1. van der Laan S, Salvetat N, Weissmann D, Molina F. Emerging RNA editing biomarkers will foster drug development. Drug Discov. Today. 2017;22:1056–1063. doi: 10.1016/j.drudis.2017.01.017.
    1. Rossetti C, et al. RNA editing signature during myeloid leukemia cell differentiation. Leukemia. 2017;31:2824–2832. doi: 10.1038/leu.2017.134.
    1. Li JB, et al. Genome-wide identification of human RNA editing sites by parallel DNA capturing and sequencing. Science. 2009;324:1210–1213. doi: 10.1126/science.1170995.
    1. Maas S, Kawahara Y, Tamburro KM, Nishikura K, A-to-I RNA. Editing and human disease. RNA Biol. 2006;3:1–9. doi: 10.4161/rna.3.1.2495.
    1. Picardi E, Horner DS, Pesole G. Single-cell transcriptomics reveals specific RNA editing signatures in the human brain. RNA. 2017;23:860–865. doi: 10.1261/rna.058271.116.
    1. Filippini A, Bonini D, La Via L, Barbon A. The good and the bad of glutamate receptor RNA editing. Mol. Neurobiol. 2017;54:6795–6805. doi: 10.1007/s12035-016-0201-z.
    1. Smith RM, et al. Whole transcriptome RNA-Seq allelic expression in human brain. BMC Genom. 2013;14:571. doi: 10.1186/1471-2164-14-571.
    1. Grohmann M, et al. Alternative splicing and extensive RNA editing of human TPH2 transcripts. PloS ONE. 2010;5:e8956. doi: 10.1371/journal.pone.0008956.
    1. Karanovic J, et al. Effect of childhood general traumas on suicide attempt depends on TPH2 and ADARB1 variants in psychiatric patients. J. Neural Transm. 2017;124:621–629. doi: 10.1007/s00702-017-1677-z.
    1. Jeon SW, Kim YK. Inflammation-induced depression: Its pathophysiology and therapeutic implications. J. Neuroimmunol. 2017;313:92–98. doi: 10.1016/j.jneuroim.2017.10.016.
    1. Kang HJ, et al. Gene expression profiling in postmortem prefrontal cortex of major depressive disorder. J. Neurosci. 2007;27:13329–13340. doi: 10.1523/JNEUROSCI.4083-07.2007.
    1. Niculescu AB, et al. Understanding and predicting suicidality using a combined genomic and clinical risk assessment approach. Mol. Psychiatry. 2015;20:1266–1285. doi: 10.1038/mp.2015.112.
    1. Hodes GE, Kana V, Menard C, Merad M, Russo SJ. Neuroimmune mechanisms of depression. Nat. Neurosci. 2015;18:1386–1393. doi: 10.1038/nn.4113.
    1. Steiner J, et al. Immunological aspects in the neurobiology of suicide: elevated microglial density in schizophrenia and depression is associated with suicide. J. Psychiatr. Res. 2008;42:151–157. doi: 10.1016/j.jpsychires.2006.10.013.
    1. Barnes J, Mondelli V, Pariante CM. Genetic contributions of inflammation to depression. Neuropsychopharmacology. 2017;42:81–98. doi: 10.1038/npp.2016.169.
    1. Jansen R, et al. Gene expression in major depressive disorder. Mol. Psychiatry. 2016;21:339–347. doi: 10.1038/mp.2015.57.
    1. Simon NM, et al. A detailed examination of cytokine abnormalities in major depressive disorder. Eur. Neuropsychopharmacol. 2008;18:230–233. doi: 10.1016/j.euroneuro.2007.06.004.
    1. Wohleb ES, Franklin T, Iwata M, Duman RS. Integrating neuroimmune systems in the neurobiology of depression. Nat. Rev. Neurosci. 2016;17:497–511. doi: 10.1038/nrn.2016.69.
    1. Holmans P, et al. Genetics of recurrent early-onset major depression (GenRED): final genome scan report. Am. J. Psychiatry. 2007;164:248–258. doi: 10.1176/ajp.2007.164.2.248.
    1. Levinson DF, et al. Genetics of recurrent early-onset major depression (GenRED): significant linkage on chromosome 15q25-q26 after fine mapping with single nucleotide polymorphism markers. Am. J. Psychiatry. 2007;164:259–264. doi: 10.1176/ajp.2007.164.2.259.
    1. Godbout JP, Berg BM, Krzyszton C, Johnson RW. Alpha-tocopherol attenuates NFkappaB activation and pro-inflammatory cytokine production in brain and improves recovery from lipopolysaccharide-induced sickness behavior. J. Neuroimmunol. 2005;169:97–105. doi: 10.1016/j.jneuroim.2005.08.003.
    1. Leonard BE. Inflammation and depression: a causal or coincidental link to the pathophysiology? Acta Neuropsychiatr. 2018;30:1–16. doi: 10.1017/neu.2016.69.
    1. Neves-Zaph SR. Phosphodiesterase diversity and signal processing within cAMP signaling networks. Adv. Neurobiol. 2017;17:3–14. doi: 10.1007/978-3-319-58811-7_1.
    1. Murthy VS, Mangot AG. Psychiatric aspects of phosphodiesterases: an overview. Indian J. Pharmacol. 2015;47:594–599. doi: 10.4103/0253-7613.169593.
    1. Aston C, Jiang L, Sokolov BP. Transcriptional profiling reveals evidence for signaling and oligodendroglial abnormalities in the temporal cortex from patients with major depressive disorder. Mol. Psychiatry. 2005;10:309–322. doi: 10.1038/sj.mp.4001565.
    1. Morse DP, Aruscavage PJ, Bass BL. RNA hairpins in noncoding regions of human brain and Caenorhabditis elegans mRNA are edited by adenosine deaminases that act on RNA. Proc. Natl. Acad. Sci. USA. 2002;99:7906–7911. doi: 10.1073/pnas.112704299.
    1. Bonaccorso S, et al. Depression induced by treatment with interferon-alpha in patients affected by hepatitis C virus. J. Affect. Disord. 2002;72:237–241. doi: 10.1016/S0165-0327(02)00264-1.
    1. Gagnidze K, Rayon-Estrada V, Harroch S, Bulloch K, Papavasiliou FN. A new chapter in genetic medicine: RNA editing and its role in disease pathogenesis. Trends Mol. Med. 2018;24:294–303. doi: 10.1016/j.molmed.2018.01.002.

Source: PubMed

3
Subskrybuj