Skip to main content
  • Research article
  • Open access
  • Published:

Olanzapine-induced methylation alters cadherin gene families and associated pathways implicated in psychosis

Abstract

Background

The complex aetiology of most mental disorders involves gene-environment interactions that may operate using epigenetic mechanisms particularly DNA methylation. It may explain many of the features seen in mental disorders including transmission, expression and antipsychotic treatment responses. This report deals with the assessment of DNA methylation in response to an antipsychotic drug (olanzapine) on brain (cerebellum and hippocampus), and liver as a non-neural reference in a rat model. The study focuses on the Cadherin/protocadherins encoded by a multi-gene family that serve as adhesion molecules and are involved in cell-cell communication in the mammalian brain. A number of these molecules have been implicated in the causation of schizophrenia and related disorders.

Results

The results show that olanzapine causes changes in DNA methylation, most specific to the promoter region of specific genes. This response is tissue specific and involves a number of cadherin genes, particularly in cerebellum. Also, the genes identified have led to the identification of several pathways significantly affected by DNA methylation in cerebellum, hippocampus and liver. These included the Gα12/13 Signalling (p = 9.2E-08) and Wnt signalling (p = 0.01) pathways as contributors to psychosis that is based on its responsiveness to antipsychotics used in its treatment.

Conclusion

The results suggest that DNA methylation changes on the promoter regions of the Cadherin/protocadherin genes impact the response of olanzapine treatment. These impacts have been revealed through the identified pathways and particularly in the identification of pathways that have been previously implicated in psychosis.

Background

Cadherins are involved in neurodevelopment, nerve cell migration and cell-to-cell interaction and therefore it follows that they may regulate the pathogenesis of psychosis [1]. For example, previous studies suggested the importance of investigating epigenetic control of PcdhXY with respect to psychiatric disorders [2]. Interestingly, previous studies have suggested that epigenetic modifications regulate the expression of clustered protocadherin genes (Pcdhs) [3]. In mammals, more than 50 Pcdh genes are categorized into three clusters Pcdh-α, Pcdh-β and Pcdh-γ encoding diversified proteins mainly expressed in the nervous system [35]. Some protocadherins (Pcdh1 and Pcdh7) are regulated by Methyl-CpG-binding protein, MeCP2, which is a protein that is strongly associated with epigenetic regulation [6].

Aberrant promoter methylation of cadherin family genes has been implicated in complex human disorders [7]. For example, Pcdh10, a novel tumor suppresser gene is found to be downregulated by hypermethylation in various cancer cases [810] corroborating the evidence that epigenetics plays a role in complex disorders. Further, the high expression of Pcdhαv8 in mouse neuroblastoma cell lines was found to be associated with demethylation of the 5’ regulatory region of the gene [11]. In the same study, a dynamic change in methylation of Pcdh genes was observed. The 5’ region of the Pcdhαv4 gene was 99% methylated in neuroblastoma cells and ~35% methylated in the cortex of mice. On the contrary, the 5’ region of the Pcdhαv8 was reported to be 99.1-100% methylated in neuroblastoma cell lines and 5% methylated in the cortex of the mouse strain C57BL/6 [11].

Although it is possible that aberrant metabolomics may be due to mutation in a single gene or be the effect of a single environment, emerging literature argues that most often the development of complex diseases and disorders represents a complex interaction of multiple genes and multiple environments [12]. This is true in most behavioral disorders that have complex aetiology involving several genes and environment [13]. Ultimately, the two contributors enact their effect by altering cellular metabolomics through changes in gene expression via epigenetic modifications such as DNA methylation [14]. However, an assessment of the mechanistic basis of the effect of environment has been arduous as an environment may cause aberration via its effect on the gene(s). This offers renewed hope for the treatment of complex disorders, as it may be possible to manipulate DNA methylation, and subsequently alter gene expression to affect metabolomic aberrations and facilitate a cure or decreased severity of symptoms. Also, this paradigm of disease causation is expected to be instrumental in identifying mechanisms of actions of antipsychotics, such as olanzapine.

Mental disorders including psychosis are difficult to treat. Antipsychotics have variable responses and their mechanism of action is imprecise. We have assessed the mechanisms of action of olanzapine, a commonly used antipsychotic medication. The results argue that olanzapine may improve the mental outcome of patients by altering the methylation profile of genes implicated in disease-specific pathways. Specifically, our studies reported changes in the methylation of genes implicated in the dopamine pathways in the hippocampus and cerebellum in a rat model [15]. Interestingly, in a genome-wide assessment, our studies showed methylation changes in the genes of a number of pathways that may alter metabolomics leading to the efficacy as well as side effects of olanzapine [15]. These results offer a novel paradigm to reveal critical genes and pathways by identifying changes in methylation in response to drugs that offer relief or cure for the disease symptoms. Here we will present data on genes of the cadherin and protocadherin (Pcdh) family that are preferentially affected by olanzapine. The clustered protocadherin (Pcdh) genes [16] encode a group of diverse cadherin-related transmembrane proteins, and are expressed in the brain [17]. They generate vast single-cell diversity in the brain [18]. They represent Ca2+-dependent cell adhesion molecules that are critical in vertebrate nervous development [19] and maintenance of neural circuitry [20]. The expression of this cluster of genes is regulated by a complex mechanism at the level of transcription [21] that may include gene specific DNA methylation [22]. Given the critical role of cadherins in brain development and function it is not surprising that genome-wide analysis for a number of mental disorders have implicated defects in cadherin genes. These include complex disorders, such as intellectual disability [23] autism [24] schizophrenia [25, 26] and bipolar disorders [27]. This report adds to this complex literature suggesting that a commonly used antipsychotic drug, olanzapine, affects methylation of the promoter regions of cadherin family genes in the rat brain. The effect of olanzapine is tissue specific, particularly affecting several gene promoters in cerebellum and may have implications in the therapeutic response.

Methods

Rats

Adult male Sprague-Dawley rats of 12 weeks of age (250 - 300 g) were purchased from Charles River, PQ, Canada. Upon arrival, rats were placed (2 per cage) in standard acrylic cages and subjected to the treatment protocol. They housed in controlled humidity and temperature on a 12-hour light/dark cycle (lights on at 7:00 a.m.). They were fed standard rat chow (LabDiet) and tap water ad libitum.

Ethical approval

The Institutional Animal Care Committee of the University of Western Ontario had approved all animal-related procedures used in this study following the Canadian and National Institute of Health Guides on animal experimentation. Also, the manuscript reporting followed the ARRIVE (Animal Research: Reporting of In Vivo Experiments) guidelines.

Olanzapine treatment

Before the commencement of olanzapine treatment, animals were weighed and divided into two treatment groups with comparable means of weight. Their stress-induced locomotor activity (following a 5 min tail pinch) was recorded for 30 min using an automated open-field activity chamber (San Diego Instruments, San Diego, CA, USA). A computer that detects the disruption of photocell beams recorded the number of beam breaks per five minutes for half an hour as the animal moves. Rats then received injections of olanzapine (Zyprexa, Lilly, IN, USA; 2.5 mg/kg, i.m.; n = 8) or vehicle (phosphate buffered saline (PBS); n = 8) between 1:30 pm and 3:00 pm daily for 19 days. On day 20, eighteen hours after the last olanzapine/vehicle injection, rats were subjected to stress-induced locomotor activity to assess the therapeutic efficacy of chronic olanzapine. Significantly reduced locomotor activity of olanzapine treated rats indicated the therapeutic efficacy of the drug administered, which was comparable to the dosing paradigm employed in previous studies [28].

Genomic DNA extraction

Rats were decapitated without anaesthesia, brain tissues were micro-dissected promptly in ice-cold PBS; and three random biopsy punches through cerebellum, hippocampus, and liver were obtained. Individual samples (three random biopsy punches of each tissue composed one sample) from each rat were kept separately and flash frozen in liquid nitrogen. The genomic DNA was isolated from olanzapine-treated (n = 2) and saline control (n = 2) samples of cerebellum, hippocampus and liver. Genomic DNA was isolated from the interphase layer of TRIzol using sodium citrate, followed by ethanol precipitation and purification using the QIAamp® DNA Micro Kit (QIAGEN, Valencia, CA). DNA was then quantified using a NanoDrop ND-1000 spectrophotometer (Thermo Fisher Scientific Inc., Wilmington, DE) and all samples had OD260/OD280 nm ratios of 1.8–2.0 and OD260/OD230 nm ratios of 2.0–2.4.

Methylated DNA immunoprecipitation

This study has used methylated DNA immunoprecipitation (MeDIP) to investigate genome-wide DNA methylation in a rat model [29] The MeDIP sample labeling, hybridization, and processing were performed at Arraystar Inc. (Rockville, Maryland, USA). The immunoprecipitated DNA was eluted and purified by phenol chloroform extraction and ethanol precipitation. The total input and immunoprecipitated DNA were labeled with Cy3- and Cy5-labeled random 9-mers, respectively, and hybridized to NimbleGen RN34 Meth 3×720 K CpG plus Promoter array, which is a single array design containing all known CpG Islands annotated by UCSC and all well-characterized RefSeq promoter regions (from about -3.88 kb to +0.97 kb of the transcription start sites [TSSs]) covered by ~720,000 probes. Scanning was performed with the Axon GenePix 4000B microarray scanner. Therefore, the array design would capture the global changes in methylation.

Data analysis involved the comparison of differentially enriched regions between drug exposed (E) and control (C) rats, the log2-ratio values were averaged and then used to calculate the M’ value [M’ = Average (log2 MeDIPE/InputE) – Average (log2 MeDIPC/InputC)] for each probe. A NimbleScan sliding-window peak-finding algorithm was run on this data to find the differential enrichment peaks (DEP). Using an R script program, a hierarchical clustering analysis was completed. The probe data matrix was obtained by using PeakScores from differentially methylated regions selected by DEP analysis. This analysis used a “PeakScore” ≥2 to define the DEPs, which is equivalent to the average p-value ≤0.01, for all probes within the peak. In this study, we have specifically analyzed 17 Cadherin family genes and their methylation status in cerebellum and hippocampus with liver as a non-neural cell types following olanzapine treatment of experimental rats (Table 1).

Table 1 List of the Cadherin genes with their promoter regions affected by olanzapine-induced methylation

Pathway analysis of array results

The affected Cadherin family genes were then subjected to ingenuity pathway analysis (Ingenuity System Inc, CA, USA) [http://www.ingenuity.com/]. Transcription factor binding sites of Cdh13 that showed increased methylation in cerebellum were identified using CTCFBSDB 2.0 [30]. This analysis was done as a representative for all cadherin genes affected by olanzapine-induced DNA methylation.

Results

Recently, we reported that therapeutic olanzapine causes an increase in 1294 genes and a decrease in 565 genes in the cerebellum of treated rats [15]. This response is tissue specific and only 10% of the genes affected were shared between cerebellum and hippocampus. With respect to cadherin genes, we report that the promoter regions of 11 Cadherin family genes have olanzapine-induced DNA methylation changes in the cerebellum as compared to matched controls (Table 1). Of those genes, five showed significantly increased methylation, while six of them showed decreased methylation (p < 0.01). In comparison to the results on cerebellum, three cadherin genes (Cdh17, Cdh23, Pcdh7) had increased methylation and one gene (Pcdhga8) showed decreased methylation in hippocampus. Also, two cadherin genes (Cdh5 and Cdh11) had increased methylation and two genes (Pcdhga8 and Pcdhgb7) showed decreased methylation in the liver. The cadherin genes in the rat genome that are affected by olanzapine-induced DNA methylation are dispersed over different chromosomes including chromosome 2, 3, 15, 18, 19, 20 and X.

For the identification of each methylation difference, we compared the Manhattan plot of individual cadherin genes. It is shown in Figure 1 using Cdh13 as a model. In this figure, the peak of differential methylation (Figure 1A), the genomic region of Cdh13 on the rat genome (Figure 1B) and the sequence depicting the promoter region (Figure 1C) are shown. It has allowed us to identify regulatory features including a CTCF binding site. As shown in Figure 1C, the methylation changes were observed in the surrounding Cs and CpGs suggesting the global effect of olanzapine. The sequence presented in the figure showed differential methylation, which included the identified CTCF binding site in the promoter region of the gene.

Figure 1
figure 1

A representative figure depicting the genomic region of Cadherin 13 gene affected by olanzapine. A) Manhattan plot showing the significance of methylation enriched regions. Red box shows DNA methylation enriched region containing subset of CpG sites overlapping with the promoter region of Cdh13 on Chromosome 19, in olanzapine treated cerebellum compared to control (upper panel), while there was no methylation enriched subset of CpGs at that particular genomic region in the control cerebellum compared to treated cerebellum sample (lower panel) B) Red box shows the gene name and the promoter region affected by olanzapine-induced DNA methylation, obtained from Rat Genome Database, C) Part of the promoter sequence entirely affected by olanzapine-induced DNA methylation, and the CTCF binding site sequence is highlighted in red.

Next, we used pathway analysis to identify major pathways and systems (Table 2) that may be affected by the observed olanzapine-induced methylation changes in the cadherin family genes in hippocampus, cerebellum and liver listed in Table 1. Interestingly, pathway analysis involving these genes revealed the most significant pathways including the Gα12/13 signalling (p = 9.2E-08) (Figure 2A), RhoGDI signalling (p = 4.8E-07) (Figure 2B) and Wnt/β-catenin signalling (p = 1.2E-02) pathways (Figure 2C). They affect molecular and cellular functions associated with cell-to-cell signalling and interactions (p = 6.13E-04 - 3.01E-02) and may affect tissue, nervous system dynamics and behaviour. In doing so, differential methylation of genes involved in the pathway suggested that olanzapine affects the physiological cadherin system (p = 6.13E-04 - 9.76E-03) (Table 2).

Table 2 Top pathways, networks and molecular and cellular functions identified by pathway analysis using the Cadherin family genes affected by olanzapine
Figure 2
figure 2

The most significant pathways identified by Ingenuity pathway analysis. A) Gα12/13 signalling (p = 9.2E-08), B) RhoGDI signalling (p = 4.8E-07), C) Wnt/β-catenin signalling (p = 1.2E-02) significantly affected by DNA methylation of the Cadherin genes.

Discussion

Cell-cell communication is an essential feature of multicellular organisms. It is particularly critical in the mammalian brain, where molecules involved in cell-cell interactions such as adhesion and exchange of information provide the foundation for neurodevelopment as well as proper functioning of the neural network. Not surprisingly, mammalian brain is rich in such molecules. Also, the exceptional richness and diversity of these molecules is often accounted for by their evolution, by multi-gene families and by diversity of regulatory mechanisms. For example, adhesion molecules such as cadherins that constitute a super-family of transmembrane receptors and mediate Ca2+ -dependent cell to cell hemophilic interactions are encoded by ~100 genes in humans [31]. Also, the organization of these genes allows differential expression including gene specific DNA methylation and differential splicing to facilitate expression of specific cadherins in different cells and cell types [32]. As it stands, the molecular basis of the cadherins is still unclear. What is known is that cells expressing a given cadherin adhere preferentially to those expressing the same cadherin subtype [33]. Also, the combination of qualitative and quantitative differences likely confers cell type-specific adhesion affinities on individual cells [34]. They provide the foundation for target recognition, synapse formation and signalling, which are critical features of any neural network [35].

The results included in this study show that olanzapine affects the methylation status of the cadherin gene promoters. Given the involvement of cadherin genes in a number of mental disorders including psychosis, it is understandable that several cadherin genes are differentially methylated in response to olanzapine. Also, this difference is prominent in the cerebellum that is involved in cognitive processing and emotional control including attention, executive control, language, working memory, pain, emotion, and addiction in addition to its role in motor coordination [36]. The results of this study have identified that the methylation differences are specific to the promoter region of individual genes (Table 1). Also, the sequence features of these regions contain regulatory signals (Figure 1) and cellular metabolomics. The associated cellular changes are compatible with anomalies in behavioral disorders via three specific pathways: Gα12/13 signalling, RhoGDI signalling and Wnt signaling (Figure 2). Gα12/13 are units of heterotrimeric G proteins that regulate cell processes including cell remodeling through the use of guanine nucleotide exchange factors [37] while Rho family proteins regulate a wide diversity of cellular processes, including adhesion, cytokinesis, cell-cell progression, membrane trafficking and signal transduction [38]. Interestingly, the Wnt signaling pathway has been implicated in the pathogenesis of schizophrenia [39] as well as negative symptom severity in psychosis [40] and it has been previously shown that E-cadherin and β-catenin affect Wnt Signaling pathway [41]. Further, aberrant expression of transcripts of the PCDH11X/Y gene was reportedly implicated in anomalous cerebral asymmetry [42]. This has led to the possibility that the variation relating to psychosis and psychological disorders may not only be sequence based, but may also be mediated by antipsychotic-induced epigenetic changes [39, 42]. The results on olanzapine-induced differential methylation of cadherin genes implicated in the three pathways add further support to this still evolving paradigm. Of special interest is the Pcdh8 gene. It has been implicated in schizophrenia [20] and has increased methylation both in hippocampus and liver in this study, suggesting that the expression of the gene is likely to be compromised due to the antipsychotic drug. The observed hypermethylation of the gene could potentially repress its transcription [22]. Moreover, there are indications that epigenetic changes affect not only a single candidate gene promoter but also intragenic sequences located farther from the transcriptional start sites [43]. The observed methylation changes are tissue-specific, and it is noted that the direction of methylation change (either increased or decreased methylation) is not consistent across tissues, suggesting the possibility that the changes in brain tissues are likely related to the efficacy of the drug while effects in liver may be related to the adverse consequences of the drug. Olanzapine-induced DNA methylation changes in genes involved in the identified pathways may alter the associated network functions (Table 2). However, further study is required to specifically analyze the effects (on a protein level) of gene-specific methylation changes on each identified network to unravel the specific mechanism of action of olanzapine. Also, we acknowledge that the prefrontal cortex and nucleus accumbens that are also implicated in psychosis [44, 45] may need to be investigated for the effect of antipsychotics on methylation, in future studies.

Conclusion

The identified pathways suggest that olanzapine-induced DNA methylation changes on the promoter regions of the Cadherin/protocadherin genes may have an important role in response to olanzapine treatment in psychosis. Further, the approach outlined in this study may be applied to other disorders of complex aetiology.

References

  1. Sasaki T, Billett E, Petronis A, Ying D, Parsons T, Macciardi FM, Meltzer HY, Lieberman J, Joffe RT, Ross CA, McInnis MG, Li SH, Kennedy JL: Psychosis and genes with trinucleotide repeat polymorphism. Hum Genet. 1996, 97 (2): 244-246. 10.1007/BF02265274.

    Article  CAS  PubMed  Google Scholar 

  2. Giouzeli M, Williams NA, Lonie LJ, DeLisi LE, Crow TJ: ProtocadherinX/Y, a candidate gene-pair for schizophrenia and schizoaffective disorder: a DHPLC investigation of genomic sequence. Am J Med Genet. 2004, 129B (1): 1-9. 10.1002/ajmg.b.30036.

    Article  PubMed  Google Scholar 

  3. Toyoda S, Kawaguchi M, Kobayashi T, Tarusawa E, Toyama T, Okano M, Oda M, Nakauchi H, Yoshimura Y, Sanbo M, Hirabayashi M, Hirayama T, Hirabayashi T, Yagi T: Developmental epigenetic modification regulates stochastic expression of clustered protocadherin genes, generating single neuron diversity. Neuron. 2014, 82 (1): 94-108. 10.1016/j.neuron.2014.02.005.

    Article  CAS  PubMed  Google Scholar 

  4. Kohmura N, Senzaki K, Hamada S, Kai N, Yasuda R, Watanabe M, Ishii H, Yasuda M, Mishina M, Yagi T: Diversity revealed by a novel family of cadherins expressed in neurons at a synaptic complex. Neuron. 1998, 20 (6): 1137-1151. 10.1016/S0896-6273(00)80495-X.

    Article  CAS  PubMed  Google Scholar 

  5. Wu Q, Zhang T, Cheng JF, Kim Y, Grimwood J, Schmutz J, Dickson M, Noonan JP, Zhang MQ, Myers RM, Maniatis T: Comparative DNA sequence analysis of mouse and human protocadherin gene clusters. Genome Res. 2001, 11 (3): 389-404. 10.1101/gr.167301.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  6. Miyake K, Hirasawa T, Soutome M, Itoh M, Goto Y, Endoh K, Takahashi K, Kudo S, Nakagawa T, Yokoi S, Taira T, Inazawa J, Kubota T: The protocadherins, PCDHB1 and PCDH7, are regulated by MeCP2 in neuronal cells and brain tissues: implication for pathogenesis of Rett syndrome. BMC Neurosci. 2011, 12: 81-2202-12-81.

    Article  Google Scholar 

  7. Gu X, Xue JQ, Zhu X, Ye MS, Zhang WH: Aberrant promoter methylation of the CHD1 gene may contribute to the pathogenesis of breast cancer: a meta-analysis. Tumour Biol. 2014, 35 (7): 6783-6790. 10.1007/s13277-014-1905-1.

    Article  Google Scholar 

  8. Narayan G, Scotto L, Neelakantan V, Kottoor SH, Wong AH, Loke SL, Mansukhani M, Pothuri B, Wright JD, Kaufmann AM, Schneider A, Arias-Pulido H, Tao Q, Murty VV: Protocadherin PCDH10, involved in tumor progression, is a frequent and early target of promoter hypermethylation in cervical cancer. Genes Chromosomes Cancer. 2009, 48 (11): 983-992. 10.1002/gcc.20703.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  9. Yu J, Cheng YY, Tao Q, Cheung KF, Lam CN, Geng H, Tian LW, Wong YP, Tong JH, Ying JM, Jin H, To KF, Chan FK, Sung JJ: Methylation of protocadherin 10, a novel tumor suppressor, is associated with poor prognosis in patients with gastric cancer. Gastroenterology. 2009, 136 (2): 640-51.e1. 10.1053/j.gastro.2008.10.050.

    Article  CAS  PubMed  Google Scholar 

  10. Tang X, Yin X, Xiang T, Li H, Li F, Chen L, Ren G: Protocadherin 10 is frequently downregulated by promoter methylation and functions as a tumor suppressor gene in non-small cell lung cancer. Cancer Biomark. 2012, 12 (1): 11-19.

    CAS  PubMed  Google Scholar 

  11. Sugino H, Toyama T, Taguchi Y, Esumi S, Miyazaki M, Yagi T: Negative and positive effects of an IAP-LTR on nearby Pcdaalpha gene expression in the central nervous system and neuroblastoma cell lines. Gene. 2004, 337: 91-103.

    Article  CAS  PubMed  Google Scholar 

  12. Reynolds JN, Weinberg J, Clarren S, Beaulieu C, Rasmussen C, Kobor M, Dube MP, Goldowitz D: Fetal alcohol spectrum disorders: gene-environment interactions, predictive biomarkers, and the relationship between structural alterations in the brain and functional outcomes. Semin Pediatr Neurol. 2011, 18 (1): 49-55. 10.1016/j.spen.2011.02.006.

    Article  PubMed  Google Scholar 

  13. Erhardt A, Spoormaker VI: Translational approaches to anxiety: focus on genetics, fear extinction and brain imaging. Curr Psychiatry Rep. 2013, 15 (12): 417-013-0417-9.

    Article  Google Scholar 

  14. Dempster E, Viana J, Pidsley R, Mill J: Epigenetic studies of schizophrenia: progress, predicaments, and promises for the future. Schizophr Bull. 2013, 39 (1): 11-16. 10.1093/schbul/sbs139.

    Article  PubMed Central  PubMed  Google Scholar 

  15. Melka MG, Laufer BI, McDonald P, Castellani CA, Rajakumar N, O'Reilly R, Singh SM: The effects of olanzapine on genome-wide DNA methylation in the hippocampus and cerebellum. Clin Epigenetics. 2014, 6 (1): 1-7083-6-1.

    Article  Google Scholar 

  16. Wu YQ, Zhang WG, Tian Z, Zhang ZY: The expression of E-cadherin-catenin complex in human salivary adenoid cystic carcinoma. Shanghai Kou Qiang Yi Xue. 1999, 8 (3): 163-165.

    PubMed  Google Scholar 

  17. Yagi T: Clustered protocadherin family. Dev Growth Differ. 2008, 50 (Suppl 1): S131-S140.

    Article  CAS  PubMed  Google Scholar 

  18. Morishita H, Yagi T: Protocadherin family: diversity, structure, and function. Curr Opin Cell Biol. 2007, 19 (5): 584-592. 10.1016/j.ceb.2007.09.006.

    Article  CAS  PubMed  Google Scholar 

  19. Krishna K, Redies C: Expression of cadherin superfamily genes in brain vascular development. J Cereb Blood Flow Metab. 2009, 29 (2): 224-229. 10.1038/jcbfm.2008.123.

    Article  Google Scholar 

  20. Bray NJ, Kirov G, Owen RJ, Jacobsen NJ, Georgieva L, Williams HJ, Norton N, Spurlock G, Jones S, Zammit S, O'Donovan MC, Owen MJ: Screening the human protocadherin 8 (PCDH8) gene in schizophrenia. Genes Brain Behav. 2002, 1 (3): 187-191. 10.1034/j.1601-183X.2002.10307.x.

    Article  CAS  PubMed  Google Scholar 

  21. Wang X, Su H, Bradley A: Molecular mechanisms governing Pcdh-gamma gene expression: evidence for a multiple promoter and cis-alternative splicing model. Genes Dev. 2002, 16 (15): 1890-1905. 10.1101/gad.1004802.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  22. Kawaguchi M, Toyama T, Kaneko R, Hirayama T, Kawamura Y, Yagi T: Relationship between DNA methylation states and transcription of individual isoforms encoded by the protocadherin-alpha gene cluster. J Biol Chem. 2008, 283 (18): 12064-12075. 10.1074/jbc.M709648200.

    Article  CAS  PubMed  Google Scholar 

  23. Tucci V, Kleefstra T, Hardy A, Heise I, Maggi S, Willemsen MH, Hilton H, Esapa C, Simon M, Buenavista MT, McGuffin LJ, Vizor L, Dodero L, Tsaftaris S, Romero R, Nillesen WN, Vissers LE, Kempers MJ, Vulto-van Silfhout AT, Iqbal Z, Orlando M, Maccione A, Lassi G, Farisello P, Contestabile A, Tinarelli F, Nieus T, Raimondi A, Greco B, Cantatore D, et al: Dominant beta-catenin mutations cause intellectual disability with recognizable syndromic features. J Clin Invest. 2014, 124 (4): 1468-1482. 10.1172/JCI70372.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  24. Shishido E: Autism spectrum disorder and genes for synaptic proteins. Brain Nerve. 2012, 64 (1): 65-70.

    CAS  PubMed  Google Scholar 

  25. Pedrosa E, Stefanescu R, Margolis B, Petruolo O, Lo Y, Nolan K, Novak T, Stopkova P, Lachman HM: Analysis of protocadherin alpha gene enhancer polymorphism in bipolar disorder and schizophrenia. Schizophr Res. 2008, 102 (1–3): 210-219.

    Article  PubMed Central  PubMed  Google Scholar 

  26. Kalmady SV, Venkatasubramanian G: Evidence for positive selection on Protocadherin Y gene in Homo sapiens: implications for schizophrenia. Schizophr Res. 2009, 108 (1–3): 299-300.

    Article  PubMed  Google Scholar 

  27. Lachman HM, Petruolo OA, Pedrosa E, Novak T, Nolan K, Stopkova P: Analysis of protocadherin alpha gene deletion variant in bipolar disorder and schizophrenia. Psychiatr Genet. 2008, 18 (3): 110-115. 10.1097/YPG.0b013e3282fa1838.

    Article  PubMed  Google Scholar 

  28. Bardgett ME, Humphrey WM, Csernansky JG: The effects of excitotoxic hippocampal lesions in rats on risperidone- and olanzapine-induced locomotor suppression. Neuropsychopharmacology. 2002, 27 (6): 930-938. 10.1016/S0893-133X(02)00376-7.

    Article  CAS  PubMed  Google Scholar 

  29. Mohn F, Weber M, Schubeler D, Roloff TC: Methylated DNA immunoprecipitation (MeDIP). Methods Mol Biol. 2009, 507: 55-64. 10.1007/978-1-59745-522-0_5.

    Article  CAS  PubMed  Google Scholar 

  30. Bao L, Zhou M, Cui Y: CTCFBSDB: a CTCF binding site database for characterization of vertebrate genomic insulators. Nucleic Acids Res. 2008, 36: D83-D87. 10.1093/nar/gkn273.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  31. Redies C, Hertel N, Hubner CA: Cadherins and neuropsychiatric disorders. Brain Res. 2012, 1470: 130-144.

    Article  CAS  PubMed  Google Scholar 

  32. Hirano S, Takeichi M: Cadherins in brain morphogenesis and wiring. Physiol Rev. 2012, 92 (2): 597-634. 10.1152/physrev.00014.2011.

    Article  CAS  PubMed  Google Scholar 

  33. Foty RA, Steinberg MS: Cadherin-mediated cell-cell adhesion and tissue segregation in relation to malignancy. Int J Dev Biol. 2004, 48 (5–6): 397-409.

    Article  CAS  PubMed  Google Scholar 

  34. Nose A, Nagafuchi A, Takeichi M: Expressed recombinant cadherins mediate cell sorting in model systems. Cell. 1988, 54 (7): 993-1001. 10.1016/0092-8674(88)90114-6.

    Article  CAS  PubMed  Google Scholar 

  35. Halbleib JM, Nelson WJ: Cadherins in development: cell adhesion, sorting, and tissue morphogenesis. Genes Dev. 2006, 20 (23): 3199-3214. 10.1101/gad.1486806.

    Article  CAS  PubMed  Google Scholar 

  36. Strick PL, Dum RP, Fiez JA: Cerebellum and nonmotor function. Annu Rev Neurosci. 2009, 32: 413-434. 10.1146/annurev.neuro.31.060407.125606.

    Article  CAS  PubMed  Google Scholar 

  37. Wang M, Gong B, Gu J: Expression of G-protein gamma 7 in patients with extrahepatic cholangiocarcinoma. Zhonghua Zhong Liu Za Zhi. 2006, 28 (10): 771-772.

    PubMed  Google Scholar 

  38. Ridley AJ: Rho family proteins: coordinating cell responses. Trends Cell Biol. 2001, 11 (12): 471-477. 10.1016/S0962-8924(01)02153-5.

    Article  CAS  PubMed  Google Scholar 

  39. Freyberg Z, Ferrando SJ, Javitch JA: Roles of the Akt/GSK-3 and Wnt signaling pathways in schizophrenia and antipsychotic drug action. Am J Psychiatry. 2010, 167 (4): 388-396. 10.1176/appi.ajp.2009.08121873.

    Article  PubMed Central  PubMed  Google Scholar 

  40. Bousman CA, Glatt SJ, Chandler SD, Lohr J, Kremen WS, Tsuang MT, Everall IP: Negative Symptoms of Psychosis Correlate with Gene Expression of the Wnt/beta-Catenin Signaling Pathway in Peripheral Blood. Psychiatry J. 2013, 2013: 852930.

    Article  PubMed Central  PubMed  Google Scholar 

  41. Konze SA, van Diepen L, Schroder A, Olmer R, Moller H, Pich A, Weissmann R, Kuss AW, Zweigerdt R, Buettner FF: Cleavage of E-cadherin and beta-catenin by calpain affects Wnt signaling and spheroid formation in suspension cultures of human pluripotent stem cells. Mol Cell Proteomics. 2014, 13 (4): 990-1007. 10.1074/mcp.M113.033423.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  42. Ahn K, Huh JW, Kim DS, Ha HS, Kim YJ, Lee JR, Kim HS: Quantitative analysis of alternative transcripts of human PCDH11X/Y genes. Am J Med Genet B Neuropsychiatr Genet. 2010, 153B (3): 736-744.

    CAS  PubMed  Google Scholar 

  43. McGowan PO, Suderman M, Sasaki A, Huang TC, Hallett M, Meaney MJ, Szyf M: Broad epigenetic signature of maternal care in the brain of adult rats. PLoS One. 2011, 6 (2): e14739-10.1371/journal.pone.0014739.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  44. Santini MA, Ratner C, Aznar S, Klein AB, Knudsen GM, Mikkelsen JD: Enhanced prefrontal serotonin 2A receptor signaling in the subchronic phencyclidine mouse model of schizophrenia. J Neurosci Res. 2013, 91 (5): 634-641. 10.1002/jnr.23198.

    Article  CAS  PubMed  Google Scholar 

  45. Olszewski M, Piasecka J, Goda SA, Kasicki S, Hunt MJ: Antipsychotic compounds differentially modulate high-frequency oscillations in the rat nucleus accumbens: a comparison of first- and second-generation drugs. Int J Neuropsychopharmacol. 2013, 16 (5): 1009-1020. 10.1017/S1461145712001034.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This research was supported by grants from the Canadian Institute of Health Research (SMS) and the Ontario Mental Health Foundation (NR), and Dr. Shiva M. Singh held a Senior Research Fellowship from the Ontario Mental Health Foundation during the course of this study.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Shiva M Singh.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

MGM analyzed the data and wrote the first draft of the manuscript. CAC critically reviewed the manuscript for important intellectual contents. NR prepared the animals, provided animal tissues and required samples; NR and RO critically reviewed the manuscript for important intellectual contents. SMS co-wrote the manuscript, and critically reviewed the manuscript for important intellectual contents. All authors read and approved the final manuscript.

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.

Authors’ original file for figure 1

Authors’ original file for figure 2

Rights and permissions

Open Access  This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.

The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.

To view a copy of this licence, visit https://creativecommons.org/licenses/by/4.0/.

The Creative Commons Public Domain Dedication waiver (https://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Melka, M.G., Castellani, C.A., Rajakumar, N. et al. Olanzapine-induced methylation alters cadherin gene families and associated pathways implicated in psychosis. BMC Neurosci 15, 112 (2014). https://doi.org/10.1186/1471-2202-15-112

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1471-2202-15-112

Keywords