Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Fluoxetine Increases the Expression of miR-572 and miR-663a in Human Neuroblastoma Cell Lines

  • Mahesh Mundalil Vasu,

    Affiliation Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan

  • Ayyappan Anitha,

    Affiliation Department of Neurogenetics, Institute for Communicative and Cognitive Neurosciences (ICCONS), Kerala, India

  • Taro Takahashi,

    Affiliation Department of Child and Adolescent Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan

  • Ismail Thanseem,

    Affiliation Department of Neurogenetics, Institute for Communicative and Cognitive Neurosciences (ICCONS), Kerala, India

  • Keiko Iwata,

    Affiliation Research Center for Child Mental Development, University of Fukui, Fukui, Japan

  • Tetsuya Asakawa,

    Affiliation Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan

  • Katsuaki Suzuki

    katsuaki-suzuki@ams.odn.ne.jp

    Affiliation Department of Biofunctional Imaging, Preeminent Medical Photonics Education and Research Center, Hamamatsu University School of Medicine, Hamamatsu, Japan

Abstract

Evidence suggests neuroprotective effects of fluoxetine, a selective serotonin reuptake inhibitor (SSRI), on the developed neurons in the adult brain. In contrast, the drug may be deleterious to immature or undifferentiated neural cells, although the mechanism is unclear. Recent investigations have suggested that microRNAs (miRNA) may be critical for effectiveness of psychotropic drugs including SSRI. We investigated whether fluoxetine could modulate expressions of neurologically relevant miRNAs in two neuroblastoma SK-N-SH and SH-SY5Y cell lines. Initial screening results revealed that three (miR-489, miR-572 and miR-663a) and four (miR-320a, miR-489, miR-572 and miR-663a) miRNAs were up-regulated in SK-N-SH cells and SH-SY5Y cells, respectively, after 24 hours treatment of fluoxetine (1–25 μM). Cell viability was reduced according to the dose of fluoxetine. The upregulation of miR-572 and miR-663a was consistent in both the SH-SY5Y and SK-N-SH cells, confirmed by a larger scale culture condition. Our data is the first in vitro evidence that fluoxetine could increase the expression of miRNAs in undifferentiated neural cells, and that putative target genes of those miRNAs have been shown to be involved in fundamental neurodevelopmental processes.

Introduction

Fluoxetine is one of the most commonly prescribed selective serotonin reuptake inhibitors (SSRIs). SSRIs can alleviate a wide variety of psychiatric symptoms such as depression, anxiety, obsession and compulsion. Underlying the clinical effects of SSRIs are multiple physiological processes, including neuroplasticity, neuroprotection, and neurogenesis in the adult brain [1]. In contrast to beneficial effects on differentiated neurons in the developed brain, SSRIs may be toxic to undifferentiated or immature cells. For instance, fluoxetine decreases the proliferation and differentiation of adipose-derived stem cells [2]. Fluoxetine also induces apoptosis in hepatic cancer cells [3], and suppresses growth of various cancer cells including lung, colon, and breast cancers, glioma and neuroblastoma [4]. With regard to the brain development, animal studies have suggested prenatal in utero exposure to SSRI is associated with adverse neurodevelopmental outcomes in the offspring [5, 6]. Furthermore, epidemiologic studies [7, 8] have suggested that the use of SSRIs by mother during pregnancy increases their children’s risk for autism spectrum disorder (ASD), a neurodevelopmental disorder characterized by social interaction deficits and restricted pattern of behaviours and interests [9]. These findings suggest that SSRI can affect neurodevelopment of the fetal brain, although the underlying mechanisms remain unclear.

MicroRNAs (miRNA) are small non-coding RNA molecules that post-transcriptionally regulate gene expression through sequence-specific interaction with its target messenger RNAs (mRNAs). miRNAs bind to the 3′ untranslated regions (UTR) of mRNAs to inhibit protein translation or to cause mRNA degradation [10, 11]. miRNAs have been predicted to modulate the expression of more than half of all protein coding genes[12]. Currently, 2,588 mature human miRNAs have been documented and verified (http://www.mirbase.org/cgi-bin/browse.pl?org=hsa). Approximately 50% of these miRNAs are abundantly or exclusively expressed in the brain [13], where they could be involved in fundamental processes such as the neurogenesis, neurodevelopment, and synaptic plasticity [1419]. Concordant with these observations, aberrant miRNA expression has been detected in several neuropsychiatric disorders including ASD [2023].

We have recently found that expressions of a group of miRNAs in the sera from children with ASD were abnormal compared with control [24]. The targets of these miRNAs were genes known to be involved in crucial neurological pathways and functions. These findings led us to assume a possible effect of SSRI on the expression of miRNAs in immature neural cells. In this in vitro study using undifferentiated human neuroblastoma cells, we examined whether fluoxetine could modulate the expression of miRNAs that were up- or down-regulated in children with ASD in our previous study [24].

Materials and Methods

Cell Culture

SK-N-SH cells (ATCC, Manassas, VA) were cultured and grown in Dulbecco's Modified Eagle's Medium (DMEM; Sigma-Aldrich, St. Louis, MD) supplemented with 10% Fetal Bovine Serum (FBS; Invitrogen, Carlsbad, CA). SH-SY5Y cells (ATCC) were maintained in 1:1 mixture of Hams F12 medium (Sigma-Aldrich) and Minimum Essential Medium (MEM, Sigma-Aldrich) supplemented with 15% FBS, 2 mM L-glutamine (Life Technologies, Tokyo, Japan) and 1% non-essential amino acids (NEAA, Sigma-Aldrich). Cells were grown at 37°C and under 5% CO2. Cells from the 3rd passage were used for drug experiments.

Drug Treatment

Fifteen millimolar stock of fluoxetine hydrochloride (Sigma-Aldrich) was prepared in dimethyl sulfoxide (DMSO). Cells, seeded at a density of 5 × 104 cells/well in 24 well plates, were treated with fluoxetine hydrochloride at 1-, 5-, 10-, 25-μM concentrations (3 wells each) at 50% confluency. An equal volume of DMSO was added to the control wells. The cells were harvested after 24 hours for miRNA analysis. Optimum drug concentration was chosen after assessing cell viability using trypan blue exclusion test [25].

RNA extraction and cDNA synthesis

Total RNA, including miRNA, was extracted from harvested cells by using miRNeasy Kit (QIAGEN GmbH, Hilden, Germany) in accordance with the manufacturer’s protocol. The RNA preparation was further purified using RNeasy Mini Kit (QIAGEN). The quality and quantity of RNA were estimated using a NanoDrop 1000 Spectrophotometer (Thermo Scientific, Yokohama, Japan).

Complementary DNA (cDNA) was synthesized using the miScript II RT kit (QIAGEN). The reverse-transcription reaction mix (20 μl) was prepared using Hispec buffer for selective conversion of mature miRNAs into cDNA. The reaction mixture was incubated for 60 min at 37°C, followed by denaturation for 5 min at 95°C. Each cDNA preparation was further diluted to 220 μl with RNase-free water and stored at −20°C until use.

Quantitative Real–Time Polymerase Chain Reaction (qPCR)

SYBR Green qPCR, performed on an ABI PRISM 7900 SDS (Applied Biosystems, Foster City, CA, USA), was used for the quantification of 13 miRNAs shown in Table 1. Ten microliters of qPCR reaction mix was prepared using the corresponding primer assays (forward primer) and a universal primer (reverse primer). All the qPCR reactions were performed in triplicate with the following cycling conditions: 95°C →15 min, followed by 40 cycles of 94°C →15 sec, 55°C →30 sec and 70°C → 30 sec. Expression of individual miRNAs was normalized against the expression of SNORD95 (QIAGEN, Accession ID: MS00033726). Fold change in gene expression between control and drug-treated cells was determined by the ΔΔCt method of relative quantification.

thumbnail
Table 1. miRNAs selected for the study with accession ID and mature sequence.

https://doi.org/10.1371/journal.pone.0164425.t001

Expression of SLC6A4 in SK-N-SH and SH-SY5Y

Expression of SLC6A4 protein was examined in SK-N-SH and SH-SY5Y cells was examined by Western blot analysis using anti-SERT (SAB4200039, Sigma-Aldrich, St. Louis, MO, USA) antibody [26]. The amount of total proteins in cell homogenate was measured by BCA protein assay (Rockford, Illinois, USA). Fluorescent-labelled (IRDye 800CW) anti-rabbit secondary antibody (Li-cor Biosciences, Lincoln, Nebraska) was used for the detection of protein band with an Odyssey Infrared Imaging System (Li-cor Bioscience).

qPCR was used to examine the expression of SLC6A4 mRNA in the cells. qPCR was performed in ABI PRISM 7900 Sequence Detection System by SYBR Green protocol. 16 μl reaction mixture containing 1μl cDNA (synthesized by using RT kit from Qiagen), 8 μl SYBR Green PCR Master Mix (Qiagen), 1 μl each of forward primer (5’-CTAGCAAAAGCCAGCAGGAC -3’) and reverse primer (5’-GTGACAGCCACCTTCCCTTA-3’) and 5 μl RNase free water, was used for qPCR. Reactions were performed in triplicate with the following cycling conditions: 95°C →15 min, followed by 40 cycles of 94°C →15 sec, 60°C →30 sec and 72°C → 30 sec.

Statistical analysis

Any differential expression of miRNAs between the control and drug-treated cells was determined by Student’s t-test. All statistical calculations were performed using PASW Statistics 18 (IBM, Tokyo, Japan).

Results

Expression of SLC6A4 mRNA and the protein in the neuroblastoma cell lines

mRNA expression of SLC6A4 in the neuroblastoma cell lines was examined by qPCR (threshold cycle, Ct range = 30–32) and the protein expression was confirmed by Western blot (Fig 1).

thumbnail
Fig 1. Western blot showing expression of SLC6A4 protein in SK-N-SH and SH-SY5Y cells at ~83 kDa (SAB4200039).

Abbreviations: M, molecular weight marker; Sk, SK-N-SH cells; Sh, SH-SY5Y cells. Concentration of sample per lane is 1.9 ug/μl.

https://doi.org/10.1371/journal.pone.0164425.g001

Preliminary microRNA screening

Among the 13 miRNAs screened except miR-433, which was omitted from further analysis since the amplification was not proper in the cell lines studied, three miRNAs [miR-489 (p = 0.01 at 1μM), miR-572 (p = 0.009 at 1μM; p = 0.0004 at 5μM; p = 0.024 at 10μM); miR-663a (p = 0.032 at 5μM; p = 0.058 at 10μM)] were up-regulated in fluoxetine-treated (1-, 5- and 10-μM) SK-N-SH cells, while four miRNAs [miR-320a (p = 0.004 at 10μM), miR-489 (p = 0.037 at 10μM), miR-572 (p = 0.0002 at 5μM; p = 0.0004 at 10μM); miR-663a (p = 0.006 at 10μM)] were up-regulated in fluoxetine-treated SH-SY5Y cells compared to corresponding DMSO-treated control cells (Fig 2). Up-regulation of miRNA expression was observed at all concentrations of fluoxetine in both the cells. miR-489, miR-572 and miR-663a were consistently up-regulated in fluoxetine-treated SK-N-SH and SH-SY5Y cells. See S1 and S2 Files for supporting data.

thumbnail
Fig 2. Up-regulated expression of miRNAs (miR-320a, miR-489, miR-572 and miR-663a) in (a) SK-N-SH cells and (b) SH-SY5Y cells treated with various concentrations of fluoxetine compared to DMSO-treated control cells.

Error bars represents standard error. *p < 0.05.

https://doi.org/10.1371/journal.pone.0164425.g002

Cell viability

In both the cell lines, 90 ± 3% cell viability for fluoxetine was detected for concentrations 1-, 5- and 10-μM by trypan blue viability test, while at 25-μM fluoxetine the SH-SY5Y cells found 60 ± 3% cell viability but for SK-N-SH cells this was found lethal. Therefore, a concentration of 10-μM Fluoxetine was used in the subsequent validation experiments mentioned below.

Validation

The expression of miR-320, miR-489, miR-572 and miR-663a were further compared between 10 μM fluoxetine (6 wells each) treated SK-N-SH and SH-SY5Y cells and their control cells. Up-regulated expression of miR-572 (ΔΔCt = 2.161, p = 4.58E-06) and miR-663a (ΔΔCt = 1.911, p = 9.75E-06) were confirmed in fluoxetine-treated SK-N-SH cells, while the up-regulated expression of miR-489 (ΔΔCt = 1.2, p = 0.007), miR-572 (ΔΔCt = 3.392, p = 3.81E-07) and miR-663a (ΔΔCt = 1.94, p = 1.77E-06) were confirmed in fluoxetine-treated SH-SY5Y cells (Fig 3). See S3 and S4 Files for supporting data.

thumbnail
Fig 3. Up-regulated expression of miR-572 and miR-663a in 10 μM fluoxetine-treated (a) SK-N-SH cells and (b) SH-SY5Y cells compared to DMSO-treated control cells.

Error bars represent standard error. *p < 0.05.

https://doi.org/10.1371/journal.pone.0164425.g003

Discussion

The essential finding emerged from the current study was that fluoxetine (1–10 μM) up-regulated the expression of miR-572 and miR-663a consistently in two neuroblastoma cell lines SK-N-SH and SH-SY5Y. The up-regulating effect of fluoxetine was dose-dependent, and higher dose (25-μM) of fluoxetine was toxic to both cell lines. After standard therapeutic doses (40 mg per day, 30 days) administered orally, plasma levels of fluoxetine reach about 1 μM [27, 28], which is much lower than the concentration that has shown to induce upregulations of miRNAs in our study. In the brain, however, fluoxetine has ability to accumulate in the tissue due to its lipophilic property, and its concentration could reach higher levels in the brain than in plasma [29, 30]. Therefore, it is likely that the dose 5–10 μM of fluoxetine is within the clinically relevant doses. In the clinical setting, psychiatrists have used fluoxetine for treatment of patients for extended periods of time at doses higher than 100 mg per day without significant side-effects [31]. These clinical data could support our contention that SSRI fluoxetine is beneficial, but not toxic, on differentiated neurons in the developed brain.

miR-572 has been implicated in the pathogenesis of several neurological disorders such as ASD [20, 24], schizophrenia [32] and multiple sclerosis [33]. The expression of miR-572 has been found to be associated with cognitive dysfunction through down-regulation of one of its target, neuronal cell adhesion molecule 1 (NCAM1) [34]. NCAM1 is known to play an important role in neuronal cell variability, axonal proliferation, and neuronal plasticity [35, 36]. Yu et al (2015) have shown that increased miR-572 expression may lead to reduced downstream NCAM1 expression and loss of neuronal protection in patients with postoperative cognitive dysfunction [34]. Our result may be inconsistent with results by Choi et al (2012) study, in which fluoxetine regulated neuronal plasticity and neurite outgrowth by phosphorylating and activating cAMP response element-binding protein (CREB) via NCAM-induced activation of MAPK pathway, probably because they used differentiated neuron-like cells from glioma cell line. Some other target genes of miR-572 including CDKN1A, DICER1, WNT7A are also reported to be associated with vital neural functions, which might be down-regulated by fluoxetine-induced upregulation of miR-572 expression [37]. miR-663 has been reported as a primate-specific miRNA largely expressed in the brain [38]. miR-663a has been reported to suppress neuronal differentiation in human neural stem cells [39]. Further, it modulates the expression of genes such as TGFβ1, DICER1, PTEN and VEGFA which regulate crucial neuronal functions such as neurogenesis, neurodevelopment and synaptic plasticity [40]. In addition to miR-572 and miR-663a, the expression of miR-489 was up-regulated in fluoxetine-treated SH-SY5Y cells. miR-489, a brain specific-miRNA [41, 42], has been implicated in the pathophysiology of various neuropsychiatric disorders [4346, 24]. Alteration of miR-489 and its target genes have been observed in postmortem brain samples from schizophrenic [43] and depressed suicidal patients [44]. The importance of miR-489 in synaptic transmission has been demonstrated in a mouse model of Alzheimer's disease [45]. In our previous study, target genes of miR-489 have been identified and predicted to be involved in important signalling systems such as MAPK, ErbB, TGF-beta and hedgehog signalling [24]. Nevertheless, whether the changes of miRNAs after fluoxetine exposure will lead to a significant alteration of respective targets and their biological functions requires further investigation.

The mechanism by which fluoxetine caused upregulation of miRNA expression was unknown. Since two lines of neuroblastoma cells we used in this study expressed serotonin transporter detectable at mRNA and protein levels, the simplest explanation for the upregulation of miRNA expression by fluoxetine would be binding of the drug with serotonin transporter. This is however unlikely, because the reported affinity (Ki) of fluoxetine for serotonin transporter is on the order of 1 nM [47], the value is far lower than the 5–10 μM concentration that we found a significant upregulation of miRNA expression. Although mechanism of antidepressant action of fluoxetine is associated with the inhibition of serotonin transporters, non-serotoninergic effects of fluoxetine have been reported. For instance, Stepulak et al. (2008) reported that fluoxetine inhibited the growth of lung and colon cancer cells as a result of inhibition of ERK1/2 activation [4]. It has also been reported that various antidepressants including fluoxetine were able to induce translocation of glucocorticoid receptor to nucleus and modulate transcription in fibroblast cells, human lymphocytes and neural cell line [48, 49]. Further study using experimental animals that examine expression levels of miRNAs in offspring from mothers having treated with SSRI during pregnancy would be warranted in this regard.

To the best of our knowledge, our data is the first in vitro evidence that fluoxetine could increase the expression of some miRNAs implicated in ASD. Target genes of these miRNA will be a focus of future studies, and it would be also important to validate the current findings in primary neurons and in vivo models of ASD.

Supporting Information

S1 File. Preliminary qPCR experimental data file of SH-SY5Y cells with three different concentrations (1-, 5-, 10- and 25-μM) of Fluoxetine against equal concentration of DMSO control.

https://doi.org/10.1371/journal.pone.0164425.s001

(XLSX)

S2 File. Preliminary qPCR experimental data file of SK-N-SH cells with three different concentrations (1-, 5- and 10-μM) of Fluoxetine against equal concentration of DMSO control.

https://doi.org/10.1371/journal.pone.0164425.s002

(XLSX)

S3 File. Confirmation qPCR experimental data with four miRNAs (miR-320, miR-489, miR-572 and miR-663a) of SH-SY5Y cells with 10-μM Fluoxetine and equal concentration of DMSO control.

https://doi.org/10.1371/journal.pone.0164425.s003

(XLSX)

S4 File. Confirmation qPCR experimental data with four miRNAs (miR-320, miR-489, miR-572 and miR-663a) of SK-N-SH cells with 10-μM Fluoxetine and equal concentration of DMSO control.

https://doi.org/10.1371/journal.pone.0164425.s004

(XLSX)

Acknowledgments

We thank Mses. Rie Gonda and Emiko Hatano for their technical assistance.

Author Contributions

  1. Conceptualization: MMV AA KS.
  2. Data curation: MMV AA KS TT.
  3. Formal analysis: MMV AA KS.
  4. Investigation: MMV.
  5. Methodology: MMV AA IT TT KI.
  6. Project administration: MMV AA KS.
  7. Resources: MMV KI TA TT.
  8. Software: MMV TT TA.
  9. Supervision: AA KS.
  10. Validation: MMV AA KS.
  11. Visualization: MMV AA KS.
  12. Writing – original draft: MMV AA IT KS.
  13. Writing – review & editing: MMV AA TT IT KI TA KS.

References

  1. 1. Krishnan V, Nestler EJ. The molecular neurobiology of depression. Nature. 2008;455(7215):894–902. pmid:18923511
  2. 2. Sun BK, Kim JH, Choi JS, Hwang SJ, Sung JH. Fluoxetine Decreases the Proliferation and Adipogenic Differentiation of Human Adipose-Derived Stem Cells. Int J Mol Sci. 2015;16(7):16655–68. pmid:26204837
  3. 3. Mun AR, Lee SJ, Kim GB, Kang HS, Kim JS, Kim SJ. Fluoxetine-induced apoptosis in hepatocellular carcinoma cells. Anticancer Res. 2013;33(9):3691–7. pmid:24023297
  4. 4. Stepulak A, Rzeski W, Sifringer M, Brocke K, Gratopp A, Kupisz K, et al. Fluoxetine inhibits the extracellular signal regulated kinase pathway and suppresses growth of cancer cells. Cancer Biol Ther. 2008;7(10):1685–93. pmid:18836303
  5. 5. Vorhees CV, Acuff-Smith KD, Schilling MA, Fisher JE, Moran MS, Buelke-Sam J. A developmental neurotoxicity evaluation of the effects of prenatal exposure to fluoxetine in rats. Fundam Appl Toxicol. 1994;23(2):194–205. pmid:7982528
  6. 6. Maciag D, Simpson KL, Coppinger D, Lu Y, Wang Y, Lin RC, et al. Neonatal antidepressant exposure has lasting effects on behavior and serotonin circuitry. Neuropsychopharmacology. 2006;31(1):47–57. pmid:16012532
  7. 7. Man KK, Tong HH, Wong LY, Chan EW, Simonoff E, Wong IC. Exposure to selective serotonin reuptake inhibitors during pregnancy and risk of autism spectrum disorder in children: a systematic review and meta-analysis of observational studies. Neurosci Biobehav Rev. 2015;49:82–9. pmid:25498856
  8. 8. Boukhris T, Sheehy O, Mottron L, Bérard A. Antidepressant Use During Pregnancy and the Risk of Autism Spectrum Disorder in Children. JAMA Pediatr. 2016;170(2):117–24. pmid:26660917
  9. 9. American Psychiatric Association: Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition. Arlington, VA, USA. American Psychiatric Association, 2013.
  10. 10. Ambros V. The functions of animal microRNAs. Nature 2004;431(7006):350–5. pmid:15372042
  11. 11. Huntzinger E and Izaurralde E. Gene silencing by microRNAs: contributions of translational repression and mRNA decay. Nat Rev Genet. 2011;12(2):99–110. pmid:21245828
  12. 12. Yates LA, Norbury CJ, Gilbert RJ. The long and short of microRNA. Cell. 2013;153(3):516–9. pmid:23622238
  13. 13. Xu B, Karayiorgou M, Gogos JA. MicroRNAs in psychiatric and neurodevelopmental disorders. Brain Res. 2010;1338:78–88. pmid:20388499
  14. 14. Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell. 2009;136(2):215–33. pmid:19167326
  15. 15. Forero DA, van der Ven K, Callaerts P, Del-Favero J. miRNA genes and the brain: implications for psychiatric disorders. Hum Mutat. 2010;31(11):1195–204. pmid:20725930
  16. 16. McNeill E, Van Vactor D. MicroRNAs shape the neuronal landscape. Neuron. 2012;75(3):363–79. pmid:22884321
  17. 17. Mellios N, Sugihara H, Castro J, Banerjee A, Le C, Kumar A, et al. miR-132, an experience-dependent microRNA, is essential for visual cortex plasticity. Nat Neurosci. 2011;14(10):1240–2. pmid:21892155
  18. 18. O'Connor RM, Dinan TG, Cryan JF. Little things on which happiness depends: microRNAs as novel therapeutic targets for the treatment of anxiety and depression. Mol Psychiatry. 2012;17(4):359–76. pmid:22182940
  19. 19. Siegel G, Saba R, Schratt G. microRNAs in neurons: manifold regulatory roles at the synapse. Curr Opin Genet Dev. 2011;21(4):491–7. pmid:21561760
  20. 20. Abu-Elneel K, Liu T, Gazzaniga FS, Nishimura Y, Wall DP, Geschwind DH, et al. Heterogeneous dysregulation of microRNAs across the autism spectrum. Neurogenetics 2008;9(3):153–61. pmid:18563458
  21. 21. Talebizadeh Z, Butler MG, Theodoro MF. Feasibility and relevance of examining lymphoblastoid cell lines to study role of microRNAs in autism. Autism Res 2008;1(4):240–50. pmid:19360674
  22. 22. Sarachana T, Zhou R, Chen G, Manji HK, Hu VW. Investigation of post-transcriptional gene regulatory networks associated with autism spectrum disorders by microRNA expression profiling of lymphoblastoid cell lines. Genome Med 2010;2(4):23. pmid:20374639
  23. 23. Ghahramani Seno MM, Hu P, Gwadry FG, Pinto D, Marshall CR, Casallo G, et al. Gene and miRNA expression profiles in autism spectrum disorders. Brain Res 2011;1380:85–97. pmid:20868653
  24. 24. Mundalil Vasu M, Anitha A, Thanseem I, Suzuki K, Yamada K, Takahashi T, et al. Serum microRNA profiles in children with autism. Mol Autism. 2014;5:40. pmid:25126405
  25. 25. Strober W. Trypan blue exclusion test of cell viability. Curr Protoc Immunol. 2001 May;Appendix 3:Appendix 3B. pmid:18432654
  26. 26. Riksen EA, Stunes AK, Kalvik A, Gustafsson BI, Snead ML, Syversen U, et al. Serotonin and fluoxetine receptors are expressed in enamel organs and LS8 cells and modulate gene expression in LS8 cells. Eur J Oral Sci. 2010;118(6):566–73. pmid:21083617
  27. 27. Kristensen JH, Ilett KF, Hackett LP, Yapp P, Paech M, Begg EJ. Distribution and excretion of fluoxetine and norfluoxetine in human milk. Br J Clin Pharmacol. 1999;48(4):521–7. pmid:10583022
  28. 28. Cheer SM, Goa KL. Fluoxetine: a review of its therapeutic potential in the treatment of depression associated with physical illness. Drugs. 2001;61(1):81–110. pmid:11217873
  29. 29. Karson CN, Newton JE, Livingston R, Jolly JB, Cooper TB, Sprigg J, et al. Human brain fluoxetine concentrations. J Neuropsychiatry Clin Neurosci. 1993;5(3):322–9. pmid:8369643
  30. 30. Bolo NR, Hodé Y, Nédélec JF, Lainé E, Wagner G, Macher JP. Brain pharmacokinetics and tissue distribution in vivo of fluvoxamine and fluoxetine by fluorine magnetic resonance spectroscopy. Neuropsychopharmacology. 2000;23(4):428–38. pmid:10989270
  31. 31. Stoll AL, Pope HG Jr, McElroy SL. High-dose fluoxetine: safety and efficacy in 27 cases. J Clin Psychopharmacol 1991 Jun;11(3):225–6. pmid:2066466
  32. 32. Lai CY, Yu SL, Hsieh MH, Chen CH, Chen HY, Wen CC, et al. MicroRNA expression aberration as potential peripheral blood biomarkers for schizophrenia. PLoS One. 2011;6(6):e21635. pmid:21738743
  33. 33. Mancuso R, Hernis A, Agostini S, Rovaris M, Caputo D, Clerici M. MicroRNA-572 expression in multiple sclerosis patients with different patterns of clinical progression. J Transl Med. 2015;13:148. pmid:25947625
  34. 34. Yu X, Liu S, Li J, Fan X, Chen Y, Bi X, et al. MicroRNA-572 improves early post-operative cognitive dysfunction by down-regulating neural cell adhesion molecule 1. PLoS One. 2015;10(2):e0118511. pmid:25680004
  35. 35. Li S, Leshchyns'ka I, Chernyshova Y, Schachner M, Sytnyk V. The neural cell adhesion molecule (NCAM) associates with and signals through p21-activated kinase 1 (Pak1). J Neurosci. 2013;33(2):790–803. pmid:23303955
  36. 36. Dallérac G, Zerwas M, Novikova T, Callu D, Leblanc-Veyrac P, Bock E, et al. The neural cell adhesion molecule-derived peptide FGL facilitates long-term plasticity in the dentate gyrus in vivo. Learn Mem. 2011;18(5):306–13. pmid:21508096
  37. 37. Choi MR, Oh DH, Kim SH, Jung KH, Das ND, Chai YG. Fluoxetine increases the expression of NCAM140 and pCREB in rat C6 glioma cells. Psychiatry Investig. 2012;9(2):180–6. pmid:22707970
  38. 38. Shi Y, Chen C, Zhang X, Liu Q, Xu JL, Zhang HR, et al. Primate-specific miR-663 functions as a tumor suppressor by targeting PIK3CD and predicts the prognosis of human glioblastoma. Clin Cancer Res. 2014;20(7):1803–13. pmid:24523440
  39. 39. Shu R, Wong W, Ma QH, Yang ZZ, Zhu H, Liu FJ, et al. APP intracellular domain acts as a transcriptional regulator of miR-663 suppressing neuronal differentiation. Cell Death Dis. 2015;6:e1651. pmid:25695604
  40. 40. Fan H, Niu W, He M, Kong L, Zhong A, Zhang Q, et al. [Bioinformatics analysis of differently expressed microRNAs in anxiety disorder] [Article in Chinese]. Zhonghua Yi Xue Yi Chuan Xue Za Zhi. 2015;32(5):641–6. pmid:26418982
  41. 41. Kloosterman WP, Steiner FA, Berezikov E, de Bruijn E, van de Belt J, Verheul M, et al. Cloning and expression of new microRNAs from zebrafish. Nucleic Acids Res. 2006;34(9):2558–69. pmid:16698962
  42. 42. Olsen L, Klausen M, Helboe L, Nielsen FC, Werge T. MicroRNAs show mutually exclusive expression patterns in the brain of adult male rats. PLoS One. 2009;4(10):e7225. pmid:19806225
  43. 43. Beveridge NJ, Gardiner E, Carroll AP, Tooney PA, Cairns MJ: Schizophrenia is associated with an increase in cortical microRNA biogenesis. Mol Psychiatry 2010;15(12):1176–89. pmid:19721432
  44. 44. Smalheiser NR, Lugli G, Rizavi HS, Torvik VI, Turecki G, Dwivedi Y. MicroRNA expression is down-regulated and reorganized in prefrontal cortex of depressed suicide subjects. PLoS One. 2012;7(3):e33201. pmid:22427989
  45. 45. Barak B, Shvarts-Serebro I, Modai S, Gilam A, Okun E, Michaelson DM, et al. Opposing actions of environmental enrichment and Alzheimer's disease on the expression of hippocampal microRNAs in mouse models. Transl Psychiatry. 2013;3:e304. pmid:24022509
  46. 46. De Santis G, Ferracin M, Biondani A, Caniatti L, Rosaria Tola M, Castellazzi M, et al. Altered miRNA expression in T regulatory cells in course of multiple sclerosis. J Neuroimmunol. 2010;226(1–2):165–71. pmid:20637509
  47. 47. Owens MJ, Morgan WN, Plott SJ, Nemeroff CB. Neurotransmitter receptor and transporter binding profile of antidepressants and their metabolites. J Pharmacol Exp Ther. 1997;283(3):1305–22. pmid:9400006
  48. 48. Herr AS, Tsolakidou AF, Yassouridis A, Holsboer F, Rein T. Antidepressants differentially influence the transcriptional activity of the glucocorticoid receptor in vitro. Neuroendocrinology. 2003;78(1):12–22. pmid:12869795
  49. 49. Pariante CM, Kim RB, Makoff A, Kerwin RW. Antidepressant fluoxetine enhances glucocorticoid receptor function in vitro by modulating membrane steroid transporters. Br J Pharmacol. 2003;139(6):1111–8. pmid:12871829