Open Access

Melatonin regulation of transcription in the reversal of morphine tolerance: Microarray analysis of differential gene expression

  • Authors:
    • Yu‑Che Cheng
    • Ru‑Yin Tsai
    • Yen‑Tseng Sung
    • Ing‑Jung Chen
    • Tzu‑Yi Tu
    • Ya‑Yuan Mao
    • Chih‑Shung Wong
  • View Affiliations

  • Published online on: December 18, 2018     https://doi.org/10.3892/ijmm.2018.4030
  • Pages: 791-806
  • Copyright: © Cheng et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Tolerance and associated hyperalgesia induced by long‑term morphine administration substantially restrict the clinical use of morphine in pain treatment. Melatonin, a neurohormone released by the pineal gland, has been demonstrated to attenuate anti‑nociceptive morphine tolerance. The present study investigates differentially expressed genes in the process of morphine tolerance and altered gene expression subsequent to melatonin treatment in chronic morphine‑infused ratspinal cords. Morphine tolerance was induced in male Wistar rats by intrathecal morphine infusion (the MO group). Melatonin (the MOMa group) was administered to overcome the effects derived by morphine. The mRNA collected from L5‑S3 of the spinal cord was extracted and analysed by rat expression microarray. Principal component analysis and clustering analysis revealed that the overall gene profiles were different in morphine and melatonin treatments. Subsequent to Gene Ontology analysis, the biological processes of differentially expressed genes of MO and MOMa compared with the control group were constructed. Furthermore, a panel of genes exclusively expressed following melatonin treatment and another panel of genes with inverse expression between the MO and MOMa group were also established. Subsequent to PANTHER pathway analysis, a group of genes with inverse expression following melatonin administrated compared with morphine alone were identified. The expression levels of genes of interest were also confirmed using a reverse transcription‑quantitative polymerase chain reaction. The gene panel that was constructed suggests a potential signaling pathway in morphine tolerance development and is valuable for investigating the mechanism of morphine tolerance and the regulatory gene profiles of melatonin treatment. These results may contribute to the discovery of potential drug targets in morphine tolerance treatments in the future.

Introduction

Morphine is a powerful analgesic agent used for treating acute and chronic pain in surgical interventions or in hospice care (1). However, long-term administration of morphine induces tolerance and hyperalgesia. Furthermore, adverse effects, including addiction, dependence, constipation and respiratory depression limit its clinical usefulness (2,3). The physiological responses of morphine tolerance include opioid receptor uncoupling, endocytosis/desensitization (4), increased binding of β-arrestin to opioid receptors, glutamatergic receptor activation and neuroinflammation (5). Melatonin is a neurohormone derived from serotonin and is released from the pineal gland (6). It is used for sleep modulation and relieves the stress caused by sleep disturbance (1). It has previously been revealed that melatonin treatment partially reverses morphine tolerance by inhibiting microglia activation though a heat shock protein 27 (HSP27)-associated pathway (7). Furthermore, melatonin co-treatment was revealed to prevent morphine-induced hyperalgesia and tolerance in rats, potentially by inhibiting protein kinase C-associated pathways (8,9). A report also demonstrated that decreased mitochondrial DNA copy numbers in the hippocampus during opiate addiction were mediated by autophagy and may be reversed by melatonin (10). Additionally, melatonin was revealed to enhance the reward behaviour of morphine via the nitric oxidergic pathway (11). Raghavendra and Kulkarni initially reported that the systemic administration of melatonin reversed morphine-induced tolerance in mice (12). Song et al (8) identified that daily intraperitoneal melatonin treatment reduced morphine tolerance in rats via the regulation of the N-methyl-D-aspartate receptor subunit 1. Furthermore, Garmabi et al (13) observed a reduction of melatonin levels in rats under constant light exposure; those animals also presented a high morphine consumption and severe morphine withdrawal syndrome. Fan et al (14) further reported a substantial decrease of serum melatonin and melatonin receptor 1 mRNA subsequent to chronic morphine infusion in rats. Previously, not only was it revealed that melatonin treatment partially reversed morphine tolerance by inhibiting microglia activation though a HSP27-associated pathway (7), but preliminary examinations additionally revealed that chronic morphine treatment resulted in transcriptomics changes. All studies noted that melatonin participates in the morphine tolerance pathway. Although melatonin was demonstrated to diminish morphine tolerance, the transcriptomic changes derived from melatonin treatment in opiate tolerance remain undetermined. To search whole genome expression profiles disturbed by long-term morphine administration and clarify the gene alterations caused by melatonin, an expression array was used in the present study to examine the effects of melatonin treatment on morphine-induced tolerance in rats. The results may provide insight on and contribute to deciphering the detailed mechanisms of morphine tolerance.

Materials and methods

Construction of intrathecal catheters

The intrathecal (i.t.) catheters were constructed by inserting a 3.5 cm Silastic tube (Corning Incorporated, Corning, NY, USA) into an 8 cm polyethylene tube (0.008 inch internal diameter, 0.014 inch outer diameter; Spectranetics, Colorado Springs, CO, USA) and sealing the joint with epoxy resin and silicon rubber as previously described (15).

Animal preparation and intrathecal drug delivery

The use of rats in the present study adhered to the Guiding Principles in the Care and Use of Animals of the American Physiology Society (16) and was ethically approved by the National Defense Medical Center Animal Care and Use Committee (Taipei, Taiwan). A total of 27 Male Wistar rats (350-400 g), each rat (with 12 weeks of age) was housed individually at a room temperature at 25°C, at 1 atm, with water and food freely as wish. The rats were anaesthetized with phenobarbital (65 mg/kg, intraperitoneally) and two i.t. catheters were implanted. The catheters were inserted via the atlantooccipital membrane down to spinal cord segments L5, L6 and S1-S3, which are associated with the tail-flick reflex (17). One catheter was connected to a mini-osmotic pump (Alzet, Cupertino, CA, USA) for an infusion of saline or morphine (15 µg/h) for 7 days at a rate of 1 µl/h. Subsequent to cath-eterization (day 0), the rats were returned to their home cages and maintained in a 12 h light/dark cycle with ad libitum access to food and water. Rats with neurological deficits were excluded. On day 7, by which time a morphine tolerance had developed, the catheter used for saline or morphine infusion was cut and blocked with a metal metal plug to prevent CSF leakage. The rats were injected i.t. via the second catheter with 5 µl either with vehicle (10% ethanol) or melatonin (50 µg in 10% ethanol), then, 30 min later, a single dose of morphine (15 µg in 5 µl saline, i.t.) was injected and the antinociceptive effect measured. The protocol is presented in Fig. 1A. There were four experimental groups used in the present study, as follows: Controls, melatonin-treated, morphine-treated and those treated with melatonin and morphine combined. For the control group, the animals were infused with saline for 7 days and infused with vehicle injection for 30 min, and subsequently injected with saline. For the morphine group, the animals were infused with morphine for 7 days, injected with vehicle injection for 30 min and subsequently injected with morphine. For the melatonin group, the animals were infused with morphine for 7 days and injected with melatonin for 30 min, and subsequently injected with saline. For the melatonin and morphine group, the animals were infused with morphine for 7 days, injected with melatonin for 30 min and subsequently injected with morphine.

Figure 1

Experimental procedure and effect of melatonin on the antinocieptive effect in morphine-tolerant rats. (A) Experimental procedure for drug administration. Male Wistar rats were implanted with two i.t. catheters, one of which was connected to a mini-osmotic pump for the infusion of morphine or saline for 7 days. On day 7, subsequent to morphine tolerance development, the catheter was cut, and 3 h later the rats were injected intrathecally with either vehicle or melatonin via the second catheter. A total of 30 min later, a single dose of morphine (15 µg) was injected intrathecally and its antinociceptive effect measured. (B) Melatonin reverses the antinociceptive effect of morphine in morphine-tolerant rats. Antinociception of morphine was assessed on day 7 following intrathecal infusion of saline or morphine. At 3 h subsequent to the discontinuation of infusion, the rats were injected intrathecally with 10% ethanol (as vehicle) or 50 µg melatonin. After 30 min, the rats underwent a 15 µg morphine administration, then tail-flick latency was measured every 30 min for 120 min. All data are presented as the mean ± standard error of the mean for at least 5 rats. **P<0.01 vs. the Ma group; ##P<0.01 vs. the MO group. C, control (saline infusion/vehicle injection/saline challenge); MO, morphine (morphine infusion/vehicle injection/morphine challenge); Ma, melatonin (morphine infusion/melatonin injection/saline challenge); MOMa, (morphine infusion/melatonin injection/morphine challenge); i.t., intrathecal; qPCR, quantitative polymerase chain reaction.

The dose of morphine selected was based on a previous study (18). For i.t. injection, melatonin was dissolved in ethanol (50 µg/5 µl in 10% ethanol maximum). All drugs were purchased from Sigma-Aldrich (Merck KGaA, Darmstadt, Germany) and were delivered i.t., followed by the flushing of the catheter with 5 µl saline. Preliminary results revealed no abnormal motor function subsequent to i.t. injection of the test drugs (data not shown).

Antinociception test

Tail-flick latency was measured using the hot water immersion test (52±0.5°C). Baseline latency was ~2±0.38 sec, and a cutoff time of 10 sec was used. Rats were placed in plastic restrainers for drug injection and antinociception testing.

Spinal cord sample collection from rats with different treatments

Spinal cord sample collection was performed as previously described (7), and morphine tolerance in rats was confirmed by the time-course of tail-flick latency over a 7-day period. Prior to day 4, the rats with morphine infusions demonstrated a reduction of tail-flick latency compared with the saline-infused group, which exhibited no changes in latency during the period. Substantial morphine tolerance was developed on day 7 as determined by a significant reduction of the antinociceptive effect of morphine compared with day one, with a reduction of the tail-flick latency of ~60%. And then, rats were i.t. injected with either 10% ethanol (as a vehicle) or melatonin via the externalized i.t. catheter. A total of 30 min later, a single dose of morphine (15 µg) was injected i.t. to confirm morphine tolerance. In contrast, melatonin pretreatment attenuated morphine tolerance, melatonin pretreatment was done by administering melatonin on day 7, at 30 min prior to morphine intrathecal injection. The lumbar enlargement segment was removed from 4 rat spinal cords from each group for differential gene expression analysis.

Spinal cord sample preparation

Following drug treatment, the rats were sacrificed by exsanguination under anaesthesia with isoflurane (Abbott Pharmaceutical Co. Ltd., Lake Bluff, IL, USA) and a laminectomy was performed at the lower edge of the 12th thoracic vertebra. Subsequently, the lumber enlargement (L5-S3) of the spinal cord was immediately collected for subsequent analysis.

Rat expression microarray

Following the tail-flick test, the rats were sacrificed, and lumber enlargement (L5-S3) of the spinal cord was immediately collected. There were 4 samples tested in each group. Total mRNAs were extracted using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA). RNA concentration and purity were assessed using an Agilent 2100 Bioanalyzer (Agilent Technologies, Inc., Santa Clara, CA, USA) with a criteria of OD260/OD280 (>1.8) and OD260/OD230 (>1.6). Next, the RNAs were labelled with Cy5 dye by an indirect NHS ester labelling kit (GE Healthcare, Chicago, IL, USA) according to the manufacturer’s protocol. The labelled RNAs were hybridized with a Rat OneArray® microarray (Phalanx Biotech Group, Hsinchu, Taiwan), which contains 24,358 rat genome probes and 980 experimental control probes. All the probes correspond to annotated genes in RefSeq and Ensembl databases. The hybridization procedure was performed at 50°C in a Phalanx Hybridization System (Phalanx Biotech Group). A total of 16 h after hybridization, non-specific binding targets were washed away using three sequential washing steps by 2X saline-sodium citrate buffer (SSC) contained 0.2% SDS solution for 5 min at 42°C. Then, the slide was spun dry with a centrifuge for 1 min at room temperature. The images of the microarray were scanned using an Agilent G2505C scanner (Agilent Technologies, Inc.). The Cy5 fluorescence intensities of each spot were analysed by GenePix 4.1 software (Molecular Devices, LLC, Sunnyvale, CA, USA).

Microarray analysis

Microarray spot analysis was resolved by the Rosetta Resolver System® (Rosetta Biosoftware, Seattle, WA, USA). Control probes data were calculated, and the reproducibility of each microarray slide was assessed using Pearson’s correlation coefficient calculations with a criterion of R-value >0.975. Normalized spot intensities were transformed to gene expression log2 ratios in each group. For further analysis, the spots with a log2 ratio ≥1 or a log2 ratio ≤−1 or undetectable log2 ratios but with differences in intensity between the two samples of >1,000 and a P<0.05 were selected according to the method of Pirooznia et al (19). Principal Component Analysis (PCA) was performed to evaluate any differences among biological replicates and their treatment conditions using FDA released ArrayTrack™ HCA-PCA Standalone Package (20). PCA uses an orthogonal transformation to convert a set of observations of possibly correlated variables into a set of values of uncorrelated variables called principal components. For advanced data analysis, intensity data were pooled and calculated to identify differentially expressed genes based on the threshold of fold-change and P-value. The correlation of expression profiles between samples and treatment conditions was demonstrated by unsupervised hierarchical clustering analysis. Average linkage clustering was performed to visualize the correlations among the replicates and varying sample conditions using and open source software, Java Treeview (21). Up and downregulated genes are represented in red and green colors, respectively.

Gene ontology (GO) enrichment analysis

The gene IDs of interest were uploaded to the Gene ontology Enrichment analysis website (22). The database and analysis services were funded by the National Human Genome Research Institute in the U.S. And right now the website were maintained and updated by the Gene Ontology Consortium (GOC). The names of the genes with interested were paste to the query column in the website and set the GO aspect as molecular function for the analysis. The database search was confined to Rattus norvegicus database.

Gene pathway mapping by PANTHER

The gene IDs of interest were uploaded to the PANTHER Classification System website (http://pantherdb.org). PANTHER is a comprehensive, curated database of protein families, trees, subfamilies, functions and ontology (23). The search parameter was set to ‘molecular function’, and the database search was confined to only the Rattus norvegicus database. The keywords used were the gene names of interest and the access date were December 12 and 19, 2017.

RNA extraction and reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Tissues were collected as described above. RNA was extracted within 1 h at room temperature using TRIzol reagent following manufacturer’s protocol (Invitrogen; Thermo Fisher Scientific, Inc.). mRNA were reverse transcribed to cDNA using the SuperScript III First-Strand Synthesis System (Invitrogen; Thermo Fisher Scientific, Inc.). cDNA were amplified and subjected to optical analysis to verify the integrity of extracted RNA. The expression of target genes were quantified for all experimental groups using LightCycler system (Roche Diagnostics, Basel, Switzerland). RT-qPCR analysis thermocycling conditions were: 95°C for 10 min and then the cycling conditions were set as 95°C for 10 sec, 60°C for 20 sec, 72°C for 40 sec for 50 cycles. The method of quantification for RT-qPCR products were followed Livak and Schmittgen et al (24) The relative abundance of transcripts were normalized to the constitutive expression of GAPDH. The primers of each genes used in RT-qPCR were listed in Table I.

Table I

Primers used for reverse transcription-quantitative polymerase chain reaction analysis.

Table I

Primers used for reverse transcription-quantitative polymerase chain reaction analysis.

Target geneGene IDPositionSequence
G protein subunit β 1 (Gnb1)NM_030987.2F:262-281 5′-tccagtgggaagaatccaaa-3′
R:317-337 5′-ccagtgcatggcataaatctt-3′
Cholecystokinin B receptor (Cckbr)NM_013165.2F:1799-1819 5′-cccgtttgacttcattattgc-3′
R:1842-1861 5′-tgaaaggcgtgtggttgata-3′
5-hydroxytryptamine receptor 1A (Htr1a)NM_012585.1F:1054-1072 5′-ggcaccttcatcctctgct-3′
R:1110-1128 5′-gtggcagctgctttcacag-3′
RAS protein activator like 1 (Rasal1)NM_001108335.1F:408-429 5′-ggagtacactgttcaccttcca-3′′
R:451-470 5′-tcctcatccagcacgtagaa-3′
General transcription factor 2A subunit 1 like (Gtf2a1l)NM_001012136.1F:1222-1242 5′-gaggatcccctaaattctgga-3′
R:1267-1289 5′-ttatctgtgtcaaacaggtctgg-3′
Period circadian clock 1 (Per1)NM_001034125.1F:1986-2008 5′-tcctaacacaaccaagcgtaaat-3′
R:2043-2062 5′-ccctctgcttgtcatcatca-3′
Methionine adenosyltransferase 2A (Mat2a)NM_134351.1F:149-168 5′-tgtaggggaaggtcatccag-3′
R:204-222 5′-cctgctgaaggtgtgcatc-3′
Collagen type V α 3 chain (Col5a3)NM_021760.1F:634-652 5′-cggggaggagtcttttgag-3′
R:673-693 5′-gcctgagggtctggaattaac-3′
Inositol 1,4,5-trisphosphate receptor, type 3 (Itpr3)NM_013138.1F:8362-8381 5′-taggggatgcaagttctcca-3′
R:8403-8422 5′-ccactgagaaatgccagtca-3′
Diacylglycerol kinase ζ (Dgkz)NM_031143.1F:330-347 5′-ctttgggcacaggaaagc-3′
R:410-429 5′-gatctgccgctcagattcac-3′
LIM zinc finger domain containing 2 (Lims2)NM_001012163.1F:966-985 5′-tcatgtgattgagggtgacg-3′
R:1032-1051 5′ccaccaggagaacagactgg-3′

[i] F, forward; R, reverse.

Data and statistical analyses

All data are presented as the mean ± standard error of the mean. Statistical analysis was performed using SigmaStat 3.0 software (SYSTAT Software Inc., San Jose, CA, USA). Tail-flick latencies were analyzed using two-way (time and treatment) analysis of variance (ANOVA), followed by one-way ANOVA with a post hoc Student-Newman-Keuls test. The RT-qPCR results were analyzed using a Student’s t-test. P<0.05 was considered to indicate a statistically significant difference.

Results

Experimental design and procedure

The experimental procedure for drug administration was depicted in Fig. 1. Male Wistar rats were implanted with two i.t. catheters and connected to a mini-osmotic pump for morphine or saline infusion for 7 days for morphine tolerance induction. On day 7, subsequent to the development of tolerance, the catheter was cut, and 3 h later, the rats received an i.t. injection of either vehicle or melatonin via the other catheter. A total of 30 min later, at the tolerance expression phase, a single dose of morphine (15 µg) was injected i.t., and the antinociceptive effect was measured. Tail flick tests were performed in every experimental group and the results were presented in Fig. 1B. There was a significant reduction of morphine tolerance subsequent to melatonin addition compared with the control group (P<0.01). Following a tail-flick test, the rats were sacrificed, and the L5 to S3 region of the spinal cords were collected for further analysis.

Differential gene expression among morphine tolerance, melatonin treatment and morphine tolerance combined with melatonin treatment groups

To determine the alterations in gene expression caused by morphine and reversed by melatonin treatment in rat spinal cords, rat global gene expression profiles of four independent RNA samples from each group were selected for microarray analysis. PCA and clustering analysis revealed that the overall gene profiles derived from microarray analysis were separated based on morphine or melatonin treatment. The result revealed that 162 genes were upregulated and 16 genes were downregulated in the morphine-tolerant group (MO group, n=7) compared with the control group (C group, n=5); 476 genes were upregulated and 71 genes were downregulated in the melatonin treatment group (Ma group, n=5) compared with the control group (C group), and 290 genes were upregulated and 15 genes were downregulated in the morphine with acute melatonin treatment group (MOMa group, n=10) compared with the control group (Table II). All genes selected using the criteria of log2|Fold change|≥ 1 and P<0.05 or undetectable log2 ratios but with differences in intensity between the two samples of >1,000. Statistical significance was used to avoid confounding due to variation amongst the animals and addressed additional evidence that the transcriptional profiles of morphine tolerance and melatonin treatment in vivo are different. The present study compared the number of upregulated genes between the MO and MOMa groups; it was identified that the number of upregulated genes in the MOMa group was greater compared with the number in the MO group, which indicated that melatonin restored the antinociceptive effect of morphine, which was accomplished with multiple gene expression alterations.

Table II

Number of differentially expressed genes.

Table II

Number of differentially expressed genes.

Group comparisonUpregulatedDownregulated
MO/C16216
MOMa/C29015
Ma/C47671

[i] Standard selection criteria to identify differentially expressed genes are as follows: i) log2 |Fold change|≥1 and P<0.05; ii) log2 ratios=‘NA’ and the differences of intensity between the two samples ≥1000. Detail gene lists were be provided by request. C, control (saline infusion/vehicle injection/saline challenge); Mo, morphine (morphine infusion/vehicle injection/morphine challenge); Ma, melatonin (morphine infusion/melatonin injection/saline challenge); MOMa, (morphine infusion/melatonin injection/morphine challenge).

Gene ontology (GO) analysis of the altered genes from the MO or MOMa group

The differentially expressed genes were then subjected to GO analysis based on molecular function (Table III). Numerous GO terms were identical between the two groups; however, opsonin binding, actin binding, calcium ion binding, sugar binding, oxidase activity, deaminase activity, protein complex binding and oxidoreductase activity were not identified in the MO group. On the other hand, GTPase activity, phospholipase inhibitor activity, cytokine activity, GTP binding, guanyl-nucleotide and ribonucleotide binding, immunoglobulin (Ig)G receptor activity, IgE binding and protein dimerization activity were not identified in the MOMa group, implying the potential regulatory mechanism of melatonin treatment. From the GO terms identified between the MO and MOMa groups, it was revealed that a number of notable pathways were altered. It has been previously reported that the morphine tolerance process involves inflammation (25). Immune-associated processes, including cytokine activity, IgG receptor activity and IgE binding, were missing following melatonin treatment, which indicates that melatonin treatment may participate in the downregulation of these cellular process. On the other hand, the gene expression for actin binding was present following melatonin treatment; this result implies that cytoskeleton reconstitution may be activated. Additionally, genes involving calcium ion binding, sugar binding, NADPH oxidase activity, deaminase activity and protein complex binding pathways appeared subsequent to melatonin treatment, indicating the requirement for the metabolic activity that emerged following melatonin treatment. The gene expression data for IgG receptor expression and actin binding were selected and provided by request.

Table III

Identified gene ontology terms of the MO and MOMa groups compared with the C group.

Table III

Identified gene ontology terms of the MO and MOMa groups compared with the C group.

Geneset nameMO/C
MOMa/C
No. of genes in genesetNo. of genes in overlapP-valueNo. of genes in genesetNo. of genes in overlapP-value
GO:0001846-opsonin bindingN.I.73<0.01
GO:0001871-pattern binding1167<0.0111613<0.01
GO:0001872-zymosan binding320.02320.04
GO:0003924-GTPase activity9840.05N.I.
GO:0003779-actin bindingN.I.233100.01
GO:0004857-enzyme inhibitor activity23810<0.0123811<0.01
GO:0004859-phospholipase inhibitor activity620.05N.I.
GO:0004866-endopeptidase inhibitor activity14860.0114870.02
GO:0005125-cytokine activity11050.01N.I.
GO:0005506-iron ion binding28970.03289100.03
GO:0005525-GTP binding31280.02N.I.
GO:0005509-calcium ion bindingN.I.672200.01
GO:0005529-sugar bindingN.I.21590.02
GO:0005539-glycosaminoglycan binding10250.0110211<0.01
GO:0008009-chemokine activity325<0.013240.01
GO:0008201-heparin binding7240.02728<0.01
GO:0016175-superoxide-generatingN.I.73<0.01
NADPH oxidase activity
GO:0016814~hydrolase activity, acting on carbon-nitrogen (but not peptide) bonds, in cyclic amidines2230.01224<0.01
GO:0019239-deaminase activityN.I.2130.04
GO:0019001-guanyl nucleotide binding32680.02N.I.
GO:0019763-immunoglobulin receptor activity73<0.0173<0.01
GO:0019770-IgG receptor activity420.03N.I.
GO:0019834-phospholipase A2 inhibitor activity320.02320.04
GO:0019863-IgE binding420.03N.I.
GO:0019864-IgG binding64<0.0164<0.01
GO:0019865-immunoglobulin binding125<0.01125<0.01
GO:0019955-cytokine binding8740.04877<0.01
GO:0020037-heme binding14850.0414870.02
GO:0030246-carbohydrate binding33710<0.0133720<0.01
GO:0030247-polysaccharide binding1167<0.0111613<0.01
GO:0030414-peptidase inhibitor activity1597<0.0115980.01
GO:0032403-protein complex bindingN.I.22212<0.01
GO:0032561-guanyl ribonucleotide binding32680.02N.I.
GO:0042379-chemokine receptor binding335<0.013340.01
GO:0042802-identical protein binding588110.02588180.01
GO:0042803-protein homodimerization activity31880.0231815<0.01
GO:0046906-tetrapyrrole binding15450.0415470.03
GO:0046983-protein dimerization activity528100.0352820<0.01
GO:0048020-CCR chemokine receptor binding320.02N.I.
GO:0050664-oxidoreductase activity, acting on NADH or NADPH, with oxygen as acceptorN.I.1130.01

[i] N.I., not identified; Ig, immunoglobulin; GO, Gene Ontology; C, control (saline infusion/vehicle injection/saline challenge); Mo, morphine (morphine infusion/vehicle injection/morphine challenge); Ma, melatonin (morphine infusion/melatonin injection/saline challenge); MOMa, (morphine infusion/melatonin injection/morphine challenge).

Venn diagram and genes exclusively expressed in the MOMa group

In order to clarify the differential gene expression panels among the three groups, Venn diagram analysis was performed, and the results depicted the overlap of differentially expressed genes between the MO, Ma and MOMa groups (Fig. 2). In total, 48 genes were upregulated and 8 were genes downregulated exclusively in the MOMa group. These genes were the candidates that participated in the reversal of morphine tolerance. It was also identified that 20 genes were upregulated and 13 genes were downregulated exclusively in the MO group; these genes were not altered by melatonin treatment, so these genes were not involved in the melatonin reversal effect in morphine tolerance. All the genes in Venn diagram analysis are listed in Table IV-A and -B. Genes expressed exclusively in the MOMa group are notable as they may be the targets for the reversal of morphine tolerance associated with melatonin in future studies. From Table IV-B, the myocilin gene demonstrated the greatest fold change in upregulation; and myocilin has been reported to mediate myelination in the peripheral nervous system (26). Furthermore, microtubule-associated protein 9 expression was decreased in the MOMa group, and this gene has been reported to serve a role in mitotic spindle formation and mitosis progression (27), implying the potential involvement of melatonin.

Table IV

Top 20 exclusively upregulated and downregulated genes in each group.

Table IV

Top 20 exclusively upregulated and downregulated genes in each group.

A, Exclusively expressed genes in MO group
Gene symbolDescriptionGene IDFold-change
Ddx60DEAD (Asp-Glu-Ala-Asp) box polypeptide 60, probable ATP-dependent RNA helicase DDX60-like1003608011.61 Up
Lgals3bpLectin, galactoside-binding, soluble, 3 binding protein2459551.54 Up
Oas1a2′-5′ oligoadenylate synthetase 1A1922811.47 Up
Isg15ISG15 ubiquitin-like modifier2986931.46 Up
Slamf9SLAM family member 92892351.29 Up
Usp18Ubiquitin specific peptidase 183126881.28 Up
Smim5Small integral membrane protein 56899261.26 Up
Casp4Caspase 4, apoptosis-related cysteine peptidase1145551.2 Up
Cd33CD33 molecule6904921.2 Up
Pik3ap1 Phosphoinositide-3-kinase adaptor protein 12940481.14 Up
Ccl7Chemokine (C-C motif) ligand 72875611.13 Up
Apol9aApolipoprotein L 9a5031641.11 Up
DptDermatopontin2891781.09 Up
CryaaCrystallin, αA242731.06 Up
IrgmImmunity-related GTPase family, M3030901.05 Up
Irf7Interferon regulatory factor 72936241.04 Up
Gpr160G protein-coupled receptor 1604995881.03 Up
Uba7Ubiquitin-like modifier activating enzyme 73010001.03 Up
Vwa5b1Von Willebrand factor A domain containing 5B13136531.03 Up
Olr104Olfactory receptor 1042932431.02 Up
LOC689064β-globin689064−1 Down
Fras1Fraser syndrome 1289486−1.01 Down
Zfp597Zinc finger protein 597266774−1.06 Down
LOC681849Similar to protein C6orf142 homolog681849−1.09 Down
Alas2Aminolevulinate, delta-, synthase 225748−1.13 Down
LOC500300Similar to hypothetical protein MGC6835500300−1.2 Down
Hspa1bHeat shock 70 kD protein 1B (mapped)294254−1.21 Down
Ccdc77Coiled-coil domain containing 77312677−1.3 Down
Oas1e2′-5′ oligoadenylate synthetase 1E494201−1.4 Down
Pmp2Peripheral myelin protein 2688790−1.47 Down
Fkbp6FK506 binding protein 6288597−1.98 Down
PrxPeriaxin78960−2.16 Down
MpzMyelin protein zero24564−2.92 Down
B, Exclusively expressed genes in MOMa group
Gene symbolDescriptionGene IDFold-change
MyocMyocilin815232.47 Up
Samsn1SAM domain, SH3 domain and nuclear localization signals, 11706371.48 Up
ScinScinderin2989751.45 Up
NcanNeurocan589821.35 Up
TaglnTransgelin251231.34 Up
AplnrApelin receptor835181.31 Up
Nlrc4NLR family, CARD domain containing 42987841.28 Up
S1pr3 Sphingosine-1-phosphate receptor 33067921.27 Up
Mxra8Matrix-remodelling associated 83137701.26 Up
Sptbn5Spectrin, β, non-erythrocytic 52960901.24 Up
Plin2Perilipin 22981991.23 Up
EpycEpiphycan3147721.23 Up
ChdhCholine dehydrogenase2905511.22 Up
HlxH2.0-like homeobox3640691.19 Up
CenpfCentromere protein F2576491.19 Up
AoahAcyloxyacyl hydrolase (neutrophil)4987571.17 Up
Spta1Spectrin, α, erythrocytic 1 (elliptocytosis 2)2892571.15 Up
Trim47Tripartite motif-containing 476903741.14 Up
Abi3ABI family, member 33034761.13 Up
Ssc5dScavenger receptor cysteine rich domain containing (5 domains)3083411.13 Up
Epm2aip1EPM2A (laforin) interacting protein 1316021−1.02 Down
LOC691921Hypothetical protein LOC691921691921−1.04 Down
Klhl11Kelch-like 11 (Drosophila)287706−1.09 Down
Ppargc1bPeroxisome proliferator-activated receptor γ, coactivator 1 β291567−1.11 Down
Pcdhb6Protocadherin β6291653−1.14 Down
Tox2TOX high mobility group box family member 2311615−1.22 Down
Map9 Microtubule-associated protein 9310544−1.26 Down
RGD1309108Similar to hypothetical protein FLJ23554315578−1.55 Down
C, Exclusively expressed genes in Ma group
Gene symbolDescriptionGene IDFold-change
Defb3β-defensin 36416233.52 Up
RT1-DaRT1 class II, locus Da2942692.60 Up
RT1-BaRT1 class II, locus Ba3096212.59 Up
Pxmp4Peroxisomal membrane protein 42826342.46 Up
RT1-BbRT1 class II, locus Bb3096222.21 Up
Ccl11Chemokine (C-C motif) ligand 11293972.08 Up
Tmem252Transmembrane protein 2523617442.07 Up
AurkbAurora kinase B1145921.99 Up
Cd74Cd74 molecule, major histocompatibility complex, class II invariant chain255991.89 Up
Birc5Baculoviral IAP repeat-containing 5640411.81 Up
Lmcd1LIM and cysteine-rich domains 14940211.80 Up
Fam111aFamily with sequence similarity 111, member A4993221.79 Up
RSA-14-44RSA-14-44 protein2971731.77 Up
Kif11Kinesin family member 111713041.70 Up
Vdac1Voltage-dependent anion channel 1835291.70 Up
Hmgn3High mobility group nucleosomal binding domain 31139901.69 Up
NalcnSodium leak channel, non-selective2667601.69 Up
Tnfrsf14Tumor necrosis factor receptor superfamily, member 143665181.67 Up
Pex11aPeroxisomal biogenesis factor 11 α852491.61 Up
RGD1564664Similar to LOC387763 protein4998391.61 Up
LOC100911604CD99 antigen-like protein 2-like, similar to MIC2L1500410−1.14 Down
Serpinb1bSerine (or cysteine) peptidase inhibitor, clade B member 1b, leukocyte elastase inhibitor A-like306891−1.15 Down
MgllMonoglyceride lipase29254−1.15 Down
LrtomtLeucine rich transmembrane and 0-methyltransferase domain containing308868−1.16 Down
Tas2r145Taste receptor, type 2, member 145100363053−1.18 Down
Kcnip3Kv channel interacting protein 3, calsenilin65199−1.19 Down
Fbll1Fibrillarin-like 1363563−1.19 Down
LOC100910054NF-κ-B-repressing factor-like100910054−1.20 Down
Ttll1Tubulin tyrosine ligase-like family, member 1362969−1.21 Down
Negr1Neuronal growth regulator 159318−1.22 Down
Ttll11Tubulin tyrosine ligase-like family, member 11689746−1.24 Down
Mgam Maltase-glucoamylase312272−1.25 Down
Apba1Amyloid β(A4) precursor protein-binding, family A, member 183589−1.26 Down
Hoxb5Homeo box B5497987−1.26 Down
Zfp238Zinc finger protein 23864619−1.27 Down
Ddx6DEAD (Asp-Glu-Ala-Asp) box helicase 6500988−1.29 Down
LOC310902Similar to Alcohol dehydrogenase 1A (alcohol dehydrogenase α subunit)310902−1.30 Down
Fgf13Fibroblast growth factor 1384488−1.32 Down
Tnnc2Troponin C type 2 (fast)296369−1.33 Down
PdynProdynorphin29190−1.43 Down

[i] log2 (Ratio) Mo, morphine (morphine infusion/vehicle injection/morphine challenge); Up, upregulated; Down, downregulated.

Reversed gene expression panel between MO/C and MOMa/C groups

In order to clarify which genes were altered by melatonin treatment in morphine-tolerant rats, genes from the microarray data with inverted gene expression profiles between MO/C and MOMa/C groups were selected. Hierarchical clustering analysis was performed to construct a reversed gene expression heatmap between the MO/C and MOMa/C groups (Fig. 3). Genes were selected from the microarray data with the criteria of log2 ratio ≥1 or ≤−1 and P<0.05, and the expression of the genes was reversed between the MO/C and MOMa/C groups. The constructed panel according to the heatmap of the reversed genes was listed in Table V. The panel with inverted gene expression may be used to investigate potential pathways derived by melatonin treatment.

Table V

Genes with inverted expressions between Mo and MOMa group

Table V

Genes with inverted expressions between Mo and MOMa group

A, Genes with upregulated expression in the MO group but downregulated expression in the MOMa group
Gene symbolDescriptionGene IDFold MOFold MOMA
Glra1Glycine receptor, α1256742.100.65
Olr1595Olfactory receptor 15953049901.700.66
Ndufaf1NADH dehydrogenase (ubiquinone) complex I, assembly factor 12960861.420.72
RGD1563091Similar to OEF25000111.400.79
Gnb1Guanine nucleotide binding protein (G protein), βpolypeptide 1244001.400.76
Ccdc122Coiled-coil domain containing 1221003607521.390.61
CstaCystatin A (stefin A)2880671.350.70
BlvraBiliverdin reductase A1165991.330.70
MGC114464Similar to expressed sequence AI8360035009251.320.75
LOC100910655 Paralemmin-2-like1009106551.310.64
Dync1i1Dynein cytoplasmic 1 intermediate chain 1295641.310.79
LOC685007Similar to unc-93 homolog A6850071.300.68
CckbrCholecystokinin B receptor257061.290.78
Art4 ADP-ribosyltransferase 43128061.290.72
Zbtb41Cytochrome C oxidase assembly factor 55032521.280.75
Cnga4Cyclic nucleotide gated channel α4852581.280.78
Thg1ltRNA-histidine guanylyltransferase 1-like (S. cerevisiae)3030671.260.82
Mettl11bMethyltransferase like 11B2891671.250.68
LOC100911235Mediator of RNA polymerase II transcription subunit 7-like1009112351.250.81
Htr1a5-hydroxytryptamine (serotonin) receptor 1A, G protein-coupled244731.240.75
Pou3f2POU class 3 homeobox 2295881.240.77
B, Genes with downregulated expression in the MO group but upregulated expression in the MOMa group
Gene symbolDescriptionGene IDFold MOFold MOMA
Itpr3Inositol 1,4,5-trisphosphate receptor, type 3256790.591.82
Mocs1Molybdenum cofactor synthesis 13012210.691.66
Col5a3Collagen, type V, α3603790.731.66
Mrc1Mannose receptor, C type 12913270.661.65
Lims2LIM and senescent cell antigen like domains 23613030.651.63
Gtf2a1lGeneral transcription factor IIA, 1-like3167110.761.58
Olr1147Olfactory receptor 11473004080.601.55
Ccnt2Cyclin T23047580.821.51
Map3k6Mitogen-activated protein kinase kinase kinase 63130220.641.45
Tnfrsf25Tumor necrosis factor receptor superfamily, member 255005920.711.43
Slc41a1Solute carrier family 41, member 13639850.831.43
Mvb12bMultivesicular body subunit 12B3621180.801.42
AcacaAcetyl-CoA carboxylase α605810.771.38
Ryr3Ryanodine receptor 31705460.771.38
Col27a1Collagen, type XXVII, α12981010.831.36
Rasal1RAS protein activator like 1 (GAP1 like)3608140.721.36
Rasal1RAS protein activator like 1 (GAP1 like)3608140.721.36
Mat2aMethionine adenosyltransferase II6905100.791.29
er1Period circadian clock 12874220.631.25
DgkzDiacylglycerol kinase ζ818210.771.24

[i] Mo, morphine (morphine infusion/vehicle injection/morphine challenge); MOMa, (morphine infusion/melatonin injection/morphine challenge).

PANTHER pathway mapping and RT-qPCR analysis

The genes listed in Table V-A and -B were used for enrichment analysis by the PANTHER algorithm provided by the GO Consortium. The PANTHER pathway mapped 4 out of 29 genes for the genes listed in Table V-A and 10 out of 66 genes for the genes listed in Table V-B. The PANTHER-mapped pathways and associated genes are listed in Table VI-A and -B. Guanine nucleotide binding protein β polypeptide 1 (Gnb1) was identified to participate in numerous cellular functions, including neuron-associated functions, including glutamatergic, cholinergic, GABAergic, dopaminergic, serotonergic and sympathetic neuron functions. Furthermore, Gnb1 has also been reported to participate in other pathways, including histamine H1 and H2 receptors and several hormone receptor signals. Gnb1 was upregulated 1.4-fold in the MO group and downregulated 1.3-fold in the MOMA group; this result implies the potential of a melatonin-mediated pathway via the repression of Gβ expression and signaling. On the other hand, the genes mapped in Table VI-B mainly participated in cell proliferation and migration in addition to cytoskeleton reconstruction. For example, a gene named inositol 1,4,5-trisphosphate receptor, type 3 (Itpr3) was downregulated 1.7-fold in the MO group but was upregulated 1.8-fold in the MOMA group. A number of pathways associated with Itpr3, including inflammatory, cell proliferation and migration, G protein mediated and vaso-relaxation pathways, were suggested. The genes mentioned in Table VI-A and VI-B were selected and their gene expressions were verified using RT-qPCR (Fig. 4). All the genes selected here demonstrated an accordance with the expression trends of the microarray results. The results indicate the potential signaling pathways of melatonin in the rat spinal cord for the restoration of cell proliferation and migration through cytoskeleton reconstruction.

Table VI

PANTHER pathway mapped cellular functions of four selected genes from Table V-A and -B.

Table VI

PANTHER pathway mapped cellular functions of four selected genes from Table V-A and -B.

A, PANTHER pathway mapped cellular functions of four selected genes from Table V-A
Gene nameCellular functionsPathways
Guanine nucleotide binding protein, β polypeptide 1 (Gnb1)Neuron
Pain_Relief_anagelsiaOpioid proopiomelanocortin pathway
Pain_Relief_anagelsiaOpioid proenkephalin pathway
Pain_Relief_anagelsiaEnkephalin release
Pain_Relief_anagelsiaCortocotropin releasing factor receptor signaling pathway
Pain_Relief_anagelsiaOpioid prodynorphin pathway
GlutamertergicMetabotropic glutamate receptor group II pathway
GlutamertergicHeterotrimeric G-protein signaling pathway-rod outer segment phototransduction
GlutamertergicMetabotropic glutamate receptor group III pathway
CholinergicMuscarinic acetylcholine receptor 1 and 3 signaling pathway
Cholinergicβ1 adrenergic receptor signaling pathway
CholinergicMuscarinic acetylcholine receptor 2 and 4 signaling pathway
GABAergic GABA-B_receptor_II_signaling
GABAergic Endogenous_cannabinoid_signaling
DopaminergicDopamine receptor mediated signaling pathway
Serotonergic5HT2 type receptor mediated signaling pathway
Serotonergic5HT4 type receptor mediated signaling pathway
DepolarizationNicotine pharmacodynamics pathway
Sympatheticβ3 adrenergic receptor signaling pathway
G protein mediated pathwayβ2 adrenergic receptor signaling pathway
Inflammation
G protein mediated pathwayHistamine H1 receptor mediated signaling pathway
G protein mediated pathwayHistamine H2 receptor mediated signaling pathway
G protein mediated pathwayHeterotrimeric G-protein signaling pathway-Gq a and Go α mediated pathway
G protein mediated pathway Thyrotropin-releasing hormone receptor signaling pathway
Others
Signaling pathwayGonadotropin releasing hormone receptor pathway
Signaling pathwayPI3 kinase pathway
Signaling pathwayWnt signaling pathway
AngiogenesisAngiotensin II-stimulated signaling through G proteins and β-arrestin
Muscle contractionOxytocin receptor mediated signaling pathway
Cell migrationCCKR signaling pathway
Cholecystokinin B receptor (Cckbr)Cell migrationCCKR signaling pathway
5-hydroxytryptamine (serotonin) receptor 1A Neuron_Serotonergic5HT1 type receptor mediated signaling pathway
G protein-coupled (Htr1a)G protein mediated pathwayHeterotrimeric G-protein signaling pathway-Gi α and Gs α mediated pathway
Dynein cytoplasmic 1 intermediate chain 1 (Dync1i1)NeuronHuntington disease
B, PANTHER pathway mapped cellular functions of four selected genes from Table V-B
Gene nameCellular functionsPathways
Inositol 1,4,5-trisphosphate receptor type 3 (Itpr3)Inflammation and immunityInflammation mediated by chemokine and cytokine signaling pathway
Histamine H1 receptor mediated signaling pathwayB cell activation
Cell proliferation and migrationGonadotropin releasing hormone receptor pathway
Wnt signaling pathway
Muscarinic acetylcholine receptor 1 and 3 signaling pathway
PDGF signaling pathway
G protein mediated pathwayAngiotensin II-stimulated signaling through
G proteins and β-arrestin
Heterotrimeric G-protein signaling pathway-Gq α and Go α mediated pathway
Vaso relaxationEndothelin signaling pathway
RAS protein activator like 1 (Rasal1)Cell proliferation and migrationFGF signaling pathway
EGF receptor signaling pathway
PDGF signaling pathway
General transcription factor subunit 1-like (Gtf2a1l)Transcription regulationTranscription regulation by bZIP transcription 2A
General transcription regulation
Period circadian proteinBiochemical oscillatorCircadian clock system
homolog 1 (Per1)Cell proliferation and migrationGonadotropin releasing hormone receptor pathway
S-adenosylmethionine synthase isoform type-2 (Mat2a)Enzyme activityS adenosyl methionine biosynthesis
Diacylglycerol kinase ζ (Dgkz)Cell proliferation and migrationGonadotropin releasing hormone receptor pathway
α4 type V collagen (Col5a3)CytoskeletonIntegrin signaling pathway
Collagen α-1(XXVII) chain (Col27a1)CytoskeletonIntegrin signaling pathway
LIM and senescent cell antigen-like-containing domain protein (Lims2)CytoskeletonIntegrin signaling pathway
Mitogen-activated protein kinase kinase kinase 6 (Map3k6)Cell proliferation and migrationFGF signaling pathway

Discussion

By using microarray analysis of rat spinal cords from different treatment groups, the gene expression alterations in different conditions were identified. Panels of gene expression with upregulation in the MO group but downregulation in the MOMa group and also inverted gene expression profiles between the MO/C and MOMa/C groups were constructed. Among them, a number of notable genes were identified following PANTHER pathway mapping. For example, Gnb1 is one of the three subunits of heterotrimeric guanine nucleotide binding proteins (G proteins), which integrate signals between G protein coupled receptors (GPCR) (28). GPCR signaling is initiated when a ligand-bound receptor activates heterotrimeric G proteins on the inner leaflet of the plasma membrane by catalyzing the exchange of GDP for GTP on G protein α subunit (Gα), causing it to release the Gβγ subunits. The GTP-bound Gα and free Gβγ subunits transmit the signal by engaging intracellular effector molecules until GTP is hydrolysed and the β subunits are recoupled to the α subunit to terminate signal transduction receptors and effectors (29). According to a study by Klein et al (30), Gnb1 belongs to a group of genes that is night/day differentially expressed in the pineal body; this result implies the potential involvement of Gnb1 in melatonin treatment. Furthermore, β-arrestin-2 mediated desensitization of the µ-opioid receptor is involved in morphine tolerance (31,32). In the present data, Gnb1 was upregulated in the MO group but downregulated by melatonin treatment; this result indicates the potential of a role of melatonin in the activation and desensitization of GPCR.

Another gene, Itpr3, was downregulated in the MO group but upregulated in the MOMA group. It was also produced following PANTHER pathway mapping. Itpr3 is the receptor for inositol 1,4,5-trisphosphate (IP3), which mediates the release of intracellular calcium (33). Following IP3 binding, Itpr3 permits calcium flow out of the endoplasmic reticulum (34) and results in the activation of transient receptor potential cation channel subfamily M member 5, which results in membrane depolarization (35). In the case of melatonin treatment with Itpr3 upregulation, it was speculated that depolarization in certain nerve cells in the spinal cord may participate in the melatonin-derived attenuation of antinociceptive morphine tolerance.

In the present model, long-term morphine administration did not affect opioid receptor expression potentially due to the alteration of signal transduction and receptor-G protein coupling. It has been demonstrated that the downregulation of opioid receptors following chronic agonist exposure induces tolerance (36,37). However, a controversial report did not observe the downregulation of opioid receptors in tolerant animals (38). On the other hand, studies suggest that β-arrestin-2 (Arrb2) binding causes OPR desensitization, and OPR endocytosis and recycling are required for receptor resensitization. This result suggests the potential involvement of Arrb2 with morphine tolerance (31). However, in the present data, the expression of Arrb2 between the groups was similar; the results did not support the potential involvement of Arrb2 with morphine tolerance. Combining the previous discussion with the present data, it was postulated that the expression changes of opioid receptor and Arrb2 are not mandatory for morphine tolerance mechanisms.

There are limitations in descriptive microarray studies. The first limitation is that sequences in the microarray will be refined in newer databases and will result in different outcomes. Secondly, the associations between mRNAs may be different between mice and humans (39). The third limitation is that the gene expression detected by microarrays is descriptive and may not reflect protein expression and subsequent post-transcriptional modifications (40). However, identifying changes in gene expression in tissues with a high-throughput approach remains a good option as it can be performed in one experiment. Even though the present study uses descriptive microarray analysis, a panel of genes that are specifically expressed in morphine-tolerant animals with or without melatonin treatment was produced. As it is impossible to collect the spinal cord from patients, therefore future studies will use drug databases to identify drugs which target the genes of interest in the present study and use the drugs in the same rat models as in the present study to assess the dosage and efficacy of the drug in the treatment of relieving the morphine tolerance. Following that, the drugs with efficacy in the rat model will be used in humans. Next, patients who are under chronic treatment and with morphine tolerance may be recruited to assess the efficacy of the drug in order to ascertain the results of the present and provide novel treatment methods. From the present microarray analysis, novel insight into the molecular profiles associated with morphine tolerance and the effects of melatonin was provided. The present study offers a foundation for future specific hypotheses testing on potential therapeutic targets derived from melatonin treatment in patients with long-term morphine exposure.

Funding

The present study was supported by research grants from Ministry of Science and Technology (grant no. 105-231 4-B-281-003-MY2) and Cathay General Hospital (grant no. CGH-MR-A10518) in Taiwan.

Authors’ contributions

YCC and RYT wrote the manuscript, were responsible for conception and design, data analysis and interpretation. YTS performed the microarray data analysis and interpretation. IJC was in charge of the animal experiments. TYT and YYM performed the RT-qPCR analysis and interpretation. CSW was responsible for conception and design, financial support, administrative support, final approval of manuscript.

Ethics approval and consent to participate

The use of rats in this study adhered to the Guiding Principles in the Care and Use of Animals of the American Physiology Society and was ethically approved by the National Defense Medical Center Animal Care and Use Committee (Taipei, Taiwan).

Patient consent for publication

Not applicable.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

The authors would like to thank Dr. Chih-Cheng Chien for his advice about the research.

References

1 

Chen WW, Zhang X and Huang WJ: Pain control by melatonin: Physiological and pharmacological effects. Exp Ther Med. 12:1963–1968. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Pasternak GW: When it comes to opiates, just say NO. J Clin Invest. 117:3185–3187. 2007. View Article : Google Scholar : PubMed/NCBI

3 

Günther T, Dasgupta P, Mann A, Miess E, Kliewer A, Fritzwanker S, Steinborn R and Schulz S: Targeting multiple opioid receptors-improved analgesics with reduced side effects. Br J Pharmacol. 175:2857–2868. 2018. View Article : Google Scholar

4 

Martini L and Whistler JL: The role of mu opioid receptor desensitization and endocytosis in morphine tolerance and dependence. Curr Opin Neurobiol. 17:556–564. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Rosenblum A, Marsch LA, Joseph H and Portenoy RK: Opioids and the treatment of chronic pain: Controversies, current status, and future directions. Exp Clin Psychopharmacol. 16:405–416. 2008. View Article : Google Scholar : PubMed/NCBI

6 

Pang CS, Tsang SF and Yang JC: Effects of melatonin, morphine and diazepam on formalin-induced nociception in mice. Life Sci. 68:943–951. 2001. View Article : Google Scholar : PubMed/NCBI

7 

Lin SH, Huang YN, Kao JH, Tien LT, Tsai RY and Wong CS: Melatonin reverses morphine tolerance by inhibiting microglia activation and HSP27 expression. Life Sci. 152:38–43. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Song L, Wu C and Zuo Y: Melatonin prevents morphine-induced hyperalgesia and tolerance in rats: Role of protein kinase C and N-methyl-D-aspartate receptors. BMC Anesthesiol. 15:122015. View Article : Google Scholar : PubMed/NCBI

9 

Xin W, Chun W, Ling L and Wei W: Role of melatonin in the prevention of morphine-induced hyperalgesia and spinal glial activation in rats: Protein kinase C pathway involved. Int J Neurosci. 122:154–163. 2012. View Article : Google Scholar

10 

Feng YM, Jia YF, Su LY, Wang D, Lv L, Xu L and Yao YG: Decreased mitochondrial DNA copy number in the hippocampus and peripheral blood during opiate addiction is mediated by autophagy and can be salvaged by melatonin. Autophagy. 9:1395–1406. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Yahyavi-Firouz-Abadi N, Tahsili-Fahadan P, Riazi K, Ghahremani MH and Dehpour AR: Melatonin enhances the anti-convulsant and proconvulsant effects of morphine in mice: Role for nitric oxide signaling pathway. Epilepsy Res. 75:138–144. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Raghavendra V and Kulkarni SK: Reversal of morphine tolerance and dependence by melatonin: Possible role of central and peripheral benzodiazepine receptors. Brain Res. 834:178–181. 1999. View Article : Google Scholar : PubMed/NCBI

13 

Garmabi B, Vousooghi N, Vosough M, Yoonessi A, Bakhtazad A and Zarrindast MR: Effect of circadian rhythm disturbance on morphine preference and addiction in male rats: Involvement of period genes and dopamine D1 receptor. Neuroscience. 322:104–114. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Fan Y, Liang X, Wang R and Song L: Role of endogenous melatoninergic system in development of hyperalgesia and tolerance induced by chronic morphine administration in rats. Brain Res Bull. 135:105–112. 2017. View Article : Google Scholar : PubMed/NCBI

15 

Chou KY, Tsai RY, Tsai WY, Wu CT, Yeh CC, Cherng CH and Wong CS: Ultra-low dose (+)-naloxone restores the thermal threshold of morphine tolerant rats. J Formos Med Assoc. 112:795–800. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Guiding principles in the care and use of animals of the American physiology society

17 

Grossman ML, Basbaum AI and Fields HL: Afferent and efferent connections of the rat tail flick reflex (a model used to analyze pain control mechanisms). J Comp Neurol. 206:9–16. 1982. View Article : Google Scholar : PubMed/NCBI

18 

Tsai RY, Chou KY, Shen CH, Chien CC, Tsai WY, Huang YN, Tao PL, Lin YS and Wong CS: Resveratrol regulates N-methyl-D-aspartate receptor expression and suppresses neuroinflammation in morphine-tolerant rats. Anesth Analg. 115:944–952. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Pirooznia M, Nagarajan V and Deng Y: GeneVenn-A web application for comparing gene lists using Venn diagrams. Bioinformation. 1:420–422. 2007. View Article : Google Scholar : PubMed/NCBI

20 

Xu J, Kelly R, Fang H and Tong W: ArrayTrack: A free FDA bioinformatics tool to support emerging biomedical research-an update. Hum Genomics. 4:428–434. 2010.PubMed/NCBI

21 

Saldanha AJ: Java Treeview-extensible visualization of microarray data. Bioinformatics. 20:3246–3248. 2004. View Article : Google Scholar : PubMed/NCBI

22 

Gene ontology Enrichment analysis website.

23 

Mi H, Muruganujan A and Thomas PD: PANTHER in 2013: Modeling the evolution of gene function, and other gene attributes, in the context of phylogenetic trees. Nucleic Acids Res 41 (Database Issue). D377–D386. 2013.

24 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar

25 

Roeckel LA, Le Coz GM, Gavériaux-Ruff C and Simonin F: Opioid-induced hyperalgesia: Cellular and molecular mechanisms. Neuroscience. 338:160–182. 2016. View Article : Google Scholar : PubMed/NCBI

26 

Kwon HS, Johnson TV, Joe MK, Abu-Asab M, Zhang J, Chan CC and Tomarev SI: Myocilin mediates myelination in the peripheral nervous System through ErbB2/3 signaling. J Biol Chem. 288:26357–26371. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Fontenille L, Rouquier S, Lutfalla G and Giorgi D: Microtubule-associated protein 9 (Map9/Asap) is required for the early steps of zebrafish development. Cell Cycle. 13:1101–1114. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Downes GB and Gautam N: The G protein subunit gene families. Genomics. 62:544–552. 1999. View Article : Google Scholar

29 

Thal DM, Vuckovic Z, Draper-Joyce CJ, Liang YL, Glukhova A, Christopoulos A and Sexton PM: Recent advances in the determination of G protein-coupled receptor structures. Curr Opin Struct Biol. 51:28–34. 2018. View Article : Google Scholar : PubMed/NCBI

30 

Klein DC, Bailey MJ, Carter DA, Kim JS, Shi Q, Ho AK, Chik CL, Gaildrat P, Morin F, Ganguly S, et al: Pineal function: Impact of microarray analysis. Mol Cell Endocrinol. 314:170–183. 2010. View Article : Google Scholar

31 

Bohn LM, Gainetdinov RR, Lin FT, Lefkowitz RJ and Caron MG: Mu-opioid receptor desensitization by beta-arrestin-2 determines morphine tolerance but not dependence. Nature. 408:720–723. 2000. View Article : Google Scholar : PubMed/NCBI

32 

Zuo Z: The role of opioid receptor internalization and beta-arrestins in the development of opioid tolerance. Anesth Analg. 101:728–734, table of contents. 2005. View Article : Google Scholar : PubMed/NCBI

33 

Nagaleekar VK, Diehl SA, Juncadella I, Charland C, Muthusamy N, Eaton S, Haynes L, Garrett-Sinha LA, Anguita J and Rincón M: IP3 receptor-mediated Ca2+ release in naive CD4 T cells dictates their cytokine program. J Immunol. 181:8315–8322. 2008. View Article : Google Scholar : PubMed/NCBI

34 

Taylor CW and Tovey SC: IP(3) receptors: Toward understanding their activation. Cold Spring Harb Perspect Biol. 2:a0040102010. View Article : Google Scholar : PubMed/NCBI

35 

Liu D and Liman ER: Intracellular Ca2+ and the phospholipid PIP2 regulate the taste transduction ion channel TRPM5. Proc Natl Acad Sci USA. 100:15160–15165. 2003. View Article : Google Scholar : PubMed/NCBI

36 

Dang VC and Christie MJ: Mechanisms of rapid opioid receptor desensitization, resensitization and tolerance in brain neurons. Br J Pharmacol. 165:1704–1716. 2012. View Article : Google Scholar :

37 

Stafford K, Gomes AB, Shen J and Yoburn BC: mu-Opioid receptor downregulation contributes to opioid tolerance in vivo. Pharmacol Biochem Behav. 69:233–237. 2001. View Article : Google Scholar : PubMed/NCBI

38 

Polastron J, Meunier JC and Jauzac P: Chronic morphine induces tolerance and desensitization of mu-opioid receptor but not down-regulation in rabbit. Eur J Pharmacol. 266:139–146. 1994. View Article : Google Scholar : PubMed/NCBI

39 

Wang L, Liu H, Jiao Y, Wang E, Clark SH, Postlethwaite AE, Gu W and Chen H: Differences between mice and humans in regulation and the molecular network of collagen, type III, alpha-1 at the gene expression level: Obstacles that translational research must overcome. Int J Mol Sci. 16:15031–15056. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Brodsky AN, Caldwell M and Harcum SW: Glycosylation and post-translational modification gene expression analysis by DNA microarrays for cultured mammalian cells. Methods. 56:408–417. 2012. View Article : Google Scholar :

Related Articles

Journal Cover

February-2019
Volume 43 Issue 2

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Cheng YC, Tsai RY, Sung YT, Chen IJ, Tu TY, Mao YY and Wong CS: Melatonin regulation of transcription in the reversal of morphine tolerance: Microarray analysis of differential gene expression . Int J Mol Med 43: 791-806, 2019
APA
Cheng, Y., Tsai, R., Sung, Y., Chen, I., Tu, T., Mao, Y., & Wong, C. (2019). Melatonin regulation of transcription in the reversal of morphine tolerance: Microarray analysis of differential gene expression . International Journal of Molecular Medicine, 43, 791-806. https://doi.org/10.3892/ijmm.2018.4030
MLA
Cheng, Y., Tsai, R., Sung, Y., Chen, I., Tu, T., Mao, Y., Wong, C."Melatonin regulation of transcription in the reversal of morphine tolerance: Microarray analysis of differential gene expression ". International Journal of Molecular Medicine 43.2 (2019): 791-806.
Chicago
Cheng, Y., Tsai, R., Sung, Y., Chen, I., Tu, T., Mao, Y., Wong, C."Melatonin regulation of transcription in the reversal of morphine tolerance: Microarray analysis of differential gene expression ". International Journal of Molecular Medicine 43, no. 2 (2019): 791-806. https://doi.org/10.3892/ijmm.2018.4030