Zum Hauptinhalt springen

Abnormal expression of rno_circRNA_014900 and rno_circRNA_005442 induced by ketamine in the rat hippocampus.

Mao, J ; Li, T ; et al.
In: BMC psychiatry, Jg. 20 (2020-01-02), Heft 1, S. 1
Online academicJournal

Abnormal expression of rno_circRNA_014900 and rno_circRNA_005442 induced by ketamine in the rat hippocampus 

Background: Recent studies have shown that circular RNA (circRNA) is rich in microRNA (miRNA) binding sites. We have previously demonstrated that the antidepressant effect of ketamine is related to the abnormal expression of various miRNAs in the brain. This study determined the expression profile of circRNAs in the hippocampus of rats treated with ketamine. Methods: The aberrantly expressed circRNAs in rat hippocampus after ketamine injection were analyzed by microarray chip, and we further validated these circRNAs by quantitative reverse-transcription PCR (qRT-PCR). The target genes of the different circRNAs were predicted using bioinformatic analyses, and the functions and signal pathways of these target genes were investigated by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. Results: Microarray analysis showed that five circRNAs were aberrantly expressed in rat hippocampus after ketamine injection (fold change > 2.0, p < 0.05). The results from the qRT-PCR showed that one of the circRNAs was significantly increased (rno_circRNA_014900; fold change = 2.37; p = 0.03), while one was significantly reduced (rno_circRNA_005442; fold change = 0.37; p = 0.01). We discovered a significant enrichment in several GO terms and pathways associated with depression. Conclusion: Our findings showed the abnormal expression of ketamine-induced hippocampal circRNAs in rats.

Keywords: Rat; Ketamine; CircRNA; Hippocampus

Jing Mao and Tianmei Li contributed equally to this work.

Background

The most commonly used antidepressants today effectively improve symptoms of depression, but require at least 2 weeks to take therapeutic effect. In addition, about two-thirds of depressive patients do not respond to the currently available antidepressants and are prone to relapse [[1]]. Therefore, the development of new, fast-acting antidepressants is particularly important for patients with depression-induced suicidal tendencies [[2]].

Recent observations suggest that sub-anesthetic doses of ketamine produce rapid therapeutic effects in depressed patients and in animal models of depression [[3]–[6]]. These effects are characterized by a rapid onset of action (within hours) after a single dose, a lasting effect (1 week), as well as an efficacy in patients resistant to traditional antidepressant drugs [[7]].

In a prior investigation, we found that the antidepressant effect of ketamine was related to the regulation of multiple microRNAs (miRNAs) in neurons [[9]]. Recent studies have shown that circular RNA (circRNA) is rich in miRNA binding sites. CircRNA plays the role of an miRNA sponge in cells, thereby relieving the inhibitory effect of the miRNAs on their target genes and increasing the expression of these genes. The mechanism by which circRNA inhibits miRNAs to increase the expression of their target genes is called the competitive endogenous RNA (ceRNA) mechanism [[10]].

Based on the previous reports, the aim of this study was to determine the effect of an antidepressant dose of ketamine on the expression of circRNAs in the hippocampus of rats and examine a possible circRNA-mediated mechanism for ketamine's action. This work may provide new perspectives on the development of circRNA as a possible drug target.

Methods

Animals

These experiments were conducted in male Sprague-Dawley rats (50 days old, 150–200 g), provided by Chengdu Dashuo biological technology Co., Ltd., China (experimental animal production license: SCXK Chengdu 2013–17). These rats were housed 5 per cage in standard cages (42 × 20 × 20 cm) in a room. Animals had access to food and water ad libitum during the experiment. The room was maintained at 25–26 °C with about 65% relative humidity, on a 12-h dark/light cycle (lights on at 7 am). All experiments were performed according to National Institute of Health (NIH) guidelines and approved by the ethics committee of Southwest Medical University (Approval number:20180306038).

Experimental design and procedure

After 1 week of adaptation, the rats were randomly divided into control and experimental group (12 rats/group). The rats in control group were daily injected with 0.9% saline, whereas the rats in experimental group received ketamine (15 mg/kg). All injections were done intraperitoneally for three consecutive days (the volume of injection was 1 ml/kg). The dose of ketamine used in this study was based on our previous study [[9]]. On day 4, approximately 24 h after the last ketamine or saline injection, the rats were sacrificed by cervical dislocation, and their hippocampus tissues were dissected out for circRNA microarray analysis (n = 3/group) and for qRT-PCR (n = 6 /group). The hippocampus of 3 remaining rats per group were spare specimen according to the quality of hippocampus removed from rats.

CircRNAs analysis from microarray chip

Relevant circRNAs were analyzed according to our previous approach [[12]]. Hippocampus tissues(n = 3/group) were used for microarray assay to determine differentially expressed circRNA between the two groups. The microarray hybridization was performed based on the Arraystar's standard protocols, including purifying RNA, transcribing into fluorescent cRNA, and hybridizing onto the rat circRNA Arrays (Arraystar). Finally, the hybridized slides were washed, fixed and scanned to images by the Agilent Scanner G2505C. The Agilent Feature Extraction software (version 11.0.1.1) were used to analyze the acquired array images. The raw data were normalized and data analysis was further performed with the R software Limma package (Agilent Technologies). The statistical significance of differentially regulated circRNAs between the two groups was identified through screening fold change ≥2.0, P < 0.05 and FDR < 0.05.

Quantitative real-time PCR validation

Hippocampus tissues(n = 6/group) were used for qRT-PCR validation. After RNA isolation, M-MLV reverse transcriptase (Invitrogen, USA) was used for synthesizing cDNA according to the manufacturer's instructions. Subsequently, we performed qRT-PCR using the ViiA 7 Real-time PCR System (Applied Biosystems, Foster City, CA, USA) in a total reaction volume of 10 μl, including 2 μl cDNA, 5 μl 2 × Master Mix, 0.5 μl PCR Forward Primer (10 μM), 0.5 μl PCR Reverse Primer (10 μM) and 2 μl double distilled water. The protocol was initiated at 95 °C for 10 min, then at 95 °C (10 s), 60 °C (60 s) for a total 40 cycles. β-actin was used as a reference. Results were harvested in three independent wells. For quantitative results, the relative expression level of each circRNA was calculated using 2−ΔΔCt method.

Competing endogenous RNA analysis of differentially expressed circRNAs

The candidate miRNA binding sites were searched on the sequences of circRNAs and mRNAs, and the circRNA-miRNA-mRNA interaction were found by the overlapping of the same miRNA seed sequence binding site both on the circRNAs and the mRNA. The miRNA-mRNA interactions were predicted by Targetscan (http://www.targetscan.org/), while the miRNA binding sites were predicted by miRcode (http://www.mircode.org/).

GO and KEGG pathway analysis

Gene Ontology (GO) analysis (http://www.geneontology.org) were conducted to construct meaningful annotation of genes and gene products in the organisms. The ontology includes molecular functions (MFs), biological processes (BPs) and cellular components (CCs). KEGG pathway analysis were also performed to harvest pathway clusters on the molecular interaction and reaction networks in differentially regulated genes. The -log10 (p-value) denotes enrichment score representing the significance of GO term enrichment and pathway correlations among differentially expressed genes.

Statistical analysis

The statistical package for the social sciences (SPSS) 11.0 was selected for statistical analysis. All data were expressed as mean ± SEM. The data from the circRNA microarray and qRT-PCR were analyzed by one-way ANOVA or multi-factorial ANOVA followed by Tukey's post hoc test. P values less than 0.05 with statistically significant differences.

Results

The expression profile of circRNAs in the rat hippocampus using microarray analysis and secon...

The RNA concentration and purity of all samples meted the requirement (larger than 1.8 and an RNA concentration greater than 30 ng) for subsequent microarray detection of the circRNA expression profile.

As shown in Fig. 1, significantly different circRNAs were selected for hierarchical clustering analysis. The circRNA hierarchical clustering map not only displays circRNA expression, but also exhibits the expression change of a single circRNA from both groups. As shown in Table 1, four circRNAs were upregulated and one was down-regulated in the hippocampus of rats treated with ketamine from circRNA microarray analysis (p < 0.05), but only two of the five circRNAs were confirmed to be differentially expressed from qRT-PCR (Table 3, p < 0.05). As shown in Table 2, the primers for the five different circRNAs were designed using Primer software 5.0.

Graph: Fig. 1 The hierarchical clustering plot of differentially expressed circRNAs (fold change ≥1.5, p < 0.05). Red indicates circRNAs with high expression levels, and green represents circRNAs with low expression levels, the color depth (ranging from black to color) indicates different expression intensities. Each row in the figure indicates a different circRNA, and each column indicates a sample (T is the ketamine group and C is the vehicle group). The left side of the figure shows the circRNA clustering tree, whereas the top shows the hippocampal sample clustering tree

Aberrantly expressed circRNAs in rat hippocampus revealed by microarray analysis (Fold change ≥2.0, p < 0.05)

circRNA

Fold Change

Regulation

P-value

rno_circRNA_003460

5.98

up

0.001

rno_circRNA_014900

2.28

up

0.003

rno_circRNA_006565

2.11

up

0.010

rno_circRNA_013109

2.17

up

0.012

rno_circRNA_005442

2.01

down

0.012

A list of primers used for real-time PCR

Gene

Bi-directional primer sequence

Annealing temperature (°C)

Primer length (bp)

β-actin (Reference)

Forward:5′CGAGTACAACCTTCTTGCAGC 3′

Reverse: 5′ ACCCATACCCACCATCACAC 3'

60

202

rno_circRNA_014900

Forward:5′ CTTAGATGACCTGGAGAAGACCT 3′

Reverse: 5′ TGACTTGGTGCTGTTGACTTTAG 3'

60

124

rno_circRNA_013109

Forward:5′ ATTATAGAGCTAATTACAACTTCCG 3′

Reverse:5′ TTATCTGAAGCATGTTAAGACAATA 3'

60

105

rno_circRNA_006565

Forward:5′ CGACTTCAAAAGAGTTGTGGATT 3′

Reverse: 5′ TTCTCCTCGTGAGCTTTTTTCTC 3'

60

54

rno_circRNA_005442

Forward:5′ ACCCCATGAGAAAGACCAGGTC 3′

Reverse:5′ CTGCTCTCTTCAAGTGAAAGACATC 3′

60

60

rno_circRNA_003460

Forward:5′ CGCTAAGCATTTCTTTGGAA 3′

Reverse: 5′ GTAGTGGGTGTAGGGAGGAGA 3′

60

76

Predicted miRNAs sponged by the two circRNAs and their corresponding target genes

As shown in Table 3, the two circRNAs 014900 and 005442 collectively sponged ten miRNAs, namely, rno-miR-466b-5p, rno-miR-6332, rno-miR-6321, rno-miR-193a-5p, rno-miR-1224, rno-miR-323-5p, rno-miR-107-5p, rno-miR-135b-5p, rno-miR-135a-5p, and rno-miR-344b-5p. Each of these miRNAs has target genes that they endogenously regulate, as shown in Table 4, the two circRNAs could indirectly regulate numerous target genes by their endogenous competition mechanism.

qRT-PCR-confirmed expression of circRNAs in rat hippocampus and the predicted target miRNAs

circRNA

Fold Change

Regulation

P-value

Predicted target miRNAs

rno_circRNA_014900

2.37

up

0.029

rno-miR-466b-5p

rno-miR-6332

rno-miR-6321

rno-miR-193a-5p

rno-miR-1224

rno_circRNA_005442

2.72

down

0.010

rno-miR-323-5p

rno-miR-107-5p

rno-miR-135a-5p

rno-miR-135b-5p

rno-miR-344b-5p

Target genes regulated indirectly by the two differentially expressed circRNAs through miRNAs

circRNA

Sponged miRNAs

Gene Symbol

Gene Description

rno_circRNA_014900

rno-miR-193a-5p

rno-miR-1224

Nova1

neuro-oncological ventral antigen 1

rno-miR-193a-5p

rno-miR-6332

Clasp1

cytoplasmic linker associated protein 1

miR-193a-5p

rno-miR-466b-5p

Rgs4, Mixl1

regulator of G-protein signaling 4; Mix paired-like homeobox 1

rno-miR-466b-5p

rno-miR-6332

Usp37, RGD1566029, Zfp91, Zmiz1, Pbx1

ubiquitin specific peptidase 37; similar to mKIAA1644 protein; zinc finger protein 91; zinc finger, MIZ-type containing 1; pre-B-cell leukemia homeobox 1

rno-miR-466b-5p

rno-miR-1224

Calcoco1

calcium binding and coiled coil domain 1

rno-miR-466b-5p

rno-miR-6321

Brwd3, Nfat5, Cnot7, Camta1

bromodomain and WD repeat domain containing 3; nuclear factor of activated T-cells 5, tonicity-responsive; CCR4-NOT transcription complex, subunit 7; calmodulin binding transcription activator 1

rno-miR-6332

rno-miR-1224

Prpf4b, Hgs, Gtdc1, Dgkk

pre-mRNA processing factor 4B;hepatocyte growth factor-regulated tyrosine kinase substrate; glycosyltransferase-like domain containing 1;diacylglycerol kinase kappa

rno-miR-6332

rno-miR-6321

RGD1562037, Usp24,Ssbp2

similar to OTTHUMP00000046255; ubiquitin specific peptidase 24; single-stranded DNA binding protein 2

rno-miR-6332

rno-miR-6321

rno-miR-466b-5p

Ssbp2, RGD1562037

singe-stranded DNA binding protein 2; similar to OTTHUMP00000046255

rno-miR-6332

rno-miR-466b-5p rno-miR-1224

Zfp91

zinc finger protein 91

rno_circRNA_005442

rno-miR-107-5p

rno-miR-323-5p

Eps8,Rhot1

epidermal growth factor receptor pathway substrate 8, ras homolog gene family, member T1

rno-miR-107-5p

rno-miR-344b-5p

Hn1

hematological and neurological expressed 1

rno-miR-107-5p

rno-miR-135b-5p

Ptk2,Tiam1

protein tyrosine kinase 2;T-cell lymphoma invasion and metastasis 1

rno-miR-107-5p

rno-miR-135a(b)-5p

Slc8a1,Man2a1,Tnpo1,Rgl1

solute carrier family 8 (sodium/calcium exchanger), member 1; mannosidase, alpha, class 2A, member 1;transportin 1; ral guanine nucleotide dissociation stimulator,-like 1

rno-miR-323-5p

rno-miR-344b-5p

Tomm6

translocase of outer mitochondrial membrane 6 homolog (yeast)

rno-miR-323-5p

miR-135a(b)-5p

Shisa7

shisa family member 7

rno-miR-344b-5p

miR-135a(b)-5p

Arel1

apoptosis resistant E3 ubiquitin protein ligase 1

rno-miR-323-5p

rno-miR-107-5p

rno-miR-135a(b)-5p

Rhot1

ras homolog gene family, member T1

GO analysis

As shown in Fig. 2, the molecular functions (MFs) of neurons that may be regulated by the target genes of the differentially expressed circRNAs (p < 0.05, left: rno_circRNA_014900; right: rno_circRNA_005442). The classification of notable MFs was shown in Fig. 2a and b shows the order of these MFs by their GO analysis enrichment scores. Figure 2c shows the notable activities of neurons using fold enrichment. The detailed list of the genes identified as regulated by circRNA was shown in Additional files 1 and 2.

Graph: Fig. 2 The molecular functions (MFs) of neurons regulated by the target genes of the differentially expressed circRNAs (p < 0.05, left: rno_circRNA_014900; right: rno_circRNA_005442). a classifies the notable MFs and b shows these same MFs ordered by their GO analysis enrichment scores. c shows the notable activities of neurons that may be regulated by target genes predicted using fold enrichment

As shown in Fig. 3, the biological processes (BPs) in neurons that may be regulated by target genes of the two circRNAs (p < 0.05, left: rno_circRNA_014900; right: rno_circRNA_005442). The classification of notable BPs was shown in Fig. 3a and b shows the different BPs predicted by enrichment scores. Figure 3c shows the different BPs of neurons predicted using fold enrichment. The detailed list of the genes identified as regulated by circRNA was shown in Additional files 3 and 4.

Graph: Fig. 3 The biological processes (BPs) in neurons regulated by target genes of the two circRNAs (p < 0.05, left: rno_circRNA_014900; right: rno_circRNA_005442). a classifies the predicted BPs and b shows the notable BPs predicted by enrichment scores. c shows the notable BPs of neurons that may be regulated by target genes predicted using fold enrichment

As shown in Fig. 4, the cellular components (CCs) that may be regulated by target genes of the two differentially expressed circRNAs (p < 0.05, left: rno_circRNA_014900; right: rno_circRNA_005442). The classification of notable CCs was shown in Fig. 4a and b shows the different CCs predicted by enrichment scores. Figure 4c shows the different CCs of neurons using fold enrichment. The detailed list of the genes identified as regulated by circRNA was shown in Additional files 5 and 6.

Graph: Fig. 4 The cellular components (CCs) that may be regulated by target genes of the two differentially expressed circRNAs (p < 0.05, left: rno_circRNA_014900; right: rno_circRNA_005442). a classifies the predicted CCs and b shows the notable CCs predicted by enrichment scores. c shows the notable CCs of neurons that may be regulated by target genes predicted using fold enrichment

KEGG pathway analysis

Tables 5 and 6 shows the signaling pathways that may be regulated by target genes of the differentially expressed rno_circRNA_014900 and rno_circRNA_005442 (p < 0.05). The identified pathways were involved in the regulation of central nervous system functions, such as Wnt signaling, long-term depression, PI3K-Akt signaling, dopaminergic synapse activity, mTOR signaling, p53 signaling, apoptosis, TGF-beta signaling, axon guidance, hippo signaling, and MAPK signaling.

The signaling pathways that may be regulated by targeted genes of the differentially expressed rno_circRNA_014900(p < 0.05)

PathwayID

Definition

Fisher-Pvalue

Enrichment_Score

rno04510

Focal adhesion - Rattus norvegicus (rat)

4.05765E-06

5.391726

rno05205

Proteoglycans in cancer - Rattus norvegicus (rat)

0.000263739

3.578826

rno05200

Pathways in cancer - Rattus norvegicus (rat)

0.000839484

3.075988

rno04216

Ferroptosis - Rattus norvegicus (rat)

0.000947662

3.023347

rno04015

Rap1 signaling pathway - Rattus norvegicus (rat)

0.001242467

2.905715

rno00564

Glycerophospholipid metabolism - Rattus norvegicus (rat)

0.001767738

2.752582

rno04310

Wnt signaling pathway - Rattus norvegicus (rat)

0.002533546

2.596271

rno05165

Human papillomavirus infection - Rattus norvegicus (rat)

0.003213976

2.492957

rno04140

Autophagy - animal - Rattus norvegicus (rat)

0.00464105

2.333384

rno05222

Small cell lung cancer - Rattus norvegicus (rat)

0.005031787

2.298278

rno04070

Phosphatidylinositol signaling system - Rattus norvegicus (rat)

0.006461245

2.189684

rno00561

Glycerolipid metabolism - Rattus norvegicus (rat)

0.00791785

2.101393

rno04213

Longevity regulating pathway - multiple species - Rattus norvegicus (rat)

0.00791785

2.101393

rno04730

Long-term depression - Rattus norvegicus (rat)

0.00791785

2.101393

rno04151

PI3K-Akt signaling pathway - Rattus norvegicus (rat)

0.00874211

2.058384

rno04512

ECM-receptor interaction - Rattus norvegicus (rat)

0.009516369

2.021529

rno04137

Mitophagy - animal - Rattus norvegicus (rat)

0.009920558

2.003464

rno04728

Dopaminergic synapse - Rattus norvegicus (rat)

0.01248382

1.903653

rno04211

Longevity regulating pathway - Rattus norvegicus (rat)

0.01286284

1.890663

rno04136

Autophagy - other - Rattus norvegicus (rat)

0.01346501

1.870793

rno04150

mTOR signaling pathway - Rattus norvegicus (rat)

0.01358861

1.866825

rno04115

p53 signaling pathway - Rattus norvegicus (rat)

0.01402018

1.853246

rno04215

Apoptosis - multiple species - Rattus norvegicus (rat)

0.01492946

1.825956

rno05214

Glioma - Rattus norvegicus (rat)

0.01496318

1.824976

rno04270

Vascular smooth muscle contraction - Rattus norvegicus (rat)

0.02428063

1.61474

rno01521

EGFR tyrosine kinase inhibitor resistance - Rattus norvegicus (rat)

0.02549639

1.593521

rno05219

Bladder cancer - Rattus norvegicus (rat)

0.02580699

1.588263

rno05224

Breast cancer - Rattus norvegicus (rat)

0.02850884

1.54502

rno05166

HTLV-I infection - Rattus norvegicus (rat)

0.02925304

1.533829

rno04066

HIF-1 signaling pathway - Rattus norvegicus (rat)

0.03188594

1.496401

rno04068

FoxO signaling pathway - Rattus norvegicus (rat)

0.03490339

1.457132

rno04144

Endocytosis - Rattus norvegicus (rat)

0.03599259

1.443787

rno04720

Long-term potentiation - Rattus norvegicus (rat)

0.03856442

1.413813

rno04978

Mineral absorption - Rattus norvegicus (rat)

0.04045532

1.393024

rno04725

Cholinergic synapse - Rattus norvegicus (rat)

0.04272807

1.369287

rno05030

Cocaine addiction - Rattus norvegicus (rat)

0.04327309

1.363782

rno04010

MAPK signaling pathway - Rattus norvegicus (rat)

0.04346333

1.361877

rno04120

Ubiquitin mediated proteolysis - Rattus norvegicus (rat)

0.04667443

1.330921

rno00062

Fatty acid elongation - Rattus norvegicus (rat)

0.04768946

1.321578

rno05211

Renal cell carcinoma - Rattus norvegicus (rat)

0.04775832

1.320951

rno01522

Endocrine resistance - Rattus norvegicus (rat)

0.04798442

1.3189

The signaling pathways that may be regulated by targeted genes of the differentially expressed rno_circRNA_005442 (p < 0.05)

PathwayID

Definition

Fisher-Pvalue

Enrichment_Score

rno04350

TGF-beta signaling pathway - Rattus norvegicus (rat)

0.002056745

2.68682

rno04960

Aldosterone-regulated sodium reabsorption - Rattus norvegicus (rat)

0.002806053

2.551904

rno05231

Choline metabolism in cancer - Rattus norvegicus (rat)

0.0039363

2.404912

rno05412

Arrhythmogenic right ventricular cardiomyopathy (ARVC) - Rattus norvegicus (rat)

0.005000479

2.300988

rno04550

Signaling pathways regulating pluripotency of stem cells - Rattus norvegicus (rat)

0.005251424

2.279723

rno04218

Cellular senescence - Rattus norvegicus (rat)

0.005747859

2.240494

rno04360

Axon guidance - Rattus norvegicus (rat)

0.005747859

2.240494

rno05225

Hepatocellular carcinoma - Rattus norvegicus (rat)

0.005747859

2.240494

rno04144

Endocytosis - Rattus norvegicus (rat)

0.006773032

2.169217

rno00510

N-Glycan biosynthesis - Rattus norvegicus (rat)

0.007410522

2.130151

rno04390

Hippo signaling pathway - Rattus norvegicus (rat)

0.009694587

2.013471

rno04068

FoxO signaling pathway - Rattus norvegicus (rat)

0.01587784

1.799209

rno04371

Apelin signaling pathway - Rattus norvegicus (rat)

0.02058289

1.686494

rno04520

Adherens junction - Rattus norvegicus (rat)

0.02501545

1.601792

rno05210

Colorectal cancer - Rattus norvegicus (rat)

0.02501545

1.601792

rno04973

Carbohydrate digestion and absorption - Rattus norvegicus (rat)

0.02580168

1.588352

rno04010

MAPK signaling pathway - Rattus norvegicus (rat)

0.03810185

1.419054

rno05410

Hypertrophic cardiomyopathy (HCM) - Rattus norvegicus (rat)

0.04204027

1.376335

rno05414

Dilated cardiomyopathy (DCM) - Rattus norvegicus (rat)

0.04832562

1.315823

Discussion

There are about 340 million patients suffering from depression around the world, and it is estimated that up to 1 million people die by depression-induced suicide every year. Therefore, depression has become a global public health problem [[13]]. The major clinical drawback of currently available antidepressants is their slow onset (14 days on average) and poor effects [[1]]. Therefore, the development of rapid-onset antidepressants is particularly important for patients suffering from major depressive disorder with suicidal tendency. The antidepressant effect of ketamine initiates quickly and is also effective in treating refractory depression [[7]]. However, the psychosocial side effects induced by ketamine have restricted its use in the treatment of depression [[14]]. An in-depth understanding of the mechanism of ketamine's rapid antidepressant effect can provide new targets for similar antidepressants.

Recent many researches have shown that many transcriptional products come from the non-coding RNA (ncRNA), including the microRNA (miRNA), long non-coding RNA (lncRNA), and circular RNA (circRNA), these ncRNAs can regulate gene expression at the DNA level, pre-transcriptional level, transcriptional level, post-transcriptional level, translational level, and post-translational level [[15]]. Previous study from our group found that ketamine induced abnormal expression of miRNAs in the brain, and subsequent target gene and functional analysis revealed that this abnormal expression of miRNAs was closely related to the antidepressant effect of ketamine, but the therapeutic effect of antidepressants focusing on a single mechanism is poor due to the complex mechanism of major depressive disorder [[9]]. Therefore, we believe that a single miRNA plays relatively weak role in the regulation of a specific target gene. It was hypothesized that a substance in the body that could gather several miRNAs regulated the same specific target gene, then the regulatory action on the specific target gene would be obviously increased. It was interesting that circRNAs, a novel type of non-coding RNAs [[15]], were perceived as a rare curiosity to having a central regulatory role in RNA metabolism, and accumulating evidences suggested that circRNAs could function as miRNA sponges (the ceRNA mechanism), therefore, circRNA could relieve the inhibitory effect of miRNA on its target gene and increase the expression of the target gene [[10]–[12]].

Our results showed that the expression of rno_circRNA_014900 was significantly increased by ketamine, while the expression of rno_circRNA_005442 was obviously decreased. Since these two circRNAs were not investigated in previous studies on depression, we further investigated their regulated target genes and signaling pathways. Competing endogenous RNA analysis found that the rno_circRNA_014900 could sponge rno-miR-466b-5p, rno-miR-6332, rno-miR-6321, rno-miR-193a-5p and rno-miR-1224, whereas the rno_circRNA_005442 had the binding sites in rno-miR-323-5p, rno-miR-107-5p, rno-miR-135a-5p, rno-miR-135b-5p and rno-miR-344b-5p. In a prior investigation, we found that miR-206 was a critical novel gene for the expression of BDNF (brain-derived neurotrophic factor) induced by ketamine [[9]], our experimental results were expected to be that the circRNA identified in this study would target the miRNA-206, but structural prediction analysis showed that the miRNAs sponged by rno_circRNA_014900 and rno_circRNA_005442 did not include miRNA-206, we thought rno_circRNA_014900 and rno_circRNA_005442 did not regulate the expression of miRNA-206, how ketamine affects the expression of miRNA-206 needs to be further explored in future research. Further prediction analysis revealed that rno_circRNA_014900 and rno_circRNA_005442 may inhibit up to four miRNAs closely related to depression. For example, the predicted target genes related to depression include NOVA1 (regulated by miR-193-5p and miR-1224) [[16]], Rgs4 (regulated by miR-193-5p and miR-466b-5p) [[17]–[20]], zinc-finger protein (regulated by miR-6332, miR-466b-5p, and miR-1224) [[21]], PBX1 (regulated by miR-466b-5p and miR-6332) [[22]], SLC8A1 (regulated by miR-107-5p, miR-135a(b)-5p, and miR-135b-5p) [[23]], PTK2 (regulated by miR-107-5p and miR-135b-5p) [[24]], and Tiam1 (regulated by miR-107-5p and miR-135b-5p) [[25]]. Therefore, we believed rno_circRNA_014900 or rno_circRNA_005442 could relieve the inhibitory effect of miRNA on its target gene and increase the expression of the target gene, the phenomenon needs to be further confirmed in future studies.

The results from GO analysis found that the the target genes of the two circRNAs regulated many molecular functions (including protein phosphatase binding, SUMO binding, Wnt-protein binding, etc.), biological processes (including adherens junction assembly, neuron projection morphogenesis, neurological processes, etc.), and cellular components (including dendritic spines, AMPA-glutamate receptor binding, neuronal cell body, etc.). The results from KEGG pathway prediction showed that the signaling pathways regulated by target genes of the two circRNAs included Wnt signaling, long-term depression, PI3K-Akt signaling, dopaminergic synapses, mTOR signaling, p53 signaling, apoptosis, MAPK signaling, TGF-beta signaling, axon guidance, Hippo signaling, etc.. These signaling pathways may be involved in the occurrence and development of depression, because some researches found that the Wnt signaling pathway played important roles in the depression-like behaviors [[26]–[28]], the PI3K-Akt signaling pathway was related to the rapid antidepressant-like effects of some drugs [[29]–[33]].

As we analyzed in other study about circRNAs [[12]], novel therapies should include multiple genes and pathways due to the complex mechanisms of major depression disorder. Because many pathophysiological processes of stress-related depression were regulated by several miRNAs, therefore these miRNAs may be commom targets of antidepressant therapies. It was not difficult to understand that the increase in relevant circRNAs expression would enhance the translation of their target genes due to miRNA sponges, a down-expression in relevant circRNAs would result in the obvious silencing of downstream target genes. Therefore, in the future, artificial circRNA drugs will be developed to restore normal transcriptional regulation.

Conclusions

In summary, we found that ketamine treatment resulted in the abnormal expression of the two circRNAs in the hippocampus of rats, and these two circRNAs may be associated with stress-related depression disorders. CircRNAs should remain the focus of researches investigating antidepressant targets because they have considerable potential in the clinical treatment of stress-related depression. As an invaluable topic for future biomedical studies, we plan to screen for specific circRNAs in the context of depression and to examine their potential value in the diagnosis and treatment of this debilitating disorder.

Funding

This work was supported by the Projects of the National Natural Science Foundation of China (Grant No. 81271478), Department of Science &Technology of Sichuan Province (Grant No. 14JC0093), Department of Education of Sichuan Province (Grant No. 14ZA0142) and Department of Science &Technology of Luzhou City (Grant No. 2015-S-46). The funding bodies had no role in the design of the study or collection, analysis, and interpretation of data or in writing the manuscript.

Acknowledgments

We would like to thank LetPub (http://www.letpub.com) for providing linguistic assistance during the preparation of this manuscript.

Authors' contributions

JM, TL, DF, HZ, JF, LL and XW made substantial contribution to conception of the intervention and design of the study and/or were involved in acquisition of data. JM, TL, DF and HZ made the first draft of the manuscript and LL, CZ and XW critically revised the manuscript. All authors read and approved the final manuscript.

Availability of data and materials

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

Ethics approval and consent to participate

All experimental procedures involving animals were performed in accordance with the NIH Guide for the Care and Use of Laboratory Animals and the Chinese Society for Neuroscience and Behavior recommendations for animal care. All of the experiments involving surgeries and treatments were approved by the ethics committee of Southwest Medical University (Ethics number: 20180306038).

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Supplementary information

Graph: Additional file 1. MF results for circRNA01490. The detailed list of the genes identified as regulated by the circRNA 01490, MF: molecular function.

Graph: Additional file 2. MF results for circRNA005442. The detailed list of the genes identified as regulated by the circRNA005442, MF: molecular function.

Graph: Additional file 3. BP results for circRNA01490. The detailed list of the genes identified as regulated by the circRNA 01490, BP: biological process.

Graph: Additional file 4. BP results for circRNA005442. The detailed list of the genes identified as regulated by the circRNA005442, BP: biological process.

Graph: Additional file 5. CC results for circRNA01490. The detailed list of the genes identified as regulated by the circRNA 01490, CC: cellular component.

Graph: Additional file 6. CC results for circRNA005442. The detailed list of the genes identified as regulated by the circRNA005442, CC: cellular component.

• BPs

  • Biological processes

• CCs

  • Cellular components

• ceRNA

  • Competitive endogenous RNA

• circRNA

  • Circular RNA

• GO

  • Gene Ontology

• i.p.

  • Intraperitoneal

• KEGG

  • Kyoto Encyclopedia of Genes and Genomes

• lncRNA

  • Long non-coding RNA

• MFs

  • Molecular functions

• miRNA

  • MicroRNA

• ncRNA

  • Non-coding RNA

• qRT-PCR

  • Quantitative reverse-transcription PCR

• SEM

  • Standard error of the mean
Supplementary information

Supplementary information accompanies this paper at 10.1186/s12888-019-2374-2.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

References 1 Duman RS, Aghajanian GK. Synaptic dysfunction in depression: potential therapeutic targets. Science. 2012; 338; 6103: 68-72. 10.1126/science.1222939 2 Machado-Vieira R, Salvadore G, Diazgranados N, Zarate CA. Ketamine and the next generation of antidepressants with a rapid onset of action. Pharmacol Ther. 2009; 123; 2: 143-150. 10.1016/j.pharmthera.2009.02.010 3 Berman RM, Cappiello A, Anand A, Oren DA, Heninger GR, Charney DS. Antidepressant effects of ketamine in depressed patients. Biol Psychiatry. 2000; 47; 4: 351-354. 10.1016/S0006-3223(99)00230-9 4 Wang XB, Chen YL, Zhou X, Liu F, Zhang T, Zhang C. Effects of propofol and ketamine as the combined anesthesia for electroconvulsive therapy in patients with depressive disorder. J ECT. 2012; 28; 2: 128-132. 10.1097/YCT.0b013e31824d1d02 5 Garcia LS, Comim CM, Valvassori SS, Réus GZ, Barbosa LM, Andreazza AC. Acute administration of ketamine induces antidepressant-like effects in the forced swimming test and increases BDNF levels in the rat hippocampus. Prog Neuropsychopharmacol. 2008; 32; 1: 140-144. 10.1016/j.pnpbp.2007.07.027 6 Sun HL, Zhou ZQ, Zhang GF, Yang C, Wang XM, Shen JC. Role of hippocampal p11 in the sustained antidepressant effect of ketamine in the chronic unpredictable mild stress model. Transl Psychiatry. 2016; 6: e741. 10.1038/tp.2016.21 7 Yang JJ, Wang N, Yang C, Shi JY, Yu HY, Hashimoto K. Serum interleukin-6 is a predictive biomarker for ketamine's antidepressant effect in treatment-resistant patients with major depression. Biol Psychiatry. 2015; 77; 3: e19-e20. 10.1016/j.biopsych.2014.06.021 8 Murrough JW, Iosifescu DV, Chang LC, Jurdi RK, Green CE, Perez AM. Antidepressant efficacy of ketamine in treatment resistant major depression: a two-site randomized controlledtrial. Am J Psychiatry. 2013; 170; 10: 1134-1142. 10.1176/appi.ajp.2013.13030392 9 Yang X, Yang Q, Wang X, Luo C, Wan Y, Li J. MicroRNA expression profile and functional analysis reveal that miR-206 is a critical novel gene for the expression of BDNF induced by ketamine. NeuroMolecular Med. 2014; 16; 3: 594-605. 10.1007/s12017-014-8312-z Van Rossum D, Verheijen BM, Pasterkamp RJ. Circular RNAs: novel regulators of neuronal development. Front Mol Neurosci. 2016; 9: 744999478 Tay Y, Rinn J, Pandolfi PP. The multilayered complexity of ceRNA crosstalk and competition. Nature. 2014; 505; 7483: 344-352. 10.1038/nature12986 Wang MZ, Su PX, Liu Y, Zhang XT, Yan J, An XG. Abnormal expression of circRNA_089763 in the plasma exosomes of patients with post operative cognitive dysfunction after coronary artery bypass grafting. Mol Med Rep. 2019; 20: 2549-25626691254 Ferrari AJ, Charlson FJ, Norman RE, Patten SB, Freedman G, Murray CJ. Burden of depressive disorders by country, sex, age, and year: findings from the global burden of disease study. PLoS Med. 2010; 10; 11: e1001547. 10.1371/journal.pmed.1001547 Newcomer JW, Farber NB, Jevtovic-Todorovic V, Selke G, Melson AK, Hershey T. Ketamine-induced NMDA receptor hypofunction as a model of memory impairment and psychosis. Neuropsychopharmacology. 1999; 20; 2: 106-118. 10.1016/S0893-133X(98)00067-0 Jeck WR, Sorrentino JA, Wang K, Slevin MK, Burd CE, Liu J. Circular RNAs are abundant, conserved, and associated with ALU repeats. RNA. 2013; 19; 2: 141-157. 10.1261/rna.035667.112 Malki K, Tosto MG, Jumabhoy I, Lourdusamy A, Sluyter F, Craig I. Integrative mouse and human mRNA studies using WGCNA nominates novel candidate genes involved in the pathogenesis of major depressive disorder. Pharmacogenomics. 2013; 14; 16: 1979-1990. 10.2217/pgs.13.154 Kim J, Lee S, Kang S, Jeon TI, Kang MJ, Lee TH. Regulator of G-protein signaling 4 (RGS4) controls morphine reward by glutamate receptor activation in the nucleus Accumbens of mouse brain. Mol Cells. 2018; 41; 5: 454-4645974622 Zeng D, He S, Yu S, Li G, Ma C, Wen Y. Analysis of the association of MIR124-1 and its target gene RGS4 polymorphisms with major depressive disorder and antidepressant response. Neuropsychiatr Dis Treat. 2018; 14: 715-723. 10.2147/NDT.S155076 Stratinaki M, Varidaki A, Mitsi V, Ghose S, Magida J, Dias C. Regulator of G protein signaling 4 [corrected] is a crucial modulator of antidepressant drug action in depression and neuropathic pain models. Proc Natl Acad Sci U S A. 2013; 110; 20: 8254-8259. 10.1073/pnas.1214696110 Mirnics K, Middleton FA, Stanwood GD, Lewis DA, Levitt P. Disease-specific changes in regulator of G-protein signaling 4 (RGS4) expression in schizophrenia. Mol Psychiatry. 2001; 6; 3: 293-301. 10.1038/sj.mp.4000866 Nätt D, Johansson I, Faresjö T, Ludvigsson J, Thorsell A. High cortisol in 5-year-old children causes loss of DNA methylation in SINE retrotransposons: a possible role for ZNF263 in stress-related diseases. Clin Epigenetics. 2015; 7: 91. 10.1186/s13148-015-0123-z Grados MA, Specht MW, Sung HM, Fortune D. Glutamate drugs and pharmacogenetics of OCD: a pathway-based exploratory approach. Expert Opin Drug Discov. 2013; 8; 12: 1515-1527. 10.1517/17460441.2013.845553 Douglas LN, McGuire AB, Manzardo AM, Butler MG. High-resolution chromosome ideogram representation of recognized genes for bipolar disorder. Gene. 2016; 586; 1: 136-147. 10.1016/j.gene.2016.04.011 Gao L, Gao Y, Xu E, Xie J. Microarray analysis of the major depressive disorder mRNA profile data. Psychiatry Investig. 2015; 12; 3: 388-396. 10.4306/pi.2015.12.3.388 Benoist M, Palenzuela R, Rozas C, Rojas P, Tortosa E, Morales B. MAP 1B- dependent Rac activation is required for AMPA receptor endocytosis during long-term depression. EMBO J. 2013; 32; 16: 2287-2299. 10.1038/emboj.2013.166 Ni X, Liao Y, Li L, Zhang X, Wu Z. Therapeutic role of long non-coding RNA TCONS_00019174 in depressive disorders is dependent on Wnt/β-catenin signaling pathway. J Integr Neurosci. 2018; 17; 2: 203-215. 10.3233/JIN-170052 Martin PM, Stanley RE, Ross AP, Freitas AE, Moyer CE, Brumback AC. DIXDC1 contributes to psychiatric susceptibility by regulating dendritic spine and glutamatergic synapse density via GSK3 and Wnt/β-catenin signaling. Mol Psychiatry. 2018; 23; 2: 467-475. 10.1038/mp.2016.184 Zhou WJ, Xu N, Kong L, Sun SC, Xu XF, Jia MZ. The antidepressant roles of Wnt2 and Wnt3 in stress-induced depression-like behaviors. Transl Psychiatry. 2016; 6; 9: e892. 10.1038/tp.2016.122 Cunha MP, Budni J, Ludka FK, Pazini FL, Rosa JM, Oliveira Á, Lopes MW. Involvement of PI3K/Akt signaling pathway and its downstream intracellular targets in the antidepressant-like effect of Creatine. Mol Neurobiol. 2016; 53; 5: 2954-2968. 10.1007/s12035-015-9192-4 Shi HS, Zhu WL, Liu JF, Luo YX, Si JJ, Wang SJ. PI3K/Akt signaling pathway in the basolateral amygdala mediates the rapid antidepressant-like effects of trefoil factor 3. Neuropsychopharmacology. 2012; 37; 12: 2671-2683. 10.1038/npp.2012.131 Lima IVA, Almeida-Santos AF, Ferreira-Vieira TH, Aguiar DC, Ribeiro FM, Campos AC. Antidepressant-like effect of valproic acid-possible involvement of PI3K/Akt/mTOR pathway. Behav Brain Res. 2017; 329: 166-171. 10.1016/j.bbr.2017.04.015 Pazini FL, Cunha MP, Rosa JM, Colla AR, Lieberknecht V. Creatine, similar to ketamine, counteracts depressive-like behavior induced by Corticosterone via PI3K/Akt/mTOR pathway. Mol Neurobiol. 2016; 53; 10: 6818-6834. 10.1007/s12035-015-9580-9 Zhou W, Dong L, Wang N, Shi JY, Yang JJ, Zuo ZY. Akt mediates GSK-3β phosphorylation in the rat prefrontal cortex during the process of ketamine exerting rapid antidepressant actions. Neuroimmunomodulation. 2014; 21; 4: 183-188. 10.1159/000356517

By Jing Mao; Tianmei Li; Di Fan; Hongli Zhou; Jianguo Feng; Li Liu; Chunxiang Zhang and Xiaobin Wang

Reported by Author; Author; Author; Author; Author; Author; Author; Author

Titel:
Abnormal expression of rno_circRNA_014900 and rno_circRNA_005442 induced by ketamine in the rat hippocampus.
Autor/in / Beteiligte Person: Mao, J ; Li, T ; Fan, D ; Zhou, H ; Feng, J ; Liu, L ; Zhang, C ; Wang, X
Link:
Zeitschrift: BMC psychiatry, Jg. 20 (2020-01-02), Heft 1, S. 1
Veröffentlichung: London : BioMed Central, [2001-, 2020
Medientyp: academicJournal
ISSN: 1471-244X (electronic)
DOI: 10.1186/s12888-019-2374-2
Schlagwort:
  • Animals
  • Computational Biology methods
  • Excitatory Amino Acid Antagonists toxicity
  • Male
  • RNA, Circular genetics
  • Rats
  • Rats, Sprague-Dawley
  • Real-Time Polymerase Chain Reaction methods
  • Signal Transduction drug effects
  • Signal Transduction physiology
  • Gene Expression Profiling methods
  • Hippocampus drug effects
  • Hippocampus metabolism
  • Ketamine toxicity
  • RNA, Circular biosynthesis
Sonstiges:
  • Nachgewiesen in: MEDLINE
  • Sprachen: English
  • Publication Type: Journal Article; Research Support, Non-U.S. Gov't
  • Language: English
  • [BMC Psychiatry] 2020 Jan 02; Vol. 20 (1), pp. 1. <i>Date of Electronic Publication: </i>2020 Jan 02.
  • MeSH Terms: Gene Expression Profiling / *methods ; Hippocampus / *drug effects ; Hippocampus / *metabolism ; Ketamine / *toxicity ; RNA, Circular / *biosynthesis ; Animals ; Computational Biology / methods ; Excitatory Amino Acid Antagonists / toxicity ; Male ; RNA, Circular / genetics ; Rats ; Rats, Sprague-Dawley ; Real-Time Polymerase Chain Reaction / methods ; Signal Transduction / drug effects ; Signal Transduction / physiology
  • References: Mol Psychiatry. 2018 Feb;23(2):467-475. (PMID: 27752079) ; Front Mol Neurosci. 2016 Aug 26;9:74. (PMID: 27616979) ; J Integr Neurosci. 2018;17(2):125-132. (PMID: 29036835) ; Behav Brain Res. 2017 Jun 30;329:166-171. (PMID: 28408298) ; Mol Cells. 2018 May 31;41(5):454-464. (PMID: 29754475) ; Biol Psychiatry. 2000 Feb 15;47(4):351-4. (PMID: 10686270) ; RNA. 2013 Feb;19(2):141-57. (PMID: 23249747) ; Pharmacol Ther. 2009 Aug;123(2):143-50. (PMID: 19397926) ; Neuropsychopharmacology. 2012 Nov;37(12):2671-83. (PMID: 22828749) ; Gene. 2016 Jul 15;586(1):136-47. (PMID: 27063557) ; Nature. 2014 Jan 16;505(7483):344-52. (PMID: 24429633) ; Mol Psychiatry. 2001 May;6(3):293-301. (PMID: 11326297) ; Transl Psychiatry. 2016 Feb 23;6:e741. (PMID: 26905413) ; PLoS Med. 2013 Nov;10(11):e1001547. (PMID: 24223526) ; Pharmacogenomics. 2013 Dec;14(16):1979-90. (PMID: 24279853) ; Psychiatry Investig. 2015 Jul;12(3):388-96. (PMID: 26207134) ; Proc Natl Acad Sci U S A. 2013 May 14;110(20):8254-9. (PMID: 23630294) ; Transl Psychiatry. 2016 Sep 13;6(9):e892. (PMID: 27622936) ; Expert Opin Drug Discov. 2013 Dec;8(12):1515-27. (PMID: 24147578) ; Prog Neuropsychopharmacol Biol Psychiatry. 2008 Jan 1;32(1):140-4. (PMID: 17884272) ; J ECT. 2012 Jun;28(2):128-32. (PMID: 22622291) ; Mol Neurobiol. 2016 Jul;53(5):2954-2968. (PMID: 25943184) ; Mol Med Rep. 2019 Sep;20(3):2549-2562. (PMID: 31524256) ; Clin Epigenetics. 2015 Sep 04;7:91. (PMID: 26339299) ; Neuromolecular Med. 2014 Sep;16(3):594-605. (PMID: 24839168) ; Science. 2012 Oct 5;338(6103):68-72. (PMID: 23042884) ; Neuroimmunomodulation. 2014;21(4):183-8. (PMID: 24504086) ; Neuropsychopharmacology. 1999 Feb;20(2):106-18. (PMID: 9885791) ; EMBO J. 2013 Aug 14;32(16):2287-99. (PMID: 23881099) ; Am J Psychiatry. 2013 Oct;170(10):1134-42. (PMID: 23982301) ; Biol Psychiatry. 2015 Feb 1;77(3):e19-e20. (PMID: 25104172) ; Neuropsychiatr Dis Treat. 2018 Mar 08;14:715-723. (PMID: 29563799) ; Mol Neurobiol. 2016 Dec;53(10):6818-6834. (PMID: 26660117)
  • Grant Information: 14JC0093 International Department of Science &Technology of Sichuan Province; 81271478 International the Projects of the National Natural Science Foundation of China
  • Contributed Indexing: Keywords: CircRNA; Hippocampus; Ketamine; Rat
  • Substance Nomenclature: 0 (Excitatory Amino Acid Antagonists) ; 0 (RNA, Circular) ; 690G0D6V8H (Ketamine)
  • Entry Date(s): Date Created: 20200104 Date Completed: 20201019 Latest Revision: 20201019
  • Update Code: 20240513
  • PubMed Central ID: PMC6939336

Klicken Sie ein Format an und speichern Sie dann die Daten oder geben Sie eine Empfänger-Adresse ein und lassen Sie sich per Email zusenden.

oder
oder

Wählen Sie das für Sie passende Zitationsformat und kopieren Sie es dann in die Zwischenablage, lassen es sich per Mail zusenden oder speichern es als PDF-Datei.

oder
oder

Bitte prüfen Sie, ob die Zitation formal korrekt ist, bevor Sie sie in einer Arbeit verwenden. Benutzen Sie gegebenenfalls den "Exportieren"-Dialog, wenn Sie ein Literaturverwaltungsprogramm verwenden und die Zitat-Angaben selbst formatieren wollen.

xs 0 - 576
sm 576 - 768
md 768 - 992
lg 992 - 1200
xl 1200 - 1366
xxl 1366 -