2022 Volume 45 Issue 11 Pages 1616-1626
The dysregulation of certain long non-coding RNAs (lncRNAs) has been considered to be involved in neuropsychiatric disorders such as depression, implying the vital role of these transcripts. We have previously identified many differentially expressed lncRNAs in chronic unpredictable mild stress (CUMS) induced mice. Among them, lncRNA Gm16638-201 was highly expressed in the hippocampus (HIP) of CUMS, but the specific role and the underlying mechanisms remain unclear. Here, we reported that lncRNA Gm16638-201 was highly expressed in the prefrontal cortex (PFC) of CUMS induced depressive mice. Bioinformatic analysis shows that Gm16638-201 is mainly located in the cytoplasm. Nine neurological-related genes (Elmo2, Satb1, Hnrnpul1, Sipa1l3, Mapt, Tada3, Sgip1, IL-16, and StarD5) were predicted to be regulated in cis or trans by Gm16638-201 and involved into the 14-3-3Ɛ neurotrophic signaling pathway. We further confirmed the down-regulation of 14-3-3Ɛ and the nine predicted target genes in the PFC of CUMS mice except for Sgip1 and IL-16. In addition, they were also down-regulated in the primary cortical cell cultures with overexpression of Gm16638-201 constructed using an adenoviral-medicated gene expression system. In conclusion, we found that overexpression of Gm16638-201 negatively regulated several target genes and inhibited the 14-3-3Ɛ pathway in the PFC of CUMS induced depressive mice. This promising result suggests that Gm16638-201 may be a potential novel therapeutic target for depression.
Depression is a common mental disorder characterized by persistent or recurrent apathy, world-weariness, anxiety, sleep disturbances, changes in appetite or weight, and impaired concentration,1) accompanied by restlessness, suicide, and other psychomotor functional disorders,2) which seriously damages the QOL and body function of patients, and increases the economic burden of the family. According to the WHO report in 2017, about 340 million people of all ages in the world are suffering from depression.3) More than 90 million people suffer from depression in China, which becomes the second-largest disease of burden.4) Given the great social impact of depression, scientists have conducted a lot of research on the pathogenesis and mechanism and proposed several related theories including the monoaminergic theory,5) the neurotrophic factors,6) and the inflammatory factors related hypotheses,7) epigenetic regulation,8) gut-brain axis modulation9)and other hypotheses till now. However, the specific pathogenesis of depression is still not clear.
Long non-coding RNAs (LncRNAs) are non-coding transcripts with over 200 nucleotides, which are highly expressed in specific brain regions such as the cortex and prefrontal cortex (PFC) of embryos and adult mammals. Recent studies have shown that lncRNAs have not only involved in the regulation of stem cell pluripotency and cancer10) but also involved in the development of the mammalian nervous system to maintain the homeostasis and function of the brain, including the maintenance of neuronal cell self-renewal, synaptic plasticity, synapse formation,11) and memory formation.12,13)
The chronic unpredictable mild stress (CUMS) mice model mimics the variable and unpredictable physiological and psychological stressors encountered in human daily life and produces various behavioral changes.14) These changes are thought to reflect some of the core symptoms of depression (such as weightlessness, anxiety, and hopelessness). It is considered to be an effective rodent model to introduce behavioral and neuronal disturbances evoked by stresses to study the molecular mechanism of depression. As we all know, the PFC and hippocampus (HIP) play important roles in learning, memory, and decision-making.15) The neuronal projections from the HIP to PFC, referred to as the HIP-PFC circuit, participate in cognitive and emotional disturbances in several psychiatric disorders.16) Studies on depression revealed that the PFC and HIP display structural and functional abnormalities in both individuals with depression and rodent depressive animals.17,18) Therefore, much effort is needed to study the candidate causal genes and molecular mechanisms underlying depression in the PFC and HIP regions. As reported, the diagnosis of depression in older people is more difficult than the younger due to its various clinical symptoms and age-related,19) thus reduction of the prevalence and misdiagnosis of geriatric depression partly relies on the dissection of its specific molecular mechanism. In the previous study, we explored the lncRNAs expression profiles at the whole transcriptome level by RNA-seq in the HIP of CUMS depressed mice at 13 months old representing the late life correlated to humans.20) Interestingly, we found that lncRNA Gm16638-201 encoding transcript RP24-252B21.2 is abnormally highly expressed in the HIP of depressed mice and it might regulate the expression of interleukin-16 (IL-16) and other proteins in cis or trans by bioinformatic prediction.20) As lncRNA Gm16638-201 (ENSMUST00000148687.1) is known to be located on chromosome 7 and consists of 4 exons with 1865 bps, but its function has not been reported currently. Therefore, in the present study, we aimed to find out the specific function of Gm16638-201 and the molecular mechanism underlying depression through over-expression of Gm16638-201 in primary cortical neuronal cultures.
The BALB/c male mice (n = 20), 13 months old (weighing 24.8 g on average) representing the late-life of humans according to the mouse lifespan,21) purchased from the Animal Experiment Center of Inner Mongolia Medical University (license number SYXK (Mongolia) 2015-0001) were fed with ad libitum food under the standard conditions at 22 ± 1 °C with 12 h light and dark cycles. After 2 weeks, the mice were randomly divided into two groups and used for further experiments. All the protocols used were approved by the Academic Ethics Committee of Inner Mongolia Medical University (the Experimental Ethics No. YKD2016024) by the Guide for Care and Use of Laboratory Animals.
Induction Protocol of CUMS Depressive MiceTwenty BALB/c male mice were randomly divided into the CUMS (n = 15) and normal control (NOR, n = 5) groups and housed separately. Mice in the CUMS group were subjected to one chronic unpredictable mild stimulation daily, including circadian rhythm reversal, humid environment, deprivation of food and water, noise stimulation, physical restraint, cage tilt, unpredictable shocks, cold water swimming, shaking stress, bedding materials deprivation, tail clipping, and intermittent flash.22) The detailed stimulation protocol was listed in Table 1. After stimulation under all the stresses, the mice in CUMS group were then received one type of variable stimulation randomly daily and lasted for 8 weeks. Finally, the behavioral tests including tail suspension test (TST), sucrose preference test (SPT), forced swimming test (FST),20,23,24) marble-burying test,25) were performed as reported and body weights were measured. After the behavior tests, all the animals were decapitated and their brains were quickly removed, frozen on dry ice, and kept in a freezer at −80 °C for further use.
Stimulus type | Operation method | Duration |
---|---|---|
Circadian rhythm reversal | Close the thick curtains during the day to keep the room dark; Keep the light on at night. | 24 h |
Humid environment | The padding is watered, but there is no ponding. | 12 h |
Deprivation of food and water | Remove the water bottle or food from the cage. | 12 h |
Noise stimulation | Intermittent playback | 30 min |
Physical restraint | The mice were put into the self-made restraint. | 4 h |
Cage tilt | Cage tilt 45° | 12h |
Unpredictable shocks | 5 mA, one shock/5 s, 10 s duration | 5 min |
Cold water swimming | Water temperature is 5. The mice were blotted dry with absorbent paper after swimming. | 5 min |
Shaking stress | Placing the mouse cage onto the shocker shaker with160 rpm. | 1 h |
Bedding materials deprivation | The rat cage bedding was withdrawn. | 12 h |
Tail clipping | A long tail clip clamps the mouse tail. | 1 min |
Intermittent flash | Intermittent glare | 2 s |
Total RNAs were extracted from the PFC of mice in both groups according to the manufacturer's instructions (TransGen, Beijing, China). First-strand cDNA was synthesized with 1 µg of total RNA using Prime-Script™ II cDNA Synthesis Kit and random primers (TaKaRa, Japan). RT was performed according to the manufacturer’s recommended protocols. Semi-quantitative PCR conditions were used as follows: Thirty cycles of denaturation at 94 °C for 30 s, annealing at 55 °C for 30 s, and extension at 72 °C for 1 min. The primers were used for amplification are listed in Table 2. PCR products were run on 2% agarose gels and visualized by UV illumination.
Primer name | Direction | Primer sequences (5′→3′) | PCR product size (bps) |
---|---|---|---|
lncRNA Gm16638-201 | Forward | ATCCAGCAGACAGCACTATG | 298 |
Reverse | GTTGCCTCTGTGTTCAGAAG | ||
Elmo2 | Forward | GCTGGGCTTCACCAACCATA | 258 |
Reverse | ATGGGGTGGTAGTCATTGCG | ||
Satb1 | Forward | AACCGAACTCAGGGATTGCT | 170 |
Reverse | GCTGCGTTCAAGCTCCTTTC | ||
Hnrnpul1 | Forward | TGGATAAGATGCGGGTGATGG | 261 |
Reverse | TTCGCTTTACTGTGCGGTCT | ||
Sipa1l3 | Forward | ACCCCAGACTCCTCCACAAT | 127 |
Reverse | AGCCGTTGGTCATCACCTTT | ||
Mapt | Forward | CCAACTGCTGAAGACGTGAC | 104 |
Reverse | TCGGCTGTAATTCCTTCTGGG | ||
Tada3 | Forward | ATTGTGCCGACATCACCAGT | 240 |
Reverse | GCAGGGCATCCACATCTTTTG | ||
Sgip1 | Forward | TTAACTCTGTCTCAGCAGCCC | 112 |
Reverse | CATTTGGTGCCCCATTGCTT | ||
StarD5 | Forward | GAAGAAGTGTCGGGAAGGCA | 217 |
Reverse | GCTCACACACAGCATATCCG | ||
Gapdh | Forward | AGGTCGGTGTGAACGGATTTG | 123 |
Reverse | TGTAGACCATGTAGTTGAGGTCA |
Quantitative RT-PCR was performed with TB Green® Premix Ex Taq™ II (Tli RNaseH Plus, TaKaRa, Japan) in a 7500 fast real-time PCR cycler (AB instrument, U.S.A.) according to the manufacturer's instructions. The glyceraldehyde-3-phosphate dehydrogenase (Gapdh) gene was used as an internal standard.
Bioinformatic AnalysisThe subcellular localization of Gm16638-201 was predicted by lncLocator (long non-coding RNA subcellular localization predictor, (http://www.csbio.sjtu.edu.cn/bioinf/lncLocator/). We obtained the nucleotide sequence of lncRNA Gm16638-201 (NR_166697.1) from the GenBank (https://www.ncbi.nlm.nih.gov/) and searched the RNALocate database by uploading it. The percentage score for subcellular localization of Gm16638-201 was then calculated and quantified. The target genes regulated by Gm16638-201 in cis or trans were predicted by searching the 10k or 100k upstream and downstream regions of lncRNAs and followed by their function analysis.26) Besides, the expressed Pearson's correlation coefficient between lncRNAs and coding genes with R function “cor.test” was calculated. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses of predicted target genes were performed using the Database for Annotation, Visualization and Integrated Discovery (DAVID) online tool (https://david.ncifcrf.gov/home.jsp). The possible interaction among the predicted target genes was predicted with confidence > 0.4 using the STRING database (https://cn.string-db.org/) and subsequently, the protein–protein interaction (PPI) network was analyzed and visualized using Cytoscape software (https://cytoscape.org/).
Primary Culture of Mouse Cortical NeuronsThe newborn mice (p = 0) were sacrificed and the neuronal cortex was removed carefully. After being scissored in a pre-cold Dulbecco’s modified Eagle’s medium (DMEM) medium, the neuronal cortex was digested in 2 mg/mL of papain (Solarbio, Beijing, China) containing deoxyribonuclease (DNase) at a concentration of 0.05 mg/mL (Solarbio, Beijing, China) and incubated at room temperature for 20–30 min. Then 1 mL of horse serum (Sangon, Shanghai, China) was added to terminate the digestion. The dissociated cells were plated to a 6-well plate precoated with 10% poly-D-lysine (Sangon, Shanghai, China) and then incubated at 37 °C with 5% CO2 for 4 h. After culturing for 4–8 h, the medium was replaced with a freshly made Neurobasal™-A (with 2% synthetic B-27 serum-free additive, 0.5 mmol/L L-glutamine, Thermo Fisher Scientific, U.S.A.) at 37 °C with 5% CO2.
Immunocytochemical StainingThe primary cultured neuronal cortical cells were fixed in 4% paraformaldehyde solution for 20 min and permeabilized with 0.5% TritonX100 for 20 min. Then the cells were incubated with the primary antibody (NF-L, 1 : 50, Proteintech, Wuhan, China) for 1 h at 37 °C. After rinsing with phosphate buffered saline (PBS), the secondary antibody (CY3 goat anti-rabbit, immunoglobulin G (IgG) 1 : 50, TransGen, Beijing, China) was applied for 30 min. Finally, the stained cells were mounted with Fluoroshield™ with 4′-6-diamidino-2-phenylindole (DAPI) mounting medium (Sigma, U.S.A.) and observed under an inverted fluorescence microscope (OLYMPUS IX73, Japan).
Adenoviral Expression Vector Construction and InfectionThe adenoviral recombinant vector of mouse Gm16638-201 transcript was constructed and adenoviral particles were produced by Genechem, Shanghai, China. Briefly, the full-length of mouse Gm16638-201 transcript (NR_166697) was constructed into the AgeI and NheI cloning site of adenoviral vector GV315 which contains enhanced green fluorescent protein (EGFP) tag (Genechem, Shanghai, China) and subsequently sequenced. Then the sequence-verified recombinant vector and packaging vector pBHG lox ΔE1, 3 Cre (Microbix. Canada) were co-transfected into HEK293 cells. After 10–15 d of co-transfection, the adenoviral particles were amplified and purified by Adeno-X™ Virus Purification Kit (BD Biosciences, Clontech) for further infection. At day in vitro (DIV) 2, the adenoviral particles (multiplicity of infection, MOI = 100) were added to infect the primary mouse neuronal cortical cells (Primary-N) at 37 °C, 5% CO2 for 12 h and removed after 4 d. The infection efficiency was observed under an inverted fluorescent microscope (Olympus IX73, Japan). The Primary-N cells infected with the adenoviral particles with Gm16638-201 overexpression were referred to as Gm16638-201-Exp. The Primary-N cells infected with the adenoviral particles with the GV315 vector-only were used as negative control (NC).
Enzyme-Linked Immunosorbent Assay (ELISA)IL-16 levels of the PFC of mice were measured using a standard quantitative sandwich ELISA (MEIMIAN, China). All analyses and calibrations were performed in duplicate. Optical densities (OD) were determined using an absorbance microplate reader (Elx808™ Bio-Tek Instruments, Winooski, VT, U.S.A.) at 450 nm. Gen5 Data Analysis software (Bio-Tek Instruments) was used to analyze all data and depict the standard curve.
Western BlotThe PFC of the mice and primary neuronal cells were harvested and total proteins were isolated. Briefly, the tissues and cells were lysed for 30 min with radio immunoprecipitation assay (RIPA) lysis buffer and centrifuged at 12000 rpm at 4 °C for 5 min. The samples were resolved by 10% sodium dodecylsulfate-polyacrylamide gel electrophoresis (SDS-PAGE), transferred to nitrocellulose membranes, and blotted using primary antibodies (14-3-3Ɛ, 1 : 1000, MAPT, 1 : 1000, BAD, 1 : 1000, Proteintech, Wuhan, China). Anti-GAPDH (1 : 10000) and β-Tubulin (1 : 1000) was used as an internal control. The membranes were washed with Tris Buffered Saline with 0.1% Tween® 20 (TBST) three times and then incubated with the IRDye 800CW goat anti-rabbit secondary antibody (1 : 10000, LICOR, U.S.A.). After stripping, the same membrane was used for the detection of other proteins. The densities of bands were measured using the image analysis software (Image studio ver3.1) of Odyssey CLX, and changes in protein levels were presented as folds of those in the control group.
Statistical AnalysisData were presented as means ± standard error of the mean (S.E.M.) and analyzed using GraphPad Prism 9 software (https://www.graphpad-prism.cn/). The unpaired Student’s t-test was used. The gene expression data were compared by the one-way ANOVA followed by a post-hoc test among three groups. The qRT-PCR results were analyzed by the 2−△△CT quantitative method to determine differences in gene expression. Statistical differences were considered significant at p < 0.05.
To verify the CUMS effectiveness, behavioral tests and body weight measurement were performed on both groups of mice (CUMS and NOR) after the eight weeks’ stimulation. As shown in Fig. 1A, compared with the NOR group, the mice in the CUMS model group showed significant body weight loss (p = 0.0121 < 0.05). The sucrose preference test found that the sucrose intake in the CUMS group was significantly lower than that in the normal group (p < 0.0001, Fig. 1B), indicating that the CUMS-induced mice developed depression-like symptoms. The anxiety behaviors were assessed by TST and FST. Compared with the NOR, the struggling time in the CUMS group was significantly reduced (Fig. 1C, p < 0.0001), and immobility time in the CUMS group was longer (Fig. 1D, p = 0.0027 < 0.01). These results indicated that the mice manifested anxiety and despair after exposure to CUMS. In the marble burying test, as shown in Fig. 1E, the mice in the CUMS group showed lower attention to unfamiliar glass marbles, and the number of marbles buried with bedding was significantly less than that of the mice in the NOR group (p = 0.0488 < 0.05), indicating that the mice showed anxiety-like behavior.
A. Compared with normal mice, the body weight of mice after CUMS stimulation was significantly reduced. B. The preference for glucose in mice induced by CUMS stimulation was significantly lower than that in the normal group; C, D. The period of immobility was significantly increased in TST (C) and FST (D). E. The number of bead-embedded mice after CUMS stimulation was significantly less than that of mice in the normal group. F. Expression of Gm16638-201 in the PFC of mice was significantly increased compared with normal mice (n = 3, p = 0.0071 < 0.01). NOR: the normal mice group, and CUMS: CUMS induced depressive mice group. * means p < 0.05, ** means p < 0.01, **** means p < 0.0001.
Our previous study found that Gm16638-201 was highly expressed in the HIP of CUMS mice,20) and the PFC has emerged as one of the regions most consistently impaired in major depressive disorder (MDD).27) Here we examined the expression of Gm16638-201 in the PFC of CUMS model mice by qPCR. As we expected, the results showed that the expression level of Gm16638-201 was also significantly increased in CUMS mice compared to NOR mice (Fig. 1F, p = 0.0071 < 0.01, Supplementary Fig. A), consistent with that in the HIP of CUMS.
Bioinformatic Analysis of the Subcellular Localization of Gm16638-201 and Functional Annotation of the Predicted Genes Regulated by Gm16638-201 in cis or transAlthough lncRNA does not encode a protein, most of these are the same as mRNA being transcribed, capped, and polyadenylated by RNA polymerase II,28) which indicates that lncRNA is similar to mRNA and will play different functions due to different subcellular localization. On the one hand, lncRNA in the nucleus can regulate the transcriptional program through interaction and remodeling with chromatin,29,30) on the other hand, it is also involved in the maintenance of nuclear chamber spatial structure.31) While lncRNA in cytoplasm not only can compete with endogenous RNA (ceRNA) and miRNA to regulate the translation and stability of mRNA32,33) but also be involved in mediating signal transduction pathway34) and protein post-translational modification.35) Therefore, bioinformatics predicted the subcellular localization of Gm16638-201 manifested that it is mainly located in the cytoplasm, with a small amount located in the nucleus, ribosome, cytoplasm, and exosomes (Fig. 2A). The result suggests that Gm16638-201 which is abnormally highly expressed in PFC, may play an important biological role in the cytoplasm of neuronal cells.
A. The subcellular localization of Gm16638-201 was predicted and analyzed by lncLocator. B. GO enrichment analysis of Gm16638-201 and predicted regulatory genes. The X-axis is the enrichment index, the Y-axis represents the GO classification, dot size represents the number of genes involved. C. PPI network of possible interactions for target genes. Nine genes including Elmo2, Satb1, Hnrnpul1, Sipa1l3, Mapt, Tada3, Sgip1, StarD5 and IL-16 were used for searching the possible interactions in the String database. PPI with confidence >0.4 was selected for further analysis and visualized by Cytoscape. Nodes represent the interacting proteins. Node size represents the PPI degree, and line thickness represents the strength of the data support.
In our previous study, 19 proteins were predicted to be co-localized or co-expressed (in cis or trans acting) with Gm16638-20120) (Table 3). Based on the review of the literature published and bioinformatics analysis, 9 genes (Elmo2, Satb1, Hnrnpul1, Sipa1l3, Mapt, Tada3, Sgip1, StarD5 and IL-16) among them were pointed out to be related to neurobiological processes or pathways and related diseases and selected to further study. GO analyses predicted that these genes are located in the nuclear periphery and intracellular part (Cellular Component, CC) and might participate in the process of response to external stimulus and DNA transcription and regulation (Biological Process, BP), and function in the SH3 domain binding and protein domain specific binding (Molecular Function, MF) (Fig. 2B). KEGG analysis revealed these target genes were in association with 14-3-3Ɛ-mediated neurotrophic signaling pathways (data not shown). Next, we searched the possible interactions among these nine genes in the STRING database and constructed a PPI network, and visualized it by Cytoscape (confidence > 0.4, Fig. 2C). Some of the interactions showed strong PPI evidence, such as Mapt and 14-3-3Ɛ, Mapt and Sgip1. Moreover, PPI network prediction revealed that 14-3-3Ɛ (encodes the tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein epsilon polypeptide/ the 14-3-3 epsilon protein, 14-3-3Ɛ) can interact with the nine target genes regulated by Gm16638-201 and play a central role in the interaction network. Several studies showed that 14-3-3Ɛ can interact with Microtubule-associated protein tau (MAPT) and BAD, and play an important role in neurobiological processes and pathways.36,37) Overall, these results indicated that Gm16638-201 might regulate target proteins, such as MAPT, and affect the 14-3-3Ɛ neurotrophic pathway in depression.
Ensembl transcript ID | Gene symbol | Gene description | In cis or trans-acting | Pearson-correlation | p-Value |
---|---|---|---|---|---|
ENSMUST00000075249 | Plekhg3 | Pleckstrin homology domain containing, family G (with RhoGef domain) member 3 | In trans | 0.97638511 | 5.56E-08 |
ENSMUST00000163147 | Epb41l5 | Erythrocyte membrane protein band 4.1 like 5 | In trans | 0.964954921 | 3.93E-07 |
ENSMUST00000225991 | Fam149b | Family with sequence similarity 149, member B | In trans | 0.977382285 | 4.49E-08 |
ENSMUST00000138016 | Mgat4c | MGAT4 family, member C | In trans | 0.977947447 | 3.96E-08 |
ENSMUST00000095585 | Adgrv1 | Adhesion G protein-coupled receptor V1 | In trans | 0.958358076 | 9.19E-07 |
ENSMUST00000064376 | Arhgap4 | Rho GTPase activating protein 4 | In trans | 0.986833555 | 3.05E-09 |
ENSMUST00000063869 | Cd1d1 | CD1d1 antigen | In trans | 0.965107126 | 3.84E-07 |
ENSMUST00000138315 | Acsbg1 | Acyl-CoA synthetase bubblegum family member 1 | In trans | 0.970712439 | 1.62E-07 |
ENSMUST00000164012 | Crhr2 | Corticotropin releasing hormone receptor 2 | In trans | 0.955207694 | 1.32E-06 |
ENSMUST00000071699 | Elmo2 | Engulfment and cell motility 2 | In trans | 0.990420307 | 6.25E-10 |
ENSMUST00000144331 | Satb1 | Special AT-rich sequence binding protein 1 | In trans | 0.992934348 | 1.37E-10 |
ENSMUST00000043765 | Hnrnpul1 | Heterogeneous nuclear ribonucleoprotein U-like 1 | In trans | 0.960188721 | 7.37E-07 |
ENSMUST00000182780 | Sipa1l3 | Signal-induced proliferation-associated 1-like protein 3 | In trans | 0.979684218 | 2.63E-08 |
ENSMUST00000145227 | Mapt | Microtubule-associated protein tau | In trans | 0.970206954 | 1.76E-07 |
ENSMUST00000099118 | Tada3 | Transcriptional adapter 3 | In trans | 0.952695079 | 1.72E-06 |
ENSMUST00000072481 | Sgip1 | SH3-domain GRB2-like (endophilin) interacting protein 1 | In trans | 0.953008545 | 1.67E-06 |
ENSMUST00000075418 | StarD5 | StAR-related lipid transfer (START) domain containing 5 | In cis | — | — |
ENSMUST00000001792 | IL-16 | Interleukin 16 | In cis | — | — |
ENSMUST00000039317 | Tmc3 | Transmembrane channel-like gene family 3 | In cis | — | — |
To further understand the role of the high expression of Gm16638-201 in the PFC of CUMS depressive mice, we cultured the murine primary neuronal cortical cells (Fig. 3A). Immunocytochemical staining showed the neuronal-specific gene neurofilament-L (NF-L)38) was expressed in over 90% of cultured cells and glial fibrillary acidic protein (GFAP)-positive cells were rarely observed due to limited cultured cells (Fig. 3B, Supplementary Fig. B). Therefore, this result indicated that primary neuronal cultures of the murine cortex were successfully obtained. The primary neuronal cells were then infected with the adenovirus carrying Gm16638-201 recombinant plasmid (Fig. 4C). Subsequently, the expression level of Gm16638-201 was examined by RT-PCR and it was significantly increased (Fig. 3C, F(2, 6) = 1204, p < 0.0001).
A. Primary culture of the murine cortical neuron cells, scale bar is 100 µM. B. Immunocytochemical staining of NF-L in the primary cortical neurons. Scale bar: 10 µM. C. Overexpression of Gm16638-201 by adenovirus infection of the primary prefrontal cortical neurons. Scale bar: 100 µM. BF, bright field; EGFP, enhanced GFP protein tag. D. Gm16638-201 was successfully overexpressed in the primary cells of prefrontal cortical neurons compared with the negative control group (F(2, 6) = 1204, p < 0.0001). ### means p < 0.001, **** means p < 0.0001. Primary-N, the primary cultured prefrontal cortex neuron cells, NC, the negative control group without Gm16638-201 expression, and Gm16638-201-Exp, Gm16638-201 over-expressed Primary-N cells.
A. Gm16638-201 and predicted target genes were all expressed in mouse PFC. B. Elmo2, Satb1, Hnrnpul1, Sipa1l3, Mapt, Tada3, and StarD5 except Sgip1 were all expressed and downregulated after overexpression of Gm16638-201 in Prime-N cells (F(2,6) = 78.89, F(2,6) = 78.89, F(2,6) = 56.15, F(2,6) = 84.06, F(2,6) = 65.35, F(2,6) = 159.9, F(2,6) = 72.27, p < 0.01). C and D. IL-16 expression was increased in CUMS group and Prime-N cells overexpressed by Gm16638-201, relative to NOR and Prime-N control cells, respectively (p < 0.01). # means p < 0.05, ## means p < 0.01, ### means p < 0.001, #### means p < 0.0001, ** p < 0.01, *** p < 0.001, **** p < 0.0001.
Nine genes (Elmo2, Satb1, Hnrnpul1, Sipa1l3, Mapt, Tada3, Sgip1, StarD5, IL-16) were predicted to be regulated by Gm16638-201 in cis or trans and implicated in neuronal development. Firstly, we examined the expression of 9 target genes and found that these 9 genes were all expressed in mouse PFC (Fig. 4A). Due to the tissue amount limitation, we secondly determined the expressions of these genes in primary neuronal cells with overexpression of Gm16638-201(Gm16638-201-Exp) which mimics its high expression in the PFC of CUMS by semi-quantitative RT-PCR. The results showed that the expression levels of Elmo2, Satb1, Hnrnpul1, Sipa1l3, Mapt, Tada3, and StarD5 except for Sgip1 were all down-regulated in the Gm16638-201-Exp cells compared with Primary-N and NC groups (Fig. 4B, F(2,6) = 78.89, F(2,6) = 78.89, F(2,6) = 56.15, F(2,6) = 84.06, F(2,6) = 65.35, F(2,6) = 159.9, F(2,6) = 72.27, p < 0.01). Cytokines including interleukins (IL), lymphokines, interferons (IFN), chemokines, or tumor necrosis factors (TNF) have modulatory effects on various psychiatric disorders including schizophrenia, depression, and Alzheimer’s disease.39) In our previous study, IL-16 was predicted to be regulated by Gm16638-201 in cis in CUMS mice. In addition, markedly elevated levels of IL-16 expression were observed in mental illnesses such as MDD, bipolar disorder (BP), and schizophrenia.40,41) Therefore, we investigated the IL-16 expression levels in the PFC of CUMS depressed mice and the Gm16638-201 over-expressed primary neuronal cells by ELISA. As a result, IL-16 expression was elevated in both tissue and cells (Figs. 4C, D, p = 0.0095 < 0.01; F(2,6) = 68.09, p < 0.01).
The Gm16638-201 Induces Mouse Depression by Inhibiting 14-3-3Ɛ-Mediated Signaling in the PFCTo further clarify the mechanism of Gm16638-201 in depression, we examined the expression of 14-3-3Ɛ and MAPT in the Gm16638-201 over-expressed primary neuronal cells by the immunocytochemical staining. We found that 14-3-3Ɛ was expressed in both the nucleus and cytoplasm of primary neuronal cells (Supplementary Fig. C). Interestingly, when Gm16638-201 was overexpressed, the expression of 14-3-3Ɛ in the cytoplasm was dramatically decreased (Supplementary Fig. C), while not in the nucleus. This result indicated that Gm16638-201 could inhibit 14-3-3Ɛ expression in the cytoplasm of the neuronal cells, and demonstrated that Gm16638-201 mainly functions in the cytoplasm of neurons. We also detected the expression of 14-3-3Ɛ and MAPT in the PFC of CUMS mice. As shown in Fig. 5A, the expression of Mapt (also known as Tau) was down-regulated in the PFC of CUMS mice compared to NOR mice (p = 0.0023 < 0.05), and the expression of 14-3-3Ɛ was not statistically changed (p = 0.9881 < 0.05) at the transcript level. Furthermore, we examined the 14-3-3Ɛ and MAPT expressions at the translation level and found that they were all down-regulated in the PFC of CUMS mice (Fig. 5B, p = 0.0112 < 0.05; p = 0.0345 < 0.05). Consistently, their expression was also decreased in the primary cortical neurons with overexpression of Gm16638-201 (Fig. 5C, F(2,6) = 57.08, F(2,6) = 14.96, p < 0.01). In addition, we also checked the expression of BAD, a proapoptotic factor in the PFC of CUMS mice by Western blot since BAD could bind to 14-3-3Ɛ in the ventrolateral orbital cortex (VLO).36) As a result, we found that BAD was also down-regulated in both the PFC of CUMS mice (Fig. 5B, p = 0.0218 < 0.05) and the primary cortical neurons with overexpression of Gm16638-201 (Fig. 5C, F(2,6) = 63.25, p < 0.0001). These results indicated that the elevated expression of Gm16638-201 inhibited the activity of the 14-3-3Ɛ signaling pathway in the PFC and affected depression.
A. The expression of Mapt was down-regulated in the PFC of CUMS mice compared to the NOR group (p = 0.0023 < 0.05). The expression of 14-3-3Ɛ was not statistically changed in the PFC of CUMS mice compared to the NOR group (p = 0.9881 > 0.05). B. Western blot showed that the expressions of 14-3-3Ɛ, MAPT, and BAD were decreased in the PFC of CUMS depressed mice (n = 3, p = 0.0112 < 0.05; p = 0.0345 < 0.05; p = 0.0218 < 0.05). C. The expression of 14-3-3Ɛ, MAPT, and BAD is reduced in primary cortical neuronal cells after overexpression of Gm16638-201 by Western blot analysis (F(2,6) = 57.08, F(2,6) = 14.96, F(2,6) = 63.25, p < 0.01). ## means p < 0.01, ### means p < 0.001, #### means p < 0.0001, * p < 0.05, ** p < 0.01, *** p < 0.001.
In this study, we used CUSM depressed mice and primary neuronal cells to investigate the molecular mechanism by which Gm16638-201 regulates PFC target genes and affects the occurrence of depression. Due to the dysfunction of the PFC in depression, qRT-PCR results showed that the Gm16638-201 was expressed highly in the PFC compared with normal mice. We also cultured the primary prefrontal cortical neuronal cells in vitro and over-expressed Gm16638-201 using an adenovirus-mediated expression system to study its function. At present, there is no relevant functional annotation for this lncRNA, and this study will provide experimental evidence for the functional annotation of Gm16638-201 in depression.
Bioinformatic analysis identified 19 possible genes regulated by Gm16638-201, and among them, nine genes were reported to be related to neurological processes or diseases. Therefore, we investigated these genes’ expression in the primary cultured neurons in that Gm16638-201 was over-expressed. Interestingly, the expression levels of these genes (Elmo2, Satb1, Hnrnpul1, Sipa1l3, Mapt, Tada3, and StarD5), except Sgip1, were found down-regulated while Gm16638-201 was highly expressed.
The down-regulated genes involve phagocytosis and cell motility protein 2 (Engulfment and cell motility protein 2, Elmo2) which can regulate the stability of microtubule dynamics by interacting with the RHO-related GTP-binding protein RhoG and the integrin-linked protein kinase ILK.42) This result suggests that Gm16638-201 may affect the stability of microtubules by reducing the expression of Elmo2, and then leading to the occurrence of depression. Another down-regulated gene is Stab1. Stabilin-1 (Stab1) can be regulated by lncRNA nuclear-enriched transcript 1 (NEAT1) and its high expression can promote Schwann cells proliferation and migration in the peripheral nervous system, enabling axonal growth and functional recovery after peripheral nerve injury.43) The expression of heterogeneous nuclear ribonucleoprotein U-like protein 1 (Hnrnpul1) was found reduced in Alzheimer’s disease,44) but its specific function has not been reported yet. Signal-induced proliferation-associated 1-like protein 3 (Sipa1l3) is one of the most abundant Rap GTPase-activating proteins (Gap GAPs) in the brain, which tightly regulates Rap signaling in postsynaptic neurons, and its C-terminus is required for postsynaptic localization.45) This study found that the expression of Sipa1l3 was inhibited when the Gm16638-201 was overexpressed in the PFC, suggesting that the Gm16638-201 may affect the signaling of postsynaptic neurons by inhibiting the expression of Sipa1l3. Transcriptional adapter 3 (Tada3) is a component of the Spt-ada-Gcn5 acetyltransferase (SAGA) complex that links the nuclear transcription of histone acetylation, which regulates the transcription of nearly all genes in human cells.46) The report from Galvan et al. has shown that Tada3 interacts with doublecortin-like kinase 3 (DCLK3) in striatal neurons and plays an important role in transcriptional regulation and/or chromatin remodeling.47) In our study, the expression of Tada3 decreased when Gm16638-201 was overexpressed, suggesting that Gm16638-201 might play an important role in gene transcription and chromatin remodeling. Taken together, our findings suggested that increased expression of Gm16638-201 in the PFC may affect neuronal cell microtubule stability, neuronal cell growth, synaptic signaling, and gene transcription and chromatin remodeling. However, further research is needed to be done to clarify the specific mechanism of lncRNA modulation in depression.
GO and KEGG pathway analysis of the 7 differentially expressed genes revealed that they were related to the 14-3-3Ɛ-mediated neurotrophin signaling pathway. 14-3-3Ɛ is a highly conserved, multifunctional protein that is most abundantly expressed in the brain, regulating cell cycle, apoptosis and autophagy by binding to various signaling proteins such as kinases and transcription factors. For example, it can interact with BAD to rescue the BAD-mediated apoptosis to prevent depression caused by chronic stress.36,48–50) In addition, 14-3-3Ɛ binds to MAPT specifically and non-specifically to regulate the aggregation of MAPT.37) MAPT not only promotes the assembly and stability of microtubules but also participates in the establishment and maintenance of neuronal polarity in the central nervous system.51) In addition, chronic stress can make pro-inflammatory cytokines such as TNF-α and IL-1β, IL-6, IL-16, etc. release, leading to the occurrence of depression.7,52) Our previous study predicted that the transcript RP24-252B21.2 of Gm16638-201 could cis-regulate IL-16 and trans-regulate Mapt expression. Therefore, we examined the expression changes of IL16, MAPT, 14-3-3Ɛ, and BAD proteins and found that compared with normal mice and Prime-N cells, the expression of IL16 was increased whereas MAPT 14-3-3Ɛ and BAD proteins expression was decreased in both CUMS depressed mice and the Prime-N cells overexpressed with Gm16638-201. Our findings suggested that the up-regulated Gm16638-201could lead to the release of IL-16 and promote an inflammatory response.
In summary, our study has found that Gm16638-201 was up-regulated in the PFC of CUMS mice and its overexpression could inhibit 14-3-3Ɛ signal pathway activity by regulating the expression of Elmo2, Satb1, Hnrnpul1, Sipa1l3, Mapt, Tada3, StarD5 and IL-16 genes in the PFC and induced depression. The specific function of Gm16638-201 and its molecular mechanism underlying depression need to be explored in future studies.
We thank all the technicians of the Key Laboratory of Molecular Pathology, Inner Mongolia Medical University, China for their detailed guidance on the experiment conduction process.
This work was supported by Scientific Research Projects of Universities in Inner Mongolia Autonomous Region (NJZY111) and Natural Scientific Research Projects of Inner Mongolia Autonomous Region (2020MS03060).
The authors declare no conflict of interest.
This article contains supplementary materials.