Identification and bioinformatic analysis of dysregulated microRNAs in human oligodendroglial cells infected with borna disease virus

  • Authors:
    • Hong Zhang
    • Peng He
    • Rongzhong Huang
    • Lin Sun
    • Siwen Liu
    • Jingjing Zhou
    • Yujie Guo
    • Deyu Yang
    • Peng Xie
  • View Affiliations

  • Published online on: October 13, 2016     https://doi.org/10.3892/mmr.2016.5842
  • Pages: 4715-4722
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

MicroRNAs (miRNAs) are recognized as important regulators of gene expression via translational depression or mRNA degradation. Previously, dysregulated miRNAs have been found in neurodegenerative and neuropsychiatric disorders. Borna disease virus (BDV) is a neurotropic, negative single‑stranded RNA virus, which may be a cause of human neuropsychiatric disease. BDV is regarded as an ideal model to analyze the molecular mechanisms of mental disorders caused by viral infection. In the present study, 10 miRNAs were dysregulated in human oligodendrocytes (OL cells) infected with the BDV strain, Hu‑H1 (OL/BDV). The predicted target genes of those different miRNAs were closely associated with DNA binding, receptor activity, cytoplasm and membrane, biopolymer metabolic process and signal transduction, which were ranked highest using Gene Ontology (GO) analysis, and were predominantly involved in ‘Immune system and adaptive Immune system pathways’ on pathway analysis. Reverse transcription‑quantitative polymerase chain reaction analysis confirmed that seven miRNAs (miR‑1290, miR‑1908, miR‑146a‑5p, miR‑424‑5p, miR‑3676‑3p, miR‑296‑3p and miR‑7‑5p) were significantly downregulated in the OL/BDV cells, whereas two miRNAs (miR‑1244 and miR‑4521) showed no significant differences between the two groups. The present study revealed for the first time, to the best of our knowledge, the miRNA profile of BDV Hu‑H1‑infected human OL cells. Based on GO and pathway analyses, further investigation of the signaling processes in BDV‑infected oligodendrocytes may offer particular promise in improving understanding of the neuropathogenesis of BDV.

Introduction

Borna disease virus (BDV), a neurotropic, non-cytolytic, non-segmented RNA virus, is an enveloped virus of ~8.9 kb with six open reading frames (1,2), which infects a wide variety of mammalian species, including horses, sheep and dogs (3). BDV has been widely investigated in neuroscientific fields on account of its numerous unique attributes causing neurobehavioral diseases (4) and the ability to introduce its RNA transcripts into host genomes (5). Previous epidemiological studies have shown that there may be a latent association between BDV infection and human neuropsychiatric diseases (6), encephalitis and other brain diseases (3,712). In our previous study, BDV infection was reported in Chinese neuropsychiatric patients and health care professionals (13,14), which supported the hypothesis that BDV can infect humans and may be a pathogen in certain mental disorders, although the underlying molecular mechanism remains to be fully elucidated. However, certain studies have found no direct evidence of BDV infection in schizophrenia, bipolar disorder or major depressive disorder (1517). The controversy requires resolution prior to use as a diagnostic method to ensure reliability. BDV-associated functional disturbances of neuron and glial cells have been evidenced (1821) and its potential effects cannot be ignored.

MicroRNAs (miRNAs) are small (~22 nucleotides in length), non-coding, single-stranded RNAs (22). They regulate gene expression by binding to the complementary sequence in the 3′-untranslated region (3′-UTR) of target mRNAs, resulting in inhibited protein synthesis or destabilizing of target mRNA translation at the post-transcriptional level. miRNAs have been shown to be pervasive in several biological processes, including cell death, cell proliferation, the function of immune cells, hematopoiesis and patterning of the nervous system (23). A wide range of studies have revealed that miRNAs are associated with several human diseases, including cancer, chronic inflammation and viral diseases (2426). Previous studies have also indicated that miRNAs have an effect on neurodegenerative and neuropsychiatric disorders (2730), which suggests the possibility to associate the mechanisms of BDV infection with miRNA dysregulation.

In the present study, miRNA arrays were used to identify the differences in miRNA expression between oligodendrocytes (OL cells) infected with the Hu-H1 BDV strain (OL/BDV cells) and non-infected OL cells. The differentially expressed miRNAs were then bioinformatically analyzed using Gene Ontology (GO) and pathway analyses, in order to determine their biological function and localization. Finally, reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis was performed to validate the expression of the differentially expressed miRNAs. The aim of the present study was to determine which miRNAs are dysregualted in OL/BDV cells, and to facilitate further investigation of the role of miRNAs in BDV infection.

Materials and methods

Cell line and preparation of BDV Hu-H1 strain solution

The BDV Hu-H1 strain (passages 75–76 in OL cells), originally isolated from PBMCs of a patient with bipolar disorder, and a human fetal-derived OL cell line, were provided by Professor Hanns Ludwig (Free University of Berlin, Berlin, Germany) (31). Dulbecco's modified Eagle's medium (DMEM), fetal bovine serum (FBS), penicillin-streptomycin solution, phosphate-buffered saline (PBS), 0.25% trypsin-EDTA and L-glutamine were purchased from GE Healthcare Life Sciences (Logan, UT, USA). The human OL cell line infected with the BDV Hu-H1 strain was cultured with DMEM in 10% FBS and 100 U/ml penicillin/streptomycin in a humidified incubator (5% CO2; 37°C). The preparation and viral titration of the BDV Hu-H1 solution were performed, as described previously (32). The cells in 20 10-cm dishes (density, 107) were washed twice with PBS, and 1 ml fresh growth medium was added when the cells in the dishes reached 90% confluence. The cell solution was then frozen (−80°C) and thawed (25°C) for 15 min, and repeated three times. The lysate was then centrifuged at 3,000 × g for 10 min at room temperature. The resulting supernatant, which contained infectious viral particles, was used as the stock viral solution.

The OL cells were seeded into 96-well plates (3×104 cells/well). At 8 h post-adherence, the medium was removed and 100 µl viral solutions were added to each well. The stock viral solution was serially diluted 10-fold five times, with four replicates for each concentration. The cells were cultured for 7 days in DMEM/2% FBS, during which the cell medium was replaced once every 2 days to maintain the extracellular environment. The viral titration was assessed using immunohistochemistry. The BDV-infected OL cells were fixed in 96-well plates for 30 min at room temperature with 4% paraformaldehyde, followed by permeabilization for 10 min in 0.25% Triton X-100. The cells were then rinsed three times with PBS (5 min each time) and blocked with 5% (w/v) skimmed milk solution for 1 h at 37°C. The cells were then incubated overnight with mouse anti-BDV-specific nuclear-protein (p40) antigen primary monoclonal antibody (provided by Professor Ludwig Hanns, 1:1,000 diluted with PBS) (33) at 4°C, followed by incubation for 1 h with secondary goat anti-mouse antibody (cat. no. A0216; 1:5,000; Beyotime Institute of Biotechnogy, Shanghai, China) at room temperature. Immunofluorescence was detected using a phase-contrast microscope following three PBS washes (32).

BDV infection of OL cells

A total of 105 non-infected OL cells were seeded into four separate 6-well plates (total 24 wells) with 10% FBS in DMEM. Half of these wells were infected with Hu-H1 stock solution (as above) at a multiplicity of infection of 1.0. Specifically, following adherence of the OL cells, the medium was removed, and 150 µl Hu-H1 strain solution was added per well to produce BDV-infected OL (OL/BDV) cells. The cells were stored in a humidified incubator (5% CO2 at 37°C) for 1.5–2 h. Next, the excess viral solution was removed by suction at the edge of the plated, and the cells were cultured in fresh medium. The remaining 12 wells of OL cells were maintained as non-infected control OL cells. The two cell groups were incubated under the same conditions for the remainder of the experiment. The BDV infection was detected and observed using an immunouorescence assay, as described previously (32,34).

miRNA arrays

On day 14 post-infection, six wells of the OL and OL/BDV cells were used for miRNA arrays, respectively. Fluorescent miRNA targets were prepared from 1 or 2.5 µg total RNA samples, which were extracted from the OL/BDV and non-infected OL cells using an OneArray® Amino Allyl miRNA Amplification kit (Phalanx Biotech Group, Hsinchu, Taiwan) and Cy5 dyes (GE Healthcare, Piscataway, NJ, USA). Fluorescent targets were hybridized to the Human Whole Genome OneArray® using a Phalanx hybridization buffer on the Phalanx miRNA OneArray® Hybridization system (Phalanx Biotech Group). Following 16 h of hybridization at 50°C, non-specific binding targets were removed through three washing steps (42°C for 5 min, 42°C for 5 min and 25°C for 5 min, followed by rinsing 20 times), and the slides were dried by centrifugation at 1,000 × g for 3 min at room temperature and scanned using an Axon 4000B scanner (Molecular Devices LLC, Sunnyvale, CA, USA). The intensities of each probe were obtained using GenePix 4.1 software (Molecular Devices LLC). The probes with a log2 ratio ≥0.58 or ≤-0.58, and P<0.05 were defined as differential genes for further pathway enrichment analysis. Each experiment was repeated three times.

Prediction of target genes and bioinformatic analysis

GO analysis (www.geneontology.org) was applied to determine the functions of the intersecting genes on the basis of molecular function, cellular component and biological process. To ensure understanding of the gene expression information, the pathways (www.reactome.org/ReactomeGWT/entrypoint.html and www.genome.jp/kegg/) of the target genes of the different miRNA were also analyzed.

RT-qPCR analysis

On day 14 post-infection, the remaining six wells of OL and OL/BDV cells were used for RT-qPCR assays, respectively. Total RNA was extracted using the miRNEasy Mini kit (cat. no.217004; Qiagen, Hilden, Germany). RT-qPCR was performed on a Corbett Research Rotor-Gene 6000 thermocycler (Corbett Life sciences, Sydney, Australia). The All-in-OneTM First-Strand cDNA Synthesis kit (cat. no. AOPT-0020) and All-in-One™ miRNA qPCR Detection kit (cat. no. AOPR-0200) was purchased from GeneCopoeia, Inc. (GeneCopoeia Inc., MD, USA). All 10 pairs of miRNA primers and the U6 primers for the RT-qPCR were purchased from GeneCopoeia, Inc. (GeneCopoeia, Inc. Guangzhou, China). Briefly, the volume used for RT was 25 µl, comprising 5 µl 5X RT buffer, 1 µl 2.5 U/µl PolyA polymerase, 1 µl RTase Mix, 2,000 ng total RNA template and RNase-free water. RT was performed using a Gene Amp PCR system 9700 (Applied Biosystems Life Technologies; Thermo Fisher Scientific, Inc., Waltham, MA, USA) at 37°C for 60 min and 85°C for 5 min.

The RT-qPCR assays used a total volume of 20 µl, according to protocol, comprising 10 µl 2X All-in-One qPCR mix, 2 µl All-in-OneTM miRNAqPCR primer (2 µM), 2 µl Universal Adaptor PCR primer (2 µM) and 2 µl cDNA (diluted 1:5). All reactions were run in a Corbett Research Rotor-Gene 6000 thermocycler for 40 cycles, which consisted of 94°C for 5 min, followed by 35 cycles of 94°C for 30 sec, 58°C for 30 sec and 68°C for 30 sec. The melting analysis of the PCR products was performed as follows: Temperature was increased between 50 and 99°C (1°C increase at each step), with a 90 sec period of pre-melt conditioning in the first step, and 5 sec for each subsequent step. Each experiment was repeated three times. Values were normalized against the expression levels of U6, and ΔΔCq values were calculated. The relative abundance of each miRNA was calculated using the 2−ΔΔCq method (3537).

Statistical analysis

For all miRNA quantification experiments, quantification cycle (Cq) values >35 were excluded. Values were normalized against the expression levels of U6, and ΔΔCq values were calculated. Statistical analysis was performed using SPSS 19.9 software (IBM SPSS, Armonk, NY, USA). Student's t-test was used to analyze the differences in miRNA expression between the OL/BDV and non-infected OL cells. P<0.05 was considered to indicate a statistically significant difference. All experiments were repeated at least three times.

Results

miRNA expression profiling

To evaluate the different miRNAs between the OL/BDV cells and non-infected OL cells, the present study profiled the expression levels of 657 miRNAs in the two groups using an miRNA array. Compared with the non-infected OL cells, a total of 10 miRNAs were differentially expressed in the OL/BDV cells (four upregulated; six downregulated). miR-146a-5p showed the highest expression level in the OL/BDV cells, whereas miR-4521 and miR-3676-3p showed the lowest expression levels (Fig. 1).

GO and pathway analyses of miRNA target genes

The target genes of the differentially expressed miRNAs were predicted using the online database (http://targetscan.org/ and http://www.mirbase.org/), and then submitted for GO functional classification and pathway analysis (http://www.broadinstitute.org/gsea/msigdb/annotate.jsp). The molecular functions of the target genes were predominantly associated with ‘DNA binding and receptor activity’, and involved in certain biological process, including ‘biopolymer metabolic process’ and ‘signal transduction’ (Fig. 2A-C; Table I). In terms of pathway analysis, the immune system and adaptive immune system were the most significant pathways (Fig. 2D; Table I).

Table I.

Gene Ontology analyses and pathway analyses of microRNA target genes.

Table I.

Gene Ontology analyses and pathway analyses of microRNA target genes.

Gene functionGenes (n)P-value
Molecular function
  DNA binding6023.53E-13
  Receptor activity5838.94E-14
  Transferase activity, transferring phosphorus-containing groups4246.75E-21
  Transmembrane receptor activity4191.34E-08
  Substrate-specific transporter activity3921.22E-05
Cellular component
  Cytoplasm2,1311.88E-43
  Membrane1,9949.85E-30
  Membrane part1,6706.37E-25
  Nucleus1,4301.37E-25
  Plasma membrane1,4266.07E-21
Biological process
  Biopolymer metabolic process1,6841.98E-40
  Signal transduction1,6341.46E-36
  Nucleobase, nucleoside, nucleotide and nucleic acid metabolic process1,2445.47E-21
  Protein metabolic process1,2311.89E-32
  Cellular macromolecule metabolic process1,1316.78E-31
Pathway analyses
  Immune system9335.06E-16
  Adaptive immune system5391.03E-14
  Metabolism of lipids and lipoproteins4782.37E-11
  Hemostasis4663.29E-11
  Developmental biology3961.52E-17
Validation of differential miRNA expression using RT-qPCR analysis

RT-qPCR analysis was performed to validate the expression levels of the 10 differentially expressed miRNAs in the OL/BDV cells. The relative expression levels of miRNAs were normalized against the expression levels of U6. Of the 10 miRNAs, seven exhibited significantly lower levels of expression in the OL/BDV cells: miR-1908, miR-3676-3p, miR-296-3p, miR-146a-5p, miR-1290, miR-424-5p and miR-7-5p (Fig. 3). No differences in expression were found in miR-1244 or miR-4521 between the two groups (Fig. 3), and only one miRNA (miR-4433-3p) was undetected in the RT-qPCR analysis. Of note, RT-qPCR showed that the expression of miR-146a-5p, which was upregulated in the miRNA array, was downregulated in the OL/BDV cells.

Discussion

BDV is a neurotropic virus, which can cause central nervous system dysfunction in several mammalian species, including humans (3,36). BDV Hu-H1, originally derived from a human bipolar patient (31), can induce apoptosis and metabolic dysfunction in human OL cells in vitro (32,38). Proteomic analyses have indicated that BDV Hu-H1 can activate the downstream extracellular signal-regulated kinase (ERK)-ribosomal S6 kinase complex of the Raf/mitogen-activated protein kinase (MAPK) kinase/ERK signaling cascade in human OL cells (39). Additionally, BDV Hu-H1 can result in brain metabolic dysfunction in Sprague-Dawley rats (40). Despite these findings, the mechanisms underlying BDV Hu-H1 infection in the human brain remain to be fully elucidated. miRNAs may provide a novel approach to addressing remaining question. Thus, the present study profiled and analyzed miRNA expression in BDV Hu-H1-infected human OL cells.

An miRNA array is a high throughput and versatile screening tool for analyzing the expression of miRNA, however, its false positives cannot be ignored. Therefore, microarray data requires validation using RT-qPCR analysis, which has higher specificity and provides reliable quantity. It was reported in previous studies that preliminary results were inconsistent with the results of RT-qPCR analysis (41,42). Similarly, in the present study, miR-146a-5p, miR-1290, miR-1908 and miR-424-5p showed downregulation in expression levels using RT-qPCR analysis, which was inconsistent with the results of the miRNA array. In addition, miR-4433-3p was not detected using RT-qPCR analysis, and no differences were found in the expression of miR-1244 or miR-4521 between the BDV-infected and non-infected OL cells. However, three consistently downregulated miRNAs were found: miR-7-5p, miR-296-3p and miR-3676-3p.

In the dysregulated miRNAs, the present study focused on miR-7-5p, miR-424-5p and miR-296-3p, which are closely associated with neural cell proliferation and apoptosis. miR-7-5p has been reported in a wide range of signaling pathways, including the MAPK and phosphoinositide 3-kinase (PI3K)/Akt pathway (43,44). miR-7-5p can inhibit vascular endothelial cell proliferation via directly targeting the 3′-UTR of RAF1, an upstream element of the Ras-Raf-MAPK pathway, which has a key effect on nervous system function. However, miR-7-5p is frequently downregulated in glioblastoma microvasculature (43). miR-7 can efficiently affect cell proliferation and metastasis in hepatocellular carcinoma, through regulation of the PI3K/AKT pathway by suppressing PIK3CD, mammalian target of rapamycin and p70S6 K (44). In addition, miRNA-7-5p can inhibit melanoma cell migration and invasion by regulating insulin receptor substrate-2 (45). The functions of miR-7-5p have also been reported in other types of cancer by targeting different signaling pathways (4650). These results provide novel information for further investigating the function of miR-7-5p in BDV-infected nervous cells.

miR-424-5p is upregulated and modulates the ERK1/2 signaling pathway by targeting suppressor of cytokine induced signaling 6 in pancreatic cancer (51). However, its downregulation can lead to the progression of liver cancer, and regulate the resistance to anoikis and epithelial mesenchymal transition during the metastatic process of hepatocellular carcinoma cells by targeting inhibitor of β-catenin and T cell factor (52). In addition, previous studies have shown that activation of the ERK1/2 pathway may hinder nerve growth factor-induced cell differentiation in BDV-infected PC12 cells (19). Therefore, whether dysregulated miR-424-5p affects BDV-infected nervous cells through the ERK1/2 signaling pathway requires further investigation. Although reports of miR-296-3p are limited, miR-296-3p has been found to regulate cell growth by targeting the potassium channel, EAG1 (53), in human glioblastoma, which presents a novel view in understanding the pathogenic mechanisms of BDV infection.

Previous studies have reported that human OL cells infected with the BDV H1766 strain, isolated from a horse, revealed downregulated expression levels of miR-122 and miR-155 (54,55), which differed from the results of the BDV Hu-H1 infected and non-infected OL cells in the present study. Of note, miRNA expression profiling may be unique due to the possible divergent mechanisms of human OL cells infected with different BDV strains; this requires further investigation.

In conclusion, the present study screened 10 of 657 dysregulated miRNAs in BDV Hu-H1-infected human OL cells using an miRNA array. Of the 10 miRNAs validated using RT-qPCR, the expression levels of seven were significantly downregulated: miR-1908, miR-3676-3p, miR-296-3p, miR-146a-5p, miR-424-5p, miR-7-5p and miR-1290. The biological functions of the dysregulated miRNAs require further investigation, however, based on GO and pathway analyses, further investigation of BDV-infected OL cells offers promise in improving current understanding of the neuropathogenesis of BDV.

Acknowledgements

This study was supported by the National Natural Science Foundation of China (grant no. 31300137), the National Key Scientific Program of China (grant no. 2009CB918300) and the Medical Scientific Research Project of Chongqing Health Bureau (grant no 20142022). The authors would like to thank Professor Liv Bode and Professor Hanns Ludwig of the Robert Koch Institute and the Institute of Virology at the Free University of Berlin for providing the human OL cell line and the BDV Hu-H1 strain used in the present study. The authors would also like to thank the scientific editors at Impactys (www.impactys.com) for their editing and proofreading services.

References

1 

Briese T, Schneemann A, Lewis AJ, Park YS, Kim S, Ludwig H and Lipkin WI: Genomic organization of Borna disease virus. Proc Natl Acad Sci USA. 91:4362–4366. 1994. View Article : Google Scholar : PubMed/NCBI

2 

Cubitt B, Oldstone C and de la Torre JC: Sequence and genome organization of Borna disease virus. J Virol. 68:1382–1396. 1994.PubMed/NCBI

3 

Ludwig H and Bode L: Borna disease virus: New aspects on infection, disease, diagnosis and epidemiology. Rev Sci Tech. 19:259–288. 2000.PubMed/NCBI

4 

Ludwig H, Bode L and Gosztonyi G: Borna disease: A persistent virus infection of the central nervous system. Prog Med Virol. 35:107–151. 1988.PubMed/NCBI

5 

Horie M, Honda T, Suzuki Y, Kobayashi Y, Daito T, Oshida T, Ikuta K, Jern P, Gojobori T, Coffin JM and Tomonaga K: Endogenous non-retroviral RNA virus elements in mammalian genomes. Nature. 463:84–87. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Wang X, Zhang L, Lei Y, Liu X, Zhou X, Liu Y, Wang M, Yang L, Zhang L, Fan S and Xie P: Meta-analysis of infectious agents and depression. Sci Rep. 4:45302014.PubMed/NCBI

7 

Bode L, Reckwald P, Severus WE, Stoyloff R, Ferszt R, Dietrich DE and Ludwig H: Borna disease virus-specific circulating immune complexes, antigenemia, and free antibodies-the key marker triplet determining infection and prevailing in severe mood disorders. Mol Psychiatry. 6:481–491. 2001. View Article : Google Scholar : PubMed/NCBI

8 

Bode L, Zimmermann W, Ferszt R, Steinbach F and Ludwig H: Borna disease virus genome transcribed and expressed in psychiatric patients. Nat Med. 1:232–236. 1995. View Article : Google Scholar : PubMed/NCBI

9 

Fu ZF, Amsterdam JD, Kao M, Shankar V, Koprowski H and Dietzschold B: Detection of Borna disease virus-reactive antibodies from patients with affective disorders by western immunoblot technique. J Affect Disord. 27:61–68. 1993. View Article : Google Scholar : PubMed/NCBI

10 

Rott R, Herzog S, Fleischer B, Winokur A, Amsterdam J, Dyson W and Koprowski H: Detection of serum antibodies to Borna disease virus in patients with psychiatric disorders. Science. 228:755–756. 1985. View Article : Google Scholar : PubMed/NCBI

11 

Sauder C, Müller A, Cubitt B, Mayer J, Steinmetz J, Trabert W, Ziegler B, Wanke K, Mueller-Lantzsch N, de la Torre JC and Grässer FA: Detection of Borna disease virus (BDV) antibodies and BDV RNA in psychiatric patients: Evidence for high sequence conservation of human blood-derived BDV RNA. J Virol. 70:7713–7724. 1996.PubMed/NCBI

12 

Thakur R, Sarma S and Sharma B: Role of Borna disease virus in neuropsychiatric illnesses: Are we inching closer? Indian J Med Microbiol. 27:191–201. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Zhang L, Xu MM, Zeng L, Liu S, Liu X, Wang X, Li D, Huang RZ, Zhao LB, Zhan QL, et al: Evidence for Borna disease virus infection in neuropsychiatric patients in three western China provinces. Eur J Clin Microbiol Infect Dis. 33:621–627. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Liu X, Bode L, Zhang L, Wang X, Liu S, Zhang L, Huang R, Wang M, Yang L, Chen S, et al: Health care professionals at risk of infection with Borna disease virus-evidence from a large hospital in China (Chongqing). Virol J. 12:392015. View Article : Google Scholar : PubMed/NCBI

15 

Hornig M, Briese T, Licinio J, Khabbaz RF, Altshuler LL, Potkin SG, Schwemmle M, Siemetzki U, Mintz J, Honkavuori K, et al: Absence of evidence for bornavirus infection in schizophrenia, bipolar disorder and major depressive disorder. Mol Psychiatry. 17:486–493. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Evengård B, Briese T, Lindh G, Lee S and Lipkin WI: Absence of evidence of Borna disease virus infection in Swedish patients with Chronic Fatigue Syndrome. J Neurovirol. 5:495–499. 1999. View Article : Google Scholar : PubMed/NCBI

17 

Wolff T, Heins G, Pauli G, Burger R and Kurth R: Failure to detect Borna disease virus antigen and RNA in human blood. J Clin Virol. 36:309–311. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Scordel C, Huttin A, Cochet-Bernoin M, Szelechowski M, Poulet A, Richardson J, Benchoua A, Gonzalez-Dunia D, Eloit M and Coulpier M: Borna disease virus phosphoprotein impairs the developmental program controlling neurogenesis and reduces human GABAergic neurogenesis. PLoS Pathog. 11:e10048592015. View Article : Google Scholar : PubMed/NCBI

19 

Hans A, Syan S, Crosio C, Sassone-Corsi P, Brahic M and Gonzalez-Dunia D: Borna disease virus persistent infection activates mitogen-activated protein kinase and blocks neuronal differentiation of PC12 cells. J Biol Chem. 276:7258–7265. 2001. View Article : Google Scholar : PubMed/NCBI

20 

Hans A, Bajramovic JJ, Syan S, Perret E, Dunia I, Brahic M and Gonzalez-Dunia D: Persistent, noncytolytic infection of neurons by Borna disease virus interferes with ERK 1/2 signaling and abrogates BDNF-induced synaptogenesis. FASEB J. 18:863–865. 2004.PubMed/NCBI

21 

Kamitani W, Ono E, Yoshino S, Kobayashi T, Taharaguchi S, Lee BJ, Yamashita M, Kobayashi T, Okamoto M, Taniyama H, et al: Glial expression of Borna disease virus phosphoprotein induces behavioral and neurological abnormalities in transgenic mice. Proc Natl Acad Sci USA. 100:8969–8974. 2003. View Article : Google Scholar : PubMed/NCBI

22 

Bartel DP: MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell. 116:281–297. 2004. View Article : Google Scholar : PubMed/NCBI

23 

Ambros V: The functions of animal microRNAs. Nature. 431:350–355. 2004. View Article : Google Scholar : PubMed/NCBI

24 

Mollaie HR, Monavari SH, Arabzadeh SA, Shamsi-Shahrabadi M, Fazlalipour M and Afshar RM: RNAi and miRNA in viral infections and cancers. Asian Pac J Cancer Prev. 14:7045–7056. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Krol J, Loedige I and Filipowicz W: The widespread regulation of microRNA biogenesis, function and decay. Nat Rev Genet. 11:597–610. 2010.PubMed/NCBI

26 

Alvarez-Garcia I and Miska EA: MicroRNA functions in animal development and human disease. Development. 132:4653–4662. 2005. View Article : Google Scholar : PubMed/NCBI

27 

Liu X, Zhang L, Cheng K, Wang X, Ren G and Xie P: Identification of suitable plasma-based reference genes for miRNAome analysis of major depressive disorder. J Affect Disord. 163:133–139. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Fagan AM and Perrin RJ: Upcoming candidate cerebrospinal fluid biomarkers of Alzheimer's disease. Biomark Med. 6:455–476. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Di Y, Lei Y, Yu F, Changfeng F, Song W and Xuming M: MicroRNAs expression and function in cerebral ischemia reperfusion injury. J Mol Neurosci. 53:242–250. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Lages E, Ipas H, Guttin A, Nesr H, Berger F and Issartel JP: MicroRNAs: Molecular features and role in cancer. Front Biosci (Landmark Ed). 17:2508–2540. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Bode L, Dürrwald R, Rantam FA, Ferszt R and Ludwig H: First isolates of infectious human Borna disease virus from patients with mood disorders. Mol Psychiatry. 1:200–212. 1996.PubMed/NCBI

32 

Huang R, Gao H, Zhang L, Jia J, Liu X, Zheng P, Ma L, Li W, Deng J, Wang X, et al: Borna disease virus infection perturbs energy metabolites and amino acids in cultured human oligodendroglia cells. PLoS One. 7:e446652012. View Article : Google Scholar : PubMed/NCBI

33 

Liu X, Zhao L, Yang Y, Bode L, Huang H, Liu C, Huang R, Zhang L, Wang X, Zhang L, et al: Human borna disease virus infection impacts host proteome and histone lysine acetylation in human oligodendroglia cells. Virology 464–465. 196–205. 2014. View Article : Google Scholar

34 

Zhang L, Wang X, Zhan Q, Wang Z, Xu M, Zhu D, He F, Liu X, Huang R, Li D, et al: Evidence for natural Borna disease virus infection in healthy domestic animals in three areas of western China. Arch Virol. 159:1941–1949. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Zhang L, Liu S, Zhang L, You H, Huang R, Sun L, He P, Chen S, Zhang H and Xie P: Real-time qPCR identifies suitable reference genes for Borna disease virus-infected rat cortical neurons. Int J Mol Sci. 15:21825–21839. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Zhang L, Wang X, Zhan Q, Wang Z, Xu M, Zhu D, He F, Liu X, Huang R, Li D, Lei Y and Xie P: Evidence for natural Borna disease virus infection in healthy domestic animals in three areas of western China. Arch Virol. 159:1941–1949. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Perkins JR, Dawes JM, McMahon SB, Bennett DL, Orengo C and Kohl M: ReadqPCR and NormqPCR: R packages for the reading, quality checking and normalisation of RT-qPCR quantification cycle (Cq) data. BMC genomics. 13:2962012. View Article : Google Scholar : PubMed/NCBI

38 

Li D, Lei Y, Deng J, Zhou C, Zhang Y, Li W, Huang H, Cheng S, Zhang H, Zhang L, et al: Human but not laboratory borna disease virus inhibits proliferation and induces apoptosis in human oligodendrocytes in vitro. PLoS One. 8:e666232013. View Article : Google Scholar : PubMed/NCBI

39 

Liu X, Yang Y, Zhao M, Bode L, Zhang L, Pan J, Lv L, Zhan Y, Liu S, Zhang L, et al: Proteomics reveal energy metabolism and mitogen-activated protein kinase signal transduction perturbation in human Borna disease virus Hu-H1-infected oligodendroglial cells. Neuroscience. 268:284–296. 2014. View Article : Google Scholar : PubMed/NCBI

40 

Lei Y, Li D, Deng J, et al: Metabolomic profiling of three brain regions from a postnatal infected Borna disease virus Hu-H1 rat model. Metabolomics. 10:484–495. 2014. View Article : Google Scholar

41 

Strickland ER, Hook MA, Balaraman S, Huie JR, Grau JW and Miranda RC: MicroRNA dysregulation following spinal cord contusion: Implications for neural plasticity and repair. Neuroscience. 186:146–160. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Zhao M, Sun L, Chen S, Li D, Zhang L, He P, Liu X, Zhang L, Zhang H, Yang D, et al: Borna disease virus infection impacts microRNAs associated with nervous system development, cell differentiation, proliferation and apoptosis in the hippocampi of neonatal rats. Mol Med Rep. 12:3697–3703. 2015.PubMed/NCBI

43 

Liu Z, Liu Y, Li L, Xu Z, Bi B, Wang Y and Li JY: MiR-7-5p is frequently downregulated in glioblastoma microvasculature and inhibits vascular endothelial cell proliferation by targeting RAF1. Tumour Biol. 35:10177–10184. 2014. View Article : Google Scholar : PubMed/NCBI

44 

Fang Y, Xue JL, Shen Q, Chen J and Tian L: MicroRNA-7 inhibits tumor growth and metastasis by targeting the phosphoinositide 3-kinase/Akt pathway in hepatocellular carcinoma. Hepatology. 55:1852–1862. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Giles KM, Brown RA, Epis MR, Kalinowski FC and Leedman PJ: miRNA-7-5p inhibits melanoma cell migration and invasion. Biochem Biophys Res Commun. 430:706–710. 2013. View Article : Google Scholar : PubMed/NCBI

46 

Shi Y, Luo X, Li P, Tan J, Wang X, Xiang T and Ren G: miR-7-5p suppresses cell proliferation and induces apoptosis of breast cancer cells mainly by targeting REGγ. Cancer Lett. 358:27–36. 2015. View Article : Google Scholar : PubMed/NCBI

47 

Saydam O, Senol O, Würdinger T, Mizrak A, Ozdener GB, Stemmer-Rachamimov AO, Yi M, Stephens RM, Krichevsky AM, Saydam N, et al: miRNA-7 attenuation in Schwannoma tumors stimulates growth by upregulating three oncogenic signaling pathways. Cancer Res. 71:852–861. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Wu DG, Wang YY, Fan LG, Luo H, Han B, Sun LH, Wang XF, Zhang JX, Cao L, Wang XR, et al: MicroRNA-7 regulates glioblastoma cell invasion via targeting focal adhesion kinase expression. Chin Med J (Engl). 124:2616–2621. 2011.PubMed/NCBI

49 

Kong X, Li G, Yuan Y, He Y, Wu X, Zhang W, Wu Z, Chen T, Wu W, Lobie PE and Zhu T: MicroRNA-7 inhibits epithelial-to-mesenchymal transition and metastasis of breast cancer cells via targeting FAK expression. PLoS One. 7:e415232012. View Article : Google Scholar : PubMed/NCBI

50 

Zhao X, Dou W, He L, Liang S, Tie J, Liu C, Li T, Lu Y, Mo P, Shi Y, et al: MicroRNA-7 functions as an anti-metastatic microRNA in gastric cancer by targeting insulin-like growth factor-1 receptor. Oncogene. 32:1363–1372. 2013. View Article : Google Scholar : PubMed/NCBI

51 

Wu K, Hu G, He X, Zhou P, Li J, He B and Sun W: MicroRNA-424-5p suppresses the expression of SOCS6 in pancreatic cancer. Pathol Oncol Res. 19:739–748. 2013. View Article : Google Scholar : PubMed/NCBI

52 

Zhang Y, Li T, Guo P, Kang J, Wei Q, Jia X, Zhao W, Huai W, Qiu Y, Sun L and Han L: MiR-424-5p reversed epithelial-mesenchymal transition of anchorage-independent HCC cells by directly targeting ICAT and suppressed HCC progression. Sci Rep. 4:62482014. View Article : Google Scholar : PubMed/NCBI

53 

Bai Y, Liao H, Liu T, Zeng X, Xiao F, Luo L, Guo H and Guo L: MiR-296-3p regulates cell growth and multi-drug resistance of human glioblastoma by targeting ether-à-go-go (EAG1). Eur J Cancer. 49:710–724. 2013. View Article : Google Scholar : PubMed/NCBI

54 

Kolodziejek J, Dürrwald R, Herzog S, Ehrensperger F, Lussy H and Nowotny N: Genetic clustering of Borna disease virus natural animal isolates, laboratory and vaccine strains strongly reflects their regional geographical origin. J Gen Virol. 86:385–398. 2005. View Article : Google Scholar : PubMed/NCBI

55 

Zhai A, Qian J, Kao W, Li A, Li Y, He J, Zhang Q, Song W, Fu Y, Wu J, et al: Borna disease virus encoded phosphoprotein inhibits host innate immunity by regulating miR-155. Antiviral Res. 98:66–75. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2016
Volume 14 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang H, He P, Huang R, Sun L, Liu S, Zhou J, Guo Y, Yang D and Xie P: Identification and bioinformatic analysis of dysregulated microRNAs in human oligodendroglial cells infected with borna disease virus. Mol Med Rep 14: 4715-4722, 2016
APA
Zhang, H., He, P., Huang, R., Sun, L., Liu, S., Zhou, J. ... Xie, P. (2016). Identification and bioinformatic analysis of dysregulated microRNAs in human oligodendroglial cells infected with borna disease virus. Molecular Medicine Reports, 14, 4715-4722. https://doi.org/10.3892/mmr.2016.5842
MLA
Zhang, H., He, P., Huang, R., Sun, L., Liu, S., Zhou, J., Guo, Y., Yang, D., Xie, P."Identification and bioinformatic analysis of dysregulated microRNAs in human oligodendroglial cells infected with borna disease virus". Molecular Medicine Reports 14.5 (2016): 4715-4722.
Chicago
Zhang, H., He, P., Huang, R., Sun, L., Liu, S., Zhou, J., Guo, Y., Yang, D., Xie, P."Identification and bioinformatic analysis of dysregulated microRNAs in human oligodendroglial cells infected with borna disease virus". Molecular Medicine Reports 14, no. 5 (2016): 4715-4722. https://doi.org/10.3892/mmr.2016.5842