Abstract
Unlabelled
MicroRNAs (miRNAs) are single-stranded small RNA molecules that regulate various cellular processes. miRNA 155 (miR-155) regulates various aspects of innate and adaptive immune responses and plays a key role in various viral infections and the resulting neuroinflammation. The present study evaluated the involvement of miR-155 in modulating Japanese encephalitis virus (JEV)-induced neuroinflammation. We observed that miR-155 expression was upregulated during JEV infection of mouse primary microglia, the BV-2 microglia cell line, and in both mouse and human brains. In vitro and in vivo knockdown of miR-155 minimized JEV-induced inflammatory responses. In the present study, we confirmed targeting of the Src homology 2-containing inositol phosphatase 1 (SHIP1) 3' untranslated region (UTR) by miR-155 in the context of JEV infection. Inhibition of SHIP1 by miR-155 resulted in higher beta interferon (IFN-β) and proinflammatory cytokine production through activation of TANK-binding kinase 1 (TBK-1). Based on these observations, we conclude that miR-155 modulates the neuroinflammatory response during JEV infection via negative regulation of SHIP1 expression. Thus, modulation of miR-155 could be a novel strategy to regulate JEV-induced neuroinflammation.Importance
Japanese encephalitis virus (JEV), a member of the family Flaviviridae that causes Japanese encephalitis (JE), is the most common mosquito-borne encephalitis virus in the Asia-Pacific region. The disease is feared, as currently there are no specific antiviral drugs available. JEV targets the central nervous system, leading to high mortality and neurological and psychiatric sequelae in some of those who survive. The level of inflammation correlates well with the clinical outcome in patients. Recently, microRNA (miRNA), a single-stranded noncoding RNA, has been implicated in various brain disorders. The present study investigates the role of miRNA in JEV-induced neuroinflammation. Our results show that miRNA 155 (miR-155) targets the Src homology 2-containing inositol phosphatase 1 (SHIP1) protein and promotes inflammation by regulating the NF-κB pathway, increasing the expression of various proinflammatory cytokines and the antiviral response. Thus, miR-155 is a potential therapeutic target to develop antivirals in JE and other brain disorders where inflammation plays a significant role in disease progression.Free full text
MicroRNA 155 Regulates Japanese Encephalitis Virus-Induced Inflammatory Response by Targeting Src Homology 2-Containing Inositol Phosphatase 1
Associated Data
MicroRNAs (miRNAs) are single-stranded small RNA molecules that regulate various cellular processes. miRNA 155 (miR-155) regulates various aspects of innate and adaptive immune responses and plays a key role in various viral infections and the resulting neuroinflammation. The present study evaluated the involvement of miR-155 in modulating Japanese encephalitis virus (JEV)-induced neuroinflammation. We observed that miR-155 expression was upregulated during JEV infection of mouse primary microglia, the BV-2 microglia cell line, and in both mouse and human brains. In vitro and in vivo knockdown of miR-155 minimized JEV-induced inflammatory responses. In the present study, we confirmed targeting of the Src homology 2-containing inositol phosphatase 1 (SHIP1) 3′ untranslated region (UTR) by miR-155 in the context of JEV infection. Inhibition of SHIP1 by miR-155 resulted in higher beta interferon (IFN-β) and proinflammatory cytokine production through activation of TANK-binding kinase 1 (TBK-1). Based on these observations, we conclude that miR-155 modulates the neuroinflammatory response during JEV infection via negative regulation of SHIP1 expression. Thus, modulation of miR-155 could be a novel strategy to regulate JEV-induced neuroinflammation.
IMPORTANCE Japanese encephalitis virus (JEV), a member of the family Flaviviridae that causes Japanese encephalitis (JE), is the most common mosquito-borne encephalitis virus in the Asia-Pacific region. The disease is feared, as currently there are no specific antiviral drugs available. JEV targets the central nervous system, leading to high mortality and neurological and psychiatric sequelae in some of those who survive. The level of inflammation correlates well with the clinical outcome in patients. Recently, microRNA (miRNA), a single-stranded noncoding RNA, has been implicated in various brain disorders. The present study investigates the role of miRNA in JEV-induced neuroinflammation. Our results show that miRNA 155 (miR-155) targets the Src homology 2-containing inositol phosphatase 1 (SHIP1) protein and promotes inflammation by regulating the NF-κB pathway, increasing the expression of various proinflammatory cytokines and the antiviral response. Thus, miR-155 is a potential therapeutic target to develop antivirals in JE and other brain disorders where inflammation plays a significant role in disease progression.
Japanese encephalitis virus (JEV), a member of the family Flaviviridae, is a single-stranded, positive-sense RNA virus that causes Japanese encephalitis (JE) (1, 2). JE is endemic in most Southeast Asian countries (3, 4). The availability of an effective vaccine and an active immunization program have resulted in a reduced number of JE cases in a few countries (5). The early clinical features of JE include fever, headache, and vomiting. Two weeks after JEV infection, patients develop neurological symptoms, like seizure, tremor, photophobia, and movement disorder (6). These clinical features are not exclusive to JEV infection, and hence, laboratory diagnosis is needed to differentiate it from other neurological disorders. Detection of anti-JEV IgM antibodies in the cerebrospinal fluid (CSF) and serum is frequently used to diagnose JE (7). The fatality rate of JEV infection is ~25%. A majority of survivors (~50%) have neuropsychiatric sequelae; only ~25% recover completely (8). Hence, JEV poses major health concerns and economic burdens in the Asia-Pacific region. During the last decade, we and others have made significant progress in the understanding molecular mechanisms involved in JEV infection.
MicroRNAs (miRNAs) are small, noncoding RNAs ~19 to 22 nucleotides in length that regulate various cellular processes by binding to the 3′ untranslated region (UTR) of target proteins, resulting in either degradation of RNA or translational suppression (9, 10). Our knowledge of the role of miRNAs in various diseases is expanding rapidly. Various reports support the role of miRNAs in neuroviral infections and the resulting neuroinflammation (11). miRNA 155 (miR-155) is one of the most widely studied miRNAs in regulating inflammation (12). Silencing miR-155 ameliorates experimental autoimmune encephalomyelitis (13) and regulates inflammatory changes in astrocytes (14) and microglia (15). miR-155 plays a major role in viral infections caused by Epstein-Barr (16), Borna disease (17), and reticuloendotheliosis (18) viruses.
Previously, we showed that miR-29b regulates JEV-induced expression of inducible nitric oxide synthase (iNOS) and COX-2 in BV-2 cells (19). It has previously been reported that miR-155 targets Src homology 2-containing inositol phosphatase 1 (SHIP1) to regulate the inflammatory response (20). In the present study, we show that miR-155 is a key regulator of JEV-induced neuroinflammation. We demonstrate that during JEV infection, miR-155 inhibits SHIP1 expression by binding to its 3′ UTR. Further, in vivo treatment of JEV-infected mice with locked-nucleic-acid-modified oligonucleotide (LNA) anti-miR-155 minimizes overall neuroinflammtion by regulating TANK-binding kinase 1 (TBK-1) phosphorylation.
Ethics statement.
Animals were handled in strict accordance with good animal practice as defined by the Committee for the Purpose of Control and Supervision of Experiments on Animals (CPCSEA) and the Ministry of Environment and Forestry, Government of India. The Institutional Animal Ethics Committee (IAEC) of the National Brain Research Centre approved the study protocol.
Virus isolation and titration.
The GP78 strain of JEV was propagated in suckling BALB/c mice (postnatal day 3 or 4) of either sex until the appearance of symptoms of sickness (limb paralysis, poor pain response, and whole-body tremor). Following the onset of symptoms, the mice were sacrificed to collect the infected brains. The brains were homogenized in minimum essential medium (MEM) and centrifuged at 10,000 × g to remove cellular debris. The resulting suspension was filtered through a 0.22-μm sterile filter, and aliquots of the filtered virus suspension were stored at −80°C until further use (21).
Virus was titrated by plaque formation assay using the porcine stable kidney (PS) cell line as described previously (22). Briefly, a monolayer of PS cells was incubated with JEV (a 10-fold dilution prepared in MEM containing 1% fetal bovine serum [FBS]) at 37°C for 1 h. The viral inocula were removed from the monolayers, and MEM containing 4% FBS, 1% low-melting-point agarose, and a cocktail of antibiotic-antimycotic solutions was added. Then, the culture plates were incubated at 37°C for 72 to 96 h until the appearance of visible plaques. The plaques were fixed and stained with 0.1% crystal violet (23).
JEV was inactivated by exposure to short-wavelength (254-nm) UV radiation (UVC) at a distance of 5 cm for 10 min on ice, using a UV cross-linker (UVC 500; Hoefer Scientific, USA), as described previously (24).
BV-2 cell culture.
The BV-2 cell line (a mouse microglia cell line) was provided by Steve Levison, University of Medicine and Dentistry, New Jersey, USA. BV-2 cells were grown at 37°C in Dulbecco's modified Eagle's medium (DMEM) supplemented with 5% sodium bicarbonate (NaHCO3), 10% FBS, 100 U/ml penicillin, and 100 μg/ml streptomycin, as described previously (25).
Primary microglial culture.
Primary microglia cells were isolated from BALB/c mouse pups of either sex (postnatal days 0 to 2), as reported previously (25). Briefly, the whole brain cortex was dissected from the mouse brain, and the meninges were removed under a dissecting microscope. Tissue was digested using trypsin-DNase solution with a brief mechanical dissociation to obtain a cell suspension. The cell suspension was passed through 100-mm cell strainers and centrifuged at 400 × g for 8 min to obtain a cell pellet. After a viable-cell count, cells were plated on 75-cm2 tissue culture flasks at a density of 2 × 105 viable cells/cm2 in complete MEM (supplemented with 10% fetal bovine serum, 100 units/ml penicillin, 100 μg/ml streptomycin, 0.6% glucose, and 2 mM glutamine). The complete MEM was changed every 2 days until the mixed glial culture became confluent. On day 12, microglia cells were dislodged by shaking the mixed glial flasks on an orbital shaker (Excella E25; New Brunswick Scientific, NJ, USA) at 250 rpm for 60 to 75 min at 37°C. The media containing the nonadherent cells were removed and plated in bacteriological petri dishes. After complete adherence (60 to 90 min), the microglia cells were treated with Accutase, scraped, and centrifuged. Subsequently, the cells were plated on 2-well chamber slides (Nunc, Denmark) at 8 × 104 viable cells/cm2 and incubated at 37°C for further experiments.
Infection of BV-2 and primary microglia cells with JEV.
BV-2 cells were plated at a density of 1.5 × 106 cells in DMEM containing 10% FBS. After 15 to 18 h, the cells were incubated with serum-free medium for 4 to 5 h. The cells were infected with JEV or UV-treated JEV at a multiplicity of infection (MOI) of 5 for 1.5 h. The unbound virus was removed by washing gently with 1× phosphate-buffered saline (PBS), and the cells were further incubated in serum-free medium for different time interval studies. Primary microglia cells plated on chamber slides were treated similarly with JEV for 12 or 24 h.
Reverse transcription real-time PCR and miRNA assay.
After isolation of total RNA from treated cells using Tri Reagent (Sigma-Aldrich), cDNA was prepared using an Advantage RT-for-PCR kit (Clontech Laboratories). Amplification of SHIP1 (forward, 5′-CCA GGG CAA GAT GAG GGA GA-3′; reverse, 5′-GGA CCT CGG TTG GCA ATG TA-3′) and beta interferon (IFN-β) (forward, 5′-TTG CCA TCC AAG AGA TGC TC-3′; reverse, 5′-TCA GAA ACA CTG TCT GCT GG-3′) mRNA was carried out by quantitative PCR (qPCR) using power SYBR green PCR master mix (Applied Biosystems, Foster City, CA). The conditions used for the PCR were as follows: 95°C for 3 min (1 cycle) and 94°C for 20 s, 55°C for 30 s, and 72°C for 40 s (40 cycles). The relative mRNA abundance was determined by normalizing to glyceraldehyde-3-phosphate dehydrogenase (GAPDH) mRNA using the 2−ΔΔCt method (Ct refers to the threshold value).
miRNA was isolated using a miR-Neasy kit (Qiagen), and cDNA was prepared using a miScript II RT Kit (Qiagen) according to the manufacturer's instructions. qPCR was performed using specific miR-155 primer, 5′ UUA AUG CUA AUU GUG AUA GGG GU 3′ (Qiagen), and the miScript SYBR green PCR kit (containing the miScript Universal reverse primer). The conditions used for qPCR were as follows: 95°C for 15 min (1 cycle), 94°C for 15 s, 55°C for 30 s, and 70°C for 30 s (40 cycles). The thermal cycler ViiA 7 Real-Time PCR (Applied Biosystems) was used for qPCR, and the data were analyzed using iCycler Thermal Cycler software (Applied Biosystems). Relative miRNA abundance was determined by normalizing to U6 or SNORD 68 small nuclear RNA using the 2−ΔΔCt method.
miRNA overexpression and inhibition in BV-2 cells.
miR-155 was overexpressed in BV-2 cells by transfecting the cells with 100 pmol of miScript miR-155 mimic (Qiagen) or miRNA mimic negative control (Ambion) using commercial medium (Opti-MEM; Invitrogen) and Lipofectamine 2000 (Invitrogen). After 24 h of transfection, cells were harvested and qPCR was performed for miR-155 expression. In another set of experiments, miR-155 was overexpressed, and SHIP1 expression was analyzed by immunoblotting.
For specific knockdown of miR-155 expression, BV-2 cells were transfected with 100 pmol of miScript anti-miR-155 inhibitor (Qiagen). After 24 h of transfection, the cells were infected with JEV for 24 h and used to isolate protein and RNA. Cy3-labeled control anti-miR was used to assess transfection efficiency.
SHIP1 siRNA transfection.
BV-2 cells were transfected with 0.5 μM small interfering RNA (siRNA) targeting SHIP1 (forward, 5′-GCTAAGTGCTTTACGAACA-3′; reverse, 5′-TGTTCGTAAAGCACTTAGC-3′) or nontargeting control siRNA (Sigma) using Lipofectamine 2000 (Invitrogen) for 24 h. After 24 h of transfection, the cells were infected with JEV and used for further studies.
MicroRNA target prediction.
The bioinformatics prediction tools PicTar (http://pictar.mdc-berlin.de/) and Target Scan (version 5.2) (http://www.targetscan.org/) were used to identify the potential targets of miR-155. The miR-155 target binding sites in the 3′ UTRs of human and mouse SHIP1 transcripts were identified with Target Scan software.
Luciferase reporter assay.
BV-2 cells were transfected either with pMIR-REPORT SHIP1 3′ UTR luciferase reporter (Add Gene), pMIR-REPORT plasmid, or pMIR-REPORT-β-galactosidase (Ambion) and cotransfected with 100 pmol of miR-155 or miR-29b for 24 h. Assays were performed using a luciferase assay system (Promega, Madison, WI, USA). In another experiment, BV-2 cells were transfected with SHIP1 construct and were exposed to JEV and UV-treated JEV for 24 h. Briefly, the cells were washed with 1× PBS and lysed in reporter lysis buffer provided by the manufacturer. After a brief centrifugation (12,000 × g for 5 min at 4°C), the supernatant was collected. Luciferase assay reagent (100 μl) was dispensed into three sets of luminometer tubes, to which 20 μl of the collected supernatant was added; the readings were taken using a Sirius single-tube luminometer (Berthold Detection Systems GmBH, Germany). The luciferase units were measured as relative luciferase units (RLU) and normalized to the total protein.
Effect of LNA anti-miR-155 treatment in JEV-infected BALB/c mice.
Adult BALB/c mice of either sex were randomly assigned to three groups: group 1, the control group (CON); group 2, the JEV-infected group (JEV); and group 3, the JEV-infected and LNA anti-miR-155-treated group (JEV + LNA). Mice in groups 2 and 3 received JEV strain GP78 (3 × 105 PFU) via the tail vein, while group 1 received an equal volume of PBS. Twenty-four hours after JEV infection, mice in groups 2 and 3 received anti-miR-155 LNA (5′-TCACAATTAGCATTA-3′) or LNA scrambled anti-miR (Exiqon) (20 mg/kg body weight intravenously [i.v.]) for three consecutive days. After 4 days, mice in the JEV group developed signs of JEV infection, and on the 7th day, the animals developed restriction of movement, limb paralysis, poor pain response, whole-body tremor, piloerection, and hind limb paralysis. All the mice were euthanized, and brain samples were collected for mRNA, miRNA, protein, and immunohistochemistry studies.
Isolation of miRNA from human brain tissue sections and qPCR.
Human brain tissue sections were obtained from the Human Brain Tissue Repository, National Institute of Mental Health and Neurosciences, Bangalore, India, in accordance with the institutional scientific ethics, protecting the confidentiality of the subjects. Formalin-fixed, paraffin-embedded (FFPE) sections from the basal ganglia/frontal cortex were obtained postmortem from confirmed cases of JE. All cases had JEV IgM antibodies in the CSF, microglial nodules, and necrolytic lesions characteristic of JE. Brain tissues from individuals who succumbed to traffic accidents and had no known prior neurological disease were used as non-JEV controls. The miRNA extraction was performed using an miRNeasy FFPE Kit (Qiagen) according to the manufacturer's instructions. The 20-μm-thick FFPE sections were deparaffinized, treated with proteinase K, and processed for RNA isolation. cDNA was synthesized using an miScript II RT Kit (Qiagen) as described above. qPCR analysis was performed using primers specific for human miR-155: 5′-UUAAUGCUAAUCGUGAUAGGGGU-3′ (Qiagen) and the universal SYBR green Power Master Mix (Applied Biosystems). The relative miRNA abundance was determined by normalizing to Hs-RNU6 (Qiagen). The real-time PCR results were analyzed and expressed as fold changes over non-JEV samples using the 2−ΔΔCt method.
Immunoblotting.
Immunoblotting was carried out at the end of the treatment period as previously described (21). Briefly, control and treated BV-2 cells or mouse brain was lysed in complete lysis buffer containing 1% Triton X-100, 10 mM Tris-HCl (pH 8.0), 150 mM NaCl, 0.5% Nonidet P-40 (NP-40), 1 mM EDTA, 0.2% EGTA, 0.2% sodium orthovanadate, and protease inhibitor cocktail (Sigma-Aldrich). Fifty to 100 μg of each protein sample was electrophoresed on SDS-PAGE and transferred onto a nitrocellulose membrane. The membrane was then blocked using 5% skim milk in 1× PBST (1× PBS-Tween 20) and incubated with the following primary antibodies: SHIP1, phospho- and total interferon-regulatory factor 3 (IRF3), phospho-IRF7, phospho- and total NF-κB (Cell Signaling), phospho-TBK (Millipore), total TBK, and total IRF7 (Santa Cruz Biotechnology, CA, USA) overnight at 4°C. All primary antibodies were prepared at a dilution of 1:1,000 in 1% bovine serum albumin (BSA) in 1× PBST. The blots were labeled with goat anti-rabbit horseradish peroxidase (1:5,000; Vector Laboratories, CA, USA) in 1× PBST and developed by exposure in a Chemigenius Bio-Imaging System (Syngene; Cambridge, United Kingdom) using Gene Snap software. The images were analyzed using Gene Tools software from Syngene. The blots were then stripped and probed with anti-β-actin antibody (1:10,000; Sigma-Aldrich).
CBA.
The cytokine levels in the brains of control and JEV-infected mice were quantified using a mouse cytokine bead array (CBA) kit (BD Biosciences, San Diego, CA) according to the manufacturer's instructions. The beads, coated with interleukin 6 (IL-6), tumor necrosis factor alpha (TNF-α), and monocyte chemotactic protein 1 (MCP-1), were mixed with test lysates and standards, to which fluorescent dye was added. The mixtures were incubated for 2 h at room temperature (RT) in the dark, following which the beads were washed and resuspended in 300 μl of wash buffer and acquired using Cell Quest Pro software in a FACSCalibur (Becton, Dickinson, San Diego, CA). The data were analyzed using BD CBA software (Becton, Dickinson, San Diego, CA, USA) (26), and the concentrations of various cytokines were expressed as pg/ml.
Immunohistochemistry.
For immunohistochemical staining, brains from sacrificed mice were excised following repeated transcardial perfusion with ice-cold saline and fixed with 4% paraformaldehyde. Twenty-micrometer-thick cryosections were prepared using a Leica CM3050S cryostat and processed for immunohistochemical staining. Sections were incubated overnight with anti-SHIP1 (1:100; Cell Signaling) and anti-Iba and Neu-N (1:250; Millipore) at 4°C. After washes, the slides were incubated with fluorescein isothiocyanate (FITC) (1:200; Vector Laboratories)- or Alexa Fluor 594 (1:1,000; Molecular Probes, Eugene, OR)-conjugated secondary antibodies. After final washes, the sections were mounted with 4′-6-diamidino-2-phenylindole (DAPI) (Vector Laboratories Inc.). The sections were observed and photographed using a Zeiss Apotome microscope (Zeiss, Gottingen, Germany) at the indicated magnification.
In situ hybridization (ISH).
Fresh-frozen (FF) brain sections 20 μm thick were obtained using a cryostat, as mentioned above. FFPE sections of uninfected (control) and JEV-infected human brains were deparaffinized with xylene and hydrated using series of alcohol. The FF sections were fixed in fresh 4% neutral buffered formalin overnight. Following washes with PBS, the sections were treated with 20 mg/ml proteinase K at 37°C for 10 min. Then, the sections were thoroughly washed with PBS and incubated with hybridization buffer containing 40 nM double 5′-digoxigenin (DIG)- and -3′-DIG-labeled LNA miR-155 probe (5′-TATCACAATTAGCATTAA-3′) (Exiqon) for 60 min at 48°C in a humidified chamber. Following hybridization, the sections were washed with 5×, 1×, and 0.2× SSC 2 times each for 5 min in that order. After treatment with blocking solution, the sections were incubated with anti-DIG reagent (sheep anti-digoxigenin alkaline phosphatase [anti-DIG-AP] antibody at 1:800 dilution) for 60 min at RT in a humidified chamber and washed with PBST. Then, the sections were incubated with freshly prepared AP substrates for 2 h at 30°C in the humidifying chamber (under dark conditions). The reaction was terminated by adding KTBT buffer (an AP stop solution composed of 50 mM Tris-HCl, 150 mM NaCl, and 10 mM KCl), washed with water, dehydrated in series of alcohol, and mounted using DPX. The slides were observed under a Leica DMRXA2 microscope, and photographs were taken under the ideal magnification and conditions for each slide.
Statistical analysis.
All the experiments were performed in sets of three unless otherwise indicated, and the data generated were analyzed using one-way analysis of variance (ANOVA), followed by Bonferroni's multiple-comparison test. The results are expressed as means ± standard errors of the mean (SEM), and graphs were prepared using Graph Pad Prism version 3.0 for Windows (Graph Pad Software, San Diego, CA, USA).
miR-155 is overexpressed during JEV infection.
First, we evaluated the time kinetics expression of miR-155 in JEV-infected mouse brain. BALB/c mice were either infected with JEV or treated with PBS, and brain samples were collected at 1, 3, 5, or 7 days postinfection (p.i.). Interestingly, JEV infection showed a time-dependent increase in miR-155 expression, with maximum changes on days 5 and 7 p.i. (Fig. 1A). We also investigated the expression of miR-155 in JEV-infected human brain samples. The results were similar to those observed in JEV-infected mouse brains. Expression of miR-155 was increased in JEV-infected human brain samples compared to the uninfected control (Fig. 1B). Further, we evaluated time-dependent (0-, 3-, 6-, 12-, or 24-h) expression of miR-155 in JEV-infected BV-2 microglia cells. We observed in BV-2 cells that JEV infection resulted in a time-dependent increase in miR-155 expression that peaked at 12 and 24 h (Fig. 1C). UV-inactivated JEV treatment failed to induce miR-155 expression (Fig. 1C). To validate these results, we evaluated miR-155 expression in JEV-infected primary mouse microglia cells at 12- or 24-h intervals. In agreement with the previous data (Fig. 1C) obtained from cell line study, there was a significant increase in miR-155 expression 24 h after JEV infection (Fig. 1D). ISH of brain sections from JEV-infected mice revealed that miR-155 was localized in the cortex area (Fig. 1A). ISH of human brain biopsy samples showed a larger area with miR-155 positivity from JEV-infected brain samples than from the control (Fig. 1B). These data strongly indicate that miR-155 expression is upregulated after JEV infection.
SHIP1 is involved during in vitro and in vivo JEV infection.
Second, we searched for a potential target of miR-155 by using Pictar and Target Scan bioinformatics tools. SHIP1 is one of the potential targets of miR-155 showing an 8-mer binding site in the SHIP1 3′ UTR. To establish the involvement of SHIP1 in JEV infection, BV-2 cells were infected with JEV at different time points (0, 3, 6, 12, or 24 h) and evaluated for SHIP1 expression. JEV-infected BV-2 cells showed a time-dependent decrease in SHIP1 mRNA and protein levels (maximal at 12 and 24 h) (Fig. 2A). Interestingly, in vitro knockdown of miR-155 using specific antisense oligonucleotides rescued the JEV infection-induced decrease in SHIP1 protein expression (Fig. 2B). Treatment of anti-miR-155 in the absence of JEV infection also recorded a similar pattern of SHIP1 protein expression (Fig. 2B). Further, to evaluate the changes in expression of SHIP1 in vivo, BALB/c mice were infected with JEV, and brain samples were collected at different time points (1, 3, 5, or 7 days p.i.). A significant decrease in expression of SHIP1 mRNA was observed that was comparable to in vitro findings (Fig. 2C). Minimal expression of SHIP1 mRNA was observed on the 7th day, when the animals developed all the major signs of infection. To further verify the involvement of SHIP1 in JEV infection, we knocked down the expression of miR-155 in BALB/c mice by administrating anti-miR-155 LNA for 3 days after JEV infection. We observed a significant reduction in the brain SHIP1 mRNA (Fig. 2C) and protein expression 7 days after infection (data not shown), while anti-miR-155 LNA treatment significantly recovered the JEV infection-induced decrease in SHIP1 expression (Fig. 2D). The results were verified with immunohistochemistry staining of SHIP1 in the mouse brain cortex and subventricular zone (SVZ) regions (Fig. 2E). Data on the quality of in vitro and in vivo miR-155 inhibition/overexpression are shown in Fig. S1 in the supplemental material. Further, treatment of anti-miR-155/miR-155 mimic or LNA anti-miR-155 did not alter the viral load in JEV-infected BV2 cells or BALB/c mice (data not shown), indicating that manipulation of miR-155 expression does not alter viral replication. Further, we evaluated the effect of control anti-miR transfection on the expression of SHIP1, its immediate downstream TBK-1, and proinflammatory cytokines in JEV-infected and control BV-2 cells. Transfection of control miRNA to uninfected and JEV-infected BV-2 cells did not alter the protein levels of SHIP1 and pTBK-1 and the mRNA level of IFN-β compared to nontransfected BV-2 cells (see Fig. S2 in the supplemental material). This observation clearly indicates that the results observed here are specific to anti-miR-155 transfection.
SHIP1 is a target of miR-155 during JEV infection.
We evaluated the role of SHIP1 in modulating miR-155-dependent changes in JEV-induced neuroinflammation. SHIP1 knockdown by siRNA in BV-2 cells resulted in higher expression of inflammatory cytokines than in untreated cells. Further, cotransfection of BV-2 cells with SHIP1 siRNA and anti-miR-155 resulted in higher expression of proinflammatory cytokines than with their individual transfections (see Fig. S3 in the supplemental material). After JEV infection of BV-2 cells, SHIP1 knockdown blunted the beneficial effects of anti-miR-155 transfection (see Fig. S3 in the supplemental material). These sets of observations provide evidence for an antineuroinflammatory role of SHIP1 (see Fig. S3 in the supplemental material). As shown in Fig. 3A, we observed a significant decrease in the SHIP1 protein level by transfection with a miR-155 mimic. This observation, along with the previously mentioned inverse relationship between miR-155 and SHIP1 expression post-JEV infection, suggests that the expression level of SHIP1 is negatively regulated by miR-155. Previous reports have identified SHIP1 as a potential target of miR-155 (20). To determine whether miR-155 binds to the 3′ UTR of SHIP1, we performed a luciferase reporter assay. A pMIR-REPORT construct encompassing a firefly luciferase reporter followed by the full-length 3′ UTR of SHIP1, pMIR-REPORT SHIP1, was used. miR-155 and miR-29b mimic (as a negative/nonspecific miRNA control) oligonucleotides and an empty vector (pMIR-REPORT) were cotransfected with pMIR-REPORT SHIP1. Luciferase activity was diminished in the reporter containing the 3′ UTR of SHIP1 treated with miR-155 compared with pMIR-REPORT SHIP1 alone. In addition, when pMIR-REPORT SHIP1was cotransfected with an irrelevant miR-29b mimic, the luciferase activity did not show any significant changes (Fig. 3C). To further validate interaction between miR-155 and SHIP1 during JEV infection, BV-2 cells were transfected with a pMIR-REPORT SHIP1 construct, followed by infection with either JEV or UV-treated JEV. The luciferase activity in the JEV-treated group was significantly decreased compared with the pMIR-REPORT SHIP1 construct alone, while the UV-treated group did not show any significant changes in luciferase activity. These results clearly imply that miR-155 targets SHIP1 during JEV infection (Fig. 3D).
miR-155 regulates JEV infection of microglia and mouse brain.
Phosphorylation of TBK-1, IRF3/7, and NF-κB determines induction of inflammatory cytokines and IFN-β secretion. Therefore, we evaluated the effects of in vitro and in vivo knockdown of miR-155 on phosphorylation of TBK-1, IRF3/7, and NF-κB. In BV-2 cells and mouse brains, JEV infection increased TBK-1, IRF3/7, and NF-κB phosphorylation, whereas inhibition of miR-155 led to decreased TBK-1, IRF3/7, and NF-κB phosphorylation (Fig. 4 and and55).
Anti-miR-155 treatment reduces JEV-induced proinflammatory cytokines.
To check if the IFN-β-mediated antiviral response induced during JEV infection can be modulated by miR-155 expression, we evaluated the time kinetics expression of IFN-β in the JEV-infected mouse brain. We observed significantly increased expression of IFN-β mRNA, especially at 5 and 7 days after infection (Fig. 6A). Knockdown of miR-155 in JEV-infected mice (LNA + JEV) significantly reduced IFN-β production compared to the control group (Fig. 6B). We evaluated the JEV-induced proinflammatory response in the mouse brain by measuring protein levels of TNF-α, MCP-1, and IL-6. Time-dependent JEV infection resulted in higher induction of TNF-α, MCP-1, and IL-6 in the mouse brain, with maximum responses at 5 and 7 days (Fig. 6A). Further, in mouse brains, LNA treatment reduced TNF-α, MCP-1, and IL-6 protein expression compared to the JEV group (Fig. 6B).
In vivo anti-miR-155 treatment decreases microglia activation and rescues neurons from death.
Pathological changes, such as elevated levels of proinflammatory cytokines, microglia activation, and neuronal death, are key features of in vivo JEV infection. As shown in Fig. 6A, JEV infection of BALB/c mice resulted in higher levels of brain TNF-α, MCP-1, and IL-6 than in control mouse brain. Inhibition of miR-155 in JEV-infected mice reduced the levels of proinflammatory cytokines. Ionized calcium binding adaptor molecule 1 (Iba1) is commonly used as a marker for microglia activation, while neuronal nuclear antigen (NeuN) is a marker of mature neurons. In the present study, we employed double-immunofluorescence staining using Iba1 and NeuN antibodies to demonstrate microglia activation and associated neuronal death. As shown in Fig. 7A, JEV-infected mouse brain sections had increased numbers of activated microglia and reduced numbers of neurons. Inhibition of miR-155 reduced microglia activation and neuronal death (Fig. 7A). Further, immunoblotting revealed that JEV-infected mouse brains had increased protein levels of Iba1 and NeuN compared to control mice. Additionally, LNA miR-155-treated mouse brains had reduced levels of Iba1 and NeuN proteins compared to JEV-infected mice (Fig. 7B). In anti-miR-155-treated mice, high SHIP1 levels correlated with low inflammatory cytokine levels, and regulation of inflammation by SHIP1 could be a mechanism for the decreased microglia activation and neuronal death observed here.
Anti-miR-155 treatment improves survival and JEV-related symptoms.
The severity of JEV infection determines behavioral and physical symptoms and, ultimately, survival. Based on reduced inflammation in LNA-treated JEV-infected mice, we evaluated the effects of LNA treatment on the JEV symptom score and survival pattern. Seven days after JEV infection, mortality was ~ 60%, and all mice died by 9 days (Fig. 8A). Further, JEV-infected mice showed a progressive increase in disease-related symptoms at days 5 to 9 (Fig. 8B). However, LNA treatment reduced JEV-induced symptoms and improved survival compared to JEV-infected mice that were not treated with LNA (Fig. 8A and andBB).
The IFNs are a group of cytokines that elicit distinct antiviral effects. Type I IFNs, which include IFN-α and IFN-β, are produced in direct response to viral infections (27, 28). Induction of an interferon-mediated antiviral response could be either Toll-like receptor (TLR) dependent or independent, depending on the type of virus (29). The antiviral response during infection determines the efficiency of infection of the host system (28). Inappropriate activation of the antiviral response can lead to cytotoxicity and cell death (30). We and others have demonstrated an IFN-β-mediated antiviral response during JEV infection (31,–34). Recent studies have shown that miRNAs, such as miR-146, miR-155, and miR-203, are involved in regulating type I IFN signaling (30, 35). Based on these observations, we hypothesized that miR-155 regulates IFN-β signaling during JEV infection.
To test our hypothesis, we evaluated miR-155 expression in JEV-infected primary microglia and BV-2 cells and found a time-dependent increase in miR-155 expression. Further, in order to establish in vivo correlation between JEV infection and miR-155 expression, we evaluated miR-155 expression from JEV-infected mouse and human brain samples by qPCR and in situ hybridization. As hypothesized, JEV infection induced significantly higher expression of miR-155 in mouse and human brains. These observations established a role of miR-155 during JEV infection. RIG-I, an RNA helicase molecule is a sensor for JEV infection (31, 32). Further, RIG-I has been shown to induce miR-155 expression (35). Therefore, we speculate that the increased expression of miR-155 following JEV infection observed in the current study is possibly mediated via RIG-I. However, further detailed studies are required to test this hypothesis.
miRNAs exert their effect by binding to the 3′ UTR of the target protein, resulting in its inhibition (11). Previous reports have identified SHIP1 as a potential target of miR-155 (20, 36, 37). SHIP1 is known to repress proliferation, survival, and activation of hematopoietic cells, mainly by translocating to membranes upon stimulation. This results in hydrolyzation of the phosphatidylinositol 3-kinase (PI3K)-generated second-messenger PI-3,4,5-P3 (PIP3) to PI-3,4-P2 (38). In mice, SHIP1 has been shown to be a negative regulator of IFN-β production and lipopolysaccharide (LPS)-induced antibacterial response (39, 40). Hence, we hypothesized that during JEV infection, SHIP1 is involved in mediating miR-155-induced IFN-β signaling. To test this hypothesis, we evaluated SHIP1 expression in JEV-infected BV-2 cells and mouse brain, where SHIP1 expression was significantly lower. We also demonstrated that SHIP1 expression decreases with miR-155 overexpression in BV-2 cells and increases with miR-155 inhibition in BV-2 cells and mouse brain. Additionally, using siRNA-mediated knockdown of SHIP1, we demonstrated that SHIP1 is required for miR-155 to induce inflammation in BV-2 cells. Further, in order to establish interaction of miR-155 with the 3′ UTR of SHIP1, we performed a luciferase reporter assay and observed that BV-2 cells cotransfected with miR-155 and the pMIR-REPORT 3′ UTR/SHIP1 significantly suppressed luciferase activity compared to the pMIR-REPORT 3′ UTR alone, while cotransfection of the 3′ UTR reporter construct with an irrelevant miRNA, miR-29b, showed nonsignificant change in luciferase activity. This compelling evidence indicates that during JEV infection, miR-155 targets SHIP1 by binding to its 3′ UTR. Thus, SHIP1 is negatively regulated by miR-155 expression during JEV infection.
Viral-infection-induced immune response via pattern recognition receptors (PRRs) (TLRs and RIG-I-like receptors) (41, 42) leads to activation of transcription factors, such as NF-κB and IRFs, resulting in induction of proinflammatory cytokines and IFNs (43). The signaling cascade downstream of RIG-I includes activation of the IKK-related kinase IKKα/β and TBK-1, resulting in phosphorylation of IRFs (IRF3 and IRF7). IRF phosphorylation facilitates dimerization and nuclear translocation of the transcription factors and subsequent induction of type I IFNs (44,–46). In our study, we observed a significant upregulation of IFN-β in the JEV-infected mouse brain. To determine if induction of IFN-β during JEV infection is regulated by miR-155 expression, we inhibited miR-155 expression using anti-miR-155 LNA. Interestingly, inhibition of miR-155 significantly decreased the mRNA level of IFN-β, indicating a direct or indirect role of miR-155 in regulating the antiviral response. Further, TBK-1 plays an essential role in TLR- and RIG-I-mediated induction of IFN-α/β and host antiviral responses by regulating activation of NF-κB and IRFs (47,–49). In addition, SHIP1 negatively regulates IFN-β expression by targeting TBK-1 (40). To further correlate induction of miR-155 with regulation of SHIP1, IRFs, and NF-κB activation during JEV infection, we evaluated the in vitro and in vivo effects of inhibiting miR-155 on expression of phosphorylated and total IRF3/7, TBK-1, and phosphorylated NF-κB. The reduced phosphorylation of TBK, IRF3/7, and pNF-κB observed following miR-155 inhibition indicated positive regulation of IRFs and NF-κB activation by miR-155. In addition, inhibition of miR-155 results in higher expression of SHIP1 and subsequent reduction of inflammation. Thus, along with the inhibitory action of SHIP1 on IFN-β production, it also inhibits activation of NF-κB and subsequent synthesis of proinflammatory cytokines by inhibiting phosphorylation of NF-κB (38).
Augmented production of proinflammatory cytokines during JEV infection has been shown to result in microglia activation (50, 51). Activated microglia secrete inflammatory mediators, such as TNF-α, IL-6, RANTES, and MCP-1, which results in central nervous system (CNS) inflammation and neuronal death (52). We were interested in checking if inhibition of miR-155 expression in the in vivo system has any effect on microglia activation and the neuronal population. Brain sections from JEV-infected mice that were treated with LNA miR-155 had reduced activated microglia and neuronal death. In addition, there was a significant reduction in proinflammatory cytokines, such as TNF-α, IL-6, and MCP-1, in LNA-treated mice. In this regard, LNA-mediated reduced inflammation could have prevented microglia activation and subsequent neuronal death resulting from secretion of cytotoxic mediators from activated microglia. Since inhibition of miR-155 increased SHIP1 expression, SHIP1 reduced the expression of inflammatory cytokines by inhibiting NF-κB activation. We conclude that the beneficial effects of LNA miR-155 treatments in JEV-infected mice are likely due to reduced inhibition of SHIP1. Further, we also observed delayed appearance of JEV-related symptoms and improved survival in LNA-treated JEV-infected mice. These results can be an indirect result of reduced proinflammatory cytokine production in LNA-treated mice.
Based on these results, we conclude that during JEV infection, miR-155 promotes inflammation by inhibiting SHIP1 expression, resulting in hyperphosphorylation of TBK-1 and subsequent phosphorylation of IRF3/7 and NF-κB, leading to augmented production of IFN-β and other proinflammatory cytokines (Fig. 9). These data indicate that miR-155 is a potential therapeutic target to modulate JEV-induced neuroinflammation.
This work was supported by a core grant from the Department of Biotechnology to the National Brain Research Centre. Menaka Chanu Thounaojam is the recipient of a research associateship, and Shalini Swaroop is the recipient of a Senior Research Fellowship from the Council of Scientific and Industrial Research, Government of India.
We acknowledge Soumya Iyengar for critically analyzing the in situ hybridization data. We also acknowledge Kanhaiya Lal Kumawat and Manish Dogra for technical assistance and Sourish Ghosh for his valuable suggestions.
Published ahead of print 12 February 2014
Supplemental material for this article may be found at http://dx.doi.org/10.1128/JVI.02979-13.
Articles from Journal of Virology are provided here courtesy of American Society for Microbiology (ASM)
Full text links
Read article at publisher's site: https://doi.org/10.1128/jvi.02979-13
Read article for free, from open access legal sources, via Unpaywall: https://jvi.asm.org/content/jvi/88/9/4798.full.pdf
Citations & impact
Impact metrics
Citations of article over time
Article citations
Regulatory role of microRNAs in virus-mediated inflammation.
J Inflamm (Lond), 21(1):43, 04 Nov 2024
Cited by: 0 articles | PMID: 39497125 | PMCID: PMC11536602
Review Free full text in Europe PMC
Short-chain fatty acids abrogate Japanese encephalitis virus-induced inflammation in microglial cells via miR-200a-3p/ZBTB20/IKβα axis.
mBio, 15(7):e0132124, 13 Jun 2024
Cited by: 0 articles | PMID: 38869276 | PMCID: PMC11253640
The long noncoding RNA loc107053557 acts as a gga-miR-3530-5p sponge to suppress the replication of vvIBDV through regulating STAT1 expression.
Virulence, 15(1):2333237, 31 Mar 2024
Cited by: 1 article | PMID: 38528779 | PMCID: PMC10984138
Single-cell RNA sequencing reveals the immune features and viral tropism in the central nervous system of mice infected with Japanese encephalitis virus.
J Neuroinflammation, 21(1):76, 26 Mar 2024
Cited by: 2 articles | PMID: 38532383 | PMCID: PMC10967088
The Role of Noncoding RNA in the Transmission and Pathogenicity of Flaviviruses.
Viruses, 16(2):242, 02 Feb 2024
Cited by: 0 articles | PMID: 38400018 | PMCID: PMC10892091
Review Free full text in Europe PMC
Go to all (78) article citations
Data
Data behind the article
This data has been text mined from the article, or deposited into data resources.
BioStudies: supplemental material and supporting data
Similar Articles
To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.
MicroRNA-19b-3p Modulates Japanese Encephalitis Virus-Mediated Inflammation via Targeting RNF11.
J Virol, 90(9):4780-4795, 14 Apr 2016
Cited by: 63 articles | PMID: 26937036 | PMCID: PMC4836334
MicroRNA-15b Modulates Japanese Encephalitis Virus-Mediated Inflammation via Targeting RNF125.
J Immunol, 195(5):2251-2262, 22 Jul 2015
Cited by: 72 articles | PMID: 26202983
miR-301a Regulates Inflammatory Response to Japanese Encephalitis Virus Infection via Suppression of NKRF Activity.
J Immunol, 203(8):2222-2238, 16 Sep 2019
Cited by: 19 articles | PMID: 31527198
Pathogenicity and virulence of Japanese encephalitis virus: Neuroinflammation and neuronal cell damage.
Virulence, 12(1):968-980, 01 Dec 2021
Cited by: 43 articles | PMID: 33724154 | PMCID: PMC7971234
Review Free full text in Europe PMC