Analysis of Long Noncoding RNA and mRNA Profiling in Peripheral Blood Mononuclear Cells of Human Immunized With Rabies Virus Vaccine by RNA Sequencing

DOI: https://doi.org/10.21203/rs.3.rs-50067/v1

Abstract

Background

Rabies is still a lethal disease caused by rabies virus (RABV), and it remains a global health threaten. The epigenetic landscape of host in response to immunization with rabies virus vaccine is not yet elucidated. Evidences reveal that long noncoding RNA (lncRNA) play an important role for control of viral infection, but the expression profile of lncRNA in human immunized with rabies virus vaccine remains unclear.

Methods

lncRNA and mRNA profiles were investigated in four volunteers vaccinated with RABV vaccine by RNA sequencing.

Results

33 lnRNAs and 427 mRNAs were differentially expressed in RABV vaccine immunized volunteers. The gene oncology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were used to analysis the functional annotation, and the result suggested the lncRNAs involved in signaling pathways related to host immune induced by the RABV vaccine.

Conclusions

To the best of our knowledge, it is the first time to report the transcriptomic landscape of lncRNAs in human immunized with RABV vaccine. Our study mimics the host response of human to RABV infection and suggests that lncRNAs might be of a potential therapeutic target in anti-rabies virus strategies.

Background

Rabies is not only one of the oldest diseases in human history, also is one of the deadliest zoonosis disease[1]. Since the clinical symptoms had developed, the mortality rate is almost 100%[2]. No matter in developing or developed country, the rabies viral (RABV) post a serious threat to the publish health [3]. Annually the more than 70000 human deaths result from the rabies viral infection and Asia is the disaster stricken area among the worldwide[4].

RABV belongs to the family Rhabdoviridae, genus Lyssavirus, and it is one of the most important zoonosis affecting the central nervous of mammals [5]. RABV has an approximately 12 kb negative-sense RNA genome which encodes five structural proteins: the nucleoprotein (N), phosphoprotein (P), matrix protein (M), glycoprotein (G) and RNA-dependent RNA polymerase (L) [6]. Following infection, the RABV virus replicates locally in muscle tissue and then travelled within axons in the peripheral nervous system (PNS) via retrograde axonal [7]. Once the clinical symptoms of rabies had developed, it elicit neuronal dysfunction and lead to death[8].

During the infection, the host innate immune system sensed the microorganisms invasion and induced the cytokines secretion by pattern-recognition receptors (PRRs)[9, 10].The cytokines contains type I interferons (IFNα/β), inflammatory cytokines, and chemokines and so on. Here in the RABV infection, the IFN-mediated immune response is the most important way in antiviral state [11, 12]. The type I IFN-IFNR signal way activates Janus-activated kinase/signal transducer(JAK/STAT)[13], result in the transcription of hundreds of IFN-stimulated genes(ISGs)[14, 15]. ISGs exert diverse antiviral effects as the effector of type I IFN response [16, 17], and inhibit the IFN mediated respond against RABV infection aggravate viral infection in vitro and in vivo [18, 19]. Although some studies demonstrated the importance of IFN mediated response against RABV infection, the mechanism by which RABV causes fatal disease remains unclear.

RABV infection can induce the host immune system responds, and the long noncoding RNAs (lncRNA) affect many cellular processes as an effector in the immune system during the infection[20, 21], such as differentiation[22], apoptosis[23], development[24], and immune responses[25]. Studies had reported that viral infections such as influenza (IAV)[26], HIV[27], hepatitis B[28] could induce specific lncRNA promote or suppress antiviral responses[29]. Most ISGs and some cellular factor play a role of antiviral or proviral function and it had been shown that IFN pathway regulates the expression of some lncRNAs. Viral-induced lncRNAs should decrease the antiviral response, while cellular lncRNAs induced by infection could either induce the pathway or decrease IFN synthesis and signaling to control the duration and the strength of the antiviral response[3032]. Hironori et al., report that the lncRNA#32 silence could reduce ISG expression, exerted anti-virus infection which suggest lncRNA#32 positively regulates the host antiviral response[33].However, little is known about lncRNA expression profile after RABV infection and their regulating role in innate immune against RABV. In our previous work, we first had analysis the expression profiles of lncRNAs and mRNAs in brains of mice infected by rabies virus and found some differentially expressed lncRNA and mRNA[34]. What’s more, we are studying the mechanism of some selected lncRNA anti-viral function.

To investigate how the lncRNA respond during the RABV infection, we use a commercial RABV vaccine to develop a model to simulate the viral induced immune respond. We analyzed the lncRNA expression profile in peripheral blood mononuclear cells (PBMCs) of volunteers after RABV vaccine immunized by RNA-SEq. The data reveal that RABV vaccine induced significant expression of lncRNA and mRNA. Gene ontology (GO) and KEGG analysis suggested lncRNA involve in the host immune response induce by the RABV vaccine. To our knowledge, this is the first study to profile the lncRNA expression induced by the RABV vaccine and the finding base on the RABV vaccine would be useful for the understanding the infection between RABV and host immunity.

Methods

Experimental design

All the volunteers enrolled for this study had all signed the Informed Consent, and the study was performed in accordance with the Declaration of Helsinki and was approved by the Research Ethics Committee of Yuebei People’s Hospital, Shantou University Medical College, Shaoguan, China.

All the four healthy volunteers with no rabies virus vaccine history, were recruited to receive three doses of Rabies Vaccine (Vero Cell) for Human Use (China Food and Drug Admistration approval number: S20120016) (Changsheng Bio-technology Co., Ltd). The vaccine is a fixed rabies vaccine virus aG strain, which was inoculated into Vero cells and culture in a bioreactor microcarries. After culture, the virus solution was harvested, inactivated, purified and lyophilized with appropriate stabilizers. The titer of the rabies vaccine is not lower than 2.5 IU. The vaccinate procedure was followed the pre-exposure procedure according to the manufacturer's instructions. It contained 3 injections: 1 dose was injected on 0 days, 7 days, 21 days (Fig. 1). And 10 ml peripheral blood samples were obtained from the volunteers at day 0 before vaccinated and the 28-days, the PBMCs had been separated immediately using Ficoll-Paque (GE Healthcare) gradient centrifugation for further sequence.

Total Rna Extraction

Total RNA was extracted from PBMC according to the manufacturer’s protocol (TRIzol™ Reagent, Invitrogen). The quantity and purity of total RNA were evaluated by Nanodrop 2000. The ratio of A260/A280 should be from 1.8 to 2.0. RNA integrity was analyzed by the Bioanalyzer 2100 system (Agilent Technologies, CA, USA).

High Throughput Sequencing

Ribo-Zero rRNA removal kit (Epicentre, USA) was used to remove ribosomal RNA. The rRNA-depleted RNA with NEBNext® Ultra™ Directional RNA Library Prep Kit (NEB, USA) for Illumina was used for preparation of the RNA libraries. Library sequencing was performed on an Illumina HiSeq 2000 platform with 150 bp read length in ShenZhen Realomics Inc.

Bioinformatic Analysis

With the Illumina HiSeq 2000 platform, The sequence raw data was filtered by the low quality reads, like the adaptors, low-quality reads and poly-N reads, to obtain clean data using the SOAPnuke. The filtered contain removal of adapter reads; removal of low-quality reads (the base number of sQ < = 5 accounts for more than 50% of the whole read); removal of the base information that undetermined ratio of reads is more than 10%. The QC checks is performed on the clean data to determine the suitable data for subsequent analysis, such as the Q20, Q30, and GC information. The filtered reads were mapped to the human reference genome by Tophat2 (Version 2.1.1).Then the mapped reads were assembled by reference annotation with transcripts method Cufflink (Version 2.2.1) [35]. LncRNA transcripts exceed 200nt and do not encode proteins. According to the characteristics of the lncRNA structure and the functional characteristics, the candidate novel lncRNA is obtained should accord with the followed requirement: (1) exon number ≥ 2, (2) length > 200 nt, (3) FPKM ≥ 0.5, (4) without coding capacity, (5) don’t overlap with mRNA or annotated lncRNA. Coding ability was predicted using coding-non-coding-index (CNCI)[36], coding potential calculator (CPC)[37] and coding-potential assessment tool (CPAT)[38]. Cuffdiff (version 2.2.1) was used to analysis the expression, the function analysis was performed with the GO (www.geneontology.org) and KEGG (http://www.genome.ad.jp/kegg/). GO terms analyses were performed to identify biological processes enriched in neighboring genes of predicted co-localized lncRNA. KEGG was used to analyze the enriched pathways of co-expressed genes for the predicted co-localized lncRNA. Q values < 0.05 were considered as significantly enriched pathways.

Quantitative Real-time Pcr Validation

The surplus of the RNA sample for the RNA-Seq was used for quantitative real-time polymerase chain reaction (RT-qPCR) validation. RT-qPCR was used the Luna® Universal One-Step RT-qPCR Kit (NEB, USA) in LightCycler480 II (Roche, USA) according to the manufacturer’s instructions. The reaction conditions were set as follows: reverse transcription at 55℃ for 10 min, initial denaturation at 95℃ for 1 min, followed by 40 cycles of 95 °C for 10 s, 60 °C for 30 s, and 72℃ for 30 s. The specificity of the amplified products was evaluated by using dissociation curves. The relative expression levels of lncRNAs were calculated using the 2−ΔΔCt method and were normalized by GADPH. The tests were triplicated.

Statistical analysis

Data were analyzed using Student’s t-test or one-way ANOVA followed by Dunnett’s multiple comparison test (compare post-immunized groups to the pre-immunized group). All data are demonstrated as the means ± S.D. (*P < 0.05, **P < 0.01, ***P < 0.001). For correlation studies, a two-tailed non-parametric Spearman analysis was used. P ≤ 0.05 were considered as significant.

Results

Identification of differentially expressed lncRNAs in PBMCs of human immunized with rabies virus vaccine by RNA-seq

To investigate the lncRNA expression profile in the volunteers induced by the RABV vaccine, high throughput RNA sequencing was performed on the PBMC of volunteers immunized with RABV vaccine. We sequenced 8 rRNA-deprived total RNA samples obtained from 4 volunteers pre- and post-immunized with RABV vaccine. Average 86 million raw reads were produced from Illumina HiSeq platform. By the bioinformatic analysis, the raw data was filter out for further analysis.

Because of the specific structure and non-coding characteristics of lncRNA, 5-step scanned was used to analysis the transcripts to identify the annotated lncRNA and novel lncRNA. Finally 558 novel lncRNA were assembled by Cuffilink (Fig. 2A). CNCI, CPC and CPAT were used to evaluate the coding capacity of transcripts (Fig. 2B). Moreover, with the relative location to the coding genes, the lncRNA identified were divided into five classifications include intergenic lncRNA (74.55%), intronic lncRNA (10.22%), antisense lncRNA (6.63%), enhancer lncRNA (3.41%) and bidirectional lncRNA (5.20%) (Fig. 2C).

The lncRNA expression profiles in eight sample of four volunteers pre- and post-immunized with RABV vaccine were analyzed with hierarchical clustering. The result showed that lncRNA expression profiles were significantly modified after RABV vaccine immunized (Fig. 3A). There was 33 lncRNA were expressed significant different, including 1 upregulated and 32 downregulated (Fig. 3B). The upregulated lncRNA was ENSG00000254162 with a fold change (FC) of 4.38 compared to the post-immunized and the downregulated lncRNA had a FC range 1.75 to 7.07 (Table 1). All the differentially expressed lncRNA were listed in Table 1. The number distribution of lncRNA in all chromosomes was display in Fig. 3C and it showed that chromosome 5 had the highest number of lncRNA, while chromosome 3, 4, 6 and so on had only one lncRNA. However, several chromosomes didn’t have any significant different lncRNA like chromosome 2, 10, et.al (Fig. 3C).

Identification of differentially expressed mRNAs in PBMCs of human immunized with rabies virus vaccine by RNA-seq

We examined the different expression of mRNA after the RABV vaccine immunized, and significant difference expression of mRNA induced by RABV vaccine was showed by hierarchical cluster analysis (Fig. 4). In our study, 427 mRNA were differentially expressed after RABV vaccine immunized, including 321upregulated and 106 downregulated (Fig. 4B). In the different expression genes, the most upregulated one was HSPA1B (FC = 14.84) and the top 3 downregulated were CLC (FC = 27.00), DEFA3 (FC = 15.54) and CAMP (FC = 11.96). The top 20 differentially expressed genes were listed in Table 2. Unlike the lncRNA, the mRNA distributed to all the chromosome but were not equally scattered among chromosomes. The chromosome 1 had the most differentially expressed mRNAs and chromosome 14 and 18 had the least numbers (Fig. 4C).

Table 1. Differentially expressed lncRNAs in peripheral blood mononuclear cells of human immunized by RABV vaccine by RNA sequencing.

Gene_id

Isoform_status

Post-immunized

Pre-immunized

log2FoldChange

p_value

q_value

Dysregulated

FC

ENSG00000245164

lncRNA_annotated

5.34448

9.3865

-0.81254

0.0002

0.020468

Down

1.756297

XLOC_042681

lncRNA_Novel

2.31781

4.21036

-0.86119

5.00E-05

0.006587

Down

1.81653

ENSG00000215458

lncRNA_annotated

6.22621

11.3207

-0.86254

5.00E-05

0.006587

Down

1.818237

ENSG00000261490

lncRNA_annotated

3.0043

5.63895

-0.9084

0.00045

0.038471

Down

1.876961

ENSG00000269821

lncRNA_annotated

1.20099

2.28741

-0.92949

5.00E-05

0.006587

Down

1.904599

XLOC_051604

lncRNA_Novel

2.42931

4.71665

-0.95722

5.00E-05

0.006587

Down

1.94156

ENSG00000281103

lncRNA_annotated

4.09089

8.10076

-0.98564

0.0001

0.011848

Down

1.980194

ENSG00000266962

lncRNA_annotated

6.01747

12.0116

-0.99721

0.0005

0.041152

Down

1.996129

XLOC_018471

lncRNA_Novel

0.977134

1.95301

-0.99907

0.00015

0.016397

Down

1.998714

XLOC_024827

lncRNA_Novel

0.902814

1.94629

-1.10822

5.00E-05

0.006587

Down

2.155795

ENSG00000235314

lncRNA_annotated

3.66534

8.05664

-1.13623

0.00055

0.044236

Down

2.198059

XLOC_056429

lncRNA_Novel

1.21387

2.79502

-1.20324

0.0005

0.041152

Down

2.302562

XLOC_050760

lncRNA_Novel

2.77627

6.39987

-1.2049

5.00E-05

0.006587

Down

2.305213

XLOC_056637

lncRNA_Novel

0.509636

1.20423

-1.24057

5.00E-05

0.006587

Down

2.362919

XLOC_049174

lncRNA_Novel

0.774507

1.85106

-1.257

5.00E-05

0.006587

Down

2.389982

XLOC_041020

lncRNA_Novel

1.62414

3.8977

-1.26295

5.00E-05

0.006587

Down

2.39986

XLOC_059380

lncRNA_Novel

0.770727

1.85765

-1.26919

0.00045

0.038471

Down

2.410262

XLOC_062407

lncRNA_Novel

0.472431

1.20236

-1.3477

0.0002

0.020468

Down

2.545061

XLOC_023926

lncRNA_Novel

0.560945

1.42784

-1.3479

0.00065

0.049892

Down

2.545413

XLOC_016347

lncRNA_Novel

0.82775

2.12904

-1.36294

5.00E-05

0.006587

Down

2.572088

XLOC_051558

lncRNA_Novel

0.922673

2.38183

-1.36818

5.00E-05

0.006587

Down

2.581447

ENSG00000230733

lncRNA_annotated

0.824332

2.15159

-1.38411

0.00025

0.024656

Down

2.610109

XLOC_051602

lncRNA_Novel

0.684579

1.84391

-1.42948

5.00E-05

0.006587

Down

2.693496

XLOC_051172

lncRNA_Novel

0.654121

1.98247

-1.59967

5.00E-05

0.006587

Down

3.03074

XLOC_066116

lncRNA_Novel

0.83044

2.59132

-1.64174

5.00E-05

0.006587

Down

3.12042

ENSG00000214894

lncRNA_annotated

1.09692

3.73051

-1.76592

5.00E-05

0.006587

Down

3.400908

XLOC_003374

lncRNA_Novel

1.10497

3.8463

-1.79947

5.00E-05

0.006587

Down

3.480923

XLOC_005414

lncRNA_Novel

0.533678

1.92461

-1.85053

0.0002

0.020468

Down

3.606326

XLOC_062716

lncRNA_Novel

0.399621

1.84311

-2.20543

5.00E-05

0.006587

Down

4.61212

XLOC_005417

lncRNA_Novel

0.363757

1.69681

-2.22177

5.00E-05

0.006587

Down

4.664654

ENSG00000197182

lncRNA_annotated

0.408312

1.96928

-2.26992

0.0004

0.035132

Down

4.822964

XLOC_000161

lncRNA_Novel

0.492994

3.48758

-2.82259

5.00E-05

0.006587

Down

7.074313

ENSG00000254162

lncRNA_annotated

3.30507

0.75435

2.13138

5.00E-05

0.006587

Up

4.381364

 

Table 2. Differentially expressed mRNAs in peripheral blood mononuclear cells of human immunized by RABV vaccine by RNA sequencing

Gene_id

Gene

Isoform_status

Post-immunized

Pre-immunized

log2FoldChange

p_value

q_value

Dysregulated

FC

ENSG00000105205

CLC

mRNA

1.33294

35.9944

-4.75509

5.00E-05

0.006587

Down

27.00379

ENSG00000239839

DEFA3

mRNA

4.43212

68.8862

-3.95815

5.00E-05

0.006587

Down

15.54254

ENSG00000204388

HSPA1B

mRNA

78.3372

5.27778

3.89169

5.00E-05

0.006587

Up

14.84279

ENSG00000164047

CAMP

mRNA

0.592677

7.0903

-3.58053

0.00065

0.049892

Down

11.96319

ENSG00000125740

FOSB

mRNA

96.1971

11.1305

3.11147

5.00E-05

0.006587

Up

8.642628

ENSG00000162772

ATF3

mRNA

23.1743

3.18224

2.86441

5.00E-05

0.006587

Up

7.28238

ENSG00000111537

IFNG

mRNA

11.2211

1.54555

2.86002

0.0002

0.020468

Up

7.260254

ENSG00000276085

CCL3L3

mRNA

51.5995

7.47768

2.7867

5.00E-05

0.006587

Up

6.900496

ENSG00000090104

RGS1

mRNA

52.4469

7.78688

2.75174

5.00E-05

0.006587

Up

6.73529

ENSG00000277632

CCL3

mRNA

80.3514

12.1697

2.72303

5.00E-05

0.006587

Up

6.602581

ENSG00000141682

PMAIP1

mRNA

49.184

8.15661

2.59215

5.00E-05

0.006587

Up

6.029967

ENSG00000125538

IL1B

mRNA

62.8604

10.7286

2.55069

5.00E-05

0.006587

Up

5.859144

ENSG00000118503

TNFAIP3

mRNA

263.218

50.6895

2.3765

5.00E-05

0.006587

Up

5.192754

ENSG00000185022

MAFF

mRNA

32.1323

6.22367

2.36818

5.00E-05

0.006587

Up

5.162894

ENSG00000255398

HCAR3

mRNA

14.3272

2.78187

2.36463

5.00E-05

0.006587

Up

5.150206

ENSG00000114315

HES1

mRNA

2.92857

0.57765

2.34193

0.00015

0.016397

Up

5.069804

ENSG00000110848

CD69

mRNA

219.951

43.9565

2.32303

5.00E-05

0.006587

Up

5.00382

ENSG00000169429

CXCL8

mRNA

24.4288

4.93953

2.30614

5.00E-05

0.006587

Up

4.945581

ENSG00000143507

DUSP10

mRNA

35.2674

7.27819

2.27668

5.00E-05

0.006587

Up

4.845616

ENSG00000112149

CD83

mRNA

62.9776

13.0436

2.2715

5.00E-05

0.006587

Up

4.828249

 

Genomic Features Of Lncrnas And Mrnas

To know more about the genomic features of the lncRNA and mRNA in volunteers, we systematically analyzed the feature of lncRNA in this study and compared them with protein-coding genes. The result show that the average expression level of lncRNA were lower than mRNAs (Fig. 5A). The exons number and the lengths of lncRNA was also less and shorter than that of mRNAs (Fig. 5B, C) Furthermore, most of the mRNAs had a longer ORFs than the lncRNA (Fig. 5D).

Functional Prediction Of Rabv Vaccine Induced Co-localized Lncrnas

To better understand the functions of differentially expressed co-localized lncRNA in RABV vaccine immunized volunteers, GO term and KEGG pathway analysis was performed. The GO analysis result reveal that the genes colocate with differentially expressed lncRNA were highly enriched in biological processes like extracellular vesicular exosome, extracellular organelle and extracellular membrane-bounded organelle process. The top 20 GO terms are presented (Fig. 6A).

What’s more, KEGG was also used to investigate the involved biological pathways of the differentially expressed lncRNA. The top20 significant enriched pathways terms exhibited in Fig. 6B. Some infectious disease pathways were shown, such Chagas disease (American trypanosomiasis), malaria, tuberculosis, herpes simplex infection, hepatitis C and measles (Fig. 6B). These finding suggested that lncRNA regulate the infectious process after RABV vaccine immunized.

Validation Of Selected Differentially Expressed Lncrnas By Rt-qpcr

To valiate the rabies vaccine induced lncRNA expression changes detected by RNA-seq, five downregulated expressed lncRNA (XLOC_005414, XLOC_062716, XLOC_005417, ENSG00000197182, XLOC_000161) and one upregulated expressed lncRNA (ENSG00000254162) were selected to validate by using RT-qPCR. The results were shown in Fig. 7. Both the results of RT-qPCR were similar to the RNA-seq result, which implicated the good reliability and reproducibility of the lncRNA changes determined by RNA-seq.

Discussion

Rabies is one of the most deadly infectious diseases and was fatal once the clinical symptoms developed, and it remains a threat to public health. Currently, without certain cure, rabies can be prevented by rabies immune globulin (RIG) or rabies vaccine injection once a bite or exposure to saliva from an infected animal. As a result, annually almost 59,000 global human deaths caused by rabies and estimated 15 million people receive post-exposure prophylaxis annually for exposures [39]. Pre-exposure prophylaxis (PEP) protect the high-risk group with the presence of rabies-specific virus-neutralizing antibodies (VNAs). Unfortunately, the pre-exposure prophylaxis or post-exposure prophylaxis was not only need multiple injections but also time-consuming and expensive [40]. Now, there is an urgent need to develop a new biologicals to potentially replace existed prophylaxis with cost-effective alternatives to the human rabies elimination.

During the infection, the viral induce the host innate immune system and secretion lots cytokines, chemokine like type I IFN [9, 10]. And the type I IFN active JAK/STAT signaling pathway result in the expression of ISGs, which had diverse antiviral functions [1315]. A important feature of the RABV is that the RABV can replicate in the central nervous system (CNS) where was an immunologically privileged area of the host [41]. Therefore, when the RABV infected like bites or scratches, the RABV G protein binds to the nicotinic acetylcholine receptor and later enter the CNS. When the RABV exposed to the innate and adaptive immune responses, it induced some molecules and chemokines, like NF-κB, type I IFN-regulated responses and toll-like receptors (TLR) to against the elimination [42, 43] [44]. Though lots efforts had done to investigate the RABV biology and anti-RABV immune response, the mechanism of how the RABV escape the elimination in the host immune response is still need deep-going.

In our previous work, we found that the RABV infection can significantly change the protein-coding profile of host cell in vitro by RNA-sequencing, and identified some genes that function against viral replication, such as ISG15 and UBA7 [45, 46]. What’s more, we also investigated the host immune response during the RABV infection in a mouse model and found something interesting (unpublished data), but how the human host immune response during the infection is still nuclear. Raising data had suggested that lncRNA involve in modulated many biological process like gene epigenetic modulation, protein scaffolding, cell development and more [47]. LncRNAs also have been implicated in the pathogenesis and response to bacterial and viral infections. Reports had reveal that HIV[27], SARS-CoV[48] and some viral can induce lncRNA differentially expressed. On the other way out, the viral-induced IFN pathway can regulate the expression of several lncRNA [49]. Accumulating evidences supported that many viral infections induced specific lncRNA which in turn play an antiviral role in host immune responses [21, 50]. Though cellular lncRNA were reported in many virus infections, i.e. enterovirus, influenza, hepatitis B and C viruses, the role of lncRNA in the RABV infection remains unclear.

Here we analysis the expression profile of lncRNA and mRNA in PBMC after RABV vaccine immunized. A total of 33 lnRNA and 427 mRNA were differentially expressed in RABV vaccine immunized volunteers. In the lncRNA, only one annotated lncRNA ENSG00000254162 was upregulated with a FC of 4.38 and the other 32 lncRNA were downregulated with a FC range from 1.75 to 7.07 (Fig. 3). To be noted, five of the top 6 downregulated lncRNA were novel ones (Table 1). With the Ensembl genome browser and NCBI data base, lookuped all the differentially expressed lncRNA, we found that it involved the chromatin regulatory like ENSG00000269821 and hormonal regulatory like ENSG00000266962 .However, the only one upregulated lncRNA ENSG00000254162 was short of research and whether the novel lncRNA were relevant with RABV vaccine immunization is still need to be investigate. In the 427 differential expressed mRNAs, 321 of them were upregulated and 106 were downregulated. The top 10 differential expression mRNA contained 3 down regulated mRNA and seven upregulated mRNA with a FC range from 6.60 to 27.00 (Table 2). Like DEAF3 (GENE ID: ENSG00000239839, defensin alpha 3) and CAMP (GENE ID: ENSG00000164047, cathelicidin antimicrobial peptide) are both belong to the antimicrobial peptide. They are the first line of defense against a wide range of pathogens. Data had proved that defensin can inhibit some viral infection, like HIV, influenza A virus (IAV) and so on [5153]. Also, data showed that the mRNA expression of INF-γ and activating transcription factor 3 (ATF3) were higher than pre-immunized. What’s more, ATF3 can modulate the IFN -γ expression to regulate viral infection in mice [54] [55]. It demonstrated that RABV vaccine can induce host immune against the viral infection.

Base on the unique structure and characteristic unlike protein coding genes and microRNAs, lncRNA sequences are currently uninformative for predicting function[56]. The regulation effects of lncRNA are mainly by regulating the expression of the neighboring protein coding genes [57]. Here GO term and KEGG pathway analysis was performed to investigate the functions of differentially expressed lncRNA in RABV vaccine immunized volunteers. GO terms were significantly enriched in biological processes like extracellular vesicular exosome, extracellular organelle and extracellular membrane-bounded organelle process. The involved biological pathways of the differentially expressed lncRNA analysis by KEGG contained many infectious diseases like Chagas disease (American trypanosomiasis), malaria, tuberculosis, herpes simplex infection, hepatitis C and measles. What’s more, the viral induced immune responses pathway like the toll-like receptor signaling pathway and TGF-beta signaling pathway were significant enriched pathways. Taking together, it suggested that lncRNA take part in host immune response during the RABV vaccine immunized through various pathways.

Conclusions

In a brief, it is first to reveal the lncRAN expression profile and function of differentially expressed lncRNA in RABV vaccine immunized model which mimic the RABV infection in human. In the present study, some lncRNA are identified and the data suggested lncRNA play an important role in regulating gene expression after RABV vaccine immunized and exert important biological effects. It may be very valuable for the development of new effective therapeutic drugs for rabies or provide novel strategies for vaccine designation.

Abbreviations

RABV

rabies virus; lncRNA:long noncoding RNA; GO:gene oncology; KEGG:Kyoto Encyclopedia of Genes and Genomes; JAK/STAT:Janus-activated kinase/signal transducer; ISGs:IFN-stimulated genes; PBMCs:peripheral blood mononuclear cells; RT-qPCR:quantitative real-time polymerase chain reaction; FC:fold change; RIG:rabies immune globulin; PEP:Pre-exposure prophylaxis; VNAs:virus-neutralizing antibodies; TLR:toll-like receptors; CNS:central nervous system.

Declarations

Authors’ contributions

The study was designed by PZ. PZ, KH and PZ drafted the manuscript. WJ, ZP, and GH contributed to the field research. PZ and KH completed the experiment. PZ, KH, PZ, SY and XX collected and analyzed the data. All authors assisted in reviewing and editing the manuscript. All authors read and approved the final manuscript.

Funding

This study was supported by The National Key Research and Development Program of China (Grant No.: 2016YFD0500405 to Dr. Pingsen Zhao), The National Key Research and Development Program of China (Grant No.: 2017YFD0501705 to Dr. Pingsen Zhao) and Natural Science Foundation of Guangdong Province, China (Grant No.: 2016A030307031 to Dr. Pingsen Zhao).

Availability of data and materials

All data analyzed during the study are included in this published article (and its supplementary information files) and are available from the included studies, which are fully referenced.

Ethical conduct of research

The authors state that they have obtained appropriate institutional review board approval or have followed the principles outlined in the Declaration of Helsinki for all animal experimental investigations. All animal experiments were performed following the National Institute of Health Guide for the Care and Use of Laboratory Animals, and the experimental protocols were approved by the Research Ethics Committee of Yuebei People’s Hospital, Shantou University Medical College, Shaoguan, China.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Author details

1Department of Laboratory Medicine, Yuebei People’s Hospital, Shantou University Medical College, Shaoguan, China

2Shaoguan Municipal Quality Control Center for Laboratory Medicine, Shaoguan, China

3Department of Endocrine and Metabolic Diseases, Longhu Hospital, The First Affiliated Hospital of Shantou University Medical College, Shantou, China

4West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China

5Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China

References

  1. Davis BM, Rall GF, Schnell MJ. Everything You Always Wanted to Know About Rabies Virus (But Were Afraid to Ask). Annu Rev Virol. 2015;2(1):451–71.
  2. Rupprecht CE, Hanlon CA, Hemachudha T. Rabies re-examined. Lancet Infect Dis. 2002;2(6):327–43.
  3. Dato VM, Campagnolo ER, Long J, et al. A Systematic Review of Human Bat Rabies Virus Variant Cases: Evaluating Unprotected Physical Contact with Claws and Teeth in Support of Accurate Risk Assessments. PLoS One. 2016;11(7):e0159443.
  4. Knobel DL, Cleaveland S, Coleman PG, et al. Re-evaluating the burden of rabies in Africa and Asia. Bulletin of the World Health Organization. 2005;83(5):360.
  5. Rupprecht CE, Hanlon CA, Hemachudha T. Rabies re-examined. Lancet Infectious Diseases. 2002;2(6):327–43.
  6. Wunner WH, Larson JK, Dietzschold B, et al., The molecular biology of rabies viruses, Rev Infect Dis. 10 Suppl 4 (1988) S771-784.
  7. Coulon P, Derbin C, Kucera P, et al. Invasion of the peripheral nervous systems of adult mice by the CVS strain of rabies virus and its avirulent derivative AvO1. J Virol. 1989;63(8):3550–4.
  8. Lafon M. Evasive strategies in rabies virus infection. Adv Virus Res. 2011;79:33–53.
  9. Iwasaki A. A Virological View of Innate Immune Recognition. Annual Review of Microbiology. 2012;66(66):177.
  10. Goubau D, Deddouche S, Reis, eSC. Cytosolic sensing of viruses. Immunity. 2013;38(5):855–69.
  11. Ivashkiv LB, Donlin LT. Regulation of type I interferon responses. Nat Rev Immunol. 2014;14(1):36–49.
  12. Faul EJ, Wanjalla CN, Suthar MS, et al. Rabies virus infection induces type I interferon production in an IPS-1 dependent manner while dendritic cell activation relies on IFNAR signaling. Plos Pathogens. 2013;6(7):e1001016.
  13. Levy DE Jr. DJ, STATS: Transcriptional control and biological impact. Nature Reviews Molecular Cell Biology. 2002;3(9):651–62.
  14. Macmicking JD. Interferon-inducible effector mechanisms in cell-autonomous immunity. Nat Rev Immunol. 2012;12(5):367–82.
  15. Schoggins JW, Wilson SJ, Panis M, et al. A diverse range of gene products are effectors of the type I interferon antiviral response. Nature. 2011;472(7344):481–5.
  16. Schoggins JW, Wilson SJ, Panis M, et al. A diverse range of gene products are effectors of the type I interferon antiviral response. Nature. 2011;472(7344):481–5.
  17. Schoggins JW, MacDuff DA, Imanaka N, et al. Pan-viral specificity of IFN-induced genes reveals new roles for cGAS in innate immunity. Nature. 2014;505(7485):691–5.
  18. Zhao P, Jiang T, Zhao L, et al. Inhibition of rabies virus replication by interferon-stimulated gene 15 and its activating enzyme UBA7. Infection Genetics Evolution. 2017;56:44.
  19. Chopy D, Detje CN, Lafage M, et al. The type I interferon response bridles rabies virus infection and reduces pathogenicity. J Neurovirol. 2011;17(4):353–67.
  20. Li X, Wu Z, Fu X, et al. Long Noncoding RNAs: Insights from Biological Features and Functions to Diseases. Med Res Rev. 2013;33(3):517–53.
  21. Zhang Q, Jeang KT. Long noncoding RNAs and viral infections. Biomedicine. 2013;3(1):34–42.
  22. Wang P, Xue Y, Han Y, et al. The STAT3-binding long noncoding RNA lnc-DC controls human dendritic cell differentiation. Science. 2014;344(6181):310–3.
  23. Huarte M, Guttman M, Feldser D, et al. A large intergenic noncoding RNA induced by p53 mediates global gene repression in the p53 response. Cell. 2010;142(3):409–19.
  24. Sauvageau M, Goff LA, Lodato S, et al. Multiple knockout mouse models reveal lincRNAs are required for life and brain development. Elife. 2013;2:e01749.
  25. Heward JA, Lindsay MA. Long non-coding RNAs in the regulation of the immune response. Trends Immunol. 2014;35(9):408–19.
  26. Winterling C, Koch M, Koeppel M, et al. Evidence for a crucial role of a host non-coding RNA in influenza A virus replication. RNA Biol. 2014;11(1):66–75.
  27. Zhang Q, Chen CY, Yedavalli VS, et al., NEAT1 long noncoding RNA and paraspeckle bodies modulate HIV-1 posttranscriptional expression, MBio. 4 (1) (2013) e00596-00512.
  28. Du Y, Kong G, You X, et al. Elevation of highly up-regulated in liver cancer (HULC) by hepatitis B virus X protein promotes hepatoma cell proliferation via down-regulating p18. J Biol Chem. 2012;287(31):26302–11.
  29. Fortes P, Morris K. Long noncoding RNAs in viral infections. Virus Res. 2016;212:1–11.
  30. Krishnamurthy M, Beihua D, Michael G, et al. Small self-RNA generated by RNase L amplifies antiviral innate immunity. Nature. 2007;448(7155):816.
  31. Barriocanal M, Carnero E, Segura V, et al. Long Non-Coding RNA BST2/BISPR is Induced by IFN and Regulates the Expression of the Antiviral Factor Tetherin. Frontiers in Immunology. 2014;5:655.
  32. Ouyang J, Zhu X, Chen Y, et al, NRAV, a Long Noncoding RNA, Modulates Antiviral Responses through Suppression of Interferon-Stimulated Gene Transcription, Cell Host & Microbe. 16 (5) (2014) 616–626.
  33. Nishitsuji H, Ujino S, Yoshio S, et al. Long noncoding RNA #32 contributes to antiviral responses by controlling interferon-stimulated gene expression. Proceedings of the National Academy of Sciences of the United States of America. 2016;113(37):10388–93.
  34. Zhao P, Liu S, Zhong Z, et al. Analysis of expression profiles of long noncoding RNAs and mRNAs in brains of mice infected by rabies virus by RNA sequencing. Sci Rep. 2018;8(1):11858-.
  35. Trapnell C, Williams BA, Pertea G, et al. Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat Biotechnol. 2010;28(5):511–5.
  36. Liang S, Haitao L, Dechao B, et al. Utilizing sequence intrinsic composition to classify protein-coding and long non-coding transcripts. Nucleic Acids Res. 2013;41(17):e166–6.
  37. Lei K, Yong Z, Zhi-Qiang Y, et al. CPC: assess the protein-coding potential of transcripts using sequence features and support vector machine. Nucleic Acids Res. 2007;35:W345. (Web Server issue).
  38. Park HJ, Wang L, Wang S, et al. CPAT: Coding-Potential Assessment Tool using an alignment-free logistic regression model. Nucleic Acids Res. 2013;41(6):e74–4.
  39. Fisher CR, Streicker DG, Schnell MJ. The spread and evolution of rabies virus: conquering new frontiers. Nat Rev Microbiol. 2018;16(4):241.
  40. Rupprecht C, Kuzmin I, Meslin F. Lyssaviruses and rabies: current conundrums, concerns, contradictions and controversies, F1000research. 6 (184) (2017) 184.
  41. Lewis P, Fu Y, Lentz TL. Rabies virus entry at the neuromuscular junction in nerve–muscle cocultures. Muscle Nerve. 2015;23(5):720–30.
  42. Faul EJ, Wanjalla CN, Suthar MS, et al. Rabies virus infection induces type I interferon production in an IPS-1 dependent manner while dendritic cell activation relies on IFNAR signaling. PLoS Pathog. 2010;6(7):e1001016.
  43. Li J, Faber M, Dietzschold B, et al. The role of toll-like receptors in the induction of immune responses during rabies virus infection. Adv Virus Res. 2011;79:115–26.
  44. Prehaud C, Megret F, Lafage M, et al. Virus infection switches TLR-3-positive human neurons to become strong producers of beta interferon. J Virol. 2005;79(20):12893–904.
  45. Zhao P, Yang Y, Feng H, et al. Global gene expression changes in BV2 microglial cell line during rabies virus infection. Infect Genet Evol. 2013;20:257–69.
  46. Zhao P, Jiang T, Zhong Z, et al. Inhibition of rabies virus replication by interferon-stimulated gene 15 and its activating enzyme UBA7. Infect Genet Evol. 2017;56:44–53.
  47. Yu AD, Wang Z, Morris KV. Long noncoding RNAs: a potent source of regulation in immunity and disease. Immunology Cell Biology. 2015;93(3):277.
  48. Peng X, Gralinski L, Armour CD, et al., Unique signatures of long noncoding RNA expression in response to virus infection and altered innate immune signaling, MBio. 1 (5) (2010).
  49. Carnero E, Barriocanal M, Segura V, et al. Type I Interferon Regulates the Expression of Long Non-Coding RNAs. Frontiers in Immunology. 2014;5:548.
  50. Ouyang J, Hu J, Chen JL. lncRNAs regulate the innate immune response to viral infection. Wiley Interdisciplinary Reviews Rna. 2016;7(1):129–43.
  51. Chang TL, Vargas J, Delportillo A, et al. Dual role of alpha-defensin-1 in anti-HIV-1 innate immunity. Journal of Clinical Investigation. 2005;115(3):765–73.
  52. Salvatore M, Garcia-Sastre A, Ruchala P, et al. alpha-Defensin inhibits influenza virus replication by cell-mediated mechanism(s). Journal of Infectious Diseases. 2007;196(6):835–43.
  53. Demirkhanyan LH, Marin M, Padillaparra S, et al. Multifaceted Mechanisms of HIV-1 Entry Inhibition by Human α-Defensin. Journal of Biological Chemistry. 2012;287(34):28821–38.
  54. Rosenberger CM, Clark AE, Treuting PM, et al., ATF3 Regulates MCMV Infection in Mice by Modulating IFN-γ Expression in Natural Killer Cells, Proceedings of the National Academy of Sciences of the United States of America. 105 (7) (2008) 2544–2549.
  55. Biron CA, Brossay L. NK cells and NKT cells in innate defense against viral infections. Current Opinion in Immunology. 2001;13(4):458–64.
  56. Mercer TR, Dinger ME, Mattick JS. Long non-coding RNAs: insights into functions. Nat Rev Genet. 2009;10(3):155–9.
  57. Wilusz JE, Sunwoo H, Spector DL. Long noncoding RNAs: functional surprises from the RNA world. Genes Dev. 2009;23(13):1494–504.