scispace - formally typeset
Search or ask a question
Posted ContentDOI

A novel antiviral lncRNA EDAL shields a T309 O-GlcNAcylation site to promote EZH2 degradation

TL;DR: A model in which a neuronal lncRNA can exert an effective antiviral function via blocking a specific O-GlcNAcylation that determines EZH2 lysosomal degradation is proposed.
Abstract: The central nervous system (CNS) is vulnerable for viral infection, yet few host factors in the CNS are known to defend invasion by neurotropic viruses. We report here that multiple neurotropic viruses, including rabies virus (RABV), vesicular stomatitis virus (VSV), Semliki Forest virus (SFV) and herpes simplex virus 1 (HSV-1), elicit the neuronal expression of a host-encoded lncRNA EDAL. EDAL inhibits the replication of these neurotropic viruses in neuronal cells and RABV infection in mouse brains. EDAL binds to the conserved histone methyltransferase enhancer of zest homolog 2 (EZH2) and specifically causes EZH2 degradation via lysosomes, reducing the cellular H3K27me3 level. The antiviral function of EDAL resides in a 56-nt antiviral substructure through which its 18-nt helix-loop intimately contacts multiple EZH2 sites surrounding T309, a known O-GlcNAcylation site. EDAL positively regulate the transcription of Pcp4l1 encoding a 10 kDa peptide, which inhibits the replication of mutiple neurotropic viruses. Our findings proposed a model in which a neuronal lncRNA can exert an effective antiviral function via blocking a specific O-GlcNAcylation that determines EZH2 lysosomal degradation.

Summary (6 min read)

INTRODUCTION

  • Among infectious diseases of the central nervous system (CNS), those caused by viral pathogens-known as neurotropic viruses-are far more common than bacteria, fungi, and protozoans (2Nd & Mcgavern, 2015 , Ludlow, Kortekaas et al., 2016) .
  • Moreover, nearly half of all emerging viruses are neurotropic viruses (Olival & Daszak, 2005) , including the Dengue and Zika viruses (Carod-Artal, 2016 , Meyding-Lamade & Craemer, 2018) .
  • It is unclear how this regulation occurs.

Identification of a host lncRNA induced by viral infection

  • The authors conducted a time-course RNA-seq analysis of cultured N2a cells that were infected with pathogenic RABV (CVS-B2c strain) or were mock infection treated.
  • This identified 1,434 differentially expressed lncRNAs (Fig 1A) .
  • QPCR analysis successfully confirmed the significantly up-regulated expression of ten of the most highly up-regulated of these lncRNAs in response to RABV infection (Fig 1B) .
  • This long intergenic non-coding RNA had no obvious annotation hits after examining its sequence using tools available with the NONCODEv5 (Fang, Zhang et al., 2017) , lncRNAdb 2.0 (Quek, Thomson et al., 2015) , or LNCipedia 5.0 (Volders, Verheggen et al., 2015) databases.

EDAL inhibits viral replication

  • The authors next transfected N2a cells with pcDNA3.1 plasmid expressing either EDAL (pcDNA-EDAL) or an EDAL-specific small interfering RNA and then verified that EDAL was appropriately expressed or specifically silenced in N2a cells (Fig EV3A and 3B) .
  • The authors also confirmed that overexpression or silencing of EDAL did not affect cell viability (Fig EV3C and 3D) .
  • Next, the authors transfected N2a cells with the EDAL expression plasmid and then infected them with RABV at 12 hours (h) post transfection.
  • The authors also analyzed the capacity of the recombinant viruses to spread between infected cells and neighboring cells, the infected N2a cells were covered by low melting agar to inhibit the virus release into the supernatant (Tian et al., 2016) .

EDAL reduces RABV pathogenicity in vivo

  • To investigate the role of EDAL in RABV infection in vivo, the authors compared the pathogenicity of rRABV, rRABV-EDAL, and rRABV-revEDAL in the C57BL/6 mouse model.
  • Mice were infected intra-nasally (i.n.) with rRABV, rRABV-EDAL, or rRABV-revEDAL (100 FFU).
  • The mice infected with rRABV and rRABV-revEDAL exhibited decreased body weights starting from 7 to 9 days post infection (dpi), and these decreases became significant between 9 and 14 dpi.
  • In contrast, the body weight of mice infected with rRABV-EDAL only exhibited a slight decrease between 10-14 dpi (Fig 3A Collectively, these results establish that EDAL can dramatically inhibit intranasal-inoculation-induced RABV infection in mice.

EDAL decreases H3K27me3 levels by promoting lysosome-mediated EZH2 degradation

  • Having demonstrated that RABV infection induces the accumulation of EDAL and established that EDAL can restrict RABV replication in vitro and in vivo, the authors were interested in potential mechanism(s) through which EDAL may exert its antiviral effects.
  • The authors have for some time been interested in the potential contributions of epigenetic regulation on host responses to neurotropic viruses, and they noted that the N2a cells transfected with the pcDNA3.1 plasmid expressing pcDNA-EDAL had significantly decreased levels of histone methylation.
  • The authors next used the recombinant viruses that they used for mice infection ( again highlighting an apparently specific contribution of EDAL to the reduced levels of H3K27me3 and its catalyst EZH2.

A 56 nt 5' segment is responsible for EDAL's antiviral activity

  • Secondary structures are thus far good candidates for identification of functional elements of lncRNAs (Bonasio & Shiekhattar, 2014 , Johnsson, Lipovich et al., 2014 , Mercer & Mattick, 2013 , Rivas, Clements et al., 2017) .
  • Seeking to identify secondary structures of EDAL that affect its specific interaction with EZH2, predictions using the RNAstructure 5.

EDAL reduces EZH2 stability by impeding an O-GlcNAcylation PTM at the T309 site

  • RNA tertiary structure prediction revealed a tertiary structure for the 56-nt antiviral RNA segment: the helix-loop tertiary structure folded by the 18-nt terminal hairpin corresponding to 125-142 of EDAL was packed on the second helix folded by the stem base-paired structure, and most of the two structural components were free for contacting other partners (Fig 6C ).
  • The authors co-transfected N2a cells with plasmids expressing wild type EZH2 and EZH2 mutant variants together with the pcDNA3.1, pcDNA-EDAL or pcDNA-revEDAL plasmids.
  • Then the authors confirmed that the specific EDAL interaction sites on EZH2 are in its N-terminal region (1-337 aa) using an RNA pull-down analysis (Fig These results support that EDAL specifically contacts T309, shielding T309 from O-GlcNAcylation.

The EZH2 inhibitor gsk126 protects neuronal cells from viral infection

  • If EDAL's antiviral effects are indeed mediated by its reduced EZH2 methyltransferase activity, then the authors could anticipate that chemical inhibition of EZH2 should cause antiviral effects.
  • The authors findings indicated that EDAL binds to EZH2 at a site different from that of lncRNA-HOTAIR binding of human EZH2 via residues in 342-368 region (Kaneko et al., 2010) .
  • Alteration of the host epigenetic dynamics by virus-elicited host lncRNAs might not be limited to EZH2 and H3K27me3 mark.
  • As a result, lnc-DC indirectly promotes STAT3 phosphorylation on tyrosine-705 and controls human dendritic cell differentiation (Wang, Xue et al., 2014) .
  • PCP4L1 display a distinct expression pattern which is dominantly expressed in the CNS, and mostly expressed in circumventricular organs and modulate the production of the cerebrospinal fluid in the adult brain (Bulfone et al., 2004) .

Cell lines, viruses, and mice

  • BV2 (murine microglia, BNCC337749) were obtained from BeNa Culture Collection.
  • Cells grown in a 37°C humidified 5% CO 2 atmosphere, growth media was DMEM or RPMI1640 supplemented with 10% (vol/vol) FBS and 1% antibiotics (penicillin and streptomycin) .
  • The recombinant rRABVs were cloned from RABV strain challenge virus standard-B2c (CVS-B2c) and constructed as described previously (Tian et al., 2016) HZAUMO-2016-009).

RNA-seq library construction, sequencing and lncRNA prediction pipeline

  • Total RNA from RABV infected N2a cells or mock-infected cells were isolated by using Trizol ® reagent following the manufacturer's instructions, and then treated with RQ1 DNase to remove DNA.
  • Each RNA-seq library was prepared using 5 μg of total RNA.
  • Purified RNAs were iron fragmented at 95°C, followed by end repair and 5' adaptor ligation.
  • Reverse transcription was performed using RT primers harboring a 3' adaptor sequence and randomized hexamer.
  • The cDNAs were purified, amplified by PCR, and products 200-500 bp in length were isolated, quantified, and used for sequencing.

RNA-seq data processing and alignment

  • Raw reads containing more than two unknown (N) bases were discarded.
  • The mus musculus genome sequence (GRCm38) and annotation file (gencode.vM6 basic annotation) were obtained from the GENCODE database (Mudge & Harrow, 2015) .
  • Clean reads were aligned end-to-end to the mouse genome by TopHat2 (Kim, Pertea et al., 2013) , allowing 2 mismatches.
  • Reads that aligned to more than one genomic location were discarded, and uniquely localized reads were used to calculate the number of reads and RPKM values (RPKM represents reads per kilobase and per million) for each gene.
  • For each gene, the fold changes, p-values, and adjusted p-values (FDR) were also determined by the edgeR package.

LncRNA prediction pipeline

  • The lncRNA prediction pipeline was implemented following the methods described by Liu et al.
  • The detailed descriptions of the prediction pipeline and filtering thresholds are as follows: (1) First, using the aligned RNA-seq data (see above), transcripts were assembled by Cufflinks V2.2.1 (Trapnell et al., 2012) using default parameters.
  • After the initial assembly, transcripts with FPKM greater than or equal to 0.1 were subjected to a series of filters.
  • (3) To remove potential protein-coding transcripts, coding potential score (CPS) was evaluated using the Coding Potential Calculator (CPC) (Kong, Zhang et al., 2007) .
  • A total of 1662 novel lncRNA transcripts were identified, originating from 1377 lncRNA loci.

Quantitative real-time PCR (qPCR)

  • Total RNA was isolated from cells and tissues by using Trizol ® reagent .
  • The genomic DNA was eliminated with TURBO DNA-free TM Kit (Ambion, AM1907) as the manufacturer's instructions.
  • RNA quality was assessed by using NanoDrop 2000 (Thermo Scientific).
  • The cDNAs were synthesized by ReverTra Ace qPCR RT Master Mix (Toyobo, FSQ-201) or First-Strand cDNA Synthesis Kit (Toyobo, FSK-101).
  • QPCR was performed using SYBR Green Supermix (Bio-Rad).

Rapid amplification of cloned cDNA ends (RACE)

  • Total RNA from N2a cells was isolated by using Trizol ® reagent and 5'-or 3'-RACE was performed with SMARTer ® RACE 5'/3' Kit (Takara, 634858) following the manufacturer's instructions.
  • Primers used for 5'-or 3'-RACE was designed based on the known sequence information.

Fluorescent in situ hybridization

  • The red fluorescence labeled probe (Ribo-lncRNA FISH Probe Mix) against EDAL lncRNA was designed by Ribobio Co (Guangzhou, China) and was detected by Fluorescent In Situ Hybridization Kit (Ribobio, R11060.1) according to the manufacturer's instructions.
  • Briefly, N2a cells grown on cover slips in 24-well plates were fixed with 4% (v/v) paraformaldehyde for 10 minutes (min) at room temperature then washed three times with cold PBS.
  • And the cells were permeabilized in PBS containing 0.5% Triton X-100 for 5 min in 4°C, then blocked in pre-hybridization buffer for 30 min at 37°C.
  • After hybridization, cells were washed in the dark with washing buffer (4×SSC/2×SSC/1×SSC) then stained with DAPI for 10 min.
  • Cells were again washed three times with PBS, and then imaged with an Olympus FV10 laser-scanning confocal microscope.

EDAL specific siRNA

  • EDAL specific siRNA was designed and synthesized by Ribobio Co.
  • The target sequence was 5'-GGTAGACACCCAGTGACAA-3', and siEDAL sequence was 5'-GGUAGACACCCAGUGACAA -3'.

Cell viability assay

  • N2a cells were transfected with plasmids, siRNAs or treated with EZH2 specific inhibitor gsk126 (Apexbio, A3446) for indicated time.
  • The viability of N2a cells was evaluated by Cell Titer 96 AQueous One Solution cell proliferation assay kits (Promega, G3582) according to the manufacturer's instruction.

Construction of the recombinant RABVs (rRABV)

  • Mouse lncRNAs, reverse EDAL were amplified from the total RNA extracted from RABV-infected N2a cells using the ReverTra Ace qPCR RT Master Mix (TOYOBO, FSQ-201) with Phanta Max Super-Fidelity DNA polymerase (Vazyme, P505-d1).
  • The primer sets used were designed by Primer 6 (PREMIER Biosoft Biolabs) .
  • PCR products were digested with BsiWI and NheI (New England Biolabs) then ligated into the genome of recombinant RABV strain B2c (rB2c) digest used the same enzymes as previously described (Tian et al., 2016) .

Rescue of rRABVs

  • Recombinant RABVs were rescued as reported previously (Tian et al., 2016) .
  • Four days post transfection, supernatants was harvested and examined for the presence of rescued viruses using FITC-conjugated anti-RABV N antibodies (Fujirebio Diagnostics, Malvern, PA).

Virus titration

  • To determine rRABV and VSV titers, BSR cells were infected with serial dilutions of the viruses.
  • After 1 h incubation in 37°C, the cell supernatant was discarded and washed once with PBS, and then overlaid with DMEM containing 1% low melting point agarose (VWR, 2787C340).
  • Then the fluorescent foci were counted under a fluorescence microscope.
  • For SFV and HSV-1 titration, Vero cells were seeded in 12-well plates and infected with serial dilutions of the viruses.
  • After staining for 4 h, the plates were washed with water, and the plaques were counted.

Mouse infection

  • Eight-week-old female C57BL/6 mice were randomly divided into indicated groups and infected intranasally with rRABV, rRABV-EDAL, rRABV-revEDAL (100 FFU) or mock infected with DMEM in a volume of 20 µl.
  • When moribund, the mice were euthanized with CO 2 , and then the brains were collected for qPCR or immunohistochemistry analysis.

Immunohistochemistry analysis

  • Groups of female C57BL/6 mice were infected intranasally with rRABV or rRABV-EDAL.
  • At indicated times post infection (pi), mouse brains were harvested and fixed in 4% paraformaldehyde for 2 days at 4°C.
  • For immunohistochemistry (IHC), the sections were deparaffinized and rehydrated in xylene and ethanol.
  • After washing, sections were incubated with diaminobenzidine (ServiceBio, G1211) for color development then photographed and analyzed using an XSP-C204 microscope (CIC).

Western blotting

  • N2a cells were lysed in RIPA buffer (Beyotime, P0013B) supplemented with 1x protease inhibitor cocktail .
  • Total cell lysates were separated on 8-12% SDS-PAGE gels and transferred to PVDF membranes (Bio-Rad).
  • Membranes were blocked with TBST with 5% (w/v) non-fat dry milk for 4 h, and probed with primary antibodies which were diluted with TBST and 5% (w/v) non-fat dry milk overnight in 4°C.
  • Images were captured with an Amersham Imager 600 (GE Healthcare) imaging system.

EDAL-EZH2 interaction 3D structure modeling

  • Murine EZH2 3D structure was predicted with SWISS-MODEL (https://swissmodel.expasy.org/interactive) based on human EZH2 3D structure (PDB code: 5HYN).
  • Then amino acid sequence comparison was conducted between human EZH2 and Murine EZH2, and 98.24% similarity was calculated by Clustal2.1 (a multiple sequence alignment software, https://www.ebi.ac.uk/Tools/msa/muscle/).
  • EDAL-FD 3D structure model was predicted with RNAComposer (A automated RNA structure 3D modeling server, http://rnacomposer.ibch.poznan.pl/).
  • In order to predict the interaction between EDAL functional domain (98-153 nt) and Murine EZH2, the template-based docking method PRIME (Zheng, Kundrotas et al., 2016 ) (If a template can be found, it is often more accurate than the free docking method) was used to dock the EDAL and EZH2 monomer structures at first.
  • Two atoms between EZH2 and EDAL with distance less than 5 angstroms in the predicted complex structure are considered to have interactions.

RNA pull-down assay

  • The synthesized RNA was treated with Rnase-free DNase I (Thermo, EN0521) and then purified with MicroElute RNA Clean-Up Kit (OMEGA, R6247-01).
  • The RNA was heated to 95°C for 2 min, put on ice for 5 min and then put it at room temperature for 20 min to form secondary structure.
  • The RNA was then added to the lysed cell containing overexpressed EZH2-1-337-flag and incubated for 2 h at 4°C.
  • After being washed with wash buffer for three times, the samples were then analyzed by Western blotting.

O-GlcNAcylation labeling and detection

  • The plasmid pCAGGS-EZH2-S73/S75/S725A-flag was co-tranfected with Chromatin was eluted from the beads by two washes with 100 µl elution buffer (100 mM NaHCO 3 , 1% SDS), the Na + concentration was adjusted to 300 mM with 5 M NaCl and the crosslinks were reversed by overnight incubation in a 65°C water-bath.
  • DNA was purified by phenol extraction and ethanol precipitation.
  • For high-throughput sequencing, the libraries were prepared following the manufacturer's instructions (ThruPLEX DNA-seq 48S Kit, R400427) and analyzed using an Illumina NextSeq-500 system for 150 nt pair-end sequencing (ABlife Inc., Wuhan, China).

ChIP-seq data analysis

  • Adaptors and low quality bases were trimmed from raw sequencing reads using Cutadapt (Martin, 2011) .
  • Reads were aligned to the mouse-GRCm38 genome using Bowtie2 (Langmead & Salzberg, 2012) .
  • To evaluate the quality of ChIP-seq data, the authors performed a cross-correlation analysis, as well as FRiP and IDR analyses for the ChIP-seq data, according to the ChIP-seq guidelines provided by the ENCODE and modENCODE consortia (Kheradpour & Kellis, 2012) .
  • All peaks from each sample were clustered by BEDTools (Quinlan & Hall, 2010) .
  • To associate peaks with genes, the authors set 10000 bp as the upstream limit for the distance from the peak maximum to the TSS (transcript start site), and 3000 bp as the downstream limit for distance from the peak maximum to the TSS.

ChIP-qPCR

  • Formaldehyde crosslinking of N2a cells, chromatin sonication and immunoprecipitation were performed following the same procedures as the ChIP-seq section described above.
  • Real-time PCR was then performed using a QuantStudio 6 Flex System (ABI) according to the manufacturer's standard protocol.
  • Input was used to normalize the amount of each sample as an internal control.
  • Assays were repeated at least three times and expressed as Ct values.

Did you find this useful? Give us your feedback

Content maybe subject to copyright    Report

1
A novel antiviral lncRNA EDAL shields a T309 O-GlcNAcylation site to 1
promote EZH2 degradation 2
Baokun Sui
1,2,
*, Dong Chen
3,4,
*, Wei Liu
1,2
, Qiong Wu
1,2
, Bin Tian
1,2
, Jing 3
Hou
3,4
, Yingying Li
1,2
, Shiyong Liu
5
, Juan Xie
5
, Hao Jiang
6
, Zhaochen Luo
1,2
, 4
Lei Lv
1,2
, Fei Huang
1,2
, Ruiming Li
1,2
, Min Cui
1,2
, Ming Zhou
1,2
, Huanchun 5
Chen
1,2
, Zhen F. Fu
1,2,7
, Yi Zhang
3,4,
, Ling Zhao
1,2,
6
Running title: lncRNA EDAL inhibits neurotropic viruses 7
1
State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, 8
Wuhan, 430070, China; 9
2
Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of 10
Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; 11
3
Center for Genome analysis and
4
Laboratory for Genome Regulation and Human Health, 12
ABLife Inc., Wuhan, 430075, China 13
5
School of Physics, Huazhong University of Science and Technology, Wuhan, 430074, 14
China; 15
6
Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and 16
Pharmacy, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, 17
Ocean University of China, Qingdao, 266003, China; 18
7
Department of Pathology, University of Georgia, Athens, GA 30602, USA;
19
*
These authors contributed equally to this work. 20
certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (which was notthis version posted October 30, 2019. ; https://doi.org/10.1101/824813doi: bioRxiv preprint

2
To whom correspondence should be addressed: 21
Ling Zhao, Mailing address: State Key Laboratory of Agricultural Microbiology, Huazhong 22
Agricultural University, Wuhan, 430070, China.Tel: +86-27-8728 5016; Fax: +86-27-8728 23
2608; E-mail: zling604@yahoo.com 24
Correspondence may also be addressed to 25
Yi Zhang, Mailing address: Center for Genome analysis and Laboratory for Genome 26
Regulation and Human Health, ABLife Inc., Wuhan, 430075, China. E-mail: 27
yizhang@ablife.cc
28
29
certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (which was notthis version posted October 30, 2019. ; https://doi.org/10.1101/824813doi: bioRxiv preprint

3
Abstract 30
The central nervous system (CNS) is vulnerable for viral infection, yet few host 31
factors in the CNS are known to defend invasion by neurotropic viruses. We 32
report here that multiple neurotropic viruses, including rabies virus (RABV), 33
vesicular stomatitis virus (VSV), Semliki Forest virus (SFV) and herpes 34
simplex virus 1 (HSV-1), elicit the neuronal expression of a host-encoded 35
lncRNA EDAL. EDAL inhibits the replication of these neurotropic viruses in 36
neuronal cells and RABV infection in mouse brains. EDAL binds to the 37
conserved histone methyltransferase enhancer of zest homolog 2 (EZH2) and 38
specifically causes EZH2 degradation via lysosomes, reducing the cellular 39
H3K27me3 level. The antiviral function of EDAL resides in a 56-nt antiviral 40
substructure through which its 18-nt helix-loop intimately contacts multiple 41
EZH2 sites surrounding T309, a known O-GlcNAcylation site. EDAL positively 42
regulate the transcription of Pcp4l1 encoding a 10 kDa peptide, which inhibits 43
the replication of mutiple neurotropic viruses. Our findings proposed a model in 44
which a neuronal lncRNA can exert an effective antiviral function via blocking a 45
specific O-GlcNAcylation that determines EZH2 lysosomal degradation. 46
Key words: EZH2/lncRNA/neurotropic virus/O-GlcNAcylation/PCP4L1 47
48
49
50
51
52
53
certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (which was notthis version posted October 30, 2019. ; https://doi.org/10.1101/824813doi: bioRxiv preprint

4
INTRODUCTION 54
Among infectious diseases of the central nervous system (CNS), those 55
caused by viral pathogens—known as neurotropic virusesare far more 56
common than bacteria, fungi, and protozoans (2Nd & Mcgavern, 2015, Ludlow, 57
Kortekaas et al., 2016). Neurotropic viruses arrive to the CNS through multiple 58
routes and propagate within various cell types including astrocytes, microglia 59
and neurons, depending on the entering routes and virus types (Manglani & 60
McGavern, 2018). Infection of some neurotropic viruses can cause meningitis 61
or encephalitis and result in severe neurologic dysfunction, such as VSV, SFV, 62
HSV-1 and HIV etc. (Bradshaw & Venkatesan, 2016, Fragkoudis, 63
Dixon-Ballany et al., 2018, Gagnidze, Hajdarovic et al., 2016). Moreover, 64
nearly half of all emerging viruses are neurotropic viruses (Olival & Daszak, 65
2005), including the Dengue and Zika viruses (Carod-Artal, 2016, 66
Meyding-Lamade & Craemer, 2018). RABV is a typical neurotropic virus and is 67
the causative agent of rabies disease, a globally well-known and often lethal 68
encephalitis. Therefore, it is urgent to develop new approaches for therapies 69
as well as for cheaper and more effective vaccines against rabies (Fisher & 70
Schnell, 2018, Schnell, McGettigan et al., 2010). 71
Long non-coding RNAs (lncRNAs) are involved in the development, 72
plasticity, and pathology of the nervous system (Batista & Chang, 2013, Briggs, 73
Wolvetang et al., 2015, Fatica & Bozzoni, 2014, Sun, Yang et al., 2017). 74
Notably, around 40% of lncRNAs detected to date are expressed specifically in 75
the brain (Liu, Wang et al., 2017). Genome-wide association studies (GWASs) 76
and functional studies have associated lncRNAs with neurological diseases 77
including autism spectrum disorders (ASD), schizophrenia, Alzheimer’s 78
disease, and neuropathic pain, among others (Briggs et al., 2015). 79
Mechanistically, it has been shown that lncRNAs can regulate chromatin 80
modifications and gene expression, at both the transcriptional and the 81
certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (which was notthis version posted October 30, 2019. ; https://doi.org/10.1101/824813doi: bioRxiv preprint

5
post-transcriptional levels (Bonasio & Shiekhattar, 2014, Mercer, Dinger et al., 82
2009, Wang & Chang, 2011). LncRNAs have recently been shown to regulate 83
innate immune responses by either promoting or inhibiting viral genome 84
replication, highlighting them as a class of novel targets for developing antiviral 85
therapies (Carpenter & Fitzgerald, 2018, Fortes & Morris, 2016, Imamura, 86
Imamachi et al., 2014, Kambara, Niazi et al., 2014, Ma, Han et al., 2017, 87
Ouyang, Hu et al., 2016, Ouyang, Zhu et al., 2014). It is conceivable that 88
antiviral lncRNAs targeting none-innate immune response pathway may exist 89
in neuron cells and brains, which has not been documented yet. 90
Polycomb repressive complex 2 (PRC2) is a protein complex with an 91
epigenetic regulator function in maintaining the histone modifications that mark 92
transcriptional repression states which are established during early 93
developmental stages (Ringrose, 2017). Some lncRNAs are known to interact 94
with and direct PRC2 towards the chromatin sites of action, thusly defining a 95
trans-acting lncRNA mechanism (Jin, Lv et al., 2018, Rinn, Kertesz et al., 96
2007). The EZH2 methyltransferase enzyme is the catalytic component of 97
PRC2: it binds RNAs and catalyzes di- or tri-methylation of histone H3 lysine 98
27 (H3K27me2/3), a modification which leads to the formation of facultative 99
heterochromatin and thus to transcriptional repression (Justin, Zhang et al., 100
2016, Kasinath, Faini et al., 2018, Margueron & Reinberg, 2011). Many 101
cancers are known to feature very high EZH2 expression levels, so this protein 102
has emerged as an anticancer target for which multiple chemical inhibitors 103
have been developed (Kim & Roberts, 2016, Lee, Yu et al., 2018). It has also 104
been recently reported that inhibitors of the histone methyltransferase activity 105
of EZH2 can suppress infection by several viruses, suggesting a function of 106
EZH2 and/or PRC2 in regulating viral infection (Arbuckle, Gardina et al., 2017). 107
However, it is unclear how this regulation occurs. In general, PRC2 (EZH2) 108
binds different classes of RNAs in a promiscuous manner in vitro and in cells, 109
and some lncRNAs such as RepA RNA show in vitro specificity with PRC2 110
certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (which was notthis version posted October 30, 2019. ; https://doi.org/10.1101/824813doi: bioRxiv preprint

Citations
More filters
Journal ArticleDOI
TL;DR: Findings show that a neuronal lncRNA can exert an effective antiviral function via blocking a specific O-GlcNAcylation that determines EZH2 lysosomal degradation, rather than the traditional interferon-dependent pathway.
Abstract: The central nervous system (CNS) is vulnerable to viral infection, yet few host factors in the CNS are known to defend against invasion by neurotropic viruses. Long noncoding RNAs (lncRNAs) have been revealed to play critical roles in a wide variety of biological processes and are highly abundant in the mammalian brain, but their roles in defending against invasion of pathogens into the CNS remain unclear. We report here that multiple neurotropic viruses, including rabies virus, vesicular stomatitis virus, Semliki Forest virus, and herpes simplex virus 1, elicit the neuronal expression of a host-encoded lncRNA EDAL. EDAL inhibits the replication of these neurotropic viruses in neuronal cells and rabies virus infection in mouse brains. EDAL binds to the conserved histone methyltransferase enhancer of zest homolog 2 (EZH2) and specifically causes EZH2 degradation via lysosomes, reducing the cellular H3K27me3 level. The antiviral function of EDAL resides in a 56-nt antiviral substructure through which its 18-nt helix-loop intimately contacts multiple EZH2 sites surrounding T309, a known O-GlcNAcylation site. EDAL positively regulates the transcription of Pcp4l1 encoding a 10-kDa peptide, which inhibits the replication of multiple neurotropic viruses. Our findings show that a neuronal lncRNA can exert an effective antiviral function via blocking a specific O-GlcNAcylation that determines EZH2 lysosomal degradation, rather than the traditional interferon-dependent pathway.

28 citations

References
More filters
Journal ArticleDOI
TL;DR: It is proposed that integrating the current knowledge of noncoding RNA into a quantitative biochemical and theoretical framework for PcG and TrxG regulation has the potential to reconcile several apparently conflicting models and identifies fascinating questions for future research.
Abstract: The question of how noncoding RNAs are involved in Polycomb group (PcG) and Trithorax group (TrxG) regulation has been on an extraordinary journey over the last three decades. Favored models have risen and fallen, and healthy debates have swept back and forth. The field has recently reached a critical mass of compelling data that throws light on several previously unresolved issues. The time is ripe for a fruitful combination of these findings with two other long-running avenues of research, namely the biochemical properties of the PcG/TrxG system and the application of theoretical mathematical models toward an understanding of the system's regulatory properties. I propose that integrating our current knowledge of noncoding RNA into a quantitative biochemical and theoretical framework for PcG and TrxG regulation has the potential to reconcile several apparently conflicting models and identifies fascinating questions for future research.

31 citations


"A novel antiviral lncRNA EDAL shiel..." refers background in this paper

  • ...Note that EZH2-lncRNA interactions have been a popular model for studies of epigenetic silencing by PRC2 (Davidovich & Cech, 2015, Lee, 2012, Margueron & Reinberg, 2011, Mercer & Mattick, 2013, N, 2013, Ringrose, 2017)....

    [...]

  • ...Polycomb repressive complex 2 (PRC2) is a protein complex with an 91 epigenetic regulator function in maintaining the histone modifications that mark 92 transcriptional repression states which are established during early 93 developmental stages (Ringrose, 2017)....

    [...]

  • ...Polycomb repressive complex 2 (PRC2) is a protein complex with an epigenetic regulator function in maintaining the histone modifications that mark transcriptional repression states which are established during early developmental stages (Ringrose, 2017)....

    [...]

Journal ArticleDOI
TL;DR: Structural alignment is better than sequence alignment in identifying good templates, suitable for generating protein-RNA complexes close to the native structure, and outperforms free docking, successfully predicting complexes where the free docking fails, including cases of significant conformational change upon binding.
Abstract: Protein-RNA complexes formed by specific recognition between RNA and RNA-binding proteins play an important role in biological processes. More than a thousand of such proteins in human are curated and many novel RNA-binding proteins are to be discovered. Due to limitations of experimental approaches, computational techniques are needed for characterization of protein-RNA interactions. Although much progress has been made, adequate methodologies reliably providing atomic resolution structural details are still lacking. Although protein-RNA free docking approaches proved to be useful, in general, the template-based approaches provide higher quality of predictions. Templates are key to building a high quality model. Sequence/structure relationships were studied based on a representative set of binary protein-RNA complexes from PDB. Several approaches were tested for pairwise target/template alignment. The analysis revealed a transition point between random and correct binding modes. The results showed that structural alignment is better than sequence alignment in identifying good templates, suitable for generating protein-RNA complexes close to the native structure, and outperforms free docking, successfully predicting complexes where the free docking fails, including cases of significant conformational change upon binding. A template-based protein-RNA interaction modeling protocol PRIME was developed and benchmarked on a representative set of complexes.

30 citations

Journal ArticleDOI

30 citations


"A novel antiviral lncRNA EDAL shiel..." refers background in this paper

  • ...Moreover, nearly half of all emerging viruses are neurotropic viruses (Olival & Daszak, 2005), including the Dengue and Zika viruses (Carod-Artal, 2016, Meyding-Lamade & Craemer, 2018)....

    [...]

Journal ArticleDOI
TL;DR: Collectively, this work has unraveled a ST7-AS1/CARM1/Sox-2 signaling axis in LSCC and may have created novel interconnections between noncoding RNAs and cancer development.

25 citations

Journal ArticleDOI
TL;DR: Cloning of a novel mouse gene that encodes a polypeptide with significant sequence similarity to the Purkinje cell protein 4 gene (Pcp4) and its expression pattern during mouse development reveals that Pcp4l1 has a distinct pattern of expression.

22 citations


"A novel antiviral lncRNA EDAL shiel..." refers background in this paper

  • ...PCP4L1 display a distinct expression pattern which is dominantly expressed in the CNS, and mostly expressed in circumventricular organs and modulate the production of the cerebrospinal fluid in the adult brain (Bulfone et al., 2004)....

    [...]

  • ...PCP4L1 is a 68 amino acids polypeptide which display sequence similarity to the Purkinje Cell Protein 4 gene (Pcp4) and both of which are characterized by their C-terminal IQ domain ends (Bulfone et al., 2004)....

    [...]