scispace - formally typeset
Search or ask a question
Posted ContentDOI

A zebrafish reporter line reveals immune and neuronal expression of endogenous retrovirus

21 Jan 2021-bioRxiv (Cold Spring Harbor Laboratory)-
TL;DR: In this article, the authors identified 8 copies of the zebrafish endogenous retrovirus (zferv) and created and characterised the first in vivo ERV reporter line in any species.
Abstract: Endogenous retroviruses (ERVs) are fossils left in our genome from retrovirus infections of the past. Their sequences are part of every vertebrate genome and their random integrations are thought to have contributed to evolution. Although ERVs are mainly kept silenced by the host genome, they are found activated in multiple disease states such as auto-inflammatory disorders and neurological diseases. What makes defining their role in health and diseases challenging is the numerous copies in mammalian genomes and the lack of tools to study them. In this study, we identified 8 copies of the zebrafish endogenous retrovirus (zferv). We created and characterised the first in vivo ERV reporter line in any species. Using a combination of live imaging, flow cytometry and single cell RNA sequencing, we mapped zferv expression to early T cells and neurons. Thus, this new tool identified tissues expressing ERV in zebrafish, highlighting a potential role of ERV during brain development and strengthening the hypothesis that ERV play a role in immunity and neurological diseases. This transgenic line is therefore a suitable tool to study the function of ERV in health and diseases. Funding This work has been supported by a European Leukodystrophy Association fellowship (ELA 2016-012F4) to NH, an MRC Programme Grant (MR/M004864/1) to SAR and . JPL is supported by Agence Nationale de la Recherche (grant ANR-16-CE20-0002-03.. Imaging was carried out in the Wolfson Light Microscopy Facility, supported by an MRC grant (G0700091) and a Wellcome Trust grant (GR077544AIA). Conflict of Interest Statement The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
Figures (5)

Content maybe subject to copyright    Report

A"zebrafish"reporter"line"reveals"immune"and"neuronal"expression"of"endogenous"
retrovirus."
!
Noémie! Hamilton
1,2
*,! Amy! Clarke
1
,! Hannah!Isles
1
,! Euan! Carson
1
,!Jean-Pierre!Levraud
3
,!
Stephen!A!Renshaw
1!
!
!
1.
!
The!Bateson!Centre,!Department!of!Infection,!Immunity!and!Cardiovascular!Disease,!
University!of!Sheffield,!Sheffield,!UK!
2.!The!Institute!of!Neuroscience,!University!of!Sheffield,!Sheffield,!UK!
3.!Macrophages!et!veloppement!de!l’Immunité,!Institut!Pasteur,!CNRS!UMR3738,!25!
rue!du!docteur!Roux,!75015!Paris!!
!
*Corresponding!author:!n.m.hamilton@sheffield.ac.uk
!
"
Abstract"
"
Endogenous!retroviruses!(ERVs)!are!fossils!left!in!our!genome!from!retrovirus!infections!
of! the! past.! Their! sequences! are! part! of! every! vertebrate! genome! and! their! random!
integrations!are!thought!to!have!contributed!to!evolution.!Although!ERVs!are!mainly!kept!
silenced!by!the!host! genome,!they!are!found! a ctivated!in!mult iple!disease!stat es!such!as!
auto-in flammat ory! d isorders!and!neurological!diseases.!What!makes!defining!their!role!
in!health!and!diseases!challenging!is!the!numerous!copies!in!mammalian!genomes!and!
the! lack! of! tools! to! study! them.! In!this! study,! we! identified! 8! cop i es! of! the!zebrafish!
endogenous! retrovirus!(!"#$%).!We!created!and!characterised!the!first!&'(%&%)!ERV!reporter!
line!in!any!species.!Using!a!combination!of!live!imaging,!flow!cytometry!and!single!cell!
RNA!sequencing,!we!mapped!!"#$%!expression!to!early!T!cells! and!neurons.!Thus,!this!new!
tool!identified!tissues!expressing!ERV!in !zebrafish,!highlighting!a !p oten t ial!role!of!ERV!
during! brain! devel opment! and! strengthening! the! hypothesis! that! ERV! play! a! role! in!
immunity!and!neurological!diseases.!This!transgen ic!line!is!therefore!a!suitable!tool!to!
study!the!funct ion!of!ERV!in!health!and!diseases.!
"
Keywords"
Retroelement,!zebrafish,!endogenous!retrovirus,!reporter!line,!!"#$%,!LTR!
"
Funding"
This! work! has! been! supported! by! a! European! Leukodystrophy! Association! fellowship!
(ELA!2016-012F4)! to!NH,!an!MRC!Programme!Grant! (MR/M004864/1)!to!SAR! and!.!JPL!
is! supported! by! Agence! Nat ionale! de! la! R echerche! ( grant! ANR-16-CE20 -0002-03..!
Imaging!was!ca rried!out! i n!t he!Wolfson!Light!M i croscopy!Facility,!supported!by!an!MRC!
grant!( G0700091)!and!a!Wellcome!Trust!grant !(GR077544AIA).!!
"
Conflict"of"Interest"Statement"
The! authors!declare!that!the!research!was!conducted!in!the!absence!of!any!commercial!or!
financial!relationships !that!could!be!construed!as!a!potential!conflict!of!interest.!
!
!
"
"
"
"
"
"
.CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under a
preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in
The copyright holder for thisthis version posted January 21, 2021. ; https://doi.org/10.1101/2021.01.21.427598doi: bioRxiv preprint

Introduction"
"
Over!40%!of!the!human!genome!comprises!endogenous!transposable!elements!capable!
of!recombination!and!disruption!of!genes!and!modification!of!their!expression!(Lander!et!
al.!2001).!Endogenous!retroviruses!(ERVs)!are!transposable!elements!originating!from!
old!integrations!of!retroviruses!so!successful!that!they!have!become!part!of!all!vertebrate!
genomes!st udied.!ERVs!replicate!autonomously!using!a!copy-and-pa ste!mechanism!and!
although!they! form!a!smaller!percentage!of! all!retroelements,!they!still!represent !8%!of!
the!human!genome!(Bourque!et!al.!2018;!Lander!et!al.!2001).! The!majority!of!known!ERVs!
have! lost! their! ability! to! replicate,! but! those! most! recently! acquired! still! h ave! intac t !
genomes! with!the!ability! to!produce!viral! RNA!and!particles.! However,!these! competent!
ERVs!are!under!strict!transcriptional!suppression!by!epigenetic! mechanisms!(Maksakova,!
Mager,! and! Reiss! 2008;! Rowe! et! al.! 2010;! Turelli! et! al.! 2014),! protecting! the! host!
organisms!against!potential!retroviral!insertions!and!viral!activities.!!!
!
Although!immobilised!by!mutations!or!transcriptiona lly!repressed,!ERVs!have!a!complex!
relationship!with!the!human!genome,!which!they!can!regulate!by!providing!cis-regulatory!
elements! to! surrounding! genes! and! by! lifting! their! transcriptionally! repressed! state.!
Through!these!mechanisms,!it!is!believed!that!transposable!elements!have!fuelled!some!
of! the! necessary!genetic!changes!for!evolution! (Feschotte!2008;!Kunarso!et!al.!2010).!!
*+',+-&'./,!an!ERV!envelope!gene! essential! for!the! vascularisation!of!the!placenta,! is! at!the!
origin! of! evolutionary! diversification! of! the! placenta! (Chuong! 2018;! Mi! et! al.! 2000).!
During!human!embryogenesis,!expression!of!specific!ERV!families!have!been!associated!
with!cell!identity!and!cell!pot ency!in!ea rly!stem!cells!(Göke!et!al.!2015).!Additionally,!ERV!
expression!has!been!reported!in!healthy!human!tissues!such!as!ovary!and!testis!for!ERV-
9! (Pi! et! al.! 2004),! pancreas! (Shiroma! et! al.! 2001),! breast! (Tavakolian,! Goudarzi,! and!
Faghihl oo!2019),! stomach! and! small! intestine! (Okahara!et!al.!2004).! Mainly! based! on!
transcriptional!studies,!the!expression!of!different!families!of!ERV!is!likely!to!be!extended!
to!more!tissues,!however!their!role!in!tissue!development!and!function!remains!largely!
unknown.!!
!
ERVs! have! been! linked! directly! and! indirectly! to! the! evolution! and! functioning! of! the!
immune!system.!Enhancer!regions!of!interferon!stimulated!genes!key!to!the!interferon!
pathway,!such!as!IRF1!and!STAT1,! were!found! i ntroduced! and!amplified!by!ERV! elements,!
with!the!human! inflammasome! failing!to!form! upon! the!deletion! of!a !subset! of!ERVs!
(Chuong,!Elde,!a nd! Feschotte!2 016) .!Ad aptive!immunity!also!benefits!from!the!presence!
of!ERVs.!Indeed,!ERV!peptide!recognition!is!used! in!T!cell !selection!to!optimise!antigen!
recognition! and! T! cells! have! a! higher! sensitivity! to! exogenous! virus! infection! when!
presented !with! ERV!peptides! during! their!initial!thymic!selection ! (Mandl! et! al.! 2013;!
Young!et!al.!2012).!The!human!ERV!(HERV)!envelope!gene!contains!immunosuppressive!
domains!that!reduce!the!Th1!response!during!pregnancy!and!therefore!promote!foetal!
development!(Knerr!et!al.! 2004;!Lokossou!et!al.! 2020).!The!role!of!ERVs!in!our!immune !
system,!particula rly!in!the!training!of!our!adap tive!immunity,!can!be!a!double-edge!sword!
as!ERVs!are!linked!to!a!range!of!different!disease!states,!including!autoimmunity.!!
!
Aberrant! expression! of! ERVs! contributes! to! multiple! pathologies.! ERVs! are! found! in!
abundance!in!multiple!forms!of!cancers!and!are!considered!tumour-promoting!fa ctors!
(extensively! reviewed! in! (Bermejo! et! al.! 2020).! The! pathology! of! auto-inflammatory!
diseases! has! also! been! strongly! associated! with! ERVs.! Syst emic! l upus! erythematosus!
(SLE)! is! an! autoimmune! disorder! with! increased! level! of! autoantigen! for! an! ERV!
(Jorgensen!et!al.!2014).! Recently,! one!of!the!murine! SLE! susceptibility!locus!was!i dentified!
as!a!key!suppressor!of!ERV!expression,!consolidating!the!role!of!ERVs!in!the!pathogenesis!
of!SLE!(Treger!et!al.!2019).!A!similar!disorder!is!Acardi-Goutieres!Syndrome!(AGS),!a !type!
1!interferonopathy!which!resembl es!congenital!cytome galovirus!infection!and!is!caused!
.CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under a
preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in
The copyright holder for thisthis version posted January 21, 2021. ; https://doi.org/10.1101/2021.01.21.427598doi: bioRxiv preprint

by!mutation!in!several!genes!encoding!enzymes!responsible!for!nucleic!acid!metabolism!
(Crow!et!al.!2015).!Mutations!in!some!of!these! genes,!such! as!TREX1,!MDA5! and!ADAR1!
trigger!aberrant!presence!of!various!retroelements!and!the!upregulation!of!an!antiviral!
immune!response!(Ahmad!et!al.!2018 ;!Thomas!et!al.!2017).!Interestingly,!anti-reverse!
transcriptase!therapy!in!AGS!pa tients!can!decrease!the!IFN!response,!highl ighting!the!role!
of! aberrant! presence! of! ERVs! in!triggering!an!immune!response! (G.!Rice!et!al.!2018).!
Increased! expression! of! ERVs! has! been! found! in! brains! of! patient! suffering! from!
neurodegenerative!disea ses! such!as! Motor! Neuron!Disease!(Li! et!al.!2015)!and!multiple!
sclerosis!(Johnston!et!al.!2001;!Mameli!et!al.!2007).!Overexpression!of!a!human!ERV!in!
neurons!was!shown!to!be!neurotoxic,!suggesting!a!p otential!role!of!ERVs!in!t riggering!
neuronal!toxicity!(Li!et!al.!2015).!A!direct!link!to!the!pathology!of!these!disorders!has!yet!
to!be!made,!but!nonetheless!ERVs!appear!as!strong!causal!factors!for!autoimmune!and!
neurological!disorders.!
Although!ERV!enrichment!has!been!detected!in!neurological!pathol ogies,!little!is! known!
about!the!function!of!ERV!in!healthy!tissues.!The!exact!role!of!ERV!in!our!immune!system!
and!brain!pathologies!has!yet!to!be!understood!and!there!is!no!model!system!specifically!
looking! at!ERV!function!&'(%&%).!!Zebrafish! is!already!established!as!a!model!to!study!the!
immune! system,! with! significan t! homology! with ! mammals! in! innat e! and! ada ptive!
immunity! (Trede! e t!al.! 2004;! Renshaw! and!Trede! 2012).! The !genetic! tractability! and!
transparency!of!the!zebrafish! embryos!have!allowed!the!creation!of!transgenic!reporter!
lines,!some!of !which!have!elucidated!the!role!of!immune!cell!behaviour!&'(%&%)!(Renshaw!
et!al.!2006).!The!tractability!of!the!zebrafish!has!already!been!exploited!to!visualise!the!
expression!of!the!human!ERV-9!in!oocytes,!similarly!to!human!expression!(Pi!et!al.!2004).!
In! this! study,! we! used! the! zebrafish! as! a! tractable! in! vivo! model! to! characterise! the!
zebrafish!endogenous!retrovirus!( !"#$%).! We!identified! multiple! !"#$%! family! members,!
including!2!complete!genomes!!"#$%/0!and!!"#$%/1.!Using!the!tractability!of!the!zebrafish!
larvae! we! developed! a! reporter! line! for! !"#$%/0! and! imaged! for! the! first! time! ERV!
activation!in!healt hy!tissues! &'(%&%).!We!showed!that!!"#$%/0!is!expressed!in!the!thymus!
and! in! the! brain.! Colocalisation! analysis! and! single! cell! RNA! sequencing! revealed!
expression! of! !"#$%/0! specifica lly! in! T-cells,! suggesting! a! potential! role! for! ERV! in!
lymphocyte! development.! Brain! exp ression! of! ZFERV! appears! to! include! neuronal!
expression.!This!transgenic!line!can!be!used!as!a!tool!to!further!investigate!the!role!of!
ERVs!in!immunity!and!in!neurological!disorders.!
!
!
!
!
!
!
!
!
!
!
!
!
!
!
!
!
!
!
!
!
!
.CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under a
preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in
The copyright holder for thisthis version posted January 21, 2021. ; https://doi.org/10.1101/2021.01.21.427598doi: bioRxiv preprint

Results"
"
The"zebrafish"genome"contains"multiple"endogenous"retrovirus"integrations."
The!presence!of!an!ERV!in!zebrafish,!named!!"#$%,!has!been!reported!by!Shen!an d!Steiner!
while!screening!a!thymic!cDNA!library!(Shen!and!Steiner!2004)!(Figure"1A).!We!searched!
for!related!sequences!in!t he!most!recent!reference!zebrafish!genome!(GRCz11,!Tü!strain)!
using!BLASTN!searches,!with!the!original! !"#$%!sequence!as!a!query.!Limiting!ourselves!to!
sequences!flan ked!by!long!terminal!repeats! (LTRs)! on!both!sides,!we!retrieved!8 ! hits!
scattered!in! t he!zebra fish!genome,!but!n ot!t he! exact!!"#$%!sequence,!possibly! because!of!
strain! difference! ( Figure" 1+" Sup." Table" 1).! We! identified! 2 ! sequences! encoding!
apparently!fully!functional!ERVs,!with!91%!and!90%!identity!to!!"#$%,!that!we!respectively!
named !!"#$%/0!and!!"#$%/1!(Figure"1B).!These!t wo!ERVs!encode!almost!identical!proteins!
(95!to!97%!identity! at!the!amino-acid!level).! Their! LTRs! are!also!highly!simil a r!(95%!
identity! at! the! nucleot id e! level),! suggesting! that! t heir! promoters! probably! d rive!
expression!in!similar!cells.!An!additional!6!pseudo!!"#$%!genes!(here!call ed!!"#$%2!-!!"#$%3)!
were!identified!(Figure"1C),! !"#$%2!containing!a! frameshift,!!"#$%4!and! !"#$%5!with!large!
deletions! and! !"#$%6,! !"#$%7! and! !"#$%3! containing! a! large! insertion,! all! resulting! in! a!
degenerated!ERV!genome.!
!
!
Figure" 1.!Multiple" copies"of"zferv" are" present" in" the" zebra fish" genome.! A.! Diagram! of! the!
original!zferv!genome!found!by!Steiner!et!al.!used!as!a!r eference!for!nucleotide!identity!(ID%!nt)!
B.!Diagram! representing! the! two! closest! related! !"#$%! genome! found!in!most!recent!zebrafish!
genome!GRCz11,!name d!zferv1a!and!!"#$%/1.!C.!Diagram!of! 6!pseudo!zferv!genes!with!degenerated!
genome! (dotted! line! represents! insertions).! 5’-LTR:!5’!Long! terminal! repeat,! Rep.:!Repetitive!
element,!gag-pol:!genes!encoding!the!polyprotein!and!reverse!transcriptase,!env:!envelope!gene.!
!
!
!
.CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under a
preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in
The copyright holder for thisthis version posted January 21, 2021. ; https://doi.org/10.1101/2021.01.21.427598doi: bioRxiv preprint

zferv1a!is"actively"expressed"in"the"brain"and"in"the"thymus.!
!"#$%!expression!was!initially!found!in!the!thymus!at!5!days!post!fertilisation!(dpf)!using!
a!RNA!probe!against!the!envelope!(#'%)!gene!of!the!original!!"#$%!(Shen!and!Steiner!2004).!
To! verify! this! observation,! the! entire! !"#$%/0! genome! was! subcloned! from! a! fosmid!!
provided!by!the!Sanger!Center!and! we!used! this!sequence!to!create!an!&'( 8&-9!hybridisatio n!
(ISH)!RNA!probe!against!the!envelope!gene!(#'%)!of!!"#$%/0.!ISH!p erformed!on!a !time!
course! starting! from! 2! cell-stage! embryos! from! the! '0,$#! strain! con firmed! that! the!
strongest!signal! appeared!at!5dpf!(Figure"2).!At!5dpf,!the!thymus!was!strongly!labelled!
(Figure" 2A-B),! similarly! to! what! was! previously! reported! (Shen! and! Steiner! 2004).!
Individual!labelled!cells!were!visible!around!the!thymus!following!the!branchial!arches!
and!around!the!ear!(Figure"2B).!Additionally,!we!identified!a!clear!signal!of!the!#'%!probe!
in!the!brain!(Figure!2A,!2C)!and!the!spinal!cord!(Figure!2A).!!
!
!
Figure" 2:"Reporter"line"for"zferv1a"recapitulates"endogenous"expression.!A.!Expression!of!the!
envelope! gene!(#'%)!by!&'(8&-9(hybridisation!from!2-cell!stage!until!5dpf.!Black!box!highlighting!
strong!expression!around!the!thymus,!black!arrows!highlighting!brain!and!spinal!cord!expression.!
Scale!bar! 500µm.! B.!Zoomed!image!on! the ! thymus!area,!single!positive!cells! are!visible ! in! the!
vicinity!of! the! thymus! around! the! ear! (white! arrowheads)! and! alongside! the! branchial! arches!
(white!arrows).!Scale!bar!70µm.!C.!Dorsal!view!of!#'%!expression!in!the!brain!at!5dpf.!Scale!bar!
100µm.!!
!
!
zferv1a"reporter"line"recapitulates"endogenous"expression.!
To!follow!the!expression!of!the!zebrafish!endogenous!retrovirus!we!identified!as!!"#$%/0,!
we!took!a!transgenic!approach!ta king!adva ntage!of!the!promoter!activity!of!retroviral!
LTR.!We!used!the!5’!viral!promoter!:-$6!from!!"#$%/0!to!drive!GFP!expres sion!by!Gateway!
recombination!(Figure"3A).!Injected!embryos!showing! expression!in!their!thymus!were!
raised!and!screened!in!adulthood!for!germline!transmission.!We!observed!a!strong!and!
consistent!GFP!expression!in!the!thymus!at!5dpf!in!F1!and!F2!larvae!(Figure"3B)!from!
.CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under a
preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in
The copyright holder for thisthis version posted January 21, 2021. ; https://doi.org/10.1101/2021.01.21.427598doi: bioRxiv preprint

References
More filters
Journal ArticleDOI
15 Dec 2006-Blood
TL;DR: It is shown that inflammation is induced after transection of the tail of zebrafish larvae and that this inflammation subsequently resolves over a similar time course to mammalian systems.

898 citations

Journal ArticleDOI
16 Jun 2005-Nature
TL;DR: It is shown that an engineered human LINE-1 element can retrotranspose in neuronal precursors derived from rat hippocampus neural stem cells, indicating that neuronal genomes might not be static, but some might be mosaic because of de novo L1 retrotransposition events.
Abstract: Revealing the mechanisms for neuronal somatic diversification remains a central challenge for understanding individual differences in brain organization and function. Here we show that an engineered human LINE-1 (for long interspersed nuclear element-1; also known as L1) element can retrotranspose in neuronal precursors derived from rat hippocampus neural stem cells. The resulting retrotransposition events can alter the expression of neuronal genes, which, in turn, can influence neuronal cell fate in vitro. We further show that retrotransposition of a human L1 in transgenic mice results in neuronal somatic mosaicism. The molecular mechanism of action is probably mediated through Sox2, because a decrease in Sox2 expression during the early stages of neuronal differentiation is correlated with increases in both L1 transcription and retrotransposition. Our data therefore indicate that neuronal genomes might not be static, but some might be mosaic because of de novo L1 retrotransposition events.

870 citations

Journal ArticleDOI
27 Aug 2009-Nature
TL;DR: It is demonstrated that neural progenitor cells isolated from human fetal brain and derived from human embryonic stem cells support the retrotransposition of engineered human L1s in vitro, and a quantitative multiplex polymerase chain reaction is developed that detected an increase in the copy number of endogenous L 1s in the hippocampus, and in several regions of adult human brains.
Abstract: It is known that LINE-1 (long interspersed element-1) retrotransposons can move throughout the genomes of adult rat neural progenitor cells (NPCs) in vitro and in the mouse brain. Now it is shown that NPCs isolated from human fetal brain and derived from human embryonic stem cells also support the retrotransposition of engineered human LINE-1s in vitro. Interestingly, there is an increase in the copy number of endogenous LINE-1s in the hippocampus and elsewhere in adult human brains when compared to the copy number of endogenous LINE-1s in heart or liver genomic DNA from the same individual. This suggests that LINE-1 retrotransposition events may contribute to individual somatic mosaicism and heterogeneity of gene expression in the brain. Long interspersed element 1 (LINE-1 or L1) retrotransposons have been shown to move throughout the genomes of adult rat neural progenitor cells (NPCs) in vitro and in the mouse brain. Here, NPCs isolated from human fetal brain and derived from human embryonic stem cells are shown to support the retrotransposition of engineered human L1s in vitro, which could contribute to individual somatic mosaicism. Long interspersed element 1 (LINE-1 or L1) retrotransposons have markedly affected the human genome. L1s must retrotranspose in the germ line or during early development to ensure their evolutionary success, yet the extent to which this process affects somatic cells is poorly understood. We previously demonstrated that engineered human L1s can retrotranspose in adult rat hippocampus progenitor cells in vitro and in the mouse brain in vivo1. Here we demonstrate that neural progenitor cells isolated from human fetal brain and derived from human embryonic stem cells support the retrotransposition of engineered human L1s in vitro. Furthermore, we developed a quantitative multiplex polymerase chain reaction that detected an increase in the copy number of endogenous L1s in the hippocampus, and in several regions of adult human brains, when compared to the copy number of endogenous L1s in heart or liver genomic DNAs from the same donor. These data suggest that de novo L1 retrotransposition events may occur in the human brain and, in principle, have the potential to contribute to individual somatic mosaicism.

773 citations

Journal ArticleDOI
TL;DR: The data indicate that CRISPR/Cas13a can be used for engineering interference againstRNA viruses, providing a potential novel mechanism for RNA-guided immunity against RNA viruses and for other RNA manipulations in plants.
Abstract: CRISPR/Cas systems confer immunity against invading nucleic acids and phages in bacteria and archaea. CRISPR/Cas13a (known previously as C2c2) is a class 2 type VI-A ribonuclease capable of targeting and cleaving single-stranded RNA (ssRNA) molecules of the phage genome. Here, we employ CRISPR/Cas13a to engineer interference with an RNA virus, Turnip Mosaic Virus (TuMV), in plants. CRISPR/Cas13a produces interference against green fluorescent protein (GFP)-expressing TuMV in transient assays and stable overexpression lines of Nicotiana benthamiana. CRISPR RNA (crRNAs) targeting the HC-Pro and GFP sequences exhibit better interference than those targeting other regions such as coat protein (CP) sequence. Cas13a can also process pre-crRNAs into functional crRNAs. Our data indicate that CRISPR/Cas13a can be used for engineering interference against RNA viruses, providing a potential novel mechanism for RNA-guided immunity against RNA viruses and for other RNA manipulations in plants.

771 citations

Journal ArticleDOI
TL;DR: It is shown that species-specific transposable elements have substantially altered the transcriptional circuitry of pluripotent stem cells and have wired new genes into the core regulatory network of embryonic stem cells.
Abstract: Detection of new genomic control elements is critical in understanding transcriptional regulatory networks in their entirety. We studied the genome-wide binding locations of three key regulatory proteins (POU5F1, also known as OCT4; NANOG; and CTCF) in human and mouse embryonic stem cells. In contrast to CTCF, we found that the binding profiles of OCT4 and NANOG are markedly different, with only approximately 5% of the regions being homologously occupied. We show that transposable elements contributed up to 25% of the bound sites in humans and mice and have wired new genes into the core regulatory network of embryonic stem cells. These data indicate that species-specific transposable elements have substantially altered the transcriptional circuitry of pluripotent stem cells.

729 citations