Science - USA (2021-07-09)

(Antfer) #1

  1. C. H. Oliveroset al.,Proc. Natl. Acad. Sci. U.S.A. 116 ,
    7916 – 7925 (2019).

  2. A. H. Thornhillet al.,Aust. Syst. Bot. 32 , 29–48 (2019).

  3. E. A. Ortlund, J. T. Bridgham, M. R. Redinbo, J. W. Thornton,
    Science 317 , 1544–1548 (2007).

  4. J. G. Cecere, F. Spina, S. Jenni-Eiermann, L. Boitani,J. Ornithol.
    152 , 923–931 (2011).

  5. Y. Todaet al., Data from“Early origin of sweet perception in the
    songbird radiation,”Dryad (2021); https://doi.org/10.5061/
    dryad.xksn02vfz.


ACKNOWLEDGMENTS
We thank those who supported these experiments; see
supplementary materials for a full list.Funding:Funding for this
work was obtained from the Putnam Expedition Grant (Harvard
University) and the Max Planck Society to M.W.B. and the Japan
Society for the Promotion of Science to Y.T. (18K14427 and
20H02941), Y.I. (#18KK0166), and T.M. (#16H04918, #19H02907).


Author contributions:Y.T., Q.L., M.-C.K., E.T.M., A.R.-G., T.N.,
T.S., T.H., S.S., Y.I., T.M., P.O., J.C., S.V.E., W.B., S.M., and M.W.B.
designed the research. A.R.-G., W.B., and M.W.B. designed and
conducted field experiments with honeyeaters. J.C., P.O., Q.L., and
M.W.B. designed experiments with canaries. Q.L. conducted
behavioral trials with canaries. Q.L., J.C., P.O., and M.W.B. analyzed
behavioral data. E.T.M. performed hidden Markov modeling.
E.T.M. and M.W.B compiled results of hidden rates analyses.
M.-C.K., P.O., Y.T., T.N., and M.W.B. designed the figures.
T.N. performed homology modeling. A.S. and K.U. cloned taste
receptors from white-eyes and bulbuls under the supervision of
S.M. Y.T. and M.W.B. cloned taste receptors from remaining
species, created and tested chimeric receptors, and determined
critical residues. Y.T. collected data for dose response curves and
ancestral receptors. Y.T., M.-C.K., and M.W.B. analyzed cell assay
experiments. T.S., M.-C.K., and M.W.B. performed ancestral
reconstruction and selection tests. Y.I., T.M., W.B., S.M., and
M.W.B. acquired funding. M.W.B. wrote the manuscript with input

from all authors.Competing interests:the authors declare no
competing interests.Data and materials availability:T1R
sequences are accessioned in GenBank (accession numbers
MZ220489 to MZ220511), and alignments and other data are
available at Dryad ( 24 ).

SUPPLEMENTARY MATERIALS
science.sciencemag.org/content/373/6551/226/suppl/DC1
Materials and Methods
Supplementary Text
Figs. S1 to S14
Tables S1 to S7
References ( 25 – 62 )
MDAR Reproducibility Checklist
Movie S1
10 November 2020; accepted 19 May 2021
10.1126/science.abf6505

ANTIVIRAL DEFENSE


An isoform of Dicer protects mammalian stem cells


against multiple RNA viruses


Enzo Z. Poirier^1 , Michael D. Buck^1 , Probir Chakravarty^2 , Joana Carvalho^3 †, Bruno Frederico^1 ,
Ana Cardoso^1 , Lyn Healy^4 , Rachel Ulferts^5 , Rupert Beale5,6, Caetano Reis e Sousa^1


In mammals, early resistance to viruses relies on interferons, which protect differentiated cells but not
stem cells from viral replication. Many other organisms rely instead on RNA interference (RNAi)
mediated by a specialized Dicer protein that cleaves viral double-stranded RNA. Whether RNAi also
contributes to mammalian antiviral immunity remains controversial. We identified an isoform of Dicer,
named antiviral Dicer (aviD), that protects tissue stem cells from RNA viruses—including Zika virus
and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)—by dicing viral double-stranded
RNA to orchestrate antiviral RNAi. Our work sheds light on the molecular regulation of antiviral RNAi in
mammalian innate immunity, in which different cell-intrinsic antiviral pathways can be tailored to the
differentiation status of cells.


T


ype I and III innate interferons (IFNs)
are rapidly induced in mammalian cells
in response to virus infection. These cyto-
kines act in an autocrine and paracrine
manner to promote the transcription of
multiple interferon-stimulated genes (ISGs),
which encode a variety of viral restriction, cel-
lular arrest, and cell death factors ( 1 ). The in-
ducible protection conferred by IFN receptor
signaling is much more marked in differen-
tiated cells than in embryonic and adult stem
cells, which lack expression of components
of the pathways that lead to IFN induction
and IFN responsiveness ( 2 – 4 ). This may ensure
that infected stem cells are spared the cyto-
static and cytotoxic effects of IFN exposure.


The IFN system is absent from invertebrates
and plants, which protect themselves from
viral infection by means of RNA interference
(RNAi) ( 5 ). Antiviral RNAi starts with the pro-
tein Dicer, which recognizes and cleaves double-
stranded RNA (dsRNA) produced during RNA
virus infection to generate small interfering
RNAs (siRNAs). These guide the sequence-
specific degradation of viral RNAs by a slicing-
active Argonaute protein such as Argonaute 2
(Ago2), present in insects and mammals. Irre-
spective of infection, RNAi also has a distinct role
in regulating cellular gene expression through
microRNAs (miRNAs) produced by Dicer cleav-
age of precursor miRNAs (pre-miRNAs) ( 5 ).
Recent work suggests that mammals, like in-
vertebrates and plants, can co-opt RNAi for
antiviral immunity ( 5 ). Examples of mamma-
lian antiviral RNAi in vitro and in vivo have
been reported for Nodamura virus, human
enterovirus 71, Zika virus, and other RNA
viruses ( 6 – 14 ), although other studies have
argued against the existence of such a response
( 15 – 18 ).Partofthecontroversymayrelateto
the fact that IFN inhibits mammalian dsRNA-
mediated RNAi and the latter may therefore
only be relevant in cells that are hyporespon-

sive to IFNs, such as stem cells ( 5 , 19 , 20 ). No-
tably, stem cells can resist virus infection, which
has been partly attributed to IFN-independent
constitutive expression of restriction factors
( 21 ). Whether stem cells additionally possess
specializations that favor antiviral RNAi re-
mains unclear.
Plants and insects that use RNAi both as
a means of regulating translation of cellular
mRNAs and as an antiviral mechanism encode
multiple Dicers, each dedicated to one arm of
the pathway. In contrast, mammals possess a
singleDICERgene with one canonical protein
product, which cleaves pre-miRNA but pro-
cesses dsRNA poorly ( 22 , 23 ). Interestingly, a
truncated form of Dicer with improved anti-
viral capacity can be produced from theDicer
gene in mice, but its expression is restricted to
oocytes ( 24 ). By analogy, we hypothesized that
antiviral RNAi in mammals may involve ex-
pression of an isoform of Dicer that processes
dsRNA more efficiently than canonical Dicer.
By performing a polymerase chain reaction
(PCR) on total cDNA from mouse small in-
testine, we identified an alternatively spliced
in-frame transcript of Dicer missing exons 7
and 8 (Fig. 1A). In silico translation of this
transcript resulted in a truncated Dicer pro-
tein in which the central Hel2i domain of the
N-terminal helicase segment is absent (Fig. 1A).
For simplicity, hereafter we refer to canonical
Dicer (which includes the sequences encoded
by exons 7 and 8) as Dicer and its truncated
form as antiviral Dicer (aviD). Using a reverse
transcription quantitative PCR (RT-qPCR) as-
say that distinguishesaviDandDicermRNA,
both isoforms could be detected in mouse cells,
including neural stem cells, embryonic stem
cells (ES cells), and a 3T3 cell line, as well as
in organs from pre-weaning or adult mice (fig.
S1, A to D). TheAVIDandDICERtranscripts
were also found in human ES cells, human in-
duced pluripotent stem cells (iPSCs), and some
human cell lines (fig. S1, E to H). In general,
transcripts encoding aviD appeared to be less
abundant than transcripts encoding full-length
Dicerbyatleastafactorof10(fig.S1),explaining

SCIENCEsciencemag.org 9JULY2021•VOL 373 ISSUE 6551 231


(^1) Immunobiology laboratory, Francis Crick Institute, London
NW1 1AT, UK.^2 Bioinformatics and Biostatistics, Francis Crick
Institute, London NW1 1AT, UK.^3 Experimental Histopathology,
Francis Crick Institute, London NW1 1AT, UK.^4 Human
Embryo and Stem Cell Unit, Francis Crick Institute, London
NW1 1AT, UK.^5 Cell Biology of Infection Laboratory,
Francis Crick Institute, London NW1 1AT, UK.^6 Division of
Medicine, University College London, London WC1E 6BT, UK.
*Corresponding author. Email: [email protected] (E.Z.P.);
[email protected] (C.R.S.)†Present address: Histopathology
Scientific Platform, Champalimaud Centre for the Unknown,
1400-038 Lisboa, Portugal.
RESEARCH | REPORTS

Free download pdf