8
RNA DELIVERY Mammalian retrovirus-like protein PEG10 packages its own mRNA and can be pseudotyped for mRNA delivery Michael Segel 1,2,3,4,5 , Blake Lash 1,2,3,4,5 , Jingwei Song 1,2,3,4,5 , Alim Ladha 1,2,3,4,5 , Catherine C. Liu 1,2,3,4,5,6 , Xin Jin 2,3,4,7 , Sergei L. Mekhedov 8 , Rhiannon K. Macrae 1,2,3,4,5 , Eugene V. Koonin 8 , Feng Zhang 1,2,3,4,5 * Eukaryotic genomes contain domesticated genes from integrating viruses and mobile genetic elements. Among these are homologs of the capsid protein (known as Gag) of long terminal repeat (LTR) retrotransposons and retroviruses. We identified several mammalian Gag homologs that form virus-like particles and one LTR retrotransposon homolog, PEG10, that preferentially binds and facilitates vesicular secretion of its own messenger RNA (mRNA). We showed that the mRNA cargo of PEG10 can be reprogrammed by flanking genes of interest with Peg10s untranslated regions. Taking advantage of this reprogrammability, we developed selective endogenous encapsidation for cellular delivery (SEND) by engineering both mouse and human PEG10 to package, secrete, and deliver specific RNAs. Together, these results demonstrate that SEND is a modular platform suited for development as an efficient therapeutic delivery modality. M ore than 8% of the human genome is composed of sequences derived from long terminal repeat (LTR) retroele- ments, including retroviruses, that have integrated into mammalian genomes throughout evolution (15). Retroviruses and retrotransposons have many common mech- anistic features, including the core structural gene (known as gag); however, whereas retro- transposons replicate intracellularly, the acqui- sition of the envelope (env) gene by retroviruses has enabled intercellular replication ( 6). Most endogenous retroelements have lost their orig- inal functions, but some of their genes have been recruited for diverse roles in normal mam- malian physiology. For example, the fusogenic syncytins evolved from retroviral env proteins ( 7). The gag homolog Arc, which forms capsids and has been reported to transfer mRNA (810), is involved in memory consolidation and regu- lates inflammation in the skin (11, 12). Another gag homolog, the LTR retrotransposonderived protein PEG10, which has been reported to bind RNA and also forms capsids (13), is in- volved in mammalian placenta formation (14, 15). These examples raise the possibility that retroelement-derived proteins encoded in the mammalian genome may be harnessed to transfer specific nucleic acids, providing a potentially programmable mechanism for in- tercellular communication. Computational survey of mammalian capsid-forming gag homologs To identify genes with the potential to transfer specific nucleic acids, we focused on homologs of gag that contain the core capsid (CA) do- main, which protects the genome of both retrotransposons and exogenous retroviruses (16, 17). Previous genome analyses identified many endogenous gag homologs in mamma- lian genomes (18), and experimental efforts have validated the ability of some of these pro- teins, including Mus musculus Arc (MmArc) and MmPEG10, to form capsid-like particles that are secreted within extracellular vesicles (EVs) (10, 13). To ensure a complete list of candidates, we searched the human and mouse genomes for gag homologs. This search iden- tified 48 gag-derived genes in the human genome and 102 gag homologs in the mouse genome; for 19 human genes, an orthologous relationship between human and mouse was readily traced (in several cases, with additional mouse paralogs), whereas the remaining ones appeared to be species-specific (tables S1 and S2). Canonical genomes of both LTR retrotrans- posons and retroviruses encode a long poly- protein consisting of several conserved domains: The matrix (MA), CA, and nucleocapsid (NC) form the gag subdomain and are responsible for membrane attachment, capsid formation, and genome binding, respectively. The pol sub- domain contains the protease (PRO), which is responsible for cleaving the polyprotein; the reverse transcriptase (RT), which converts retroelement RNA into DNA; and the integrase (IN) domain, which integrates the genome into that of the host. Some families of endogenous gag-containing proteins, such as the PNMA (paraneoplastic antigen Ma) family, contain only the CA and NC subdomains of gag, whereas others, such as RTL1 and PEG10 (also known as RTL2 or Mart2), additionally include subdomains of pol , namely a PRO domain and a predicted RT-like domain (Fig. 1A and table S1). Phylo- genetic analysis of Peg10 and its homologs supports the origin of this gene from LTR ret- rotransposons (fig. S1, A and B). Among these genes, Arc is the most well studied. Drosophila Arc1 ( darc1 ) is a gag homolog that contains the MA, CA, and NC domains. It has been shown to form capsids, bind its own mRNA, and transfer it from motor neurons to muscles at the neuromuscular junction ( 9). darc1 mRNA binding is dependent on its own 3- untranslated region (3UTR), and fusion of this sequence to heterologous mRNAs can initiate their export and transfer as well. MmArc, by contrast, contains only the CA domain and has also been shown to form capsids and transfer Arc and other mRNAs across synapses (10). To narrow down the scope of our analysis, we focused on CA domaincontaining pro- teins that are conserved between human and mouse and have detectable levels of mRNA in adult human tissues, reasoning that such pro- teins were most likely to have been co-opted for important physiological roles in mammals (fig. S2). We produced mouse versions of the selected CA-containing proteins in Escherichia coli and found that a number of these formed higher molecular weight oligomers that were identified by size exclusion (Fig. 1B and fig. S3A), as previously noted for some of these proteins, such as MmArc (10). Electron micros- copy of these aggregated proteins showed that MmMOAP1, MmZCCHC12, MmRTL1, MmPNMA3, MmPNMA5, MmPNMA6a, and MmPEG10 self- assemble into capsid-like particles, many of which appear spherical (Fig. 1, C and D, and fig. S3, B and C). MmPEG10 binds and secretes its own mRNA To determine whether these proteins are secreted within an EV, we overexpressed an epitope-tagged mouse ortholog of each CA-containing gene in human embryonic kidney (HEK) 293 FT cells and harvested both the whole-cell lysate and the virus- like particle (VLP) fraction by clarification and ultracentrifugation of the culture media (Fig. 1E). We found that MmMOAP1, MmArc, MmPEG10, and MmRTL1 were all present in the VLP fraction (Fig. 1F and fig. S4A), but MmPEG10 was the most abundant protein in the VLP fraction (Fig. 1G). Additionally, endogenous MmPEG10, but not MmMOAP1 or MmRTL1, was readily detect- able in cell-free adult mouse serum (fig. S4B). We next tested whether any of the capsid- like particles formed by Gag homologs con- tained specific mRNAs using RNA sequencing. RESEARCH Segel et al., Science 373, 882889 (2021) 20 August 2021 1 of 7 1 Howard Hughes Medical Institute, Cambridge, MA 02139, USA. 2 Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA. 3 McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. 4 Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. 5 Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. 6 Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. 7 Society of Fellows, Harvard University, Cambridge, MA 02138 USA. 8 National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, USA. *Corresponding author. Email: [email protected] Downloaded from https://www.science.org at Chinese University of Hong Kong on September 09, 2021

Mammalian retrovirus-like protein PEG10 packages its own

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

RNA DELIVERY

Mammalian retrovirus-like protein PEG10 packagesits own mRNA and can be pseudotyped formRNA deliveryMichael Segel1,2,3,4,5, Blake Lash1,2,3,4,5, Jingwei Song1,2,3,4,5, Alim Ladha1,2,3,4,5,Catherine C. Liu1,2,3,4,5,6, Xin Jin2,3,4,7, Sergei L. Mekhedov8, Rhiannon K. Macrae1,2,3,4,5,Eugene V. Koonin8, Feng Zhang1,2,3,4,5*

Eukaryotic genomes contain domesticated genes from integrating viruses and mobile genetic elements.Among these are homologs of the capsid protein (known as Gag) of long terminal repeat (LTR)retrotransposons and retroviruses. We identified several mammalian Gag homologs that form virus-likeparticles and one LTR retrotransposon homolog, PEG10, that preferentially binds and facilitates vesicularsecretion of its own messenger RNA (mRNA). We showed that the mRNA cargo of PEG10 can bereprogrammed by flanking genes of interest with Peg10’s untranslated regions. Taking advantage of thisreprogrammability, we developed selective endogenous encapsidation for cellular delivery (SEND) byengineering both mouse and human PEG10 to package, secrete, and deliver specific RNAs. Together,these results demonstrate that SEND is a modular platform suited for development as an efficienttherapeutic delivery modality.

More than 8% of the human genome iscomposed of sequences derived fromlong terminal repeat (LTR) retroele-ments, including retroviruses, that haveintegrated into mammalian genomes

throughout evolution (1–5). Retroviruses andretrotransposons have many common mech-anistic features, including the core structuralgene (known as gag); however, whereas retro-transposons replicate intracellularly, the acqui-sition of the envelope (env) gene by retroviruseshas enabled intercellular replication (6). Mostendogenous retroelements have lost their orig-inal functions, but some of their genes havebeen recruited for diverse roles in normalmam-malian physiology. For example, the fusogenicsyncytins evolved from retroviral env proteins(7). The gag homolog Arc, which forms capsidsandhas been reported to transfermRNA (8–10),is involved inmemory consolidation and regu-lates inflammation in the skin (11, 12). Anothergaghomolog, the LTR retrotransposon–derivedprotein PEG10, which has been reported tobind RNA and also forms capsids (13), is in-volved in mammalian placenta formation(14, 15). These examples raise the possibilitythat retroelement-derived proteins encoded

in the mammalian genomemay be harnessedto transfer specific nucleic acids, providing apotentially programmable mechanism for in-tercellular communication.

Computational survey of mammaliancapsid-forming gag homologs

To identify genes with the potential to transferspecific nucleic acids, we focused on homologsof gag that contain the core capsid (CA) do-main, which protects the genome of bothretrotransposons and exogenous retroviruses(16, 17). Previous genome analyses identifiedmany endogenous gag homologs in mamma-lian genomes (18), and experimental effortshave validated the ability of some of these pro-teins, including Mus musculus Arc (MmArc)and MmPEG10, to form capsid-like particlesthat are secreted within extracellular vesicles(EVs) (10, 13). To ensure a complete list ofcandidates, we searched the human andmousegenomes for gag homologs. This search iden-tified 48 gag-derived genes in the humangenome and 102 gag homologs in the mousegenome; for 19 human genes, an orthologousrelationship between human and mouse wasreadily traced (in several cases, with additionalmouse paralogs), whereas the remaining onesappeared to be species-specific (tables S1 and S2).Canonical genomes of both LTR retrotrans-

posons and retroviruses encode a long poly-protein consisting of several conserved domains:The matrix (MA), CA, and nucleocapsid (NC)form the gag subdomain and are responsiblefor membrane attachment, capsid formation,and genome binding, respectively. The pol sub-domain contains the protease (PRO), which isresponsible for cleaving the polyprotein; thereverse transcriptase (RT), which convertsretroelement RNA intoDNA; and the integrase

(IN) domain, which integrates the genome intothat of the host. Some families of endogenousgag-containing proteins, such as the PNMA(paraneoplastic antigen Ma) family, containonly theCAandNCsubdomains ofgag,whereasothers, such as RTL1 and PEG10 (also known asRTL2orMart2), additionally include subdomainsof pol, namely a PRO domain and a predictedRT-like domain (Fig. 1A and table S1). Phylo-genetic analysis of Peg10 and its homologssupports the origin of this gene from LTR ret-rotransposons (fig. S1, A and B).Among these genes, Arc is the most well

studied.DrosophilaArc1 (darc1) is a gaghomologthat contains the MA, CA, and NC domains. Ithas been shown to form capsids, bind its ownmRNA, and transfer it from motor neurons tomuscles at the neuromuscular junction (9). darc1mRNA binding is dependent on its own 3′-untranslated region (3′UTR), and fusion of thissequence to heterologous mRNAs can initiatetheir export and transfer as well. MmArc, bycontrast, contains only the CA domain and hasalso been shown to form capsids and transferArc and other mRNAs across synapses (10).To narrow down the scope of our analysis,

we focused on CA domain–containing pro-teins that are conserved between human andmouse and have detectable levels of mRNA inadult human tissues, reasoning that such pro-teins were most likely to have been co-optedfor important physiological roles in mammals(fig. S2). We produced mouse versions of theselected CA-containing proteins inEscherichiacoli and found that a number of these formedhigher molecular weight oligomers that wereidentified by size exclusion (Fig. 1B and fig.S3A), as previously noted for some of theseproteins, such asMmArc (10). Electronmicros-copy of these aggregated proteins showed thatMmMOAP1,MmZCCHC12,MmRTL1,MmPNMA3,MmPNMA5, MmPNMA6a, and MmPEG10 self-assemble into capsid-like particles, many ofwhich appear spherical (Fig. 1, C and D, andfig. S3, B and C).

MmPEG10 binds and secretes its own mRNA

To determine whether these proteins are secretedwithin anEV,weoverexpressed anepitope-taggedmouse ortholog of each CA-containing gene inhuman embryonic kidney (HEK) 293 FT cells andharvested both thewhole-cell lysate and the virus-like particle (VLP) fraction by clarification andultracentrifugation of the culture media (Fig. 1E).We found that MmMOAP1, MmArc, MmPEG10,andMmRTL1were all present in theVLP fraction(Fig. 1F and fig. S4A), butMmPEG10was themostabundant protein in the VLP fraction (Fig. 1G).Additionally, endogenous MmPEG10, but notMmMOAP1 or MmRTL1, was readily detect-able in cell-free adult mouse serum (fig. S4B).We next tested whether any of the capsid-

like particles formed by Gag homologs con-tained specificmRNAs using RNA sequencing.

RESEARCH

Segel et al., Science 373, 882–889 (2021) 20 August 2021 1 of 7

1Howard Hughes Medical Institute, Cambridge, MA 02139,USA. 2Broad Institute of MIT and Harvard, Cambridge, MA02142, USA. 3McGovern Institute for Brain Research,Massachusetts Institute of Technology, Cambridge, MA02139, USA. 4Department of Brain and Cognitive Science,Massachusetts Institute of Technology, Cambridge, MA02139, USA. 5Department of Biological Engineering,Massachusetts Institute of Technology, Cambridge, MA02139, USA. 6Department of Biology, Massachusetts Instituteof Technology, Cambridge, MA 02139, USA. 7Society ofFellows, Harvard University, Cambridge, MA 02138 USA.8National Center for Biotechnology Information, NationalLibrary of Medicine, National Institutes of Health, Bethesda,MD 20894, USA.*Corresponding author. Email: [email protected]

Dow

nloaded from https://w

ww

.science.org at Chinese U

niversity of Hong K

ong on September 09, 2021

Segel et al., Science 373, 882–889 (2021) 20 August 2021 2 of 7

A

C D

MmASPRV1

MmPNMA3

MmMOAP1 MmRTL1

MmPEG10

cryo

TE

M

B

frac

tion

prot

ein

olig

omer

ized 1.0

0.5

0

Mm

Arc

Mm

RTL1

Mm

PNMA6

Mm

PNMA3

Mm

PNMA1

Mm

PEG10

Mm

PNMA5

Mm

MOAP1

Mm

ASPRV1

Mm

Zcchc

12

E F

G

Mm

ASPRV1

Mm

RTL1

Mm

PEG10

Mm

PNMA6

Mm

PNMA5

Mm

PNMA3

Mm

Zcchc

12

Mm

Arc

Mm

PNMA1

Mm

MOAP1

mChe

rry

160

50

40

kDa

HA

CD

81C

NX

CN

X

VLP

frac

tion

from

HE

K29

3FT

cel

lsW

.C.

overexpress HA-tagged capsid protein in HEK293FT

filter supernatant with 0.45µm filter andconcentrate via ultracentrifugation to obtain VLP fraction

blot VLP fraction for HA

HA

band

inte

nsity

rat

ioV

LP /

(who

le c

ell+

VLP

)

0.0

0.2

0.4

0.6

Mm

ASPRV1

Mm

MOAP1

Mm

PNMA1

Mm

Arc

Mm

ZCCHC5

Mm

PNMA3

Mm

PNMA5

Mm

PNMA6

Mm

PEG10

Mm

RTL1

********

MmASPRV1 MmMOAP1

MmPNMA3

MmZcchc12

MmPNMA5 MmPNMA6A

MmRTL1

MmPEG10

nega

tive

stai

nT

EM

MA CA NC PR RT

ENVGAG POL

IN LTRLTRretroviralgenome

Asprv1

Peg10 (Rtl2)

Rtl1 (Peg11)

* ribosomal frameshift

MA CA NC PR RT

GAG POL

IN LTRLTRLTR retrotransposon

genome

Arc

Pnma3, Pnma5,Zcchc12 (Pnma8)

Pnma1, Pnma6a, Moap1 (Pnma4)

Fig. 1. Identification of mammalian retroelement-derived Gag homologsthat form capsids and are secreted. (A) Domain architectures of selectedcapsid (CA)–containing mammalian Gag homologs compared with that of typicalretrovirus and LTR retrotransposons. Each group of Gag homologs contains adistinct combination of predicted CA, nucleocapsid (NC), protease (PR), andreverse transcriptase (RT) domains. LTR, long terminal repeat; MA, matrix; IN,integrase. (B) Fraction of the total bacterially produced protein that formsoligomers (>600 kD), as determined by size exclusion chromatography.(C) Representative negative stain transmission electron micrographs (TEMs) ofthe Mm orthologs of the CA domain–containing proteins. Scale bar, 100 nm.

(D) Representative electron micrographs using cryogenic electron microscopy(cryoTEM) of a selected subset of the identified CA domain–containing proteins.Scale bar, 50 nm. (E) Method for detecting extracellular forms of CA domain–containing homologs. (F) Representative blots of CA domain–containingproteins in the cell-free fraction. CD81 was used as loading control for theultracentrifuged cell-free fraction. Whole-cell (W.C.) and VLP fraction blots for theendoplasmic reticulum marker CALNEXIN (CNX) ensure equal loading of wholecell protein and the purity of cell-free VLP fraction. (G) Quantification ofextracellular CA domain–containing proteins [as in (F)] on the basis of n = 3replicates. ****P < 0.0001.

RESEARCH | RESEARCH ARTICLED

ownloaded from

https://ww

w.science.org at C

hinese University of H

ong Kong on Septem

ber 09, 2021

To avoid the possibility of transfected Gaghomolog expression plasmids contributingto high background signal during sequencing,we used CRISPR activation (19) to induce ex-pression of endogenous genes in mouse N2acells (Fig. 2A and fig. S5A). We performed

mRNA sequencing on whole-cell lysate andthe VLP fraction (after nuclease treatment toremove any residual, unencapsidated RNA)to identify RNA species in the VLP fraction.We found thatMmPeg10 transcriptional acti-vation led to the accumulation of appreciable

amounts of full-lengthMmPeg10mRNA tran-scripts in the VLP fraction (Fig. 2, B and C).Previous work on MmPEG10 demonstratedthat it binds a number of mRNAs insidetrophoblast stem cells, including itself (13);however, here we further show that MmPEG10

Segel et al., Science 373, 882–889 (2021) 20 August 2021 3 of 7

A B

sign

ifica

nce

(-lo

g10

q-va

lue)

5

4

3

2

1

0–10 1050–5

MmPeg10

Rn7s2

Rn7s1

NTsgRNA

MmPeg10sgRNA

differential RNA abundance inVLP fraction (log2 fold change)

activate each capsid gene in N2a cellsusing CRISPR activation

filter, concentrate, MNase treat VLP fraction

sequence total RNA

G H

E F

I

Mm

Peg

10sg

RN

AN

Tsg

RN

A W.C.

VLP

W.C.

VLP

C

D

MmPeg10

5 kbp 500

read

s

sign

ifica

nce

(-lo

g10

q-va

lue)

150

100

50

0–10 1050–5

differential RNA abundance inVLP fraction (log2 fold change)

RNA found in the VLP fraction from N2a cellsafter heterologous expression of MmPeg10

MmPeg10

CMV::mCherry CMV::MmPeg10

fold

Mm

Peg

10 m

RN

A e

nric

hmen

tGFP

Mm

Peg10

-ΔNC

(del4

16-4

29)

Mm

Peg10

Mm

Peg10

-ΔRT

(del8

61-1

037)

150

100

50

0

MmPeg10 mRNA in VLP fraction from N2acells expressing different MmPeg10 mutants

transcription &translation

proteolyticprocessing

CANC*

PRRT*

ribosome frameshiftleads to 2 polypeptides

*ribosomal frameshift

MmPeg10 gene

* RNA binding

sign

ifica

nce

(-lo

g10

q-va

lue)

1.5

1.0

0.5

00 15105

MmDdit4

enrichment of MmPEG10-boundmRNAs (log2 fold changerelative to HA-GFP cells)

MmPeg10 MmYwhag

eCLIP detection of MmPEG10-HA-boundRNAs in N2a cells

eCLIP detection of MmDdit4 mRNAbound by MmPEG10 mutants

in N2a cells

3' UTR

2000

rea

ds

0.5 kbp

5' UTR

HA-GFP

MmPEG10(ΔNC)-HA

MmPEG10-HA

MmPEG10(ΔRT)-HA

MmDdit4

3 kbp 500

read

s

5' UTR 3' UTR

eCLIP detection of MmPeg10 mRNAof P30 frontal cortex tissue fromHA-MmPeg10 knock-in mouse

HA-MmPeg10knock-in

mouse

C57BL/6wildtype

mouse

MmPeg10

RNA found in the VLP fractionfrom N2a cells after CRISPR

activation of endogenous MmPeg10

RNAseq coverage of MmPeg10after CRISPR activation in whole-cell

and VLP fractions

Fig. 2. MmPEG10 protein and mRNA are secreted in vesicles by cells in vitro.(A) Method for identifying nucleic acids that are secreted in the VLP fractionupon gene activation of CA domain–containing proteins. (B) Differential RNAabundance and significance in the VLP fraction from N2a cells after CRISPRactivation of endogenous MmPeg10. NT, nontargeting gRNA. (C) Alignment ofsequencing reads showing sequencing coverage of the MmPeg10 mRNA from(B). (D) Differential RNA abundance and significance in the VLP fraction fromN2a cells after heterologous transfection of MmPeg10. n = 3 replicates. CMV,cytomegalovirus. (E) Four domains of MmPEG10 are translated into twoisoforms. These are self-processed by the PEG10 protease into separate

domains, of which the NC and RT bind RNA. (F) Fold enrichment of MmPeg10mRNA compared with GFP in the VLP fraction from N2a cells transfected withwild-type MmPeg10 or deletions of the predicted nucleocapsid (DNC) andreverse transcriptase (DRT) domains. (G) Log2 fold change and significance ofbound RNAs from eCLIP data comparing HA-GFP with wild-type MmPEG10-HA.(H) Representative sequencing alignment histogram of the MmDdit4 locusgenerated from eCLIP of N2a cells transfected with wild-type or mutantMmPeg10. (I) Representative sequencing alignment histogram of theMmPeg10 locus generated from eCLIP data of n = 3 HA-PEG10 and n = 3untagged animals.

RESEARCH | RESEARCH ARTICLED

ownloaded from

https://ww

w.science.org at C

hinese University of H

ong Kong on Septem

ber 09, 2021

binds and secretes its ownmRNA into the VLPfraction.An important caveat of this experimentis that someof theseproteins,particularlyMmArc,are subject to regulation at the level of translation,so the lack of enrichment in the VLP fractioncould be due to low protein expression (20).To confirm our observation for MmPeg10,

we transiently transfected overexpression plas-mids of UTR-flanked MmPeg10 into N2a cellsand found only enrichment for MmPeg10mRNA in the VLP fraction (Fig. 2D) under thisoverexpression condition. PEG10 contains twoputative nucleic acid-binding domains, namelythe NC and RT, which are released from thepolypeptide upon PEG10 self-processing (21)(Fig. 2E, supplementary text 1, and fig. S5, B toD). We generated deletions of these domainsand found that mRNA export depends on theMmPEG10 NC, as loss of the nucleic acid–binding zinc finger CCHC motif (residues 416to 429) from the MmPEG10 NC substantiallyreduced export of its mRNA (Fig. 2F).To better understand the roles of the nucleic

acid–binding domains of MmPEG10 in RNAbinding, we performed enhanced cross-linkingand immunoprecipitation (eCLIP) in N2acells after transient transfection with hemag-glutinin (HA)–tagged MmPeg10 as well asthe NC and RT mutants (fig. S6, A and B).Comparedwith the control,MmPEG10 stronglybound a number of mRNAs in N2a cells, in-cluding its own mRNA (Fig. 2G). Notably, boththeNC and theRTdomains are required for thebinding of thesemRNAs byMmPEG10 (Fig. 2Hand fig. S6C). To confirm MmPEG10’s cellularrole in an in vivo context, we generated knockinmice carrying an N-terminal HA tag on theendogenous MmPEG10 protein (fig. S6D). Ex-pression of MmPeg10 in cortical neurons hasbeen demonstrated previously (fig. S6E) (22).Endogenous MmPEG10 was also found to bindits own mRNA as well as other transcriptsabundant in neurons (fig. S6, F and G); incontrast to previous datasets, we detectedstrong MmPEG10 binding in the 5′ UTR, aswell as some additional binding near theboundary between the NC and PRO codingsequences and in the beginning of the 3′UTR (Fig. 2I) (13).Binding of mRNA by MmPEG10 has been

reported to increase the cellular abundanceof target transcripts (13). To confirm this roleof MmPEG10 in its native context in vivo, weperturbed MmPeg10 gene expression in thepostnatal mouse brain and assessed the ex-pression changes of MmPEG10-bound transcripts(supplementary text 2). We found that themRNAs of 49 genes that are down-regulatedin the brain upon MmPeg10 knockout arebound to MmPEG10 in the age-matchedmouse brain (fig. S7F), suggesting that oneof the functions of MmPEG10 is to bind andstabilize mRNAs with fundamental roles inneurodevelopment.

Pseudotyped PEG10 VLPs can deliverengineered cargo mRNAs bearing RNApackaging signals from PEG10 UTRsTo reprogramMmPEG10 to bind and packageheterologous RNA, we tested whether a cargomRNA consisting of both the 5′ and 3′ UTR ofMmPeg10 flanking a gene of interest would beefficiently packaged, exported, delivered, andtranslated in recipient cells (Fig. 3A). ThisUTR grafting approach has been demonstratedfor the Ty3 retroelement and darc1 (9, 23). Wefirst used a Cre-loxP system, a highly sensitivesystem for trackingRNAexchange that has beenused previously with exosomes in vivo (24). Weflanked the Cre recombinase coding sequencewith the MmPeg10 UTRs and cotransfected itwithMmPeg10 with and without a fusogen, thevesicular stomatitis virus envelopeprotein (VSVg)(Fig. 3A). We found that MmPEG10 VLPs pseu-dotyped with VSVg are secreted within EVs thatmediate transfer of Cre mRNA, not protein, intotarget loxP–green fluorescent protein (GFP) re-porterN2a cells in a VSVg- andMmPeg10UTR–dependent manner (Fig. 3, B to D; fig. S8; andsupplementary text 3). This result suggests thataddition of the Peg10 UTRs enables the func-tional intercellular transfer of anmRNAviaVLPsand that these VLPs require a fusogenic proteinfor cell entry.We next examined whether there is a min-

imal UTR packaging signal for mediating effi-cient packaging and functional transfer. The 3′UTR of MmPeg10 is ~4 kb long, but eCLIPindicates that only portions of the 3′ UTR arebound by MmPEG10 (Fig. 2I). We createdconstructs that encode the MmPeg10 5′ UTR,Cre, and 500–base pair (bp) segments of theMmPeg10 3′UTR. We found that the proximal500 bp of the MmPeg10 3′ UTR are sufficientfor efficient functional transfer of Cre mRNAinto target reporter cells (Fig. 3E). Notably, noefficient functional mRNA transfer was ob-served for non–UTR-flanked Cre or for Crewithout the proximal 500 bp of the 3′ UTR.Henceforth, we refer to RNA cargo flankedby the MmPeg10 5′ UTR and the proximal500 bp of the 3′UTR asMm.cargo(RNA), where“(RNA)” specifies the cargo being flanked [e.g.,Mm.cargo(Cre)].Like the mouse ortholog, human PEG10

(HsPEG10) is an abundantly secreted proteinin the VLP fraction (fig. S10A). Using the sameapproach that we employed with MmPeg10,we identified that the 5′ UTR and the first500 bp of the HsPEG10 3′ UTR are sufficientto mediate functional transfer of Cre mRNA,hereafter denoted asHs.cargo(RNA) (Fig. 3F).Notably, these functional regions of the UTRsare highly conserved acrossmammals (fig. S10B).Similar to its mouse ortholog, the human systemis specific and requiresHsPEG10UTR sequencesfor functional mRNA transfer, whereas non-flanked Cre produced only minimal reportercell activity (Fig. 3F).

To further boost the packaging of a cargoRNA by PEG10, we explored the impact ofremoving any additional PEG10 cis bindingelements within theMmPeg10/HsPEG10 codingsequence. For both human and mouse ortho-logs, transferwas increasedasa result of recodingthe sequence between the NC and the PROdomains, which corresponds to the MmPEG10-bound region in the eCLIP experiments (Fig. 2Iand supplementary text 4).Combining these optimizations, we produced

VLPs with the recoded mouse and humanPEG10 (rMmPEG10 or rHsPEG10), VSVg, andthe optimized cargoRNA containing the first500 bp of the 3′UTR; we refer to this system asselective endogenous encapsidation for cellulardelivery (SEND). With SEND, we detected asubstantial (up to 60%) increase in the func-tional transfer of cargo(Cre) into N2a cells forboth human andmouse PEG10 (Fig. 3, G andH).Furthermore, we showed that VLPs producedwith rMmPEG10 can mediate the functionaltransfer of H2B-mCherry (fig. S12, A and B). Acomparison of SEND with previously devel-oped delivery vectors showed that SEND isfour to five times less potent than an integratinglentiviral vector, as assayed by digital dropletpolymerase chain reaction and functional titra-tion (fig. S12, B to E). However, given that SENDdelivers mRNA rather than integrating anoverexpression cassette, we expect it to per-form competitively against other mRNA deliv-ery vehicles.

PEG10 is a modular platform for RNA delivery

To generate a fully endogenous SEND system,we tested whether VSVg can be replaced withan endogenous fusogenic transmembrane pro-tein. Given the overlapping tissue expression ofMmPeg10/HsPEG10 and syncytin genes (sup-plementary text 5), we tested the feasibility ofpseudotyping the mouse SEND system withMmSYNA orMmSYNB compared with pseudo-typing with VSVg. Pseudotyped particles wereincubated with tail-tip fibroblasts from loxP-tdTomato reportermice, a cell type thatwe havefound amenable to transduction by thesefusogens. Based on previous reports, we addedthe transduction enhancer vectofusin-1 to thesupernatant for MmSYNA and MmSYNB par-ticles to enhance in vitro transduction (25). Inthese primary cells, both VSVg and MmSYNAenabled SEND-mediated functional transfer ofMm.cargo(Cre), whereas MmSYNB did not(Fig. 4, A and B). Again, this packaging washighly specific, as only UTR-flanked mRNA[i.e., Mm.cargo(Cre)] was functionally trans-ferred. Together with MmSYNA, SEND can beconfigured as a fully endogenous system forfunctional gene transfer.Supported by our understanding of the mini-

mal requirements for PEG10-mediated mRNAdelivery (i.e.,UTRsandanendogenous fusogen),we could begin to probe the endogenous role of

Segel et al., Science 373, 882–889 (2021) 20 August 2021 4 of 7

RESEARCH | RESEARCH ARTICLED

ownloaded from

https://ww

w.science.org at C

hinese University of H

ong Kong on Septem

ber 09, 2021

Segel et al., Science 373, 882–889 (2021) 20 August 2021 5 of 7

A UTR-mediated packaging of cargoRNA in PEG10 VLPs

transduce target cells with VLPsPeg10

cargoRNA

Peg10 5' UTRGene of Interest

Peg10 3' UTR

transfect Peg10, cargoRNA & fusogen

plasmids

Fusogen

B

F

G H

E

0 10 20 30 40 50

1 2 3 4 5 6Cre

MmPeg10 5' UTR

MmPEG10 +

mCherry +

untransfected

MmPeg10 3' UTR

33901 370 6667**

****GFP+ cells (%)

0 20 40 60 80

SEND(rMmPEG10,VSVg, Cre mRNA)

GFP+ cells (%)

***

SEND(rMmPEG10, VSVg, Mm.cargo(Cre))

0 20 40 60 80

SEND(rHsPEG10, VSVg, Cre mRNA)

GFP+ cells (%)

***

SEND(rHsPEG10, VSVg, Hs.cargo(Cre))

C D

40

10

30

20

0

GF

P+

cel

ls (

%)

CMV::MmPeg10

CMV::mCherry

CMV::VSV-G

CMV::UTR Cre

+

+

+

+

+

+

+

+

+

+

****

MmPEG10 VLP mediatedmRNA transfer and expression

CMV::MmPeg10 +CMV::UTR Cre

CMV::MmPeg10 +CMV::Cre

Cre lentivirus

GF

PD

AP

I

anti-PEG10 immunogold

1 2 3 4 5 66Cre

HsPeg10 5' UTR

HsPEG10 +

mCherry +

cargo mRNA cargo mRNAHsPeg10 3' UTR

26051 343 66180 10 20 30 40 50

********

GFP+ cells (%)

7 8 9 10

Negative

PEG10 PEG10

PEG10

Fig. 3. Flanking mRNA with MmPeg10 5 and 3 UTRs enables functionalintercellular transfer of mRNA into a target cell. (A) Schematic showingreprogramming MmPEG10 for functional delivery of a cargo RNA flankedwith the MmPeg10 5′ and 3′ UTRs [hereafter “cargo(RNA)”]. (B) Represent-ative TEMs of VLP fraction immunogold labeled for MmPEG10. Text labelsindicate transfection of cells with MmPeg10 or mock (negative). Arrowheadsindicate gold labeling. Scale bar, 50 nm. (C) Representative images ofloxP-GFP N2a cells treated with VSVg-pseudotyped MmPEG10 VLPs, whichwere produced by transfecting Mm.cargo(Cre) or Cre mRNA, and a lentivirusencoding Cre. Scale bar, 100 mm. DAPI, 4′,6-diamidino-2-phenylindole.(D) Functional transfer of RNA into loxP-GFP N2a cells mediated by VSVg-pseudotyped MmPEG10 VLPs. Data were quantified by flow cytometry 72 hours afterVLP addition, n = 3 replicates. (E) Functional transfer of RNA into loxP-GFP N2a cellsmediated by VSVg-pseudotyped VLPs that were produced with MmPeg10 or

mCherry and Mm.cargo(Cre) constructs that encoded tiles of the MmPeg103′ UTR. Data were quantified by flow cytometry 72 hours after VLP addition,n = 3 replicates. (F) Functional transfer of RNA into loxP-GFP N2a cellsmediated by VSVg-pseudotyped VLPs that were produced with HsPEG1010 ormCherry and Hs.cargo(Cre) constructs that encoded tiles of the HsPeg103′ UTR. Data were quantified by flow cytometry 72 hours after VLP addition,n = 3 replicates. (G) Functional transfer of RNA into loxP-GFP N2a cellsmediated by VSVg-pseudotyped VLPs that were produced with rMmPeg10 andMm.cargo(Cre) or Cre mRNA. Data were quantified by flow cytometry 72hours after VLP addition, n = 3 replicates. (H) Functional transfer of RNA intoloxP-GFP N2a cells mediated by VSVg-pseudotyped VLPs that were producedwith rHsPeg10 and Hs.cargo(Cre) or Cre mRNA. Data were quantified by flowcytometry 72 hours after VLP addition, n = 3 replicates. For all panels, **P < 0.01,***P < 0.001, ****P < 0.0001, one-way analysis of variance.

RESEARCH | RESEARCH ARTICLED

ownloaded from

https://ww

w.science.org at C

hinese University of H

ong Kong on Septem

ber 09, 2021

MmPEG10-mediatedMmPeg10RNAdelivery inneurons. The functional transfer of MmSYNA-pseudotyped VLPs that carry the native PEG10transcript into primary mouse cortical neurons

led to up-regulation of a number of genes in-volved in neurodevelopment (supplementarytext 6). This finding reinforces the notion thatone role of endogenous MmPeg10 delivery is

binding and stabilizing specific mRNA tran-scripts in recipient cells. RNA sequencing ofN2a cells receiving Mm.cargo(Peg10) revealedsubstantial gene expression changes upon

Segel et al., Science 373, 882–889 (2021) 20 August 2021 6 of 7

Fig. 4. SEND is a modular systemcapable of delivering gene edit-ing tools into human and mousecells. (A) Representative imagesdemonstrating functional transfer ofMm.cargo(Cre) or Cre mRNA inrMmPEG10 VLPs pseudotypedwith VSVg (V), MmSYNA (A), orMmSYNB (B) in Ai9 (loxP-tdTomato)tail-tip fibroblasts. Scale bar,200 mm. (B) Percent of tdTomato-positive cells out of the total numberof H2A-stained nuclei from highcontent imaging of n = 3 replicatesof (A). (C) Schematic representingthe retooling of SEND for genomeengineering. (D) Indels at theMmKras locus in MmKras1-sgRNA-N2a cells treated with SEND (VSVg-pseudotyped rMmPEG10 VLPs)containing SpCas9 mRNA, Mm.UTR(SpCas9), or Mm.cargo(SpCas9)and a lentivirus encoding SpCas9.Indels were quantified by NGS72 hours after VLP or lentivirusaddition, n = 3 replicates. (E) Indelsat the mouse MmKras locus in aconstitutively expressing SpCas9N2a cell line either transfected witha plasmid carrying the MmKrassgRNA or treated with SEND(rMmPEG10, VSVg, or MmKrassgRNA). Indels were quantified byNGS after 72 hours, n = 3 repli-cates. (F) Indels at the MmKraslocus in N2a cells treated with SEND(VSVg-pseudotyped rMmPeg10SEND VLPs) containing eitherSpCas9 mRNA or Mm.cargo(SpCas9) and sgRNA. Indels werequantified by NGS 72 hours afterVLP addition, n = 3 replicates.(G) Indels at the HsVEGFA locus inHEK293FT cells treated with SEND(VSVg-pseudotyped rHsPEG10VLPs) containing either SpCas9mRNA or Hs.cargo(SpCas9) and anunmodified sgRNA. Indels weredetermined by NGS 72 hours afterVLP addition, n = 3 replicates.(H) SEND is a modular deliveryplatform combining an endogenousGag homolog, cargo mRNA, andfusogen, which can be tailored forspecific contexts.

A B

D E F

H

********

20

0

40

60

80

100

inde

l at M

mK

ras

locu

s (%

)

SEND(rMm

PEG10,

VSVg, S

pCas

9 m

RNA)

SEND(rMm

PEG10,

VSVg, M

m.U

TR(SpC

as9)

)

SEND(rMm

PEG10,

VSVg, M

m.ca

rgo(

SpCas

9))

SEND(rMm

PEG10, V

SVg,

sgRNA, M

m.ca

rgo(

SpCas

9))

Lenti

viral

SpCas

9

****

fusogencargoRNAcargoRNAA fusogen

CremCherry

SpCas9

healthy gene

VSVg

SYNA

capsid

PEG10

PNMA

VSVg SynA

Mm

.car

go(C

re)

Cre

mR

NA td

Tom

ato+

cel

ls (

%)

15

10

5

0

rMmPEG10 VLPs pseudotyped withdifferent fusogens on mouse tail-tip

fibroblast cells

Fusogen

Cre mRNA

Mm.cargo(Cre)

B

+

B

+

V

+

V

+

A

+

A

+

TdTomH2A-Nuclei

SynB

SEND-mediated sgRNA and cargo(SpCas9) delivery

into N2a cell line

inde

l at M

mK

ras

locu

s (%

)

0

10

20

30

40

50

SEND(rMm

PEG10, V

SVg,

sgRNA, S

pCas

9 m

RNA)moc

kinde

latM

mK

ras

locu

s (%

)

trans

fecte

d sg

RNA

SEND (rM

mPEG10

,

VSVg, sg

RNA)

0

20

40

60

80

100

moc

k

plasmid- or SEND-mediated sgRNA delivery into SpCas9

N2a cell line

SEND-mediated delivery of SpCas9into MmKras sgRNA-expressing

N2A cell line

Fusogen

Mm.cargo(SpCas9)

Cas9cargo produce MmPEG10

VLPs carrying Cas9cargoRNA

apply VLP to MmKras1sgRNA cell line and

quantify indelPEG10

rMmPeg10

VSVg

C

tailored SEND

G

****

10

0

20

30

40

50

inde

l at H

sVE

GFA

locu

s (%

)

mock

SEND-mediated sgRNAand cargo(SpCas9)

delivery into HEK293FT cells

SEND(rHsP

EG10, V

SVg,

sgRNA, H

s.car

go(S

pCas

9))

SEND(rHsP

EG10, V

SVg,

sgRNA, S

pCas

9 m

RNA)

RESEARCH | RESEARCH ARTICLED

ownloaded from

https://ww

w.science.org at C

hinese University of H

ong Kong on Septem

ber 09, 2021

MmPeg10 delivery that were largely abrogatedwith PEG10-mediated Mm.cargo(Cre) delivery(supplementary text 7). This suggests thattransferring a reprogrammed cargo does nothave the same impact on recipient cells astransferring MmPeg10 and indicates thatMmPeg10 transcript delivery rather than thedelivery of MmPEG10 protein is responsiblefor the observed gene expression changes. Itremains unclear whether MmPEG10 VLPsare natively pseudotyped by the endogenousfusogen MmSYNA to enable cellular uptakeof PEG10 VLPs in the central nervous system.To further characterize themodularity of the

components of this system, we tested differentcargoRNAs. Using the same pipeline devel-oped for cargo(Cre), we tested whether SENDcouldmediate the functional transfer of a large~5-kb Mm.cargo(SpCas9) into N2a cell linesthat constitutively express a single guide RNA(sgRNA) againstMmKras (Fig. 4C). SEND wasable to functionally transfer SpCas9, leading to~60% insertions and deletions (indels) in reci-pient cells (Fig. 4D); similar to the results withCre, SEND is specific and only able to effi-ciently functionally transfer SpCas9 flankedby either the full-length or optimized Peg10UTR sequences.To create an all-in-one vector for delivery

of sgRNA and SpCas9, we first tested whetheran sgRNA can be efficiently delivered by SEND.We independently packaged an sgRNA target-ingKras into rMmPeg10 VLPs by coexpressingrMmPeg10 with VSVg and a U6-driven sgRNAand incubated them with Cas9-expressingN2a cells; we detected very little activity eventhough direct transfection of the guide showedrobust indel formation (Fig. 4E). We found,however, that copackaging the guide alongsideMm.cargo(SpCas9) by coexpressing Mm.cargo(SpCas9) with a U6-driven sgRNA on a sep-arate plasmid was sufficient to mediate 30%indels (Fig. 4F). To determine the reprodu-cibility of this genome-editing approach, werepeated this copackaging strategy with thehuman SEND system and were able to gen-erate ~40% indels in HEK293FT cells at theHsVEGFA locus (Fig. 4G).The development of SEND (Fig. 4H) from

an endogenous retroelement complements ex-

isting delivery approaches using lipid nano-particles (26), VLPs derived from bona fideretroviruses (27–29), and active mRNA-loadingapproaches in EVs (30, 31). Moreover, SENDmay have reduced immunogenicity comparedwith currently available viral vectors (32) becauseof its use of endogenous human proteins. Sup-porting this are gene expression data fromthe developing human thymus, which dem-onstrate that HsPEG10 is highly expressedcompared with other CA-containing genes inthe thymic epithelium (fig. S16) (33), which isresponsible for T cell tolerance induction. As amodular, fully endogenous system, SEND hasthe potential to be extended into a minimallyimmunogenic delivery platform that can berepeatedly dosed, which greatly expands theapplications for nucleic acid therapy.

REFERENCES AND NOTES

1. J. L. Goodier, H. H. Kazazian Jr., Cell 135, 23–35 (2008).2. A. F. Smit, Curr. Opin. Genet. Dev. 9, 657–663 (1999).3. M. R. Patel, M. Emerman, H. S. Malik, Curr. Opin. Virol. 1,

304–309 (2011).4. L. Guio, J. González, Methods Mol. Biol. 1910, 505–530

(2019).5. C. Feschotte, C. Gilbert, Nat. Rev. Genet. 13, 283–296 (2012).6. F. J. Kim, J.-L. Battini, N. Manel, M. Sitbon, Virology 318,

183–191 (2004).7. A. Dupressoir et al., Proc. Natl. Acad. Sci. U.S.A. 106,

12127–12132 (2009).8. C. Myrum et al., Biochem. J. 468, 145–158 (2015).9. J. Ashley et al., Cell 172, 262–274.e11 (2018).10. E. D. Pastuzyn et al., Cell 173, 275–288 (2018).11. E. Korb, S. Finkbeiner, Trends Neurosci. 34, 591–598 (2011).12. P. Barragan-Iglesias, J. B. De La Pena, T. F. Lou, S. Loerch,Cell Rep.

10.2139/ssrn.3684856 (2020). doi: 10.2139/ssrn.3684856.13. M. Abed et al., PLOS ONE 14, e0214110 (2019).14. R. Ono et al., Nat. Genet. 38, 101–106 (2006).15. C. Henke et al., Retrovirology 12, 9 (2015).16. M. Krupovic, E. V. Koonin, Proc. Natl. Acad. Sci. U.S.A. 114,

E2401–E2410 (2017).17. S. O. Dodonova, S. Prinz, V. Bilanchone, S. Sandmeyer,

J. A. G. Briggs, Proc. Natl. Acad. Sci. U.S.A. 116, 10048–10057(2019).

18. M. Campillos, T. Doerks, P. K. Shah, P. Bork, Trends Genet. 22,585–589 (2006).

19. S. Konermann et al., Nature 517, 583–588 (2015).20. C. S. Wallace, G. L. Lyford, P. F. Worley, O. Steward, J.

Neurosci. 18, 26–35 (1998).21. M. Golda, J. A. Mótyán, M. Mahdi, J. Tőzsér, Functional Study

of the Retrotransposon-Derived Human PEG10 Protease,Int. J. Mol. Sci. 21, 2424 (2020).

22. A. Saunders et al., Cell 174, 1015–1030.e16 (2018).

23. K. Clemens, V. Bilanchone, N. Beliakova-Bethell, Virus Res. 171,319–331 (2013).

24. K. Ridder et al., OncoImmunology 4, e1008371 (2015).

25. Y. Coquin, M. Ferrand, A. Seye, L. Menu, A. Galy, bioRxiv816223 [Preprint]. 24 October 2019.

26. P. S. Kowalski, A. Rudra, L. Miao, D. G. Anderson, Mol. Ther. 27,710–728 (2019).

27. U. Mock et al., Sci. Rep. 4, 6409 (2014).28. S. J. Kaczmarczyk, K. Sitaraman, H. A. Young, S. H. Hughes,

D. K. Chatterjee, Proc. Natl. Acad. Sci. U.S.A. 108,16998–17003 (2011).

29. P. E. Mangeot et al., Nat. Commun. 10, 45 (2019).30. R. Kojima et al., Nat. Commun. 9, 1305 (2018).31. M. E. Hung, J. N. Leonard, J. Extracell. Vesicles 5, 31027 (2016).32. J. L. Shirley, Y. P. de Jong, C. Terhorst, R. W. Herzog, Mol. Ther.

28, 709–722 (2020).33. J.-E. Park et al., Science 367, eaay3224 (2020).

ACKNOWLEDGMENTS

We thank D. S. Yun for electron microscopy assistance, A. Kollerfor mass spectrometry assistance, L. Wu and the Harvard GMF forthe generation of transgenic animals, A. Tang for illustrationassistance, and the entire Zhang laboratory for support and advice.Funding: This work was supported by a grant from the SimonsFoundation to the Simons Center for the Social Brain at MIT (M.S.);National Institutes of Health Intramural Research Program (E.V.K.);National Institutes of Health grants 1R01-HG009761 and 1DP1-HL141201 (F.Z.); Howard Hughes Medical Institute (F.Z.); OpenPhilanthropy (F.Z.); G. Harold and Leila Y. Mathers CharitableFoundation (F.Z.); Edward Mallinckrodt, Jr. Foundation (F.Z.);Poitras Center for Psychiatric Disorders Research at MIT (F.Z.);Hock E. Tan and K. Lisa Yang Center for Autism Research at MIT(F.Z.); Yang-Tan Center for Molecular Therapeutics at MIT (F.Z.);Lisa Yang (F.Z.); Phillips family (F.Z.); R. Metcalfe (F.Z.); and J. andP. Poitras (F.Z.). Author contributions: M.S. and F.Z. conceivedthe project. M.S., B.L., and F.Z. designed the experiments. M.S.,B.L., J.S., A.L., X.J., and C.C.L. performed the experiments. M.S.,B.L., J.S., and F.Z. analyzed the data. M.S., S.L.M., and E.V.K.performed bioinformatics analysis of Gag protein diversity. F.Z.supervised the research and experimental design with supportfrom R.K.M., and M.S., B.L., R.K.M., and F.Z. wrote the manuscriptwith input from all authors. Competing interests: M.S., B.L., andF.Z. are co-inventors on a US provisional patent application filed bythe Broad Institute related to this work. (U.S. Provisional PatentApplication no. 63/191,067) F.Z. is a cofounder of Editas Medicine,Beam Therapeutics, Pairwise Plants, Arbor Biotechnologies, andSherlock Biosciences. Data and materials availability: Expressionplasmids are available from Addgene under a uniform biologicalmaterial transfer agreement. Additional information is availablethrough the Zhang Lab website (https://zlab.bio). Next-generationsequencing data generated are available from National Centerfor Biotechnology Information Sequence Read Archive withaccession number PRJNA743280. All other data are available in thepaper and supplementary materials.

SUPPLEMENTARY MATERIALS

science.sciencemag.org/content/373/6557/882/suppl/DC1Materials and MethodsSupplementary Text 1 to 7Figs. S1 to S16Tables S1 to S4References (34–56)Data S1

17 January 2021; resubmitted 26 April 2021Accepted 6 July 202110.1126/science.abg6155

Segel et al., Science 373, 882–889 (2021) 20 August 2021 7 of 7

RESEARCH | RESEARCH ARTICLED

ownloaded from

https://ww

w.science.org at C

hinese University of H

ong Kong on Septem

ber 09, 2021

Use of think article is subject to the Terms of service

Science (ISSN ) is published by the American Association for the Advancement of Science. 1200 New York Avenue NW, Washington, DC20005. The title Science is a registered trademark of AAAS.Copyright © 2021 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claimto original U.S. Government Works

Mammalian retrovirus-like protein PEG10 packages its own mRNA and can bepseudotyped for mRNA deliveryMichael SegelBlake LashJingwei SongAlim LadhaCatherine C. LiuXin JinSergei L. MekhedovRhiannon K. MacraeEugeneV. KooninFeng Zhang

Science, 373 (6557),

Hitching a ride with a retroelementRetroviruses and retroelements have inserted their genetic code into mammalian genomes throughout evolution.Although many of these integrated virus-like sequences pose a threat to genomic integrity, some have been retooledby mammalian cells to perform essential roles in development. Segel et al. found that one of these retroviral-likeproteins, PEG10, directly binds to and secretes its own mRNA in extracellular virus–like capsids. These virus-likeparticles were then pseudotyped with fusogens to deliver functional mRNA cargos to mammalian cells. This potentiallyprovides an endogenous vector for RNA-based gene therapy. —DJ

View the article onlinehttps://www.science.org/doi/10.1126/science.abg6155Permissionshttps://www.science.org/help/reprints-and-permissions

Dow

nloaded from https://w

ww

.science.org at Chinese U

niversity of Hong K

ong on September 09, 2021