Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase...

153
UNIVERSITE DE GENEVE Département de biologie cellulaire FACULTE DES SCIENCES Professeur Didier Picard Département de Microbiologie FACULTE DE MEDECINE et Médecine Moléculaire Professeur Daniel Kolakofsky _____________________________________________________________________ SENDAI VIRUS AND THE INNATE ANTIVIRAL DEFENCE THESE présentée à la Faculté des sciences de l’Université de Genève pour obtenir le grade de Docteur ès sciences, mention biologie par Laura STRÄHLE de Versoix (GE) Thèse n° 3900 Genève 2007

Transcript of Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase...

Page 1: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

UNIVERSITE DE GENEVE Département de biologie cellulaire FACULTE DES SCIENCES

Professeur Didier Picard

Département de Microbiologie FACULTE DE MEDECINE et Médecine Moléculaire Professeur Daniel Kolakofsky _____________________________________________________________________

SENDAI VIRUS AND

THE INNATE ANTIVIRAL DEFENCE

THESE

présentée à la Faculté des sciences de l’Université de Genève pour obtenir le grade de Docteur ès sciences, mention biologie

par

Laura STRÄHLE

de Versoix (GE)

Thèse n° 3900

Genève 2007

Page 2: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

Acknowledgements

First of all I would like to thank Professor Daniel Kolakofsky for welcoming me to his laboratory, for sharing his knowledge and for his encouragement which helped during my work.

I also thank all the former and actual members of the lab and of the MMM department, for supporting and encouraging me during the period of my PhD work. A personal and big thank you to Dominique Garcin and Jean-Baptiste Marq, who were always there since the beginning and who helped me become a better scientist (and a better person too...) And thanks to Stéphane Hausmann for his “positive thinking” and support in the final part of my PhD. I will remember the aperitifs that always occurred at the right moment…and will return for many more in the future ;o)

I express my gratitude to Professor Didier Picard and Professor Klaus Conzelmann for accepting to be the examiners of the thesis jury.

Last but not least, I would like to thank my family and friends for their encouragement and support during the making of this thesis.

Page 3: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

TABLE OF CONTENTS

Summary…………………………………………………………...……………….……....….2

Résumé en Français…………………………………………………………........………..…..4

List of abbreviations …………………………………………………………...………….…13

I. GENERAL INTRODUCTION ............................................................................ 14

A. Classification of the Paramyxoviridae Family ............................................................ 15

B. SeV Structure ............................................................................................................... 16

C. SeV Genome & Encoded Proteins ............................................................................... 17

D. Life Cycle and RNA Synthesis .................................................................................... 20

E. Accessory Proteins ....................................................................................................... 23

F. Role of the C and V Viral Proteins .............................................................................. 24

G. Small Viral RNAs : Leader and Trailer ....................................................................... 27

H. Sendai Virus Strains .................................................................................................... 28

I. The Defective Interfering (DI) Genomes ..................................................................... 30

J. Reverse Genetic System ............................................................................................... 34

K. Interferon System ......................................................................................................... 36

L. Sensors of RNA Virus Infection .................................................................................. 40

M. Viral Antagonist of the Interferon System ................................................................... 43 II. PUBLICATION 1.. ............................................................................................... 46

"SENDAI VIRUS TARGETS INFLAMMATORY RESPONSES, AS WELL AS THE INTERFERON-INDUCED ANTIVIRAL STATE, IN A MULTIFACETED MANNER"

III. PUBLICATION 2................................................................................................. 62

"SENDAI VIRUS DEFECTIVE-INTERFERING GENOMES AND THE ACTIVATION OF THE INTERFERON-BETA" IV. PUBLICATION 3 . ............................................................................................... 77

"ACTIVATION OF THE IFNβ PROMOTER BY UNNATURAL SENDAI VIRUS INFECTION REQUIRES RIG-I AND IS INHIBITED BY THE VIRAL C PROTEINS "

V. ADDITIONAL DATA: . ....................................................................................... 92

VI. GENERAL DISCUSSION.................................................................................. 101

VII. REFERENCES .................................................................................................... 106

1

Page 4: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

Summary SeV genome is characterized by a non-segmented negative stranded RNA of negative

polarity, which is tightly associated with the viral nucleoprotein N, forming a very stable

helicoïdal structure called the nucleocapsid (NC). This genome is replicated via an

intermediate RNA of positive polarity, the antigenome, representing the full copy of the

genome. SeV is composed of 6 genes, which are flanked by control regions essential for

transcription and replication, namely the leader and the trailer. The leader and the trailer are

part of the genomic promoter (G/Pr) and the antigenomic promoter (AG/Pr), respectively. The

G/Pr (located at the 3’extremity of the genome) is involved in the control of replication and

transcription initiation, whereas the AG/Pr (located at the 3’end of the antigenome) is only

implicated in the replication.

An important component of the host’s innate immune response in viral infection is the

production of type I interferons (IFNs). Efforts to understand the molecular mechanisms by

which viruses and also double-stranded RNA (dsRNA) trigger the induction of IFN have led

to the identification of cellular sensors of viral infection. One type of sensor is the family of

cytosolic receptor proteins known as the retinoic acid inducible gene I (RIG-I)-like receptors.

This family includes RIG-I and Mda5, two DexD/H box helicases with CARD domains that

were found to participate in the detection of cytoplasmic RNA. dsRNA as well as single-

stranded viral RNA bearing 5’triphosphates (5’pppRNA) are thought to be products of RNA

virus infections that acts as pathogen-associated molecular patterns (PAMPs) responsible for

initiating the innate antiviral defence. RIG-I and Mda5 initiate antiviral responses by

coordinately activating several transcription factors, including NF-κB and IRF-3, that bind to

the IFNβ promoter forming an enhanceosome, which in turn activates the IFNβ gene. IFNβ is

secreted and feeds back onto cells in a paracrine manner to prime neighbouring cells for

possible infection and in a autocrine manner to induce multiple IFN stimulated genes (ISGs)

leading to the antiviral state of the cell. Many viruses including SeV have developed strategies

for counteracting the host type interferon I response and this at different levels. For that

purpose SeV mainly uses the non-structural C and V proteins to act at the level of the IFN

induction pathway by blocking RIG-I activation and also at the level of the IFN feedback loop

where C blocks Stat-1 signaling pathway.

In the first study, we have used cDNA arrays to compare the activation of various

cellular genes in response to infection with SeV that contain specific mutations. Mutations

that disrupt four distinct elements in the SeV genome (the leader RNA, two regions of the C

2

Page 5: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

protein, and the V protein) all lead to enhanced levels of IFN-β mRNA, and at least three of

these viral genes also appear to be involved in preventing activation of IL-8. Our results

suggest that SeV targets the inflammatory and adaptive immune responses as well as the IFN-

induced intracellular antiviral state by using a multifaceted approach.

SeV stocks available commercially are known to strongly induce IFNβ and are

commonly used by many laboratories. Plus, these stocks are known for a long time to contain

DI genomes. The paramyxoviruses DI genomes can be of two types: internal deletion or

copyback DI genomes. Copyback DIs have the capacity to form dsRNA by at least two ways:

1) When the level of the N protein is not sufficient, DI genomes and antigenomes can self-

anneal, and 2) Because some DI genomes contain termini that are perfectly complementary,

they are free to form dsRNA. Moreover, DIs (especially those from the copyback variety)

interfere robustly with the ND helper genome by competing for replication and consequently

reducing the production of viral proteins involved in the antiviral state. The second paper

shows evidence that the strong induction of IFNβ activation upon SeV infection (SeV stock

containing DI genomes) is mainly due to the presence of copyback DI genomes. The level of

IFNβ activation was found to be proportional to that of DI genome replication. Moreover, this

activation can be inhibited by the overexpression of the C and V proteins, whose

concentrations are reduced in DI infected cells, because of the strong interference of the DI

versus the ND genome.

In the last paper the contribution of RIG-I (and Mda-5) in the detection of SeV

infection is examined. Because 5’triphophorylated products have become new potential

targets of RIG-I, we decided to test whether SeV infections induced IFNβ activation by

producing pppRNAs as well as dsRNA. The involvement of both helicases was also analysed.

We used two different Sendai virus infections to study virus-induced IFNβ activation; 1) SeV-

DI-H4, which is composed mostly of small, copyback DI genomes, and whose infection is

likely to over-produce short 5’ tri-phosphorylated (ppp) trailer RNAs and under-produces the

viral V and C proteins, and 2) SeV-GFP(+/-), a co-infection that produces WT amounts of

viral gene products but also produces both GFP mRNA and its complement, which can form

dsRNA with capped 5’ ends. We found that 1) virus-induced signaling to IFNβ depended

predominantly on RIG-I (as opposed to mda-5) for both SeV infections, i.e., that RIG-I senses

both pppRNAs and dsRNA without 5’ tri-phosphorylated ends, and 2) that it is the viral C

protein (and not V) that is primarily responsible for countering RIG-I dependent signaling to

IFNβ.

3

Page 6: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

Résumé en Français Introduction générale

Le virus de Sendaï (SeV) a été découvert en 1953 au Japon et isolé à partir d’un nouveau né

présentant des troubles respiratoires. Ce virus n’est pas pathogène pour l’homme mais il est

extrêmement contagieux et virulent chez le rat et la souris de laboratoire. La transmission se

fait par contact direct et est suivit par une infection des voies respiratoires. SeV appartient au

genre Respirovirus de la famille des Paramyxovirus. Cette famille avec celle des

Rhabdovirus, des Filovirus et des Bornavirus constituent l’ensemble de l’ordre des

Mononegavirus.

Il existe plusieurs pathogènes humains appartenant à cette famille, comme par exemple le

virus de la rougeole, le virus des oreillons ou encore le virus Nipah, un virus apparu

récemment. Ces virus sont encore très présents dans les pays en voie de développement.

Effectivement le virus de la rougeole à lui seul tue encore plus d’un million de personnes par

année. Ainsi, SeV est considéré comme un bon modèle d’étude permettant de mieux

comprendre les mécanismes moléculaires de cette famille et de trouver des thérapies.

SeV contient une enveloppe sphérique provenant de la membrane plasmique de l’hôte dans

laquelle sont ancrées les glycoprotéines virales de fusion (F) et l’hémagglutinine-

neuraminidase (HN), toutes deux impliquées dans l’attachement, la fusion et le relâchement

des particules virales. Les protéines de la matrice (M) se trouvent contre la surface interne de

la membrane et jouent un rôle important dans la structure et l’assemblage de la particule

virale. A l’intérieur de cette particule se trouve le génome à ARN(-) de SeV étroitement

associé aux protéines de la capside (N), formant une structure hélicoïdal très stable appelée la

nucléocapside (NC) ou le complexe ribonucléoprotéique. Attachées à ce complexe, les

protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription

et de la réplication du virus.

Le génome de SeV est long de 15'384 nucléotides et se caractérise par un simple brin d’ARN,

non-segmenté et de polarité négative. SeV est composé de six gènes : N, P, M, F, HN et L.

L’ordre de ces gènes est extrêmement bien conservé parmi les paramyxovirus. Chaque gène

commence avec une courte séquence régulatrice de transcription de dix nucléotides appelée

“gene start” et se termine par une séquence de terminaison nommée « gene end ». Entre ces

deux séquences se trouve une région intergénique (IG) non transcrite de trois nucléotides. Les

extrémités flanquant les 6 gènes comportent des régions extracistoniques essentielles pour la

transcription et la réplication, nommées le leader à l’extrémité 3’ et le trailer à l’extrémité 5’.

4

Page 7: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

Ces séquences font partie des promoteurs génomiques et antigénomiques, respectivement. Les

gènes de SeV sont monocistroniques à l’exception du gène P, qui contient différents cadres de

lecture ouverts ainsi qu’un site d’editing en son milieu. De ce fait, différentes espèces de

protéines (P, V, W, C’, C, Y1, Y2 and X) peuvent être produites.

Le cycle viral et la synthèse d’ARN viral

Lors d’une infection, le virus est adsorbé par les récepteurs cellulaires situés à la surface de la

cellule, ce qui conduit à la fusion entre l’enveloppe du virus et la membrane plasmique. Suite

à la fusion, la nucléocapside hélicoïdale se trouve libérée dans le cytoplasme de la cellule

hôte, lieu de toutes les étapes du cycle de multiplication virale. La nucléocapside contenant

l’ARN génomique est la matrice pour toutes les synthèses d’ARN. Deux fonctions sont

assurées par le génome viral : la transcription des ARN messagers et la réplication de l’ARN

viral. Les protéines N, P/C/V, M, HN et L protéines sont synthétisées par le système de

traduction de la cellule, et finalement suit l’assemblage du génome viral avec les protéines de

la capside. La protéine M se place à la surface interne de la membrane plasmique alors que les

protéines de surface F et HN se trouvent au niveau des patchs créés par la protéine M,

excluant les protéines cellulaire. Une fois que les nucléocapsides sont associées à la protéine

M, les nouvelles particules se forment et sortent de la cellule en emportant avec elles une

partie de la membrane plasmique cellulaire.

Le génome viral encapsidé sert de matrice à la polymérase virale pour synthétiser dans un

premier temps les ARN messagers nécessaires pour produire les protéines virales qui sont

impliquées dans la réplication même du virus. La polymérase virale entre sur le génome à

l’extrémité 3’ et transcrit en premier le leader qui est un ARN non codant, puis commence la

transcription des six gènes en six messagers à ARN et ce d’une manière séquentielle et

polaire. Occasionnellement la polymérase oublie de réinitier le messager suivant la jonction,

en atténuant par conséquent la transcription des gènes en aval. Ainsi un gradient de messagers

synthétisés peut être observé, qui est inversement proportionnel à la distance du gène par

rapport à l’extrémité 3’ du génome. La protéine N est la protéine la plus synthétisée et la

concentration intracellulaire de sa forme « non-assemblée » est un moyen de contrôler le taux

de transcription et de réplication à partir de la matrice génomique. Lorsque la quantité de

protéinés N « non assemblées » est suffisante, la synthèse d’ARN viral est couplée avec

l’encapsidation concomitante de la chaine naissante. Dans ces conditions la polymérase

ignore toutes les jonctions, formant ainsi une copie complète du génome de polarité positive

entièrement encapsidée. Ce dernière est appelé antigénome et servira à son tour à la synthèse

5

Page 8: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

d’une nouvelle copie d’ARN génomique, qui à nouveau sera utilisée comme matrice ou alors

qui sera envoyée et assemblée dans une particule virale naissante.

Les protéines accessoires

Les protéines C et V de SeV sont désignées comme des protéines « accessoires », car elles ne

sont pas présentes dans tous les virus de la famille des Paramyxoviridae. Effectivement

certains virus possèdent les deux protéines alors que d’autres ne possèdent que l’une ou

l’autre. Les protéines C et V sont exprimées à partir du gène P qui contient 5 codons

d’initiation ainsi qu’un site d’editing en son milieu, ce qui lui permet de coder pour 8

protéines : P, V, W, C’, C, Y1, Y2 et X.

Les protéines C’, C, Y1 et Y2 sont collectivement nommées les « protéines C ». La

participation des protéines virales C et V dans la contremesure des réponses innées de la

cellule, a été intensément étudiée ces 10 dernières années. Apparemment, les Paramyxovirus

(incluant SeV) utilisent la protéine C pour cette fonction, alors que les autres membres de la

famille (Rubula-, Morbili- et Henipa-virus) utilisent la protéine V. Les protéines C de SeV

sont des protéines non-essentielles à la multiplication du virus in vitro, mais sont nécessaires à

la réplication du virus dans les souris. Cette particularité reste cependant dépendante du type

cellulaire étudié. Les protéines C contiennent une séquence qui les localise à la membrane

plasmique de la cellule mais elles sont également retrouvées dans le cytoplasme. Toutes les

protéines C interagissent physiquement avec Stat1 (Signal transducer and activator of

transcription 1), dont le rôle est de transduire le signal de l’IFN pour permettre l’expression de

protéines antivirales. Cette interactions (C/Stat1) empêche la signalisation de l’IFN par la

voie de JAK/Stat et en même temps bloque l’établissement d’un état antiviral de la cellule.

Les quatre protéines C (C’, C, Y1, Y2) partagent la même région C-terminal alors que seul les

longues protéines C (C’ et C) partagent la même région N-terminale. La partie C-terminal de

la protéine C est nécessaire et suffisante pour bloquer le signal de l’IFN. Par conséquent les

protéines Y1 et Y2 peuvent à elle seules garantir cette fonction. Par contre les longues

protéines de C (C’,C) provoquent l’instabilité de Stat1, réduisant le niveau de Stat1 tout en

augmentant celui de Stat1 sous sa forme phosphorylé. Ainsi la partie N-terminal de la protéine

C est associée avec la dégradation de Stat1 qui permet de renverser l’état antiviral induit par

l’IFN. Il a aussi été démontré que la partie C1-23 (nucléotides 1-23 de la partie N-terminal)

était suffisante pour réduire la quantité de Stat1 et agissait comme un signal ciblant la

membrane plasmique. De plus la localisation des longues protéines C à la membrane

plasmique est apparemment nécessaire à leur activité. Finalement, il a été montré également

6

Page 9: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

que la protéine C pouvait inhiber la signalisation de RIG-I, un détecteur cytoplasmique

d’ARN viral. Les protéines Y1 et Y2 (partie C terminale) seraient responsables de cette

inhibition, mais aucun résultat n’a encore été clairement montré.

Leader et Trailer

Chez les paramyxovirus le promoteur génomique et le promoteur antigénomique (G/Pr et

AG/Pr) se trouvent à l’extrémité 3’ du génome et à l’extrémité 5’ de l’antigénome,

respectivement. Chez SeV ces promoteurs ont tous deux une longueur de 96 nucléotides. Le

leader fait partie du G/Pr, incluant les 55 premiers nucléotides à l’extrémité 3’ du génome (-).

Cette séquence contient les signaux d’initiation pour la synthèse d’ARN (transcription et

réplication) par la polymérase virale. Le trailer comporte les 57 premiers nucléotides à

l’extrémité 3’ de l’antigénome. Cette séquence est essentielle pour la réplication virale. Les

transcrits du leader et du trailer sont tous deux exempt de régions codantes et ne sont ni

coiffés, ni polyadenylés. Par contre ils portent chacun à leur extrémité 5’ un triphosphate, qui

a été récemment déterminé comme une nouvelle signature virale détectée spécifiquement par

des sentinelles cytoplasmiques de la cellule. L’AG/Pr, responsable de la synthèse du génome

est plus fort que le G/Pr. Effectivement, l’AG/Pr a une plus grande affinité pour la polymérase

et la production de génome est dix fois supérieure à celle de l’antigénome. De plus, il a été

montré que le « gène start » réduisait la force du promoter de réplication du G/Pr.

Les génomes défectifs et interférants: les DIs

Des formes incomplètes de génome viral générées lors du processus de réplication du virus

standard ont été observées dans quasiment tous les virus à ARN et ADN et ont été nommées

les “génomes défectifs et interférants”. Ce phénomène apparait lorsque le virus est passé à

haute multiplicité d’infection et dépend de l’ARN polymérase virale. La fréquence des ces

événements est faible et se manifeste pendant la transcription ou la réplication du génome

non-défectif (ND). Plusieurs DIs peuvent être générés, mais seul quelques uns sont

sélectionnés selon leur capacité à interférer avec le génome ND. La notion d’interférence est

importante car elle permet aux génomes DI de se répliquer et de s’amplifier au dépend du

génome ND, qui doit rentrer en compétition avec ces derniers afin de générer les protéines

virales, elles-mêmes impliquées dans la réplication et la maturation du virus. En général, la

plupart des DIs ne sont pas capables de transcrire, ni de traduire. Ainsi, ils sont entièrement

dépendants du ND pour leur réplication. Une autre particularité de ces génomes DI est qu’ils

sont requis dans l’établissement et la maintenance d’infections persistantes.

7

Page 10: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

On peut observer principalement deux types de DI: le DI de « délétion interne » et le DI

d’extrémités symétriques ou « Copyback ». Dans le premier cas, la polymérase à ARN

commence à synthétiser le génome et à un certain moment saute en avant sur sa matrice pour

continuer la synthèse. Le DI à « délétion interne » conserve les extrémités 3’ et 5’ mais une

partie plus ou moins grande du génome manque ou est absente. Dans le cas du DI

« copyback », la polymérase commence la synthèse normalement, mais à un certain moment

se détache de sa matrice et au lieu de continuer en avant, elle commence à copier dans le sens

inverse, utilisant la chaine naissante comme matrice. Par conséquent, dans le DI

« copyback », il manque une grande partie 3’ du génome ND, mais on trouve le même AG/Pr

à chaque extrémité. Cette particularité du DI « copyback » lui permet d’avoir une meilleure

réplication et d’interférer ainsi avec le virus ND. Le fait que les DIs « copyback » n’expriment

pas de protéines ne veut pas dire qu’ils n’ont aucun rôle. Effectivement, le fait qu’ils

possèdent deux AG/Pr implique aussi la présence de deux trailers, qui ont un rôle

antiapoptotique. Ceci expliquerait en partie la mise en place d’une infection persistante, lors

d’une infection avec un stock de SeV contenant des DIs de type « copyback ».

Le système de l’Interféron

Lors de leur évolution, les cellules ont développé des défenses efficaces afin de contrecarrer

les infections virales. La première ligne de défense d’un organisme contre l’invasion d’un

pathogène se traduit principalement par la sécrétion d’interférons (IFNs). Les IFNs font partie

de la famille des cytokines identifiées par leur capacité à induire une forte résistance cellulaire

suite à une infection virale. Leur action a comme effets d’induire un état antiviral aux cellules

et tissus alentours à l’infection et d’utiliser différents moyens biologiques pour interférer avec

la réplication virale, moduler la réponse immune et réguler l’apoptose.

L’ARN double brin est connu pour être un fort inducteur de l’IFN et est potentiellement

généré lors du processus de la transcription et de la réplication du génome viral. Il existe trois

types d’IFNs : 1) L’IFN de type I, incluant l’IFNα et IFNβ. 2) L’IFN de type II (également

appelé IFNγ) comprend les IFNε,κ and ω qui ont été récemment définis. 3) L’IFN de type III

ou IFNλ est une nouvelle cytokine similaire à IL10. Les IFNs de types I et II sont tous deux

impliqués dans l’activité antivirale, mais l’IFN de type I joue un rôle plus important dans

l’immunité innée alors que l’IFNγ est plus impliqué dans l’immunité adaptative. L’IFN de

type III possède des activités et des fonctions similaires aux IFNs de type I, bien qu’il utilise

un autre complexe de récepteurs de surface. Les trois types d’IFNs induisent des réponses

8

Page 11: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

transcriptionnelles à travers la voie de signalisation de JAK-Stat qui résultent en l’activation

de multiples gènes. La régulation de l’expression des IFNs est bien caractérisée et demande

la participation de différents complexes de facteurs de transcription qui sont déjà présents

dans la cellule et qui sont activés lors d’une infection virale. Il existe quatre facteurs de

transcription connus pour lier l’enhancer activatrice de l’IFNβ: ATF-2/ C-Jun, NF-κB et deux

facteurs de transcriptions (IRFs), IRF-3 and IRF-7. La famille des protéines IRF contient 9

membres, qui sont extrêmement importants pour un grand nombre de processus incluant la

réponse immune, la signalisation de cytokines et la croissance cellulaire ainsi que

l’hématopoïèse. IRF-3 et IRF-7 résident dans le cytoplasme des cellules non infectées et qui, à

la suite d’une infection virale, sont transloquées dans le noyau. IRF-3 and IRF-7 sont

essentiels pour l’induction maximale de l’expression d’IFNα/β. Alors qu’IRF-3 est

constitutivement exprimé, IRF-7 est principalement dépendant de son induction par l’IFN. A

la suite d’une infection, IRF3 est activé et s’installe dans le noyau où il initie la synthèse

d’IFNα/β en se liant aux régions enhancer activatrice. Les IFNα/β sont alors sécrétés et se

lient à nouveau de façon paracrine sur les cellules voisines afin de les alerter d’une possible

infection ou alors de façon autocrine pour induire plusieurs gènes ISGs (IFN stimulated

genes) dans la cellule même. De cette façon la première vague de défense est mise en place

résultant en un état antiviral des cellules.

Les détecteurs d’infections de virus à ARN

Plusieurs voies de signalisation conduisant à l’induction des IFNα/β ont été découvertes

récemment. Elles incluent différents récepteurs cellulaires qui ont la particularité de détecter

la présence du virus en reconnaissant les signatures moléculaires virales. Ces signatures

virales font partie des PAMPs (pathogen associated molecular pattern) qui contiennent

beaucoup de signatures potentielles provenant de différents pathogènes, incluant les virus, les

bactéries et les champignons. Ces PAMPs sont reconnus par un large spectre de récepteurs

nommée PRRs (pattern recognition receptors) qui comprennent les « R-proteins » (pathogen-

resistance protein) chez les plantes, les récepteurs Toll-like (TLRs), les récepteurs NOD-like

(NLRs) ainsi que les récepteurs Rig-like (RLRs) chez les animaux. Les TLRs se trouvent

essentiellement associées à la membrane plasmique, soit à la surface cellulaire soit dans la

membrane des endosomes. Les NLRs et les RLRs quant à eux, sont des protéines solubles qui

surveillent le cytoplasme et qui sont prêts à détecter la présence de pathogènes à l’intérieure

de la cellule. Jusqu’à maintenant, la reconnaissance des bactéries dans la cellule reste

9

Page 12: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

spécifique aux NLRs, alors que celle des virus reste restreinte aux RLRs. Finalement il

semblerait que la coopération entre ces PRRs constitue un véritable bouclier contre les

pathogènes envahissants.

Il existe trois membres de la famille des RLRs : retinoic-acid-inducible gene 1 (RIG-1),

melanoma differentiation-associated gene 5 (MDA-5) et laboratory of genetics and

physiology 2 (LPG2). RIG-I et Mda-5 sont des hélicases, dont le rôle est de distinguer les

ARNs du soi de la cellule et les ARNs du non soi provenant des virus, et de réguler ainsi le

signal de transduction en aval. Par contre, LPG2 joue un rôle de régulateur-inhibiteur de RIG-

I et de Mda-5. Des études ont révélé que RIG-I était essentiel à la reconnaissance d’un set

spécifique de virus incluant les Paramyxoviruses, Flaviviruses, Orthomyxoviruses et

Rhabdoviruses, alors que Mda-5 était essentiel à la reconnaissance d’un différent set de virus,

incluant les Picornaviruses et les Alphaviruses. Il a été également démontré que RIG-I était

activé par de l’ARN double brin ainsi que par des ARN non-cappés, portant un triphosphate à

leur extrémité 5’. De ce fait, il semblerait que différentes voies d’activation peuvent être

activées selon le PRRs impliqué dans la reconnaissance de la signature virale et selon le type

de virus. Malgré ces différences, RIG-I et MDA-5 gardent la même voie de signalisation en

aval. Ils sont tous deux exprimés ubiquitairement dans la plupart des tissues et font partie des

ISGs. Il a été montré que la reconnaissance des PAMPs induisait un changement de

conformation de RIG-I et Mda-5, les rendant capables de se lier à une protéine liée à la

surface externe de la membrane mitochondriale, nommée Cardif. Cette interaction conduit

indirectement à l’activation de différentes kinases qui induisent l’activation de IRF-3 et NF-

κB, ce qui finalement résulte dans l’induction de l’IFN de type I et à la mise en place de l’état

antiviral de la cellule.

Papier 1 :

Le virus de Sendai cible de multiples façons les réponses inflammatoires ainsi que l’état

antiviral induit par l’IFN

Pendant leur évolution, les virus ont développé plusieurs stratégies afin de réguler et

contrecarrer les réponses innées des cellules hôtes, en particulier la production des IFNs. Pour

cela, le virus de SeV utilise ses protéines C pour combattre les réponses à l’IFN. Le premier

papier se base sur des microarrays d’ADN complémentaires afin de comparer l’activation de

différents gènes cellulaires en réponse aux infections de SeV contenant des mutations

spécifiques dans le gène C ou dans les régions promotrices du génome (qui ont apparemment

un rôle dans la prévention de l’apoptose et dans l’infection persistante). Ces analyses ont

10

Page 13: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

permis d’observer que le niveau d’activation d’environ 20 ARNm augmentait

significativement lors d’infection avec les virus SeV mutants, comparé aux infections de SeV

WT. Trois différents groupes de gènes cellulaires ont pu être mis en évidence selon les

mutations que porte SeV. Certains des gènes sont connus comme des gènes stimulés par l’IFN

(ISGs) d’autres, comme IL-6 ou IL-8 ne sont pas directement induits par l’IFN. Le gène de

l’IFNβ, qui est essentiel pour initier l’état antiviral fut également activé lors d’infection de

SeV portant des mutations. Ce travail met en avant le fait que SeV portant des mutations

spécifiques dans le gène de C, par opposition au SeV WT, active l’expression d’IL-6 et IL-8

ainsi que quelques autres ISGs. Ainsi les protéines accessoires C et V et la présence du leader

ont un rôle dans la prévention de l’expression de ces gènes cellulaires qui sont essentiels

autant pour les réponses inflammatoires, que pour les réponses adaptatives et innées de la

cellule hôte.

Papier 2:

Les génomes défectifs de SeV (DIs) et l’activation de l’Interféron-beta

Le promoteur de l’IFNβ est normalement activé lorsque les cellules sont traitées avec de

l’ARN double brin synthétique (polyI/C) ou lorsqu’elles sont infectées par des virus. SeV est

généralement utilisé à cet effet, et des stocks de SeV peuvent être facilement obtenus dans le

commerce. Cette remarquable capacité à induire l’IFN est cependant reliée au fait que ces

stocks contiennent des génomes défectifs (DIs). Dans ce travail nous avons tout d’abord

comparé la capacité d’induire l’IFN de plusieurs stocks de SeV, contenant ou non différents

génomes défectifs. Plusieurs niveaux d’induction de l’IFN ont pu être observés selon le type

de stock utilisé. Apparemment, cette propriété est particulièrement due à la présence de DI

copyback et corrèle a priori avec leur capacité à s’hybrider et à former de l’ARN double brin.

Le niveau d’activation de l’IFN semble également être proportionnel à la réplication même du

génome défectif et au ratio génomes défectifs/génomes ND lors de l’infection. Dans cette

étude, on démontre d’autre part que les protéines C et V de SeV sont aptes à bloquer

l’induction à l’IFN lors d’infection avec les DIs ou lors de traitement avec le polyI/C. Ainsi

on conclut que les infections de SeV contenant des DIs sont particulièrement de puissants

inducteurs de l’IFNβ. Effectivement il est probable qu’ils fournissent une grande quantité de

double brin et qu’ils réduisent en même temps l’expression virale des protéines C et V

(normalement responsables de contrecarrer la réponse antivirale de l’hôte) favorisant de ce

fait l’induction à l’IFN.

11

Page 14: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

Papier 3:

Lors d’infections non-naturelles de SeV la voie d’activation de l’IFNβ requiert RIG-I et est

inhibée par les protéines virales C

Sachant que SeV-WT n’active que très faiblement l’IFNβ et afin d’étudier l’activation de

l’IFNβ dans les cellules MEFs, nous avons utilisé deux différentes sorte d’infection de SeV:

1) Le stock de SeV-DI-H4 est composée en majorité de génomes non-défectifs de type

“copyback” et lors d’un infection, il produit en grande quantité les ARNs trailer contenant

trois phosphates en extrémité 5’ et, en quantité moindre, les protéines virales C et V. 2)

L’infection SeV-GFP(+/-) est une coinfection, qui génère (en plus des produits de gènes

viraux en quantité équivalente à une infection SeV-WT) des ARN messagers GFP ainsi que

leurs compléments pouvant former des ARNs double brin synthétiques (polyI/C) comportant

des extrémités 5’ cappées. Ce travail nous a permis de découvrir 1) que l’activation de l’IFN,

induit par les deux types de virus, dépendait principalement de RIG-I (et non de Mda-5). Plus

précisément, que RIG-I détectait les pppARN simple brin ainsi que les ARN double brins ne

comportant pas les trois phosphates en extrémité 5’ ; 2) que c’est la protéine C (et non la V)

qui est responsable en premier lieu de contrecarrer l’induction de l’IFN dépendante de RIG-I.

SeV n’exprimant pas spécifiquement les protéines C, ne peut pas prévenir l’activation de

l’IFN induite par du polyI/C ou par les pppARN simple brin. Cette activation se trouve même

amplifiée. D’autre part, de SeV n’exprimant pas de protéine V se comporte comme un SeV-

WT et contrecarre les effets du polyI/C ou des pppARN simple brin.

12

Page 15: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

List of abbreviations AG/Pr Antigenomic promoter

CAT Chloramphenicol acetyl transferase

DI Defective interfering

G/Pr Genomic promoter

HPIV 1,2 and 3 Human parainfluenza virus type 1,2 and 3

ISG Interferon stimulated gene

IRF Interferon regulatory factor

le Leader

Mda-5 Melanoma differentiation-associated gene 5

MeV Measles virus

MV Mumps virus

NC Nucleocapsid

ND Non defective

NDV Newcastle disease virus

NNV Nonsegmented negative stranded RNA virus

RSV Respiratory syncytial virus

RV Rabies virus

SeV Sendai virus

STAT Signal transducer and activator of transcription

SV5 Simian virus 5

RIG-1 Retinoic-acid-inducible gene 1

tr Trailer

vRNAP Viral RNA polymerase

VSV Vesicular stomatitis virus

13

Page 16: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

I. General Introduction Sendai virus (SeV) was discovered in Japan in 1953. It was isolated in the Tohuku University

Hospital from a newborn patient presenting pneumonia syndromes. SeV is also referred to as

murine parainfluenza type I virus as it was found to infect respiratory tract of mice, to cause

pneumonia and to spread to uninfected animals. SeV is currently an important respiratory

pathogen of laboratory rodents, causing terrible epidemics with high mortality during the

acute phase. It is extremely contagious and transmission occurs via contact and aerosol

infection of the respiratory tract.

SeV is an enveloped nonsegmented negative stranded RNA virus (NNV) of the

Paramyxoviridae family, subfamily Paramyxovirinae and genus Paramyxovirus (or

Respirovirus). It is considered to be a good model to study the Paramyxoviridae family

because it includes significant human pathogens of infants and children, such as Mumps virus

(MV), Measles virus (MeV), respiratory syncytial virus (RSV) and Nipah virus. Some of

these viruses are still importantly present in undeveloped countries (e.g. MeV still causes a

million deaths/year) and some others have only recently emerged. Thus, studies on SeV can

offer important information for understanding the molecular mechanisms of this virus family

and consequently offer medical treatment and therapy.

The Paramyxoviridae family along with the Rhabdoviridae, the Filoviridae and the

Bornaviridae families, are all part of the Mononegalvirales order. It is subdivided into two

subfamilies: the Paramyxovirinae containing the Respiro-, the Rubula-, the Morbilli-, the

Avula- and the Henipa- viruses; and the Pneumovirinae, containing the Pneumo- and the

Metapneumo-viruses. Emergence of new paramyxoviruses, such as Hendra and Nipah,

causing respiratory and neurological disease in cattle and human, has been observed recently

(Table 1).

The classification of these different viruses is based on morphologic criteria, the organisation

of the genome, the biological activities of the proteins, and the sequence relationship of the

encoded proteins. The Mononegalviruses share a number of fundamental characteristcs: (1)

Their genome is a single negative stranded RNA, packaged in a helical nucleocapsid (NC);

(2) nucleocapsids are enclosed within an envelope derived from the plasma membrane of the

cell; (3) a virus-coded RNA polymerase packaged in the virion synthesizes the viral mRNAs

by transcribing the RNA as part of the intact NC after it enters the cell; (4) the RNA

polymerase begins transcribing at the 3’end of the genome RNA and sequentially transcribes

5-10 genes, terminating and releasing each mRNA before starting the next one.

14

Page 17: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

Family Paramyxoviridae

Subfamily Paramyxovirinae Genus Respirovirus (Paramyxovirus)

Sendai virus (mouse parainfluenza virus type 1) Human parainfluenza virus type 1 and 3 Bovine parainfluenza virus type 3

Simian virus 10

Genus Rubulavirus

Simian virus 5 (Canine parainfluenza virus type 5) Mumps virus Human parainfluenza virus type 2, type 4a and 4b (hPIV2/4a/4b) Porcine rubulavirus

Genus Morbillivirus Measles virus Dolphin morbillivirus Canine distemper virus Peste-des-petits-ruminants virus Phocine distemper virus Rinderpest virus

Cetacean morbillivirus Genus Avulavirus

Newcastle disease virus (avian paramyxovirus 1) Genus Henipavirus Hendra virus Nipah virus

Subfamily Pneumovirinae Genus Pneumovirus

Human respiratory syncytial virus Bovin respiratory syncytial virus

Murine pneumonia virus (Pneumonia virus of mice)

Genus Metapneumovirus Avian pneumovirus

Unclassified paramyxoviruses Tupaia Paramyxovirus

Table 1: Classification of the Paramyxoviridae family (www.ncbi.nlm.nih.gov/ICTVdb/Ictv/index.htm)

15

Page 18: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

B. Virion structure of Sendai Virus

SeV virions are surrounded by a lipid bilayer envelope that is derived from the plasma

membrane of the host cell in which the virus has grown (Choppin et al., 1975). The envelope

contains two surface glycoproteins, F and HN, which mediate the entry and exit of the virus

from its host cell. The nucleocapsid (NC), which is composed of the viral RNA genome

tightly surrounded by hundreds of copies of the viral nucleoprotein (N), forms the active viral

genome inside the envelope. The phosphoprotein (P) and the large (L) protein form the viral

polymerase complex, which initiate intracellular virus replication. Between the envelope and

the core lies the viral matrix (M) protein, which interacts with itself, with the lipid bilayer,

with the NC, and with the cytoplasmic tails of the HN and F proteins (Fig.1).

Figure 1: Schematic representation of SeV particle (From P. Le Mercier)

16

Page 19: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

C. Genome & encoded proteins The genome of SeV is a nonsegmented, single stranded RNA genome of negative polarity,

which replicates entirely in the cytoplasm, and contains 15’384 nucleotides. SeV is composed

of 6 genes, the N, P/C/V, M, F, HN and the L, which are flanked by control regions essential

for transcription and replication. The 3’ extracistronic region is known as the leader (le) and

the 5’ extracistronic region is known as the trailer (tr). SeV genes are monocistronic except

for the P gene, which can produce multiple protein species (P,V,W,C’,C,Y1,Y2 and X) via

overlapping reading frames and mRNA editing. Each mRNA begins with a short transcription

regulatory sequence of ten nucleotides named the gene start (UCCCANUUNC) and

terminates with a gene end sequence (UNAUUCU5). There are also intergenic regions located

between these sequences gene boundaries that are precisely three nucleotides long containing

GAA, except for the junction between HN and L containing GGG (Fig. 2).

Figure 2: Genomic organisation of SeV (From (Lamb and Kolakofsky, 2001).

The N protein SeV N protein, also named the nucleoprotein, and genome RNA are assembled in a helicoidal

helix forming the nucleocapsid (NC), with a stoichiometry of one N protomer for six

nucleotides. This precise hexamer arrangement is required for an efficient replication and is

called “the rule of six”, that is also applied to the other Paramyxoviruses of the

Paramyxovirinae subfamily. In this rule, nucleocapsid assembly presumably begins with the

17

Page 20: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

first nucleotide at the 5’ end of the nascent chain, and continues by assembling six nucleotides

at a time until the 3’ end is reached (Calain and Roux, 1993; Kolakofsky et al., 1998). The NC

core is composed of 2564 N proteins and approximately 300 P proteins and 50 L proteins

(Lamb et al., 1976). It is remarkably stable, as it withstands the high salt and gravity forces of

cesium chloride density gradient centrifugation. Within the NC, the RNA is also resistant to

nuclease attack at any salt concentration (Lamb and Kolakofsky, 1996). For paramyxoviruses,

SeV N protein is divided into two regions; the well conserved NCORE (1-400aa) and the

hypervariable NTAIL (401-524aa) (Houben et al., 2007). The N protein associates with the P-L

polymerase during replication. In the current model for encapsidation, there is an initial

sequence specific binding of N to viral leader RNA followed by the cooperative assembly of

N on the growing chain, presumably through N-N and non-specific N-RNA interactions.

Several paramyxoviruses N proteins, including those of SeV, MeV, Newcastle disease virus

(NDV) and parainfluenza virus (PIV) 2, have been shown to self assemble onto cellular RNA

to form NC–like particles when they are expressed alone, thus providing support for a N-N

interaction (Buchholz et al., 1993; Errington and Emmerson, 1997; Nishio et al., 1999)

Sendai Virus RNA polymerase: The P and L proteins The P protein and the L protein form the core of the viral RNA dependent RNA polymerase

(vRNAP). The vRNAP is involved in the transcription and the replication, which are two

important steps in the life-cycle of the virus. For transcription, the polymerase complex is

constituted only by the P and L proteins, but for efficient replication, the N protein is required

in addition to the L and P proteins. Several studies indicate that there are two different

possible states (multiprotein complex) of the polymerase depending on whether it is

transcribing or replicating. For example, it has been recently shown that the polymerase of

Vesicular Stomatitis virus (VSV) founds itself in a replicase or in a transcriptase state,

depending on its protein composition (Qanungo et al., 2004). There is, however, no indication

that this could be the case for the Paramyxovirus RNA polymerase.

The polymerase cofactor P protein of SeV forms a tetramer and is named for its highly

phosphorylated nature. It is a modular protein with distinct functional domains (Deshpande

and Portner, 1984). It is composed of N-terminal and a C-terminal domain separated by

hypervariable hinge (Curran and Kolakofsky, 1990). The N-terminal part is a chaperone for

unassembled N proteins (N°), preventing it from binding to nonviral RNA in the infected cell

(Curran et al., 1995) and forming a complex (P-N°) whose intracellular concentration is

18

Page 21: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

believed to regulate rates of transcription and replication from genomic template (Curran et

al., 1995; Masters and Banerjee, 1988). The C-terminal part is only functional as an oligomer

and forms, along with L, the polymerase complex. It has already been shown for some other

paramyxoviruses that the association of the N and P proteins has an effect on the N

conformation (MeV) and on the virus assembly (VSV) (Das and Pattnaik, 2005; Kingston et

al., 2004).

The L protein is the largest and the least abundant protein of the structural proteins. Its gene is

also the most promoter-distal in the transcriptional map. It binds to the N:RNA template via

the P protein and contains all vRNAP catalytic activities like synthesis,

capping/polyadenylation and methylation of the nascent viral mRNA (Ogino et al., 2005). The

L protein is highly unstable when expressed alone and needs to bind P to confer a good

stability and a proper conformation, as co-expression of P and L is necessary for the

formation of an active polymerase complex (Curran et al., 1995; Horikami and Moyer, 1982;

Horikami et al., 1997). Until today, no crystal structure of the mononegalvirales L proteins is

available. However, primary structure conservation among the RNA polymerases suggests

similar protein architecture. Sequence comparisons of NNV-L proteins have identified 6

conserved regions, interrupted by variable sequences. These regions have been proposed to

correspond to functional domains of the protein (Poch et al., 1990; Sidhu et al., 1993). There

is also new evidence that the L protein could directly interact with itself and that this

interaction would help RNA synthesis (Smallwood and Moyer, 2004).

The M protein The matrix (M) protein is the most abundant protein in SeV virion. It is a quite basic and

hydrophobic protein. The M protein is considered to be the central organizer in

paramyxovirus budding and virus morphogenesis (Mottet et al., 1996; Sakaguchi et al.,

1994b). The M protein self-associates and interacts with membranes, forming patches at the

inner surface of the plasma membrane (Stricker et al., 1994). It also interacts with the

cytoplasmic tails of integral membrane proteins such as the F and HN proteins, the lipid

bilayer and the NC (Ali and Nayak, 2000; Yoshida et al., 1979). Moreover, the M protein

forms vesicles and self-releases from cells when singly expressed from cDNA (Takimoto et

al., 2001).

19

Page 22: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

The HN & F proteins The fusion of SeV requires co-expression of both HN (hemagglutinin-neuraminidase) and F

(fusion) proteins. The HN and the F proteins are integral membrane glycoproteins and are

essential for regulating morphogenesis and budding (Fouillot-Coriou and Roux, 2000;

Takimoto et al., 1998). The HN protein is involved in cell attachment and is responsible for

the adsorption of the virus to sialic acid-containing cell-surface molecules. In addition, it

mediates enzymatic cleavage of sialic acid, namely neuraminidase activity, from the surface

of virions and of infected cells. This activity prevents self-aggregation of viral particles during

budding at the plasma membrane. The F protein mediates viral penetration by fusion between

the virion envelope and the host cell plasma membrane. The F protein is synthesised as a

precursor F0 which must be proteolytically cleaved to F1 and F2 for fusion activity

(Morrison, 2003). The fusion occurs directly at the cell surface in an endosome-independent

way, suggesting that infection does not require the acid pH of endosomes to activate fusion.

The F protein has a self-release activity when expressed alone (Takimoto et al., 2001). After

infection, the F proteins expressed at the plasma membrane of infected cells can mediate

fusion with neighbouring cells to form syncytia, a cytopathic effect that can lead to tissue

necrosis in vivo and might be a mechanism of virus spread.

D. Viral life cycle All aspects of the replication of SeV happen in the cytoplasm. In cell culture, single-cycle

growth generally last for 24 hours. As the infection takes place, the virus is adsorbed to the

receptors found at the cell surface, and fusion occurs between the viral membrane and the

cellular plasma membrane. This leads to the release of the helical NCs in the cytoplasm. This

NC containing the viral RNA genome is the template for all RNA synthesis (Fig.3). Two

functions are provided by the viral RNA genome: the mRNAs transcription and the viral RNA

replication. The N, P/C/V, M, HN and L proteins are synthesised by the cellular ribosomes.

The assembly of the genomes and the N proteins takes place in the cytoplasm. The M protein

lies in the inner surface of the cytoplasmic membrane whereas the HN and the F float at the

membrane and concentrate to the M patches, excluding other cellular proteins. Finally, the

NCs associate with the M proteins and the new viral particles bud out of the cell taking a

portion of the plasma membrane (Lamb and Kolakofsky, 2001).

20

Page 23: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

Figure 3 : The life cycle of a Paramyxovirus (From Lamb and Kolakofsky, 2001).

RNA synthesis Intracellular replication begins with the transcription of the viral genome into capped and

polyadenylated mRNAs by the vRNAP. The vRNAP first transcribes the leader RNA at the

3’end of the genome, and then begins the transcription of the genes into six individual

mRNAs in a sequential and polar manner. This polymerase occasionally fails to reinitiate the

downstream mRNA at each junction, leading to the loss of transcription of further-

downstream genes, consequently a gradient of mRNA synthesis that is inversely proportional

to the distance of the gene from the 3’end of the genome is observed. The N protein is the

most abundant of the structural proteins being synthesized, and the intracellular concentration

of its unassembled state (N°) is a way of controlling the relative rates of transcription and the

replication from the genome template. When sufficient amounts of N° are present, viral RNA

synthesis becomes coupled to the concomitant encapsidation of the nascent (+) RNA chain.

Under these conditions, vRNAP ignores all the junctions, to produce an exact complementary

21

Page 24: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

antigenome (+) chain, in a fully assembled NCs. The antigenome will then be used for the

synthesis of a new RNA genome, which will be used again as a template or assembled into a

nascent viral particle. The vRNAP can also initiate RNA synthesis at the 3’end of the

antigenome in the absence of sufficient N, but only a trailer RNA is made in this case (Fig.4)

(Lamb and Kolakofsky, 2001).

Figure 4 : RNA synthesis of SeV. The viral polymerase copies the genome into the leader (le) and six individual mRNAs from the genomic promoter (G/Pr). When the N° is sufficient, the viral polymerase starts replicating the genome into full-length antigenomes, which serve as intermediates in genome replication. The viral polymerase synthesises from the antigenomic promoter (AG/Pr) new genomes and other small RNA products, the trailer (tr). Both the genome and antigenomes are tightly encapsidated with the N proteins. (From (Lamb and Kolakofsky, 2001)

Genome (-) N P/C/V) M F HN L 5’3’

G/Pr

AG/Pr

Antigenome (+) N P/C/V) M F HN L 3’5’

le

tr

mRNAs

TRANSCRIPTION

NN

5’

5’

REPLICATION

22

Page 25: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

E. Accessory Proteins The P gene of SeV expresses multiple species of proteins by means of using overlapping open

reading frames (ORFs) (Fig. 5). This gene encodes as many as eight polypeptides via these

ORFs: the P, V, W, C’, C, Y1, Y2 and X proteins. SeV P gene mRNA contain 5 start codons

near its 5’ end, four of which are used for a nested set of “C” proteins that initiate at

ACG81(C’), AUG114 (C), AUG183 (Y1) and AUG201 (Y2) and terminate at UAA726. Among

the four C proteins, the C is the major species expressed in infected cells, at a molar ratio

several fold higher than that of the other three proteins (Kurotani et al., 1998). The second

start codon, AUG104, initiates 3 proteins (P, V and W) as a consequence of cotranscriptional

mRNA editing (Lamb and Kolakofsky, 2001). The start site (AUG104) for translation of the P

protein is in a favourable context (kozak) for recognition by the ribosome; and since it is

placed right after ACG81 (which is normally not a favourable start site) it is more often used.

For SeV, AUG104, and AUG114 are initiated by leaky scanning, whereas AUG183 and AUG201

are initiated by ribosomal shunting (Curran and Kolakofsky, 1988; Gupta and Patwardhan,

1988; Latorre et al., 1998b).

23

Page 26: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

Figure 5: The P gene encodes eight proteins. There are five alternative start sites (ACG or AUG) and the numbered nucleotides (at bottom) show positions of the initiation codons of P, C', C, Y1 and Y2. Transcription stuttering occurs within the P reading frame at sequence shown in the middle, adding one or two G residues that change the open reading frame, producing V or W, respectively.

SeV P gene contains an editing site in the middle of its reading frame. At this sequence, the

vRNAP recognises the 3’-UUU UUU CCC stretch on the template and occasionally stutters.

This stuttering most likely occurs when the vRNA pauses, and the growing RNA chain slips

backward on the RNA template by one (or more) nucleotides (Hausmann et al., 1999; Pelet et

al., 1991; Vidal et al., 1992; Hausmann et al., 1999). The vRNAP then resumes elongation.

When this happens in the run of three G’s, an extra G is added in the growing chain changing

the reading frame. Addition of one G at the editing site produces an mRNA that encodes the

V protein, whereas addition of two Gs leads to the W protein. The frequency of V and W

production can vary depending on the kind of virus (Lamb and Kolakofsky, 2001).

F. Role of the C and V proteins The C proteins are relatively small (175-215 residues), highly basic proteins and non-essential

for the virus multiplication in vitro. Together with the V protein, they are referred to as

“accessory” proteins (Tapparel et al., 1997), because viruses that do not express them are still

viable in cell culture. SeV like many other members of the paramyxovirinae subfamily uses

one or more products of its P/V/C gene to modulate viral RNA synthesis and to antagonize

innate immunity. During the last decade, the involvement of the paramyxoviruses C and V

proteins in counteracting the innate immune response was intensively studied. The

Paramyxoviruses are likely to use their C proteins for this function by opposition to the

Rubula-, Morbili-, and Henipa-viruses that use their V protein.

SeV C proteins The localisation of the C proteins is likely to be at the membrane for two reasons: First, the C

proteins were found to interact with a host protein involved in apoptosis and endosomal

membrane trafficking, called Alix (Sakaguchi et al., 2005). Secondly, the C proteins contain a

specific sequence at the N-terminus that functions as a membrane targeting signal and

membrane anchor (Marq et al., 2007).

24

Page 27: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

The C proteins are also required for virus replication because they act as inhibitors of the

replication of the antigenomes in a promoter-specific fashion. More precisely, as the C

proteins slowly reach a certain concentration during the course of infection, the genomic

promoter (G/Pr) gets particularly sensitive to their presence, and the replication of the

antigenome is reduced (Cadd et al., 1996a; Tapparel et al., 1997).

As mentioned before (Fig. 5), all four C proteins (C’, C, Y1, Y2) of SeV share the same C-

terminal region and only the long proteins (C’ and C) contain the same N-terminal region.

SeV that cannot express any of the C proteins are at the limit of viability. SeV mutants that

can only express the short C proteins (Y1 and Y2) block IFN signaling like SeV-WT but are

highly debilitated. The four C proteins physically interact with signal transducer and activator

of transcription 1 (Stat1), which is a specific intracellular protein whose role is to protect and

signal pathogen invasion. Activated Stat1 is phosphorylated on tyrosine 701, and is referred to

as p-Stat1. As a consequence, these interaction between C and Stat1 will prevent IFN

signaling through the JAK/Stat pathway and at the same time block the establishment of the

antiviral state (Stark et al., 1998). Moreover, the SeV C proteins have also a role in disrupting

Stat2 phophorylation (Gotoh et al., 2003; Li et al., 2006). It was shown that the C terminal

domain of C is necessary and sufficient for blocking the IFN signaling, suggesting that the Y1

and Y2 proteins are able to guarantee this function on their own. On the other hand, only the

longer C proteins (C’, C) provoke the instability of Stat1, by reducing Stat1 levels and

inducing p-Stat1 formation in an IFN-independent manner throughout the course of infection

(Fig.6) (Garcin et al., 2003). Consequently, the pre-existing IFN-induced antiviral state is

reversed and this suggests that the N-terminal domain of the C protein is associated with the

degradation of Stat-1 in the cell (Garcin et al., 2002). Furthermore, C1-23 (23 residues at the N-

terminal of the C proteins) was shown to be sufficient for reducing Stat1 levels and to act as a

membrane targeting signal. Moreover, the activities of the longer C proteins are required for

the localisation of C at the plasma membrane (Marq et al., 2007). Finally, recent data suggest

that the C protein is able to inhibit the IFNβ signaling of RIG-I, a cytoplasmic viral sensor

involved in the induction of IFNβ, by a yet unknown mechanism. The Y protein domain

would presumably be responsible for the inhibition of RIG-I but no binding between the C

protein and RIG-I has been shown yet.

25

Page 28: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

a) C’,C,Y1,Y2

STAT 1

C term N term

IFN

1-23STAT

1

C term N term

Stat1 degradation

Only C’,Cb)

Figure 6 : SeV C proteins counteract the host interferon response by at least two mechanisms. All four C proteins (C’, C, Y1, Y2) of SeV share the same C-terminal region and only the long proteins (C’ and C) contain the same N-terminal region. These 2 regions are associated with different functions. a) The C-terminal domain is necessary for the binding to Stat1, preventing its activation in response to IFN. b) The N-terminal domain (1-23 residus) is associated with the degradation of Stat-1, in this case only the C’ and C target Stat1 for degradation. SeV V protein The V protein seems to display similar functions to the C proteins. Elimination of V by

mutating the editing site, or mutations in the C-terminal domain specific to V, reduces the

virulence of SeV in mice, indicating that V is essential for efficient virus replication and

pathogenesis in mice. Therefore, the SeV V protein seems to interfere with some host

mechanisms that reduce virus replication or spread (Sakaguchi et al., 2003). SeV V protein as

well as other V proteins of the paramyxoviruses, has been shown to limit IFNβ induction

upon synthetic dsRNA poly(rI)-poly(rC) (PolyI/C) treatment(Andrejeva et al., 2004; Childs et

al., 2007).

The role of the V protein, in inhibiting the host interferon response, has been discovered only

recently and can act differently depending on the virus. Indeed, expression of the Rubulavirus

V proteins (MV, SV5 and HPIV2) was demonstrated to induce polyubiquylation of their

target Stat (Stat1,2 or 3 depending on the virus) resulting in efficient proteasomal degradation

26

Page 29: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

(Horvath, 2004; Ulane et al., 2003; Yokosawa et al., 2002). Nipah and Hendra viruses share

the V-dependent IFN signaling evasion properties with other paramyxoviruses, but unlike the

Rubulaviruses, they do not induce Stat destabilization. Indeed, they subvert IFN responses by

sequestering Stat1 and Stat2 in high molecular weight complexes without inducing their

degradation (Rodriguez et al., 2003). Finally, MeV encodes a V protein distinct form both the

Rubula- and Henipa-viruses genera. Its expression effectively prevents both IFNα/β and

IFNγ-induced transcriptional response. It does not degrade Stat or prevent Stat

phosphorylation, but blocks IFN-induced Stat1 and Stat2 nuclear import (Palosaari et al.,

2003).

G. Leader and Trailer RNAs

The genomic and antigenomic replication promoters (G/Pr and AG/Pr) of paramyxoviruses

are found within the terminal 96 nucleotides of each RNA and are bi-partite in nature

(Murphy et al., 1998; Pelet et al., 1996; Tapparel et al., 1998) (Fig 7).

Figure 7: Primary structure of the SeV replication promoters. The 96 nt of the genomic (G/Pr) and antigenomic (AG/Pr) promoters are presented as RNA sequence in ‘hexamers’, numbered 1–16 from the 3' end (-OH 3'). In the G/Pr, the leader coding sequence is outlined, as well as the N gene transcription start signal (nt 56–65), which constitutes the first transcription start signal (N gs1) seen by the viral polymerase. In the AG/Pr, the trailer coding sequence is shown, as well as the complement of the L gene polyadenylation site (L ge). (From (Vulliemoz et al., 2005).

27

Page 30: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

SeV leader and trailer RNAs are short transcripts generated during abortive antigenome and

other hand, the trailer is believed to include the last 57 nucleotides at the 3’end of the

genome synthesis, respectively. They both contain no coding region, and are neither capped

nor polyadenylated, and carry triphosphates at their 5’ends that are believed to be potential

targets for cellular antiviral genes (Plumet et al., 2007). The leader is part of the G/Pr and

includes the first 55 nucleotides at the 3’end of the negative-strand genome RNA. Its

sequence contains signals for initiation of RNA synthesis by the vRNAP, and is also thought

to contain the encapsidation signal that direct packaging of full-length plus-strand copies of

the viral genome in NCs, as it is the case for the leader of VSV (Smallwood and Moyer,

1993).

On the

antigenome RNA. This sequence, very rich in A/U is essential for the viral transcription and

replication and also contains signals for genome packaging. Moreover, it has been shown by

Iseni et al. in 2002, that a U-rich trailer sequence, nucleotides 31-41, has been found to bind

the cellular TIAR protein, involved in the induction of apoptosis. Recombinants of SeV

expressing the TIAR binding domain from both the G/Pr and the AG/Pr, which was modified

in order to contain the U-rich sequence, exhibited a reduced cythopathic effect, and lead to

infected cells survival (Iseni et al., 2002a). This result underlines the fact that, although G/Pr

and AG/Pr carry out similar functions, they also have distinct properties. Indeed, the exchange

of these sequences has interesting effects on virus infections. One of the main differences

between the AG/Pr and the G/P is their strength: It has been shown that the AG/Pr has a

stronger affinity with the polymerase and that there is 10 times more genome than antigenome

produced (Lamb and Kolakofsky, 2001). Plus, an excess of 5-10 fold of genome over

antigenome has also been observed for VSV (Kiley and Wagner, 1972); and an excess of 20

to 50 fold of genome over antigenome has also been shown for Rabies virus (RV). Further

analysis on SeV showed the important role played by the gene start gs1 in decreasing the

strength of the replication activity of the G/Pr (Le et al., 2003). Indeed, when gs1 was

introduced in the AG/Pr sequence, the balance between genome and antigenome RNAs was

equalized by weakening AG/Pr replication. Finally, the presence of the leader (containing the

start site), instead of the trailer in the G/Pr also favours the hypothesis that the AG/Pr is

stronger than the G/Pr.

28

Page 31: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

H. Sendai Virus strains There are two known lineages of SeV: Z/H/Fushimi and Ohita M/Hamamatsu (Fujii et al.,

2002; Itoh et al., 1997). The nucleotide sequences within each lineage are 99% identical and

they are 89% identical between lineages.

Z/H/Fushimi come from viruses isolated in Japan in 1956 after an epidemic of newborn

infants and adapted to grow in embryonated chicken’s eggs (Ishida and Homma, 1978;

Skiadopoulos et al., 2002). These adapted viruses, which were continuously passaged in eggs

over a period of several decades are moderatly virulent for mice (50% lethal dose [LD50] =

103 to 104 PFU) (Sakaguchi et al., 1994a).

Ohita M (SeVM) and Hamanatsu, in contrast, are highly virulent (LD50, <102). They were both

isolated from two completely separate, very severe epidemics of animal houses in Japan and

were low-egg-“passaged”. This virus is presumably closer to the virus in its natural host, and

it is known that SeV passage in eggs attenuates its virulence in mice. For instance, in an

infectious model, in which Kiyotani et al used three-year old mice, the Hamamatsu strain was

very virulent; but when serially “passaged” 30 times in eggs, it strain became attenuated.

(Kiyotani et al., 2001).

29

Page 32: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

I. The Defective Interfering (DI) genomes General aspects: The generation of uncompleted forms of the viral genome during the viral replication process

called Defective Interfering (DI) genomes has been observed in almost all the RNA and DNA

viruses including SeV (Huang and Baltimore, 1970). A table is represented summarizing the

occurrence of DI in negative stranded RNA viruses (Table 2). The ease with which DIs are

produced varies widely within different virus groups and depends on many different factors.

These include growth conditions, multiplicities of infection, the host cell, relative rates of

standard virus replication and DI enrichment, virus strain differences, and intrinsic rates of DI

generation. The appearance of DI genomes arises during passage at high multiplicities of

infection because of the need for complementation by the helper virus. It has also been

reported that single clones from certain viruses (SeV, VSV and Influenza I) have a genetic

capacity to regularly generate the same DI species (Holland et al., 1980; Kolakofsky, 1979).

A good way to get rid of most or all DI genomes in a given virus stock is to “plaque” the virus

stock several times in a row (Baltimore and Huang, 1975). Since DI RNAs are not

antigenetically distinct from their parent virus, their biological properties are attributable to

the genome deletions that they contain. Most of the time, the DI genome can be separated by

velocity gradient, because they are smaller than the standard helper virus genome.

.

Table 2:

Virus Group Member name References

Negative Strand Rhabdo Vesicular stomatisis, rabies, others Reichmann and Schniztlein (1979) Paramyxo Sendai Kolakofsky (1979) Newcastle disease Roman and Simon (1976) Measles Rima et al. (1977) Mumps Norval (1979) Orthomyxo Influenza, fowl plague Nayak (1980) Arena Lymphocytic choriomenengitis, others Pedersen (1979) Tacariibe Gimenez and Compans (1980) Bunya Bunyavera Kascsak and Lyon (1978) Lacrosse Bishop and Shope (1980) Positive strand Picorna Poliovirus Lundquist et al. (1979) Toga Sindbis, Semliki Forest, West Nile Stollar (1979)

Corona Mouse hepatitis Robb and Bond (1979)

30

Page 33: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

The Origin of DI RNAs: DI genomes (DIs) stocks are defective viruses that contain only a fraction of genetic

information of the infectious non defective (ND) virus genome and that require homologous

parental virus as helper for replication. They also contain virus structural proteins and exhibit

the capacity to replicate preferentially at the expense of the infectious helper virus in cells

infected by both. DI RNAs originate from low frequency events during the replication or

transcription of standard virus RNAs. Many different DIs can be generated but only a few are

selected, depending on their capacity to interfere with the ND genome. By “interference”, we

mean that DI particles can replicate and amplify their genome preferentially at the expense of

the replication of the helper virus. Indeed, the latter encodes replication and encapsidation

proteins and must compete with the DIs for these gene products. Small DIs are likely to be the

majority that are selected, but defective full-size genomes can also be found to exert

interference (Roux et al., 1991). In general, most of the DI particles of RNA viruses are only

replicative entities that are not capable of transcription or translation. They can undergo

extensive mutational changes but must conserve the segments that are necessary for efficient

replication and encapsidation. It is important to underline the fact that there must be a balance

between the generation of DIs and the rate of viral production. If the DI interference is too

strong, there will be insufficient virus helper to support significant DI particle replication,

leading to an increased yield of infectious virus and to a reduced yield of DI.

The formation of DI particles is expected to be a consequence of the viral polymerase. It has

been suggested that DIs arise by virus rearrangement or recombination as a result of viral

replicase “leaping” or skipping from one virus RNA template to another or from one segment

of a template to another (Huang, 1977). During this “leaping” the RNA replicase carries

uncompleted nascent strand to a new template, then uses this nascent strand as a primer for

continuation of chain elongation at the new template elongation site. Depending on the

leaving sites or continuation sites, different DIs can be observed. There are mainly two

different forms of DIs that can be generated: the Internal deletion DI and the Copyback DI

genomes (Fig 8). In the case of the internal deletion DI the viral polymerase starts

synthesising the genome and at a certain point jumps forwards on its template. The internal

deletion DI genome still conserve the 3’ and the 5’ ends of the ND genome and lacks various

portions of the internal transcription units. It carries both the minus- and plus-strand polarity

RNAs containing both promoters enabling the DI to transcribe from the minus-strand and

replicate from both the minus- and plus-strand. In the case of the copycack DIs, the viral

31

Page 34: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

polymerase detaches itself from the template, and instead of going forwards, it initiates

copying in reverse direction using the nascent strand as template. Consequently, the copyback

DI genome lacks most of the genomic 3’ end sequence from the ND genome and in addition,

it contains complementary extremities to the genomic 5’end, as inverted repeats (Kolakofsky,

1976; Leppert et al., 1977) (Fig 8). The origin of terminal sequence complementarity in SeV

and VSV DIs is generally thought to occur by some sort of strand-switching event during

polymerisation, where the polymerase would drop off the template and start the synthesis of

the daughter or another template. In this way the features of terminal sequence

complementarity in DI genomes provide strong argument for the involvement of the viral

polymerase in the origin of DIs.

The presence of inverted complementarity sequences (not found in the standard genome) was

defined as the hall mark of the most abundant class of VSV DI (DI_011) RNAs also called the

“snapback” DI. The first evidence for this structural feature came from electronic microscopic

observations of circular DI RNAs with characteristic small panhandle or stems in SeV (e.g.

the natural copyback DI-H4), (Kolakofsky, 1976) and in VSV (Perrault and Leavitt, 1978).

The shortest inverted repeat reported for Paramyxoviruses amounts to 94 nucleotides, with a

range between 94 and 168 nucleotides (110 nucleotides for SeV DI-H4) (Calain et al., 1992;

Calain and Roux, 1993). In this case, the copy-back DI carries both the minus- and plus-

strand polarity RNAs but contains the antigenomic (AG) promoter at both the genomes and

antigenomes ends. Furthermore, copyback DIs in SeV seem to interfere more with the ND

genome than other conventional DIs, presumably because of the strength of the two strong

AG promoters they contain. As mentioned before, the replication efficiency of the copyback

DI (e.g. DI-H4) is 20 fold higher than the one of the Internal Deletion DI-E307 (Calain and

Roux, 1995). For VSV, the presence of the specific sequences at the 3’-terminus of both the

genomic and antigenomic DI RNAs may also explain in part the replicative dominance of DI

genome over the full-length genome, which contains these sequences only at the 3’-terminus

of the antigenome (Pattnaik et al., 1995). Finally, it has been observed that some DI copyback

particles could modulate the course of infection by interfering indirectly with the helper virus

by inducing IFN activation. This aspect is likely dependent on the kind of DI genomes

involved in the infection. The VSV “snapback” DI has been shown to strongly induce the IFN

and interestingly, it was the minimal multiplicity of DI infection that was required to promote

maximal effects on IFN induction (Marcus and Gaccione, 1989).

Another important aspect of DI genomes is that they are required for the establishment and/or

maintenance of persistent infections in cell culture (Holland et al., 1979). This characteristic

32

Page 35: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

can be observed in several different virus systems, such as RV (Kawai and Matsumoto 1977),

VSV (Horodyski and Holland, 1980) and NDV. For SeV infection, this property can be

explained (in part) by the fact that copyback DIs contain two trailer sequences that can bind to

TIAR and thus prevent apoptosis leading to the establishment of a persistent infection (Iseni

et al., 2002b). Even though the ability of DI genomes to modulate the intensity and the course

of viral infections has been studied widely in in vitro systems, their possible roles in natural

infections remain largely unexplored and unconfirmed (Barrett and Dimmock, 1986).

Viral polymerase

SeV Antigenome (+) N P/C/V M F HN L 3’ 5’

AG/Pr

Internal deletion (DI-E307)

5’ 3’

3’ 5’

5’

Copy-back (DI-H4)

3’

1. 2.

Uncompleted forms of viral RNA genome = DI genomes

Figure 8: A schematic representation of DI genomes synthesis.

During the replication process, the viral polymerase synthesizes new SeV genomes from the antigenomic promoter (AG/Pr) and occasionally generates uncompleted forms of the viral genome called the Defective Interfering (DI) genomes. The dotted lines below the SeV antigenome indicate the path of the viral polymerase when it generates internal deletion DI genome (e.g. E307) or copyback DI genomes (e.g. DI-H4).

33

Page 36: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

J. The reverse genetics system In contrast to the positive RNA virus, which are potentially infectious by forming directly

viral proteins, neither the genome nor the antigenome RNA of NNV are infectious. Indeed,

they need to be transcribed before they can produce active proteins, such as the polymerase or

the nucleoprotein, that are required for the synthesis of the viral proteins and the RNA

encapsidation, respectively. For this reason, the recovery of infectious virus from cDNA

(reverse genetics), which requires the co-expression of the N, P and L proteins in trans, has

been delayed compared to the one of positive stranded viruses (Racaniello and Baltimore,

1981). Influenza virus was the first NNV to be recovered from cDNA. Luytjes et al. have

described a system in which they use purified N proteins and synthetic RNA to assemble a

synthetic gene-like RNA into a biologically active ribonucleoprotein (Luytjes et al., 1989).

In contrast to influenza virus the paramyxo-ribonucleoproteins have resisted for a long time to

be assembled in a functional form in vitro, probably because they are much more tightly

structured (Baudin et al., 1994; Iseni et al., 1998). Indeed even after banding in CsCl density

gradients, they remain highly active and the RNA within this structure remains resistant to

RNase treatment at any salt concentration (Enami and Palese, 1991; Lynch and Kolakofsky,

1978). In 1991, paramyxoviridae derived RNA corresponding to a SeV minigenome was

rescued for the first time (Krystal et al., 1986; Park et al., 1991). This artificial SeV RNA,

containing the antisens coding region of the CAT gene flanked by the entire 5’ and 3’

noncoding termini of SeV, was generated in vitro by the T7 polymerase from a linearized

plasmid. When introduced into cells that was infected with SeV, this RNA construct was

transcribed, replicated, and packaged into infectious virions and significant CAT activity was

observed. This further confirmed the fact that the cis-acting sequences necessary for the

encapsidation, replication and transcription of SeV are located in the terminal sequences and

that the helper virus provides the necessary viral proteins for the RNA recognition.

The concept of replicating mini-genomes using support plasmids providing N, P, and L

proteins in trans was finally the key to the development of the technology. Indeed, the use of

mini-genome RNA transcripts carrying only a reporter gene facilitated in vivo experiments, in

which RNA encapsidation was strongly required. Moreover, a system named the Vac-T7

system, in which a recombinant Vaccinia virus encoding the bacteriophage T7 RNA

polymerase was established (Fuerst et al., 1986). Cell were infected with the recombinant

Vaccinia virus and transfected with plasmids encoding all the required viral genes and the

viral RNA genome, which were all expressed under the control of the T7 polymerase

34

Page 37: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

promoter. This approach conferred several advantages: (1) it provids the T7 polymerase,

whose function is absolutely necessary for producing RNA genomes and for increasing the

expression level of the N, P and L mRNA in the cytoplasm; (2) it allows further analysis

regarding the individual function of each viral gene involved or not in the RNA amplification.

The vac T7 system required the generation of correct viral RNA termini. Indeed, while the

exact 5’ends were determined by the position of the T7 promoter transcription start signal,

generation of the correct 3’ends required a ribozyme activity sequence derived from the

antigenomic strand of the hepatitis delta virus RNA (Gershon et al., 1991; Perrotta and Been,

1990; Shih and Been, 2002).

More refinements to the original technique have been made using stably transfected cell lines

BHK, which express the bacteriophage T7 RNA polymerase instead of vac-T7 infection.

(Buchholz et al., 1999). This approach based on the critical thought that the assembly of free

genome (or antigenome) RNA into a functional NC could be accomplished in a living cell,

has been successful for rescuing VSV and SeV DIs (Calain et al., 1992; Calain and Roux,

1993; Curran and Kolakofsky, 1991; Pattnaik and Wertz, 1990).

With time, this approach has been successful for many negative-stranded RNA viruses: RV

(Conzelmann and Schnell, 1994), VSV (Pattnaik and Wertz, 1990), SeV (Garcin et al., 1995),

RSV (Collins et al., 1991), MeV (Radecke et al., 1995), SV5 (Murphy et al., 1998),

Rinderpest virus (Baron and Barrett, 1997), Canine distemper virus (Gassen et al., 2000) and

Nipah virus (Yoneda et al., 2006).

35

Page 38: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

K. The Interferon System During evolution cells were forced to develop sophisticated defences for counteracting virus

infection. Secretion of cytokines and chemokines is one of the first responses of an organism

to pathogen infection. Among the cytokines are the interferons (IFNs). The IFNs are an

heterogeneous family of cytokines, originally identified on the basis of their ability to strongly

induce cellular resistance to viral infections (Pestka et al., 1987; Vilcek, 1996). The IFNs’

action on virus-infected cells and surrounding calls and tissues elicits an antiviral state. The

IFNs exert various biological effects such as interference with virus replication, modulation of

the immune response, antiproliferative activities and apoptosis. A potent inducer of IFNs is

intracellular double stranded RNA (dsRNA) which is generated during replication and

transcription of viral genome. There are three types of IFNs: 1) Type I IFNs includes IFNα

and IFNβ, plus additional IFNs recently defined: IFNε,κ and ω; 2) Type II IFNs, containing

IFNγ, also called “immune interferon”; 3) Type III IFNs or IFNλ a novel interleukin 10-

related cytokines (Fig. 9). IFNα is produced by peripheral blood leucocytes and many other

cell types in response to viral infection or treatment with dsRNA. IFNβ is mainly produced by

fibroblasts or epithelial cells. IFNγ is produced exclusively by immune cells. Although both

classes of IFNs exhibit potent antiviral activity, Type I IFNs play a more important role in the

innate immune response to viral infection, whereas IFNγ plays a much more significant role

in the subsequent acquired immune response (Schindler, 1999).

A common property of both IFN types is to induce immediate transcriptional responses

through a JAK-Stat signal transduction pathway (Schindler and Darnell, Jr., 1995). All type I

IFNs bind to a class II cytokine receptor composed of IFNα receptor 1 (IFNAR1) and

IFNAR2 chains (Novick et al., 1994), which are associated with the Janus kinase (JAKs)

TYK2 and JAK1 respectively (Darnell, Jr. et al., 1994). Ligand-bound, tyrosine-

phosphorylated receptor complexes bind the SH2 domains of signal transducers and activators

of transcription (Stats) 1 and 2, causing phosphorylation of the proteins on tyrosines 701 and

692, respectively. Interaction of Stats through reciprocal SH2 domain-phosphotyrosine

binding results in the formation of two distinct transcription factor complexes: 1) ISGF3, a

heteromeric complex consisting of Stat1 and Stat2 in association with a third protein, p48 or

IRF9, associates specifically with and transactivates genes containing the interferon-

stimulated response elements (ISRE) in their promoter or enhancer regions. 2) A simpler

complex, consisting solely of the Stat1 homodimer, is also active as a transcription factor and

binds to different DNA sequences, termed IFNγ activated site (GAS) elements (Katze et al.,

36

Page 39: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

2002). The receptor for IFNγ is structurally related to that for IFN type I. It consists in IFNγ

receptor 1 (IFNGR1) and IFNGR2 chains in association with Jak1 and Jak2 kinases. Once

tyrosine-phosphorylated in the presence of ligand, it binds Stat1 and causes phosphorylation

of tyrosine 701. Stat1 homodimers are formed, move to the nucleus, and regulates

transcription of promoters containing GAS sequences (Decker et al., 2002). Finally, type III

IFNs display activities and functions much like the type I IFNs and utilise a different receptor

complex to signal (IL10R2 and IFNLR1 chains).

Figure 9: Interferon signaling by the JAK-Stat pathway. (See text for details ) (Samuel, 2007)

The regulation of IFNs synthesis is well characterized and requires the participation of several

transcriptional factor complexes that already pre-exist in the cell and are activated upon virus

infection. These factors bind to regulatory sequences called positive regulatory domains,

which lie within 200 nucleotides upstream of the transcription start sites on both IFNα and

IFNβ genes. Four different transcription factors are known to bind to the IFNβ enhancer: the

ATF-2/ C-Jun heterodimer, the NF-kB heterodimer and two interferon-regulatory factors

(IRFs), IRF-3 and IRF-7. Together with general transcription factors including TFIID and

RNA polymerase II, this collection of DNA-bound proteins assembles to form an

“enhanceosome” (Thanos and Maniatis, 1995). The IFNα enhancer also contains multiple

positive regulatory domains and is activated by binding to similar transcription factors. The

IRFs are extremely important and function in a number of processes such as pathogen

37

Page 40: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

response, cytokine signaling, cell growth regulation, and hematopoiesis. IRF-3 and IRF-7 are

two of the nine members of the IRF family (Mamane et al., 1999), residing in the cytoplasm

of uninfected cells, and undergoing nuclear translocation upon viral infection. IRF-3 and IRF-

7 are crucial to maximally induce the expression of IFNα/β (Sato et al., 2000). IRF-3 is

constitutively expressed in normally growing cells, whereas IRF7 is mainly dependent on

IFNα/β signaling. Upon viral infection, IRF3 is activated by phosphorylation on its serine

residues by IKKε and TANK-binding kinase-1 (TBK-1) (Fitzgerald et al., 2003; Sharma et

al., 2003). Phosphorylated IRF-3 homodimerizes and translocates to the nucleus, where it

recruits the transcriptional coactivators p300 and CREB-binding protein (CBP) to initiate

IFNα/β mRNA synthesis by binding to the IFNα/β enhancer regions (Hiscott et al., 1999).

IFNα/β are secreted and feed back onto cells in a paracrine manner to prime neighbouring

cells for possible infection and in a autocrine manner to induce multiple IFN stimulated genes

(ISGs) including ISGF3 and IRF-7. As a result the “first wave” of IFN leading to the cellular

antiviral state is in place (Sato et al., 1998) (Fig. 10).

Figure 10: Type I IFN induction, signaling and action. Left panel: RNA, a characteristic by-product of virus replication, leads to activation of the transcription factors NF-κB, IRF-3 and AP-1. The cooperative action of these factors is required for full activation of the IFNβ promoter. IRF-3 is phosphorylated by the kinases IKK and TBK-1 which in turn are activated by the RNA-sensing complex of RIG-I, Mda5 and IPS-1/MAVS. Right panel: Newly synthesized IFNβ binds to the type I IFN receptor (IFNAR) and activates the expression of numerous ISGs via the JAK/Stat pathway. IRF-7 amplifies the IFN response by inducing the expression of several IFNβ subtypes. Mx, ISG20, OAS and PKR are examples of proteins with antiviral activity (Haller et al., 2006).

38

Page 41: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

Interferons confer the first line of defence against pathogen invasion by activating the ISGs.

The ISGs carry antiviral functions, targeting the stability or translation of viral RNA. The

three main IFN-induced systems are the following: the Mx proteins, 2’-5’oligo (A) synthetase

(OAS) and ribonuclease L, and the dsRNA-dependent protein kinase PKR. Mx (for

“myxovirus”) proteins are inducible-IFN proteins with the ability to hydrolyse GTP (Haller et

al., 2007). Expression of Mx affects virus replication by interfering with the transcription of

influenza virus and other negative-single stranded viruses. Mx proteins inhibit the activity of

the vRNAP, thus blocking viral mRNA production. They also interfere with the transport of

influenza virus RNP complexes from the cytoplasm to the nucleus. The exact function/action

of Mx is not yet understood and the role of the GTPase activity also remains unclear. The 2’-

5’oligo (A) synthetase is produced in IFN-treated cells, and is activated by binding to dsRNA

in virus infected cells. This enzyme has the sole ability to produce oligomers of ATP via a 2’,

5’ linkage in contrast to the normal 3’, 5’ linkage found in the natural RNAs. These oligomers

bind to and activate ribonuclease L, which degrades viral and cellular RNAs (Player and

Torrence, 1998). PKR is a well characterized effector of anti-viral responses in mammals.

PKR is a serine-threonine protein kinase that is present in normal cells at low levels. Like 2’-

5’oligo (A) synthetase, PKR is activated by dsRNA or stem loop RNA structures. Activated

PKR phosphorylates a variety of cellular proteins, such as the α-subunit of translation

initiation factor eIF-2, thus inhibiting the translation. When PKR is overexpressed,

programmed cell death is activated (Takizawa et al., 1996). Additionally PKR is believed to

play an important role in regulation of cellular protein synthesis in absence of virus infection

(Williams, 1999), as well as in signal transduction and cell growth (Koromilas et al., 1992).

39

Page 42: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

L. Sensors of RNA virus infection Many signaling pathways leading to the IFNα/β induction upon virus infection, have been

recently discovered. These involve specific cellular receptors that can detect the presence of

virus by recognising viral molecular signatures. These viral molecular signatures are part of

the PAMPs (Pathogen-Associated Molecular Patterns) that contain many other potential

patterns from other pathogen, including bacteria and funghi. The PAMPs are recognised by a

wide range of receptors, called PRRs (Pattern Recognition Receptors) that comprise the

Pathogen-Resistance Protein (R-protein) in plants, the Toll-Like Receptors (TLRs), the Nod-

Like Receptors (NLRs) and the recently discovered Rig-like receptors (RLRs) in animals

(Fig.11).

The R proteins were the first PRR to be discovered. These are crucial for immune defence

against all the possible pathogen-derived molecules invading plants. Considering the fact that

these R genes are of real importance for the survival of plants, it is not surprising to find that

kind of pathogen-resistance protein in other organisms (Nimchuk et al., 2003). The next PRR

to be discovered was the Toll receptor that was identified in the fruit fly, Drosophila

melanogaster, as an essential receptor for the establishment of the dorso-ventral pattern in

developing embryos. In the mid-90s, toll-mutant flies were shown to be highly susceptible to

fungal infection (Lemaitre et al., 1996; Yamamoto and Akirz, 2005). The receptor Toll was

identified as a key mediator of innate immune defences in Drosophila melanogaster. A year

later, the identification of a Toll-like receptor (TLR) in the human genome was reported and it

was later called TLR4 (Medzhitov et al., 1997). Sequencing of human and murine genomes

further allowed the identification of 11 TLRs in mice and 10 TLRs in human. The ability of

the TLRs to recognise microbes and directly initiate specific signal transduction cascades that

alert the host defences, can also be observed in the two additional families of innate receptors:

the NLRs and the RLRs. Unlike the TLRs that are essentially found at the plasma membrane,

these families consist of soluble proteins that survey the cytoplasm for signs that broadcast the

presence of intracellular invaders. Until now, the NLRs have been shown to detect bacteria

and many other pathogens, whereas the RLRs only recognise viruses (Creagh and O'Neill,

2006). NLRs share high structural and functional homology with plant R genes and search of

the human genome database revealed 22 R-gene homologous classified in two main

subclasses: the NODs (Nucleotides Oligomerization Domain leucine-rich repeat protein)

counting 5 members and the NALPs (NACHT Leucine rich domain and Pyrin-containing

40

Page 43: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

protein) containing 14 members. Some of the NLRs activation is proposed to occur via a

mechanism similar to the mechanism described for the apoptosome (Reith and Mach, 2001).

NACHT

TLRs

RLRs

Pattern recognition receptors (PRRs)

A B Helicase

TIR

NLRs (NOD1)

LRRR protein

Figure 11: Domain organization of the PRRs. All of the proteins have the NACHT-LRR configuration, with the exception of the RLRs, which contain a helicase domain. The A and B represent the Walker A and Walker B motifs. Not all of the possible N-terminal domains are represented; the caspase-recruitement domains (CARD) are specific to the RLRs and are also present in the NLRs when there are no BIR (baculoviral inhibitory repeat) or acidic domains. The Toll-IL-1 receptor or coiled-coil domains (TIR/CC) are N-terminal domains specific to the R protein or the TLRs (Adapted from (Ting and Davis, 2005). It is likely that the initiation of innate immunity involves an important cooperation between

the TLRs, the NLRs and the RLRs depending on the pathogen, providing a tightly controlled

combinatorial repertoire for triggering host defences. TLRs seem to be specifically active in

immune cells such as dendritic cells whereas NLRs and especially RLRs are expressed in

more various cell lines. The RLR family comprises three DExD/H-box-containing RNA

helicases: retinoic-acid-inducible gene 1 (RIG-1), melanoma differentiation-associated gene 5

(Mda-5) and laboratory of genetics and physiology 2 (LGP2). RIG-I and Mda-5 are both

ubiquitously expressed in most tissues and are part of the ISGs, which allows autocrine and

paracrine amplification of the sensing system. RIG-I and Mda-5 encode two caspase

recruitment domains (CARD) at the N-terminus, followed by an RNA helicase domain. The

helicase domain recognises viral RNA and regulates signal transduction in an ATPase-

dependent manner, whereas the CARD domain is involved in the signal transduction

downstream (Kang et al., 2004). LGP2 lacks completely the CARD domains and functions as

a dominant-negative regulator of RIG-I/Mda-5 mediated signaling (Rothenfusser et al., 2005;

Yoneyama et al., 2005). The recognition of the molecular viral signature via the helicase

domain of RIG-I and Mda-5 likely induces a conformationnal change enabling their CARD

41

Page 44: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

domains to interact with the CARD-like domain of a protein anchored to the outer

mitochondrial membrane called Cardif (MAVS/IPS-1/VISA) (Kawai et al., 2005; Meylan et

al., 2005; Seth et al., 2005; Xu et al., 2005). This interaction leads indirectly to the activation

of several different kinases such as TBK-1/IKKε and IKKγ and activate IRF-3 and NF-κB

respectively, inducing type I IFNs and the antiviral state of the cell (Fig. 12) (Hornung et al.,

2006; Pichlmair et al., 2006).

Figure 12: RIG-I and Mda-5 are cellular RNA helicases that, upon activation, stimulate IFN gene expression. RIG-I recognises RNAs with 5’triphosphates, whereas Mda-5 recognises dsRNA (From (Basler and Garcia-Sastre, 2007). Studies of RIG-I and Mda-5-deficient mice have revealed that RIG-I is essential for the

recognition of a set of specific ssRNA viruses, including Paramyxoviruses, Flaviviruses,

Orthomyxoviruses and Rhabdoviruses, whereas Mda-5 is important for the recognition of a

different set of RNA virus that includes Picornaviruses and alphaviruses (Basler and Garcia-

Sastre, 2007; Kato et al., 2006). In vitro studies have also shown that both RIG-I and Mda-5

can bind to polyI/C and respond to polyI/C and RNA viruses (Yoneyama et al., 2005). In

addition to this, it has been shown that RIG-I is more likely to recognise RNAs with

5’triphosphates than dsRNA, whereas Mda-5 specifically recognises dsRNA (Basler and

Garcia-Sastre, 2007). The role of these helicases is to distinguish between self RNA and non-

self RNA coming from the viruses. At the end of the year 2006, Veit Hornung et al. provided

evidence that uncapped unmodifided 5’-triphosphate RNA present in viruses known to be

recognised by RIG-I, but absent in viruses known to be detected by Mda-5, serves as PAMP

for the detection of viral infection by RIG-I in the cytosol of eukaryotic cells and that this

42

Page 45: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

property is not confined to immune cells. They further observed that RIG-I does not activate

the IFNβ activation with RNA containing 5’di- or 5’-monophosphate (Hornung et al., 2006).

Many of the RNA species in the cytosol are known to lack free 5’-triphosphate group

although all RNA transcripts generated in the nucleus of a eukaryotic cell initially contain a

5’tri-phophate. Indeed cellular self RNA escapes detection by RIG-I because they are known

to undergo several modifications before being transported to the cytoplasm: messenger RNA

acquires a 7-methyl-guanosine cap structure at its 5′-end; transfer RNA undergoes 5′ cleavage

and a series of nucleotide modifications; and ribosomal RNA undergoes several cleavages and

finally associates with ribosomal proteins (Bowie and Fitzgerald, 2007) (Fig. 13).

Figure 13: Discrimination of self and non-self RNA by RIG-I. Viral infection leads to the accumulation of non-self RNAs in the cytoplasm, such as dsRNA and 5-triphosphate RNA. Cellular RNA synthesis takes place in the nucleus by 3 different RNA polymerases. The 5’ ppp of these RNA are eventually removed or masked, thus self RNA species do not activate RIG-I (From (Yoneyama and Fujita, 2007).

M. Viral antagonists of the interferon system Many viruses including SeV have developed strategies for counteracting the host type

interferon I response and this at different levels. For that purpose viruses use most of the time

non-structural proteins which are non-essential for virus growth. These antagonists are often

multifunctional proteins that interact with multiple viral or host cell components and are

involved in regulating many different functions in infected cells. Hence the IFN system can be

triggered at different levels: 1) the IFN induction pathway comprising the IRF-3 activation, 2)

43

Page 46: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

the basic transcription level, such as the inhibition of mRNA exportation or inhibition of polII

from Flu, 3) IFN signaling pathway including the JAK-Stat activation and 4) the IFN effectors

level. Some viruses can contain more than one antagonist inhibiting one or several different

components of the IFN induction and signaling pathway. Moreover, the virus-induced IFN

response is generated in a cascade-like manner. Consequently, viral proteins blocking one

cellular component in this circuit also affect distant signaling molecules or effectors, which

amplify their inhibitory effect. Figure 14, from O. Haller et al. illustrates the range of

activities mediated by IFN antagonists of various viruses (Fig. 14). As mentioned before, SeV

mainly uses the non-structural C and V proteins to act at the level of the IFN induction

pathway by blocking RIG-I activation and also at the level of the IFN feedback loop where C

blocks Stat-1 signaling pathway. In many NNV, the phosphoprotein is the main antagonist.

The P protein of RV prevents IRF-3 phosphorylation by TBK-I (Brzozka et al., 2005). In the

same way, the P protein of Ebola Virus interferes with IRF-3 activation (Basler et al., 2003).

The P protein of Borna disease virus directly binds to TBK-I and reduces its activity

(Unterstab et al., 2005).

Figure 14: Viral inhibitors of the virus-induced IFNα/β response loop: Viral gene products interfere with the type I IFN system at all levels. For example, V of paramyxoviruses were found to be IFN antagonists at the level of the IFN induction pathway. Many others viral proteins are involved in bloking the IFN activation at different levels. For more details see review (From (Haller et al., 2006).

44

Page 47: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

Vaccinia Virus from the Poxvirus family, as well as Influenza I virus, also possess genes that

can interfere with the cellular activation pathway of IFN signaling. E3L and NS1 are viral

regulatory protein found in Vaccinia Virus and Influenza virus respectively. E3L can bind to

dsRNA and inhibit the dsRNA-stimulated enzymes, PKR and RNA-specific adenosine

deaminase. A vaccinia deficient in E3L is highly sensitive to the activity to type I IFN and

restricted growth in some cell lines such as Hela but not in other as chicken fibroblasts

(Fischer et al., 2006). NS1 from Influenza A virus can also act against the IFN type I

response. It has been suggested that NS1 can prevent IFNβ induction in several ways: by

sequestering dsRNA through its amino terminus (Donelan et al., 2003; Hatada and Fukuda,

1992), by forming a complex with RIG-I to abrogate RIG-I signaling (Pichlmair et al., 2006),

by binding to PKR (Li et al., 2006), and by inhibiting posttranscriptional processing of the

3’end of cellular specificity factor and poly(A)-binding protein II (Chen et al., 1999; Fortes et

al., 1994; Li et al., 2006).

45

Page 48: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

II. PUBLICATION 1

"SENDAI VIRUS TARGETS INFLAMMATORY

RESPONSES, AS WELL AS THE

INTERFERON-INDUCED ANTIVIRAL STATE, IN

A MULTIFACETED MANNER"

Laura Strähle, Dominique Garcin, Philippe Le Mercier, Joerg F. Schlaak

and Daniel Kolakofsky

JOURNAL OF VIROLOGY, July 2003, p. 7903–7913

46

Page 49: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

INTRODUCTION TO PAPER ONE:

Upon viral infection, cells respond by protecting themselves by inducing specific

antiviral responses, such as IFNs. IFNs induce an antiviral state via the JAK/Stat pathway that

leads to the activation of the ISGs expression. On the other hand, SeV infection counteracts

IFN action by producing several specific proteins. SeV is known to use its C protein 1) to

evade the host IFN response by binding to Stat1, preventing its activation in response to IFN,

and 2) to target Stat1 for degradation. The leader and trailer products are short promoter-

proximal products generated during abortive antigenome and genome synthesis. Moreover,

the trailer is known to play a role in the regulation of programmed cell death, which is part of

the antiviral program.

We have based our study on cDNA microarrays done in collaboration with Ian Kerr’s

laboratory in London (ICRF) (Schlaak et al., 2002) to study the cellular response to IFNα/βs.

These genes comprise known ISGs and genes of intrinsic interest that might or might not be

induced by IFNs in different cell systems. They also include genes involved in cell

proliferation, immune response, and response to a variety of cytokines. We have used these

arrays to compare the activation of various cellular genes in response to infection by different

strains and mutants of SeV in 2FTGH cells (human fibroblasts). Knowing the importance of

the C gene and the promoter region in disturbing the antiviral system of the cell, two different

SeV strains (SeVM or SeVZ) containing specific mutations in their C proteins (SeV C F170Sand

SeV C Δ1015) or in the promoter regions (SeV GP31-42 and SeV GP42) were used in our study.

The mutant SeV C F170S, represents a SeVM strain that has been isolated after 5 passages in

LLC-MK2 cells. It has three mutations and one, F170S is located in the carboxyl-terminal of

all four C proteins (C’,C Y1 and Y2) (Itoh et al., 1997). This base change is silent in the Open

reading frame of P, V and W but this phenylalanine at position 170 of C is critical for

blocking STAT1 activation, thus inhibiting the IFN signaling (Garcin et al., 2002; Gotho et al.

2001). This strain appears to be strongly attenuated in mice (LD50 of > 8.105 PFU) (Wang et

al., 1994). The mutant SeV C Δ1015 is a recombinant of SeVZ. The codons 13 to 18 of P (W and

V) and the codons 10 to 15 of C have been deleted. These deletions affect the N-terminal part

of the C protein, a region essential for Stat1 degradation. It is an attenuated strain containing

mutated C and C’ and WT Y1 and Y2 proteins. This deletion might alter the function of P, W

and V, even though the P protein seems to be fully functional. Mutants SeV GP31-42 and SeV

GP42 are SeV Z stocks carrying specific mutations in the leader region within the genomic

47

Page 50: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

promoter. Compared with the WT SeV, SeV GP42 has its first 42 nucleotides from the leader

promoter replaced into those of the trailer promoter. SeV GP31-42 has nucleotides from the

leader promoter between the 31st and 42nd changed into those of the trailer promoter (Fig.15).

Nucleotides 31-41 of the trailer promoter are important for binding to TIAR, a host RNA

binding protein important for virus-induced apoptosis. Consequently, infections with mutants

SeV GP42 and SeV GP31-42 lead to persistent infection. Finally, it is important to take into

account that this gain of function can be due to either the over-expression of the trailer or the

absence of the leader.

Figure 15: SeV mutants carrying mutations in the leader region, which is part of the genomic promoter. SeV GP42 has its first 42 nucleotides replaced by the 42 first nucleotides of the trailer region. SeV GP31-42 contains nucleotides 31 to 42 from the trailer region.

Upon infection with SeV mutant viruses (SeV CF170S, SeV CΔ10-15, SeV GP31-42 and SeV

GP42) 15 mRNA levels were found to increase significantly and three distinct patterns of

gene activation could be observed. But, the levels of these mRNAs remain unchanged upon

SeV WT infection (SeVM and SeVZ), presumably because these SeV stocks carry functions to

counteract the cellular host defence. (Fig. 16):

A) The first group includes a series of genes that have their mRNA level increased by

SeV CF170S alone. SeV CΔ10-15 and others SeV mutant infections have on the other hand

no effect on these mRNA levels. The CF170S substitution inactivates the ability of all

four C proteins (C’, C Y1 and Y2) to bind STAT1 in a stable manner and to counteract

IFN signaling. It is likely that any of the four C proteins can block the increase of

mRNA levels upon SeV infection.

B) The second group is only represented by the IL-6 gene. It is activated by SeV CF170S

as well as SeV Δ10-15, but not by the WT viruses or other promoter mutants. Consistant

48

Page 51: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

with this, the N-terminal region in the longer viral C proteins has a role in preventing

the increase of IL-6 messengers upon SeV infection.

C) The third group includes genes whose mRNA levels increase upon SeV CF170S, SeV

CΔ10-15 and SeV GP42 infection, but not upon WT viruses or the SeV GP31-42 mutant.

A function provided by either the C or the first 30 nucleotides of the genomic

promoter (le) is required to prevent activation of these genes.

Figure 16: Schematic representation of the viral mutations and their effects on the host gene activation.

The C proteins are shown as two telescoping boxes representing the longer (C’ and C) and the shorter (Y1

and Y2) C proteins. The requirement for the various WT genetic elements to prevent host gene activation

is shown (Instab: instability; sig: signaling) (From Strahle et al., 2003).

We then decided to confirm the above results by a more quantitative method, Real-time PCR

(Taqman). The following host genes activated by our mutant SeV infections were selected,

each one representative of one group: 6-16 (ISG) for group 1, IL-6 for group 2 and IL-8 for

group 3. Since 6-16 is an ISG, we used it as a positive control. The chemokines, IL-6 and IL-8

were chosen because they are not part of the ISGs (as they are not activated upon simple IFN

treatment) and are presumably involved in other different signaling pathways induced by

virus infection. We also decided to examine the IFNβ gene, because of its role in the early

host response upon viral infection and the cellular gene GAPDH as an internal control to

normalize the level of mRNAs.

49

Page 52: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

We examined the levels of each gene (as well as protein secretion in the case of IL-8) in

different conditions: 1) upon WT and mutated SeV infections to confirm the microarrays; 2)

upon dsRNA treatment or IRF-3 overexpression followed by WT and mutated SeV infections,

to observe whether the antiviral response was enhanced by these pre-treatments; 3) by

changing cell types: we used U5A and U3A cells that are 2fTGH cells, which have been

generated by X irradiation. U5A are defective in the IFNα/β receptor 2 chain (IFNAR2) and

U3A are defective in the Stat 1 gene. The use of these cells allowed us to determine whether

induction of our genes upon WT and mutated SeV infections were dependent on IFN

signaling and required Stat1; 4) Because SeV appears to use the V and C proteins to

counteract the IFNβ expression and because the influence of the presence of the leader and

the trailer RNAs is still unclear, we examined our SeV mutants as well as other recombinants

SeV for their relative activation of IFNβ: I) SeV AGP55 which transcribes two leader RNAs

from both promoters; II) SeV GP48 which transcribes two trailer RNAs and carries the same

phenotype than SeV GP1-42; III) and SeV V minus/W++ that do not expressed any V proteins.

The results of our study revealed that infection of SeV containing specific mutations in their C

proteins and leader region activates specific cellular genes. The absence of the leader (1-42),

C Δ1015, C F170S, and V-/W++ mutations appear to disrupt four distinct elements in the SeV

program to counteract the cellular antiviral response. This means that the products of the C

and V proteins together with the presence of the leader RNA transcripts prevent the

expression of these cellular genes involved in the IFN-induced antiviral state and in the

inflammatory responses (Fig. 17).

Cellular responses

Cellular antiviral

response (activation

IL-8, IFNβ...)

C protein (2 ways) V protein leader

SeV accessory functions

Virus Infection

Figure 17: Upon SeV infection, the V and C accessory proteins with the leader have a role in preventing

the expression of specific cellular genes central to the host antiviral response as well as inflammatory

response.

50

Page 53: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

JOURNAL OF VIROLOGY, July 2003, p. 7903–7913 Vol. 77, No. 140022-538X/03/$08.00�0 DOI: 10.1128/JVI.77.14.7903–7913.2003Copyright © 2003, American Society for Microbiology. All Rights Reserved.

Sendai Virus Targets Inflammatory Responses, as Well as theInterferon-Induced Antiviral State, in a Multifaceted Manner

Laura Strahle,1 Dominique Garcin,1 Philippe Le Mercier,1 Joerg F. Schlaak,2†and Daniel Kolakofsky1*

Department of Genetics and Microbiology, University of Geneva School of Medicine, CMU,CH1211 Geneva, Switzerland,1 and Imperial Cancer Research Fund,

London WC2A 3PX, United Kingdom2

Received 13 February 2003/Accepted 17 April 2003

We have used cDNA arrays to compare the activation of various cellular genes in response to infection withSendai viruses (SeV) that contain specific mutations. Three groups of cellular genes activated by mutant SeVinfection, but not by wild-type SeV, were identified in this way. While some of these genes are well known inter-feron (IFN)-stimulated genes, others, such as those for interleukin-6 (IL-6) and IL-8, are not directly inducedby IFN. The gene for beta IFN (IFN-�), which is critical for initiating an antiviral response, was also specifi-cally activated in mutant SeV infections. The SeV-induced activation of IFN-� was found to depend on IFN reg-ulatory factor 3, and the activation of all three cellular genes was independent of IFN signaling. Mutations thatdisrupt four distinct elements in the SeV genome (the leader RNA, two regions of the C protein, and the Vprotein) all lead to enhanced levels of IFN-� mRNA, and at least three of these viral genes also appear to beinvolved in preventing activation of IL-8. Our results suggest that SeV targets the inflammatory and adaptiveimmune responses as well as the IFN-induced intracellular antiviral state by using a multifaceted approach.

Alpha/beta interferons (IFN-�/�) are cytokines that act in apleiotropic manner to limit viral replication and spread (2, 57).In fibroblasts (e.g., the bronchial epithelial target of manyparamyxoviruses), the product of the single IFN-� gene isdirectly induced by viral infection, and IFN-� feeds back ontocells in an autocrine manner to induce multiple IFN-� genesand in a paracrine manner to prime neighboring cells for theirpossible infection (60). Since most viruses induce IFN-� tosome extent, intracellular double-stranded RNA (dsRNA)generated from the viral genome is traditionally assumed to bethe common signature of virus replication that sets the IFNsystem in motion (22, 32). dsRNA is thought to induce theformation of an enhanceosome at the IFN-� promoter thatincludes IFN regulatory factor 3 (IRF-3) and NF-�B (amongother transcription factors) (65). IFNs induce a cellular statethat is nonconducive for viral replication by signaling throughtheir cell surface receptor, leading to the phosphorylation ofcytoplasmic STAT proteins and their nuclear translocation.IFN-�/� responses are regulated primarily via IFN-stimulatedgene (ISG) factor 3, a heterotrimeric transcription factor com-posed of STAT1, STAT2, and IRF-9 (p48). ISG factor 3 bindsto a DNA element (IFN-stimulated response element) in thepromoters of ISGs and activates their expression (7).

The extravasation of neutrophils, eosinophils, basophils, andmononuclear cells is the salient feature of the innate responseto microorganisms in the lung. Localized and systemic pro- andanti-inflammatory cytokines thus also play an important role in

the outcome of viral infection and pathogenicity of this organ(58). The CC chemokine interleukin-8 (IL-8) is secreted fromepithelial surfaces in a polar fashion during infection withpathogenic bacteria such as Salmonella enterica serovar Typhi-murium and sets up a subepithelial chemotactic gradient di-recting neutrophils and other immune cells to the site of in-fection (27). In polarized epithelial monolayers, S. entericaserovar Typhimurium-induced IL-8 expression is controlled viathe activation of the mitogen-activated protein kinase cascadeand I�B� kinase, followed by NF-�B translocation to the nu-cleus and production of IL-8 mRNA. IL-8 secretion by primaryhuman monocytes in response to dengue virus infection is alsotightly linked to NF-�B activation (3). Sendai virus (SeV) in-fection of human embryonic kidney 293 cells induces theexpression of the CXC chemokine RANTES in an IRF-3-and NF-�B-dependent manner (23, 41). NF-�B, like IRF-3,is found in the cytoplasm of unstimulated cells, retained in acomplex with the inhibitory I�B proteins. Upon stimulationwith many inducers, including dsRNA and virus infection, I�Bis rapidly phosphorylated and degraded, resulting in NF-�Brelease and translocation to the nucleus (30, 33).

Given the importance of the host innate immune response tovirus infection, viruses have, during their coevolution withcells, developed strategies to regulate cytokine synthesis andaction. SeV, a model paramyxovirus and respiratory pathogenof mice, is known to use its C protein to evade the host inter-feron response by at least two mechanisms. (i) C binds STAT1,preventing its activation in response to IFN, and the carboxylpart of the C protein (i.e., residues 24 to 204, or the Y proteins)is sufficient for this activity. A phenylalanine at position 170 ofC is also critical for blocking STAT1 activation (18, 59). (ii) Calso targets STAT1 for degradation, and the amino-terminalresidues of the C proteins (residues 1 to 23, which are absent

* Corresponding author. Mailing address: Department of Geneticsand Microbiology, University of Geneva School of Medicine, CMU, 9Ave de Champel, CH1211 Geneva, Switzerland. Phone: 41 22 3795657. Fax: 41 22 379 5702. E-mail: [email protected].

† Present address: Department of Gastroenterology and Hepatol-ogy, University of Essen, 45122 Essen, Germany.

7903

Page 54: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

in the Y proteins) are essential for this activity (reference 17and references therein).

This paper reports that SeVs carrying specific mutations inthe C gene, in contrast to wild-type SeV (SeV-wt), activateIL-8 and IFN-� expression as well as that of several ISGs. Ourresults suggest that the products of virtually all of the viralaccessory genes (C and V proteins and leader RNA) act toprevent the expression of these cellular genes that are centralto the overall host antiviral response.

MATERIALS AND METHODS

Cells and viruses. 2C4 cells (39), 2fTGH cells (48), and their derived cell linesU3A (45) and U5A and U5A�IFNAR (43) were obtained from IM Kerr (Im-perial Cancer Research Fund, London, United Kingdom) and grown in Dulbec-co’s modified Eagle’s medium supplemented with 10% fetal bovine serum in thepresence of the relevant maintenance drug (hygromycin at 250 �g/ml or G418 at400 �g/ml). The generation of recombinant SeV (rSeV) expressing alternate Cand V (and P) proteins is described elsewhere (8, 19, 37, 38). All SeV stocks weregrown in the allantoic cavities of 10-day-old embryonated chicken eggs. Viruspresent in the allantoic fluid was analyzed by sodium dodecyl sulfate (SDS)-polyacrylamide gel electrophoresis and Coomassie blue staining after virus pel-leting. Virus titers were determined by plaquing on LLC-MK2 cells.

Virus infections. Cells were infected at a multiplicity of infection of 20 inDulbecco’s modified Eagle’s medium containing 10% fetal bovine serum. Afteran absorption period of 1 to 2 h, the inoculum was removed and replaced withfresh medium.

Plasmids, transient transfections, and luciferase assay. IRF-3 (54), IRF-3�N(41), and IRF-3-ribozyme (67) were obtained from John Hiscott and Paula Pitha.pDsRed2, expressing red fluorescent protein (RFP), was from Clontech. Thereporter plasmid with the firefly luciferase gene under the control of the humanIFN-� promoter was described by King and Goodbourn (35) and is referred tohere as p�-IFN-fl-lucter. pTK-rl-lucter, used as a transfection standard, containsthe herpes simplex virus thymidine kinase promoter region upstream of theRenilla luciferase gene (Promega). For transfections, 100,000 cells were plated insix-well plates 20 h before transfection with 1 �g of p�-IFN-fl-lucter, 0.3 �g ofpTK-rl-lucter, 1 �g of IRF-3-expressing plasmid, and 6.9 �l of Fugene (Roche)according to the manufacturer’s instructions. At 24 h posttransfection, the cellswere (or were not) infected with SeV recombinants or treated with 50 �g ofpoly(I)-poly(C) (Sigma, St. Louis, Mo.) per ml. Twenty hours later, cells wereharvested and assayed for firefly and Renilla luciferase activities (dual-luciferasereporter assay system; Promega). Relative expression levels were calculated bydividing the firefly luciferase values by those of Renilla luciferase.

RNA extraction and quantification. Total RNA was extracted with Trizolreagent (Invitrogen), and two dilutions were electrophoresed on agarose-HCHOgels. The gels were stained with ethidium bromide, and the intensities of the 18Sand 28S rRNA bands were quantified by using the ChemiDoc System (Bio-Rad)and One-D-scan software. All samples were diluted to a final concentration of1 �g/�l so that their subsequent transcription into DNA, if not quantitative,would be little influenced by this parameter.

RT and real-time PCR via TaqMan. Ten microliters of total RNA was mixedwith 0.5 �g of random hexamer primer (Promega) and subjected to a reversetranscription (RT) reaction with Superscript enzyme (Gibco), as described by themanufacturer, in a total volume of 50 �l. Two microliters of each cDNA was thencombined with 1 �l of internal control (either 20� ribosomal 18S or humanglyceraldehyde-3-phosphate dehydrogenase [GAPDH] [Applied Biosystems]),11 �l of MasterMix (Eurogentec), 20 pmol (each) of forward and reverse prim-ers, and 4.4 pmol of TaqMan probe in a total volume of 22 �l. The followingprimers and probes (Eurogentec or Microsynth) were used: for the IFN-� gene,5�-CAGCAATTTTCAGTGTCAGAAGCT-5� (forward), 5�-TCATCCTGTCCTTGAGGCAGT-3� (reverse), and 5�-CTGTGGCAATTGAATGGGAGGCTTGA-3� (probe); for the IL-8 gene, 5�-CGGTGGCTCTCTTGGCAG-3� (forward),5�-TTAGCACTCCTTGGCAAAACT-3� (reverse), and 5�-CTTCCTGATTTCTGCAGCTCTGTGTGAAGGT-3� (probe); for the IL-6 gene, 5�-TCCAGGAGCCCAGCTATGAA-3� (forward), 5�-CCCAGGGAGAAGGCAACTG-3� (re-verse), and 5�-TCCTTCTCCACAAGCGCCTTCGG-3� (probe); for the 6-16gene, 5�-CCTGCTGCTCTTCACTTGCA-3� (forward), 5�-AGCCGCTGTCCGAGCTC-3� (reverse), and 5�-TGGAGGCAGGTAAGAAAAAGTGCTGCTCGG-3� (probe); for the N gene of SeVZ, 5�-GCAATAACGGTGTCGATCACG-3�(forward), 5�-GATCCTAGATTCCTCCTACCCCA-3� (reverse), and 5�-CGAAGATGACGATACCGCAGCAGTAGC-3� (probe); and for the N gene of

SeVM, 5�-CGAAGAGGATGATGCCGC-3� (forward), 5�-GGGTCATGTATCCTAAATCCTCGT-3� (reverse), and 5�-CAGCAGCTGGGATGGGAGGAAT-3� (probe). Real-time PCR was carried out in a 7700 sequence detector(Applied Biosystems, Foster City, Calif.).

Generation of customized cDNA arrays. Macroarrays were prepared as de-scribed previously (55). 5� IMAGE clones 0.5 to 0.8 kb in length were chosen andobtained from the Human Genome Mapping Project (Hinxton, United King-dom), plated onto L agar plates and grown overnight at 37°C. Single colonieswere picked and propagated overnight in Luria-Bertani medium containing 50�g of ampicillin per ml. Bacterial lysates were generated by 1:10 dilution indistilled water. From these lysates, inserts were amplified by PCR as describedabove. After purification (QIAquick PCR purification kit; Qiagen, Crawley,United Kingdom), PCR products were sequenced (ABI Prism; Applied Biosys-tems). PCR-amplified cDNAs were transferred into 96-well plates and spottedmanually onto dry nylon membranes (Hybond N�; Amersham Pharmacia, LittleChalfont, United Kingdom) in triplicates by using 96-pin replicators (NalgeNunc, Naperville, Ill.; V&P Scientific, San Diego, Calif.). Membranes were airdried, denatured by alkaline treatment, and then neutralized. The membraneswere again air dried and UV cross-linked prior to the experiment.

Generation of labeled cDNA, hybridization, washing of membranes and anal-ysis. Radiolabeled cDNA was generated from 10 �g of total RNA by RT with400 U of reverse transcriptase (Superscript II; Gibco) in the presence of 30 �Ciof [�-33P]dCTP. After RT, residual RNA was hydrolyzed by alkaline treatmentat 70°C for 20 min. For removal of unincorporated nucleotides, the cDNA waspurified by using G-50 columns (Amersham Pharmacia) according to the instruc-tions of the manufacturer. Before hybridization to the arrays, the labeled cDNAwas mixed with 50 �g of COT-DNA (Gibco) and 10 �g of poly(A) DNA (Sigma),denatured at 95°C for 5 min, and hybridized for 1 h to minimize nonspecificbinding. The cDNA was then added directly to the membranes, which had beenprehybridized in 20 ml of hybridization buffer for at least 30 min. The membraneswere hybridized for 16 h at 65°C in hybridization bottles (Amersham Pharmacia)in a rotary hybridization oven. After hybridization, the hybridization buffer wasdiscarded and replaced by 150 ml of washing buffer. The membranes werewashed once in 2� SSC (1� SSC is 0.15 M NaCl plus 0.015 M sodium citrate)–0.1% SDS, twice in 0.2� SSC–0.1% SDS, and once in 0.1� SSC–0.1% SDS for20 min each at 65°C. The membranes were then exposed to phoshorimagescreens for 48 h and scanned with a phosphorimager (Storm; Molecular Dynam-ics, Little Chalfont, United Kingdom). For analysis, images were analyzed withImageQuant (Molecular Dynamics). Further data analysis was performed withExcel (Microsoft).

IL-8 assay. Levels of IL-8 in supernatants and in cell lysates were determinedby a sandwich enzyme-linked immunosorbent assay with paired monoclonalantibodies (Pierce) as described by the manufacturer. The IL-8 concentrationswere normalized by using total protein levels in the lysate.

RESULTS

In our experiments we have used two strains (or lineages) ofSeV whose history is relevant to studies of virulence. Thenatural host of SeV has not been clearly identified, and thisvirus is sometimes referred to as murine parainfluenza virustype I because it efficiently infects mice, causes disease, andspreads readily to uninfected animals. However, there is novirologic or serologic evidence of SeV in wild mouse popula-tions (29). There are two known lineages of SeV, Z/H/Fushimiand Ohita M/Hamanatsu (12, 31, 53, 62). The nucleotide se-quences within each lineage are 99% identical, and they are89% identical between lineages. Z/H/Fushimi comes from vi-ruses isolated in the early 1950s after an epidemic of pneumo-nitis of newborn infants in Sendai, Japan (29, 56). These SeVshave been passaged extensively in eggs in various labs since the1950s, and they are moderately virulent for mice (50% lethaldose [LD50] 103 to 104 PFU). All of the SeVZ strains usedin this study (including the wt) are recombinants.

Ohita M (SeVM) and Hamanatsu, in contrast, are highlyvirulent (LD50, 102), low-egg-passage (nonrecombinant) vi-ruses isolated from two completely separate, very severe epi-demics of animal houses in Japan. This lineage is presumably

7904 STRAHLE ET AL. J. VIROL.

Page 55: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

closer to the virus in its natural (unknown) host, and it is knownthat SeV passage in eggs attenuates its virulence in mice. SeVM

grew poorly in cell culture, and a clear-plaque variant emergedthat was avirulent (LD50, �105) and contained only two aminoacid substitutions, CF170S and LE2050A (31). When placed in therSeVZ background, the CF170S mutation was found to accountfor most or all of the loss of virulence (15). This virus isreferred to here as SeVM-CF170S. SeVM-CF170S appeared toinitiate the infection of mice normally, but the infection waslimited to the first day. This was the first evidence that the Cgene, like the SeV V gene (34), was involved in countering hostinnate defenses. Although SeVZ-wt is attenuated relative toSeVM-wt in laboratory mice, rSeVZ-wt still replicates as effi-ciently in the monkey and chimpanzee models of human re-spiratory disease as human parainfluenza virus type 1 (56), thevirus which is most closely related to SeV and which is endemicin children.

We have used a cDNA array designed to study the humancell response to IFN-�/� (55) to monitor the effects of variousSeV infections on host mRNA levels. Around 150 genes ofinterest were selected from the UniGene database. Thesegenes comprise known ISGs and genes of intrinsic interestwhich might or might not be induced by IFNs in different cellsystems. They include genes involved in cell proliferation, im-mune responses, and the responses to a variety of cytokines(see Table 1 of reference 55). We compared matched sets ofSeV carrying two different mutations in the C gene (SeVM-CF170S and SeVZ-C�10-15), whose products interact withSTAT1 in different ways (to interfere with IFN signaling and toinduce STAT1 instability, respectively) (Fig. 1D). We also usedmatched sets of SeV carrying mutations in the viral replicationpromoters (SeVZ-GP1-42 and -GP31-42) that prevent apopto-sis and lead to persistent infections (the numbers refer to thenucleotides of the genomic promoter that have been replacedwith the equivalent sequences of the antigenomic promoter)(16, 20). Promoter mutations are thought to act via mutantleader RNAs that are abundantly transcribed from the geno-mic replication promoter and which bind to cellular RNA-binding proteins that regulate mRNA fate (28).

Upon infection with SeVM-wt or SeVZ-wt, many of thesemRNA levels remain unchanged (Fig. 1A to C). This lack ofresponse is presumably due in part to active SeV countermea-sures that neutralize the cell’s antiviral response (24). Uponinfection with the mutant viruses, the mRNA levels of 15 of the150 genes examined were elevated, and three patterns of geneactivation were seen (all values are triplicates, and a twofolddifference is very significant [55]). One series of genes (group1, nine genes) (Fig. 1A) is activated by SeV-CF170S infectionalone; these mRNA levels are unchanged in SeV-C�10-15 andSeV-GP1/31-42 infections. The CF170S substitution inactivatesthe ability of all four C proteins (C�, C, Y1, and Y2) to stablybind STAT1 and to interdict IFN signaling (18, 59). Accordingto this view, any of the four C proteins may function to preventthese mRNA levels from increasing during SeV infection (Fig.1D) (14). The IL-6 gene is the sole representative of group 2;it is activated by SeV-C�10-15 as well as SeV-CF170S, but notby the promoter mutants or the wt viruses (Fig. 1B). Accordingto this view, a second function of the C gene, specific to theNH2-terminal 23 amino acids present only in the longer C

proteins, is also required for SeV to prevent IL-6 activation.The third group, consisting of five genes, is activated by SeV-C�10-15 and SeV-GP1-42 infections as well as SeV-CF170S, butnot by SeV-GP31-42 or SeV-wt infections (Fig. 1C). Appar-ently, a third function provided specifically by the first 30 nu-cleotides (nt) of the genomic promoter (or leader RNA) is alsorequired to prevent activation of genes such as that of IL-8(Fig. 1C). This third function is not the ability of mutant leaderRNA to bind TIAR, a host RNA-binding protein important forvirus-induced apoptosis, as this occurs with SeV-GP31-42 aswell (28). In summary, comparative analysis of host gene acti-vation with SeV with specific mutations has identified threegroups of cellular genes that respond differently to SeV infec-tion.

Real-time RT-PCR estimations of mRNA levels. IFN signal-ing through the JAK/STAT pathway activates many ISGs (suchas 6-16, PKR, etc.) that collectively contribute to the cellularantiviral response. SeVs that interdict IFN signaling wouldtherefore also prevent the activation of these ISGs. Whilemany of the genes activated by the mutant SeVs are wellknown ISGs, IL-6 and IL-8 are known to be non-ISGs; treat-ment of 2fTGH cells with 1,000 IU of IFN-� does not increaseIL-6 or IL-8 mRNA levels over those of the untreated control(55). We therefore continued our study of selected host genesactivated by SeV by real-time RT-PCR (TaqMan), a methodthat is more quantitative than DNA arrays. We first examinedIL-8 (a chemokine) and IFN-�, an early host response proteinwhose gene was absent in the DNA array. 2C4 cells (a 2fTGH-derived cell line) were infected with 20 PFU of the variousSeVs per cell, and the levels of various host mRNAs weredetermined, including that of GADPH as an internal control(see Materials and Methods). All four SeVs (SeVM-wt, CF170S,SeVZ-wt, and C�10-15) grow relatively well in 2C4 cells, asindicated by their accumulated N mRNAs (Fig. 2 and 3) or Nproteins (data not shown); however, the SeVM-wt infectionsaccumulated ca. 2-fold less N mRNA than the three other SeVinfections (Fig. 2C). The relative levels of virus replication arepresumably important in stimulating the host response, andSeVM-wt infections might therefore be expected to activateIL-8 and IFN-� less strongly than the other SeVs for thisreason alone. We found that IL-8 and IFN-� mRNA levelswere elevated �20-fold in SeVM-CF170S versus SeVM-C-wtinfections. A strong difference was also found between SeVZ-C�10-15 and SeVZ-C-wt infections, where N mRNAs had ac-cumulated identically (Fig. 2). Moreover, IL-8 and IFN-�mRNA levels were also elevated in other SeVM-CF170S versusSeVM-C-wt infections of 2C4 cells, where N mRNAs had ac-cumulated identically (Fig. 3). Thus, specific mutations in twodifferent regions of the SeV C proteins lead to increased acti-vation of IL-8 and IFN-�. In all cases, IL-8 and IFN-� mRNAlevels were more strongly increased by SeVM-CF170S than bySeVZ-C�10-15.

We also examined the effects of various SeV infections ofcells transfected with plasmids expressing IRF-3 (or RFP as aneutral control), and in some cases the cells were also treatedwith 50 �g of poly(I)-poly(C) (dsRNA) per ml. Elevated IRF-3levels should enhance the antiviral response of the cells to SeVinfection, and the combined treatment is thought to approxi-mate a virus infection in itself and should potentiate the anti-viral response. This appears to be so, as the levels of IL-6, IL-8,

VOL. 77, 2003 SENDAI VIRUS TARGETING OF INFLAMMATORY RESPONSES 7905

Page 56: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

FIG. 1. Comparison of host mRNA levels in 2fTGH cells infected with various SeVs. (A to C) Parallel cultures of 2fTGH cells were infectedwith 20 PFU of the various SeVs per cell. Total cytoplasmic RNA was prepared with Trizol at 24 hpi, and equal amounts (10 �g) were used asa template for oligo(dT)-primed [33P]cDNA synthesis. The [33P]cDNA was annealed to triplicate DNAs arrayed on nylon membranes, and the[33P]cDNA bound was quantitated in a PhosphorImager. The graphs show the fold increase in each mRNA relative to the mock control. (D)Schematic representation of the viral mutations and their effects on host gene activation. The C proteins are shown as two telescoping boxesrepresenting the longer (C� and C) and shorter (Y1 and Y2) C proteins, whose activities during infection, and the mutations investigated, areindicated. The promoter mutation GP42 is thought to exert its effect via mutant leader (Le) RNA. The presumed requirement for the various wtgenetic elements to prevent host gene activation is shown. The names of the mutant SeVs used are also indicated. Instab., instability; sig., signaling.

7906 STRAHLE ET AL. J. VIROL.

Page 57: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

and IFN-� mRNAs induced by SeV infection in general in-creased with increasing proresponse treatment (i.e., RFP,IRF-3, and IRF-3 plus dsRNA) (Fig. 3). IRF-3 overexpression(Fig. 3A to C, IRF-3 versus RFP) enhanced IFN-� and IL-6activation by SeV but did not enhance that of IL-8. The addi-tional dsRNA treatment had little effect on further enhancingIFN-� and IL-6 mRNA levels but strongly enhanced IL-8mRNA levels. These differences in the enhancement of IL-6and -8 and IFN-� activation upon treatment with IRF-3 with orwithout dsRNA presumably reflect different activation path-ways in response to SeV infection. It is possible that IL-8activation requires both IRF-3 and dsRNA, whereas IRF-3 issufficient for IL-6 and IFN-� activation.

SeV activation of IL-6, IL-8, and IFN-� is independent ofIFN signaling. 2fTGH human fibrosarcoma cells were chosenfor these experiments because sublines defective in specificcomponents of the IFN signaling system have been generatedfrom these cells by X irradiation (48). U5A cells, for example,are defective in the IFN-�/� receptor 2 chain, which is essentialfor IFN-�/� signaling, and these cells have been restored toIFN sensitivity by complementation with the IFNAR2 gene(U5A�IFNAR2 cells) (43). Even though IL-6 and -8 are notactivated upon simple IFN treatment of uninfected cells, IFNsecreted during SeV infection may act differently, as additionalsignaling pathways are being induced by the virus infection.Moreover, CF170S (in contrast to C�10-15) does not preventIFN signaling, and it is important to know whether this phe-notype is responsible for the activation of IL-6 and -8. Wetherefore examined the various SeV infections of U5A as wellU5A�IFNAR2 cells to determine whether activation of IL-6and IFN-� by SeV-CF170S and SeV-C�10-15 required IFNsignaling. We also examined the activation of the 6-16 gene, aknown ISG, as a positive control (10). As shown in Fig. 4, bothSeV-CF170S and SeV-C�10-15 activated IL-6, IL-8, and IFN-�in U5A cells relative to SeV-wt infection. The IFNAR2-com-

plemented cell line yielded similar results, except that theactivation of these genes was paradoxically reduced in U5A�IFNAR2 cells relative to U5A cells. In contrast to the case forIL-6, IL-8, and IFN-�, little or no activation of 6-16 occurredin SeV-C mutant-infected U5A cells, whereas a modest acti-vation was evident in U5A�IFNAR2 cells. Moreover, 6-16 wasthe only mRNA whose levels in U5A�IFNAR2 cells exceededthose in U5A cells. The activation of IL-6, IL-8, and IFN-�during SeV infection, in contrast to that of 6-16, is thus largelyindependent of IFN signaling.

STAT1-defective U3A cells. The SeV C proteins interactwith STAT1 in two ways (Fig. 1D). C and STAT1 form a stablecomplex in vitro and during SeV infection, and this complex isassociated with a loss of IFN signaling. These events areblocked by the CF170S mutation but not by C�10-15. Theshorter Y proteins are also active in this respect. The longer Cproteins alone also induce STAT1 instability, and in contrast totheir effects on IFN signaling, this effect does not require F170(Fig. 1D). To examine whether SeV-C mutant-induced activa-tion of IL-6, IL-8, and IFN-� requires STAT1, U3A cells,which are known to be defective for STAT1, were examined(45). However, we were unable to examine the companionU3A�STAT1 cells, as these cells were found to have lostSTAT1 expression. Moreover, attempts to recomplement U3Acells with STAT1 failed (data not shown).

The results of the U3A cell infections are shown in Fig. 5. Asbefore, IL-6 and -8 and IFN-� mRNA levels were all clearlyincreased in SeV-CF170S infections relative to SeV-wt infec-tions. However, in contrast to the case for U5A and 2fTGHcells, SeV-C�10-15 infections did not contain enhancedmRNA levels relative to SeV-wt infections. Given that STAT1is probably not the only gene that has been lost in U3A cellsthat have been X irradiated (which randomly destroys chro-mosomal DNA), we can conclude only that enhanced mRNAlevels due to SeV-C�10-15 infection require STAT1 and/or

FIG. 1—Continued.

VOL. 77, 2003 SENDAI VIRUS TARGETING OF INFLAMMATORY RESPONSES 7907

Page 58: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

another, unknown gene, whereas activation due to SeV-CF170S

requires neither STAT1 nor any other gene destroyed in U3Acells.

IL-8 gene activation and IL-8 secretion. IL-8 is not known toact intracellularly. We therefore examined whether the SeV-induced IL-8 gene activation in U5A/U5A�IFNAR2 cells also

led to increased IL-8 protein synthesis and secretion. WhenIL-8 protein levels in cytoplasmic extracts of the various SeV-infected cells were examined, they were found to roughly mir-ror the mRNA levels (Fig. 4E). However, when the culturesupernatants were examined, a somewhat different result wasfound (Fig. 4F). Whereas the increased IL-8 mRNA level ofSeV-C�10-15-infected U5A cells was accompanied by stronglyincreased IL-8 secretion, that of U5A�IFNAR2 cells led toonly a modest increase in IL-8 secretion. Moreover, the in-

FIG. 2. Effects of SeV C gene mutations on IFN-� and IL-8 mRNAlevels during infection. Parallel cultures of 2C4 cells were infected (ornot) in triplicate for 24 h with 20 PFU of the various SeVs per cell.Total cytoplasmic RNA was prepared from each culture, and the sameamount of RNA (ca. 1 �g) was transcribed into cDNA with randomhexadeoxynucleotides and murine leukemia virus reverse transcrip-tase. The relative amounts of IFN-� and IL-8 gene sequences, relativeto that of GADPH as an internal control, were determined by real-time PCR (see Materials and Methods). The average levels of themRNAs and their deviations in the triplicate infections are shown. Therelative SeV N mRNA levels of the SeVZ-C-wt and -C�10-15 infec-tions were also determined (SeVZ and SeVM are 10% different insequence, and their detection requires different primers and probes).

FIG. 3. Effects of IRF-3 and dsRNA treatment on SeV-inducedhost gene activation. Parallel cultures of 2C4 cells were transfectedwith pRFP or pIRF-3. The cultures were then infected (or not) with 20PFU of the various SeVs per cell at 20 h posttransfection, and some ofthe cultures were also treated with 50 �g of poly(I)-poly(C) per ml.The cells were harvested at 24 hpi. The relative amounts of IFN-�,IL-6, and IL-8 gene sequences present were determined as describedfor Fig. 2.

7908 STRAHLE ET AL. J. VIROL.

Page 59: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

creased IL-8 mRNA levels in either SeV-CF170S-infected cellline did not lead to clearly increased IL-8 secretion.

IL-8 expression can be controlled at both the transcriptionaland posttranscriptional levels. In polarized epithelial monolay-ers, S. enterica serovar Typhimurium-induced IL-8 secretionrequires not only the activation of NF-�B and production of

IL-8 mRNA but also the activation of the small, Rho familyGTPases Cdc42 and Rac1, which regulate endocytic proteintraffic from the Golgi network to the basolateral surface of thecell. In the absence of Cdc42 or Rac1 function, IL-8 mRNAlevels increase in response to bacterial infection but IL-8 is notsecreted (3), similar to the case for our SeV-CF170S-infected

FIG. 4. SeV infection of cells defective for the IFN receptor and their complemented pseudo-wt derivatives. (A to D) Parallel cultures of U5Aor U5A�IFNAR2 cells were infected (or not) in duplicate with 20 PFU of the various SeVs per cell for 24 h. The relative amounts of IFN-�, IL-6,IL-8, and 6-16 mRNAs present were determined as described for Fig. 2. (E and F) Equal samples of the culture supernatants (F) or cytoplasmicextracts of the cultures (E) were analyzed for IL-8 protein levels by enzyme-linked immunosorbent assay.

VOL. 77, 2003 SENDAI VIRUS TARGETING OF INFLAMMATORY RESPONSES 7909

Page 60: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

U5A cells. The requirement for Cdc42 and Rac1 activation,moreover, is cell type dependent (9). The CF170S and C�10-15mutations thus appear to affect IL-8 secretion differently.

SeV prevents IFN-� gene activation in several ways. Themanner in which IFN-� transcription is induced by virus infec-tion is well studied, and the activation of IRF-3 is central to thisprocess. IRF-3 is expressed constitutively and is found in thecytoplasm in an inactive, unphosphorylated state. Upon virusinfection or dsRNA treatment of cells, IRF-3 is phosphory-lated by an unknown kinase and translocates to the nucleus,

where, together with other transcription factors such as NF-�B(which is itself also directly activated by virus infection ordsRNA), it activates IFN-� transcription (41, 65, 68). Phos-phorylation of IRF-3 after viral infection is the first step in theactivation of a gene program that includes a positive feedbackloop of IFN-�/� and IRF family members (60). The resultsdescribed above suggest that the SeV C gene encodes func-tions that prevent virus-induced IFN-� transcription (directlyor indirectly). During the course of this work, it was reportedthat the SeV V protein, as well as the V proteins of therubulaviruses SV5 and hPIV2, also prevented IFN-� transcrip-tion (26, 50, 64).

Given that SeV appears to use two viral genes (C and V) toneutralize IFN-� expression, we have examined a broaderpanel of mutant rSeV infections for their relative activation ofthe IFN-� promoter compared to that of dsRNA treatment.Besides SeV-CF170S and C�10-15, we examined two promotermutants, SeV-AGP55, in which the first 55 nt of the antigeno-mic promoter is replaced with the equivalent leader sequencesof the genomic promoter (38). SeV-AGP55 transcribes leaderRNA from both promoters (and no trailer RNA). The con-verse SeV-GP48 has the first 48 nt of the genomic promoterreplaced with the equivalent trailer sequences, and SeV-GP48transcribes basically trailer RNA from both promoters (and noleader RNA; GP48 and GP1-42 are identical in this respect[data not shown]) (19, 20). Finally, we examined SeV-V�/W��, which contains a stop codon at the beginning of the Vopen reading frame (ORF), such that edited V mRNAs aretranslated into W-like proteins, and specifically no V protein isexpressed (8).

2fTGH cells were transfected with a reporter plasmid inwhich luciferase is controlled by the IFN-� promoter (pIFN�-luciferase). To determine whether virus-induced IFN-� tran-scription required IRF-3 activation, the cells were cotransfect-ed with either a dominant-negative mutant of IRF-3 (IRF-3�N),an anti-IRF-3 ribozyme, or an empty control plasmid (seeMaterials and Methods). The transfected cultures were theninfected (or not) with the various SeVs (or treated withdsRNA) 24 h later and were harvested at 20 h postinfection(hpi). As shown in Fig. 6, with the notable exception of SeV-AGP55, all of the mutant SeV infections induced the reportermore strongly than SeV-wt and as well as dsRNA treatment. Inall cases, the coexpression of IRF-3�N or an anti-IRF-3 ri-bozyme prevented the SeV-induced expression of the reporter.Thus, mutations in two regions of the C protein that carry outdifferent functions, as well as the loss of leader RNA expres-sion or the expression of the V protein, all lead to IFN-�promoter activation in an IRF-3-dependent manner. Overex-pression of the W protein cannot compensate for the lack of Vprotein, so the highly conserved Cys-rich carboxyl domain of Vis specifically required. Only the loss of trailer RNA expression(SeV-AGP55) did not result in IFN-� activation.

Leader and trailer RNAs, the promoter-proximal productsof viral RNA synthesis, are AU rich and are thought to bind tocellular RNA-binding proteins that bind AU-rich elements(28). In contrast to leader RNA, trailer RNA is expressedrelatively late in infection (upon antigenome accumulation)and would not be expected to counteract immediate-early re-sponse genes. However, many of the other viral products thatdo not have a dedicated essential role in the replication ma-

FIG. 5. SeV infection of cells defective for STAT1. Parallel cul-tures of U3A cells were infected (or not) in duplicate with 20 PFU ofthe various SeVs per cell for 24 h. The relative amounts of IFN-�, IL-6,and IL-8 mRNAs present were determined as described for Fig. 2.

7910 STRAHLE ET AL. J. VIROL.

Page 61: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

chinery per se (C and V proteins and leader RNA) appear tobe pressed into service to coordinately prevent IFN-� activa-tion.

DISCUSSION

We have used cDNA arrays to compare the activation ofvarious cellular genes in response to infection with SeVs thatcontain mutations in their C proteins or leader regions. Threegroups of cellular genes were identified in this way (Fig. 1). Ofthe mutant SeV examined by DNA arrays, only the leader(31-42) mutation failed to activate any of the cellular genes relativeto SeV-wt, and consistent with this failure, SeV-GP31-42 wasthe only one of the four mutant SeVs not to have lost virulencein mice (M. Itoh, unpublished data). The CF170S mutation isassociated with a ca. 5-log-unit increase in LD50 (31), and thismutation appears to be the most important in cell cultureinfections as well as in mice (15). This mutation activates IL-6,

IL-8, and IFN-� more strongly than C�10-15, and only thismutation activates all three groups of cellular genes that re-spond to SeV infection.

The IFN-� gene is both a primary response gene and anISG, and it plays a central role in initiating the IFN-inducedantiviral response. This is the first report that the SeV C pro-teins and leader RNA counteract the expression of this keyprimary response gene, and it confirms that the report of Pooleet al. (50) that the product of our hemagglutinin-tagged V geneis active in this respect. As expected, activation of IFN-� re-quired activation of IRF-3 (Fig. 6) and was independent of IFNsignaling (Fig. 4), but the details of how this occurs remain tobe elucidated. We have also provided evidence that the SeV Cproteins and leader RNA counteract the expression of thechemokine IL-8. Infections by other viruses, e.g., respiratorysyncytial virus (RSV) (44), dengue virus (3), hepatitis C virus(49), and human immunodeficiency virus type 1 (46, 52), areknown to induce IL-8 secretion, as do infections by bacteriaand parasites, including Mycobacterium tuberculosis (66). In arelated vein, cytomegalovirus, a large DNA virus, encodes achemokine receptor that may facilitate virus replication (13),and human herpesvirus 8/Kaposi’s sarcoma virus carries fourORFs whose products are related to chemokines (42). Viralmodulation of chemokine expression presumably representsone aspect of the continuous battle between viral parasites andantiviral, inflammatory, and immune responses of the host.SeV infection has been reported to induce the CXC chemo-kine RANTES via the activation of IRF-3 and NF-�B (23, 40).IL-8 may not have been noticed in these earlier studies, be-cause SeV-wt induces very little IL-8 (Fig. 2). Our resultssuggest that SeV targets the inflammatory and adaptive im-mune responses (IL-6 and IL-8) as well as the IFN-inducedintracellular antiviral state (IFN-� and STAT1). As IFN-� andIL-8 transcription both depend on NF-�B activation, SeV maytarget this key transcription factor as well.

The leader(1-42), C�10-15, CF170S, and V�/W�� mutationsappear to disrupt four distinct elements in the SeV program tocounteract the cellular antiviral response. The facts that theyall lead to enhanced levels of IFN-� mRNA and that at leastthree of them increase IL-8 mRNA levels suggest that SeVemploys a multifaceted approach to inhibit viral clearance byinflammatory cells as well as to prevent the IFN-induced an-tiviral state, sometimes using the same viral macromoleculesdue to its limited coding capacity. The best-studied example ofparamyxovirus-induced activation of IL-8 is that of RSV (21,69). The IL-8 promoter in A549 cells can be induced by RSVinfection in at least three distinct pathways: via tumor necrosisfactor alpha (which requires only an intact NF-�B bindingsite), directly by intracellular RSV replication (which also re-quires other transcription factor binding sites) (5), and via theinteraction of the viral F protein with Toll-like receptor 4 (inwhich IRF-3 plays an important role) (36). Measles virus Hprotein interaction with Toll-like receptor 2 also activates IL-8(1). If all three parallel cellular pathways for IL-8 expressionoperate during SeV infection of 2fTGH cells, several differentSeV products will be required to effectively prevent IL-8 acti-vation (Fig. 1D).

Rubulaviruses do not express C proteins, but their V pro-teins have recently been found to prevent IFN-� expression bypreventing the activation of IRF-3 and NF-�B, as well as in-

FIG. 6. Effect of SeV infection on IFN-� promoter reporter geneexpression. Parallel cultures of 2fTGH cells were transfected with amixture of pINF�-fl-lucter and pTK-rl-lucter along with pIRF-3-�N,pAnti-IRF-3-ribozyme, or an empty plasmid (Ctrl), as indicated inMaterials and Methods. Duplicate cultures were infected (or not) with20 PFU of the various SeVs per cell or treated with dsRNA andharvested at 20 hpi. The relative activities of the fl- and rl-luciferaseswere determined. Error bars indicate standard deviations.

VOL. 77, 2003 SENDAI VIRUS TARGETING OF INFLAMMATORY RESPONSES 7911

Page 62: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

ducing the degradation of STAT1 or STAT2 (26, 50). TheRubulavirus V proteins thus also counteract more than onearm of the innate antiviral response. The versatility of theseviral gene products continues to surprise us. The SeV C pro-teins have been more intensively studied than SeV V proteinor leader RNA. Like the influenza A virus NS1 protein (6, 63)and hepatitis C virus NS5A protein (49), the SeV C proteinsare pleiotropic polypeptides that have multiple activities dur-ing infection, presumably due to their interaction with variousviral and cellular proteins. Their multiple functions, deci-phered in large part via C gene mutations, include (i) stimu-lation of viral RNA synthesis early in infection (SeV-C�/C�

infections exhibit a 10-h delay in the accumulation of viralproducts) (37); (ii) inhibition of viral RNA synthesis in a pro-moter-specific manner late in infection, by interacting with theP4-L vRdRP (this selective inhibition may promote the switchfrom mRNA synthesis to genome replication and increase thefidelity of vRdRP promoter recognition) (4, 61); (iii) a role invirion assembly, possibly by interaction with the matrix (M)protein (SeV-4C� particles are poorly infectious and amor-phic) (25); (iv) interaction with STAT1 in two separate ways, toinhibit IFN signaling and to induce STAT1 instability (17); and(v) inhibition of the IRF-3-dependent activation of IFN-� andthe activation of IL-8 expression in an IFN signaling-indepen-dent manner (this work).

How C interacts specifically with all of its viral and cellularpartners remains an enigma and is reminiscent of acidic acti-vation domains of transcription factors that interact with mul-tiple partners. Acidic activation domains are “natively disor-dered” (11, 51), and this property apparently allows them tobind different surfaces with high specificity (multiple inducedfits) and limited stability. The NH2-terminal portion of themeasles virus P protein that contains the overlapping C proteinORF is, in fact, a recent example of such natively disorderedproteins, in accordance with the prediction of algorithms thatdetect unstructured regions (47). By using the same algorithms(PONDR), the SeV C protein is strongly predicted to be na-tively disordered, and this property is shared with the commonNH2-terminal portions of rubulavirus V, I, and P proteins (17,26). The C�10-15 deletion, moreover, is in a region of C withthe highest prediction of disorder. It will be of interest toexamine whether purified SeV C proteins are indeed nativelydisordered.

ACKNOWLEDGMENTS

We thank Ian Kerr (London, United Kingdom) for useful discus-sions and for numerous cell lines, Paula Pitha (Baltimore, Md.) andJohn Hiscott (Montreal, Canada) for the IRF-3 reagents, and theJerome Pugin lab (University of Geneva) for help with the IL-8 proteinassays.

ADDENDUM IN PROOF

Fujii et al. (Y. Fujii, T. Sakaguchi, K. Kiyotani, C. Huang, N.Fukuhara, Y. Egi, and T. Yoshida, J. Virol. 76:8540–8547,2002) have shown that mutations in the leader region specifi-cally attenuate virus virulence in mice.

REFERENCES

1. Bieback, K., E. Lien, I. M. Klagge, E. Avota, J. Schneider-Schaulies, W. P.Duprex, H. Wagner, C. J. Kirschning, V. ter Meulen, and S. Schneider-

Schaulies. 2002. Hemagglutinin protein of wild-type measles virus activatestoll-like receptor 2 signaling. J. Virol. 76:8729–8736.

2. Biron, C. A., and G. C. Sen. 2001. Interferons and other cytokines, p.321–349. In D. M. Knipe and P. M. Howley (ed.), Fields virology. Lippincott,Williams, and Wilkins, Philadelphia, Pa.

3. Bosch, I., K. Xhaja, L. Estevez, G. Raines, H. Melichar, R. V. Warke, M. V.Fournier, F. A. Ennis, and A. L. Rothman. 2002. Increased productionof interleukin-8 in primary human monocytes and in human epithelialand endothelial cell lines after dengue virus challenge. J. Virol. 76:5588–5597.

4. Cadd, T., D. Garcin, C. Tapparel, M. Itoh, M. Homma, L. Roux, J. Curran,and D. Kolakofsky. 1996. The Sendai paramyxovirus accessory C proteinsinhibit viral genome amplification in a promoter-specific fashion. J. Virol.70:5067–5074.

5. Casola, A., R. P. Garofalo, M. Jamaluddin, S. Vlahopoulos, and A. R.Brasier. 2000. Requirement of a novel upstream response element in respi-ratory syncytial virus-induced IL-8 gene expression. J. Immunol. 164:5944–5951.

6. Chen, Z., and R. M. Krug. 2000. Selective nuclear export of viral mRNAs ininfluenza-virus-infected cells. Trends Microbiol. 8:376–383.

7. Darnell, J. E. J. 1997. STATs and gene regulation. Science 277:1630–1635.8. Delenda, C., S. Hausmann, D. Garcin, and D. Kolakofsky. 1997. Normal

cellular replication of Sendai virus without the trans-frame, nonstructural Vprotein. Virology 228:55–62.

9. Denk, A., M. Goebeler, S. Schmid, I. Berberich, O. Ritz, D. Lindemann, S.Ludwig, and T. Wirth. 2001. Activation of NF-kappa B via the Ikappa Bkinase complex is both essential and sufficient for proinflammatory geneexpression in primary endothelial cells. J. Biol. Chem. 276:28451–28458.

10. Der, S. D., A. Zhou, B. R. Williams, and R. H. Silverman. 1998. Identificationof genes differentially regulated by interferon alpha, beta, or gamma usingoligonucleotide arrays. Proc. Natl. Acad. Sci. USA 95:15623–15628.

11. Feldmann, H., and H. D. Klenk. 1996. Marburg and Ebola viruses. Adv.Virus Res. 47:1–52.

12. Fujii, Y., K. Kiyotani, T. Yoshida, and T. Sakaguchi. 2001. Conserved andnon-conserved regions in the Sendai virus genome: evolution of a genepossessing overlapping reading frames. Virus Genes 22:47–52.

13. Gao, J. L., and P. M. Murphy. 1994. Human cytomegalovirus open readingframe US28 encodes a functional beta chemokine receptor. J. Biol. Chem.269:28539–28542.

14. Garcin, D., J. Curran, M. Itoh, and D. Kolakofsky. 2001. Longer and shorterforms of Sendai virus C proteins play different roles in modulating thecellular antiviral response. J. Virol. 75:6800–6807.

15. Garcin, D., M. Itoh, and D. Kolakofsky. 1997. A point mutation in the Sendaivirus accessory C proteins attenuates virulence for mice, but not virus growthin cell culture. Virology 238:424–431.

16. Garcin, D., P. Latorre, and D. Kolakofsky. 1999. Sendai virus C proteinscounteract the interferon-mediated induction of an antiviral state. J. Virol.73:6559–6565.

17. Garcin, D., J. B. Marq, S. Goodbourn, and D. Kolakofsky. 2003. The amino-terminal extensions of the longer Sendai virus C proteins modulate pY701-Stat1 and bulk Stat1 levels independently of interferon signaling. J. Virol.77:2321–2329.

18. Garcin, D., J. B. Marq, L. Strahle, P. Le Mercier, and D. Kolakofsky. 2002.All four Sendai virus C proteins bind Stat1, but only the larger forms alsoinduce its mono-ubiquitination and degradation. Virology 295:256–265.

19. Garcin, D., T. Pelet, P. Calain, L. Roux, J. Curran, and D. Kolakofsky. 1995.A highly recombinogenic system for the recovery of infectious Sendaiparamyxovirus from cDNA: generation of a novel copy-back nondefectiveinterfering virus. EMBO J. 14:6087–6094.

20. Garcin, D., G. Taylor, K. Tanebayashi, R. Compans, and D. Kolakofsky.1998. The short Sendai virus leader region controls induction of pro-grammed cell death. Virology 243:340–353.

21. Garofalo, R., M. Sabry, M. Jamaluddin, R. K. Yu, A. Casola, P. L. Ogra, andA. R. Brasier. 1996. Transcriptional activation of the interleukin-8 gene byrespiratory syncytial virus infection in alveolar epithelial cells: nuclear trans-location of the RelA transcription factor as a mechanism producing airwaymucosal inflammation. J. Virol. 70:8773–8781.

22. Geiss, G., G. Jin, J. Guo, R. Bumgarner, M. G. Katze, and G. C. Sen. 2001.A comprehensive view of regulation of gene expression by double-strandedRNA-mediated cell signaling. J. Biol. Chem. 276:30178–30182.

23. Genin, P., M. Algarte, P. Roof, R. Lin, and J. Hiscott. 2000. Regulation ofRANTES chemokine gene expression requires cooperativity between NF-kappa B and IFN-regulatory factor transcription factors. J. Immunol. 164:5352–5361.

24. Goodbourn, S., L. Didcock, and R. E. Randall. 2000. Interferons: cell sig-nalling, immune modulation, antiviral response and virus countermeasures.J. Gen. Virol. 81:2341–2364.

25. Hasan, M. K., A. Kato, M. Muranaka, R. Yamaguchi, Y. Sakai, I. Hatano, M.Tashiro, and Y. Nagai. 2000. Versatility of the accessory C proteins of Sendaivirus: contribution to virus assembly as an additional role. J. Virol. 74:5619–5628.

26. He, B., R. G. Paterson, N. Stock, J. E. Durbin, R. K. Durbin, S. Goodbourn,

7912 STRAHLE ET AL. J. VIROL.

Page 63: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

R. E. Randall, and R. A. Lamb. 2002. Recovery of paramyxovirus simianvirus 5 with a V protein lacking the conserved cysteine-rich domain: themultifunctional V protein blocks both interferon-beta induction and inter-feron signaling. Virology 303:15–32.

27. Hobert, M. E., K. A. Sands, R. J. Mrsny, and J. L. Madara. 2002. Cdc42and Rac1 regulate late events in Salmonella typhimurium-induced inter-leukin-8 secretion from polarized epithelial cells. J. Biol. Chem. 277:51025–51032.

28. Iseni, F., D. Garcin, M. Nishio, N. Kedersha, P. Anderson, and D. Kolakof-sky. 2002. Sendai virus trailer RNA binds TIAR, a cellular protein involvedin virus-induced apoptosis. EMBO J. 21:5141–5150.

29. Ishida, N., and M. Homma. 1978. Sendai virus. Adv. Virus Res. 23:349–383.30. Israel, A. 2000. The IKK complex: an integrator of all signals that activate

NF-kappaB? Trends Cell Biol. 10:129–133.31. Itoh, M., Y. Isegawa, H. Hotta, and M. Homma. 1997. Isolation of an

avirulent mutant of Sendai virus with two amino acid mutations from a highlyvirulent field strain through adaptation to LLC-MK2 cells. J. Gen Virol.78:3207–3215.

32. Jacobs, B. L., and J. O. Langland. 1996. When two strands are better thanone: the mediators and modulators of the cellular responses to double-stranded RNA. Virology 219:339–349.

33. Karin, M., and Y. Ben Neriah. 2000. Phosphorylation meets ubiquitination:the control of NF-�B activity. Annu. Rev. Immunol. 18:621–663.

34. Kato, A., K. Kiyotani, K. Sakai, T. Yoshida, and Y. Nagai. 1997. The para-myxovirus, Sendai virus, V protein encodes a luxury function required forviral pathogenesis. EMBO J. 16:578–685.

35. King, P., and S. Goodbourn. 1994. The beta-interferon promoter responds topriming through multiple independent regulatory elements. J. Biol. Chem.269:30609–30615.

36. Kurt-Jones, E. A., L. Popova, L. Kwinn, L. M. Haynes, L. P. Jones, R. A.Tripp, E. E. Walsh, M. W. Freeman, D. T. Golenbock, L. J. Anderson, andR. W. Finberg. 2000. Pattern recognition receptors TLR4 and CD14 mediateresponse to respiratory syncytial virus. Nat. Immunol. 1:398–401.

37. Latorre, P., T. Cadd, M. Itoh, J. Curran, and D. Kolakofsky. 1998. Thevarious Sendai virus C proteins are not functionally equivalent and exertboth positive and negative effects on viral RNA accumulation during thecourse of infection. J. Virol. 72:5984–5993.

38. Le Mercier, P., D. Garcin, S. Hausmann, and D. Kolakofsky. 2002. Am-bisense Sendai viruses are inherently unstable but are useful to study viralRNA synthesis. J. Virol. 76:5492–5502.

39. Lillemeier, B. F., M. Koster, and I. M. Kerr. 2001. STAT1 from the cellmembrane to the DNA. EMBO J. 20:2508–2517.

40. Lin, R., C. Heylbroeck, P. Genin, P. M. Pitha, and J. Hiscott. 1999. Essentialrole of interferon regulatory factor 3 in direct activation of RANTES che-mokine transcription. Mol. Cell. Biol. 19:959–966.

41. Lin, R., C. Heylbroeck, P. M. Pitha, and J. Hiscott. 1998. Virus-dependentphosphorylation of the IRF-3 transcription factor regulates nuclear translo-cation, transactivation potential, and proteasome-mediated degradation.Mol. Cell. Biol. 18:2986–2996.

42. Liu, C., Y. Okruzhnov, H. Li, and J. Nicholas. 2001. Human herpesvirus 8(HHV-8)-encoded cytokines induce expression of and autocrine signaling byvascular endothelial growth factor (VEGF) in HHV-8-infected primary-effusion lymphoma cell lines and mediate VEGF-independent antiapoptoticeffects. J. Virol. 75:10933–10940.

43. Lutfalla, G., S. J. Holland, E. Cinato, D. Monneron, J. Reboul, N. C. Rogers,J. M. Smith, G. R. Stark, K. Gardiner, K. E. Mogensen, et al. 1995. MutantU5A cells are complemented by an interferon-alpha beta receptor subunitgenerated by alternative processing of a new member of a cytokine receptorgene cluster. EMBO J. 14:5100–5108.

44. Mastronarde, J. G., B. He, M. M. Monick, N. Mukaida, K. Matsushima, andG. W. Hunninghake. 1996. Induction of interleukin (IL)-8 gene expressionby respiratory syncytial virus involves activation of nuclear factor (NF)-kappaB and NF-IL-6. J. Infect. Dis. 174:262–267.

45. Muller, M., C. Laxton, J. Briscoe, C. Schindler, T. Improta, J. E. Darnell,Jr., G. R. Stark, and I. M. Kerr. 1993. Complementation of a mutant cellline: central role of the 91 kDa polypeptide of ISGF3 in the interferon-alphaand -gamma signal transduction pathways. EMBO J. 12:4221–4228.

46. Ott, M., J. L. Lovett, L. Mueller, and E. Verdin. 1998. Superinduction of IL-8in T cells by HIV-1 Tat protein is mediated through NF-kappaB factors.J. Immunol. 160:2872–2880.

47. Palangat, M., and R. Landick. 2001. Roles of RNA:DNA hybrid stability,RNA structure, and active site conformation in pausing by human RNApolymerase II. J. Mol. Biol. 311:265–282.

48. Pellegrini, S., J. John, M. Shearer, I. M. Kerr, and G. R. Stark. 1989. Use of

a selectable marker regulated by alpha interferon to obtain mutations in thesignaling pathway. Mol. Cell. Biol. 9:4605–4612.

49. Polyak, S. J., K. S. A. Khabar, D. M. Paschal, H. J. Ezelle, G. Duverlie,G. N. Barber, D. E. Levy, N. Mukaida, and D. R. Gretch. 2001. HepatitisC virus nonstructural 5A protein induces interleukin-8, leading to partialinhibition of the interferon-induced antiviral response. J. Virol. 75:6095–6106.

50. Poole, E., B. He, R. A. Lamb, R. E. Randall, and S. Goodbourn. 2002. TheV proteins of simian virus 5 and other paramyxoviruses inhibit induction ofinterferon-beta. Virology 303:33–46.

51. Rochat, S., H. Komada, and D. Kolakofsky. 1992. Loss of V protein expres-sion in human parainfluenza virus type 1 is not a recent event. Virus Res.24:137–144.

52. Roux, P., C. Alfieri, M. Hrimech, E. A. Cohen, and J. E. Tanner. 2000.Activation of transcription factors NF-�B and NF-IL-6 by human immuno-deficiency virus type 1 protein R (Vpr) induces interleukin-8 expression.J. Virol. 74:4658–4665.

53. Sakaguchi, T., K. Kiyotani, M. Sakaki, Y. Fujii, and T. Yoshida. 1994. A fieldisolate of Sendai virus: its high virulence to mice and genetic divergence formprototype strains. Arch. Virol. 135:159–164.

54. Schafer, S. L., R. Lin, P. A. Moore, J. Hiscott, and P. M. Pitha. 1998.Regulation of type I interferon gene expression by interferon regulatoryfactor-3. J. Biol. Chem. 273:2714–2720.

55. Schlaak, J. F., C. M. Hilkens, A. P. Costa-Pereira, B. Strobl, F. Aberger,A. M. Frischauf, and I. M. Kerr. 2002. Cell-type and donor-specific tran-scriptional responses to interferon-alpha. Use of customized gene arrays.J. Biol. Chem. 277:49428–49437.

56. Skiadopoulos, M. H., S. R. Surman, J. M. Riggs, W. R. Elkins, M. St Claire,M. Nishio, D. Garcin, D. Kolakofsky, P. L. Collins, and B. R. Murphy. 2002.Sendai virus, a murine parainfluenza virus type 1, replicates to a level similarto human PIV1 in the upper and lower respiratory tract of African greenmonkeys and chimpanzees. Virology 297:153–160.

57. Stark, G. R., I. M. Kerr, B. R. Williams, R. H. Silverman, and R. D.Schreiber. 1998. How cells respond to interferons. Annu. Rev. Biochem.67:227–264.

58. Strieter, R. M., J. A. Belperio, and M. P. Keane. 2002. Cytokines in innatehost defense in the lung. J. Clin. Invest. 109:699–705.

59. Takeuchi, K., T. Komatsu, J. Yokoo, A. Kato, T. Shioda, Y. Nagai, and B.Gotoh. 2001. Sendai virus C protein physically associates with Stat1. GenesCells 6:545–557.

60. Taniguchi, T., K. Ogasawara, A. Takaoka, and N. Tanaka. 2001. IRF familyof transcription factors as regulators of host defense. Annu. Rev. Immunol.19:623–655.

61. Tapparel, C., S. Hausmann, T. Pelet, J. Curran, D. Kolakofsky, and L. Roux.1997. Inhibition of Sendai virus genome replication due to promoter-in-creased selectivity: a possible role for the accessory C proteins. J. Virol.71:9588–9599.

62. Wang, X.-L., M. Itoh, H. Hotta, and M. Homma. 1994. A protease activationmutant, MVCES1, as a safe and potent live vaccine derived from currentlyprevailing Sendai virus. J. Virol. 68:3369–3373.

63. Wang, X., M. Li, H. Zheng, T. Muster, P. Palese, A. A. Beg, and A. Garcia-Sastre. 2000. Influenza A virus NS1 protein prevents activation of NF-�Band induction of alpha/beta interferon. J. Virol. 74:11566–11573.

64. Wansley, E. K., and G. D. Parks. 2002. Naturally occurring substitutions inthe P/V gene convert the noncytopathic paramyxovirus simian virus 5 into avirus that induces alpha/beta interferon synthesis and cell death. J. Virol.76:10109–10121.

65. Wathelet, M. G., C. H. Lin, B. S. Parekh, L. V. Ronco, P. M. Howley, and T.Maniatis. 1998. Virus infection induces the assembly of coordinately acti-vated transcription factors on the IFN-beta enhancer in vivo. Mol. Cell1:507–518.

66. Yamada, Y., A. Nakamura, M. Hosoda, T. Kato, T. Asano, K. Tonegawa, andM. Itoh. 2001. Cytokines in pleural liquid for diagnosis of tuberculous pleu-risy. Respir. Med. 95:577–581.

67. Yeow, W. S., W. C. Au, W. J. Lowther, and P. M. Pitha. 2001. Downregula-tion of IRF-3 levels by ribozyme modulates the profile of IFNA subtypesexpressed in infected human cells. J. Virol. 75:3021–3027.

68. Yoneyama, M., W. Suhara, Y. Fukuhara, M. Fukuda, E. Nishida, and T.Fujita. 1998. Direct triggering of the type I interferon system by virus infec-tion: activation of a transcription factor complex containing IRF-3 and CBP/p300. EMBO J. 17:1087–1095.

69. Zhang, Y., B. A. Luxon, A. Casola, R. P. Garofalo, M. Jamaluddin, and A. R.Brasier. 2001. Expression of respiratory syncytial virus-induced chemokinegene networks in lower airway epithelial cells revealed by cDNA micro-arrays. J. Virol. 75:9044–9058.

VOL. 77, 2003 SENDAI VIRUS TARGETING OF INFLAMMATORY RESPONSES 7913

Page 64: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

III. PUBLICATION 2

"SENDAI VIRUS DEFECTIVE-INTERFERING

GENOMES AND THE ACTIVATION

OF THE INTERFERON-BETA"

Laura Strahle, Dominique Garcin, Daniel Kolakofsky

Virology 351 (2006) 101–111

62

Page 65: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

INTRODUCTION TO PAPER TWO:

dsRNA is known to be a common viral molecular signature that is generated during virus

replication and acts a pathogen-associated molecular pattern (PAMP). These PAMPs are

recognised in the cytoplasm by RIG-I and Mda-5, which activate TBK1 and IKKε leading to

the phosphorylation and dimerization of IRF-3 and IRF-7. Activated IRF-3 and IRF-7

translocate into the nucleus where transcription of early specific genes are activated. IFNβ is

first secreted and feeds back onto cells in an autocrine manner, to induce multiple ISGs

resulting in an antiviral state and in a paracrine manner, to prime neighbouring cells for

possible infection.

SeV stocks available commercially are known to induce IFNβ and are commonly used by

many laboratories. Plus, these stocks are known for a long time to contain DI genomes. The

paramyxoviruses DI genomes can be of two types: internal deletion or copyback DI genomes

(cf. Introduction). DI genomes have the capacity to form dsRNA by at least two ways: 1) if

the level of the N protein is not sufficient, DI genomes (and antigenomes) can self-anneal; and

2) some DI genomes contain termini that are perfectly complementary, thus are likely free to

form dsRNA. Moreover, DI genomes (especially those from the copyback variety) interfere

robustly with the ND helper genome by competing for replication and consequently reducing

the production of viral proteins involved in counteracting the antiviral state. The effect of

reduced level of SeV accessory proteins on the innate antiviral response is confirmed by the

fact that mutated SeV stocks (containing deficient V and C proteins or leader) lead effectively

to enhanced levels of IFNβ mRNA (cf. Paper one).

Because the induction of the IFNβ activation is known to be due to the presence of DI

genomes, we investigate in this study whether the induction of IFNβ is truly due to the

presence of DIs (since plaqued purified WT SeV does not activate IFNβ) and whether this

property is specific to a certain type of DI genome. Moreover, the interference between both

the DI and the ND genomes was also examined. Finally, because of the involvement of the

viral C and V proteins in the counteraction of the innate immune response, their effect on the

IFNβ activation were tested upon DI infection (Fig. 19).

63

Page 66: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

IFNβ

DI infection (Copyback or

Internal deletion ?)

Viral V and C proteins

?

Figure 19: Upon DI infection, a strong IFNβ activation is observed. Two questions are raised in this paper: (1) Is the IFNβ activation dependent on the presence of the DI genome? And what kind of DI is responsible? (2) Does overexpression of the viral C and V proteins inhibit IFNβ induction upon DI infection?

We first compared three SeV stocks containing different kind of DI genomes and their ability

to activate IFNβ in non-IFN sensitive cells (293T). Levels of IFNβ were monitored depending

on the different SeV stocks, but the copyback DI variety (DI-H4) was found to be the most

potent IFNβ inducer. Moreover, because IFNβ activation requires modification of IRF-3, the

dimerization and the phosphorylated forms of IRF-3 were analysed upon DI infection. The

formation of dimers, as well as hyper-phosphorylated forms of IRF-3 were observed in DI

infections, as opposed to SeV ND infection.

In order to estimate independently the DI and the ND genomes by RT/PCR, we generated a

tagged version of the copyback DI-H4 genome, containing a sequence from the YFP gene.

This enabled us to follow the evolution of this new stock, containing both the DI and ND

genomes (generated by multiple passages in embyonated chicken eggs), in parallel with the

activation of IFNβ. The correlation between the level of DI genomes and the level of IFNβ

activation was also observed. We found that the activation of the induction of IFNβ upon DI-

64

Page 67: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

H4 infection was proportional to DI genome replication and that the ratio of DI to ND

genomes during infection.

Finally, we used IFN competent cells (2fTGH) to study the general aspect of the DI-H4

infection. Since the C and V proteins can block dsRNA signaling, we examined the effect of

their overexpression upon DI-H4 infection, considering that DI infection produces less viral

proteins. Indeed, overexpression of C and V proteins was effective in blocking the DI-H4-

induced activation of IFNβ as well as the poly(I/C) activation. This result further confirms the

idea that DI-H4 induces IFNβ via dsRNA.

This study provides evidence that the strong induction of IFNβ activation upon SeV infection

(SeV stock containing DI genomes) is mainly due to the presence of copyback DI genomes.

The level of IFNβ activation was found to be proportional to that of DI genome replication.

This activation can be inhibited by the overexpression of the C and V proteins, whose

concentrations are reduced in DI infected cells, because of the strong interference of the DI

versus the ND genome.

65

Page 68: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

6) 101–111www.elsevier.com/locate/yviro

Virology 351 (200

Sendai virus defective-interfering genomes and theactivation of interferon-beta

Laura Strahle, Dominique Garcin, Daniel Kolakofsky ⁎

Department of Microbiology and Molecular Medicine, University of Geneva School of Medicine, 11 Ave de Champel, CH1211 Geneva, Switzerland

Received 17 January 2006; returned to author for revision 23 February 2006; accepted 14 March 2006Available online 2 May 2006

Abstract

The ability of some Sendai virus stocks to strongly activate IFNβ has long been known to be associated with defective-interfering (DI)genomes. We have compared SeV stocks containing various copyback and internal deletion DI genomes (and those containing only nondefective(ND) genomes) for their ability to activate reporter genes driven by the IFNβ promoter. We found that this property was primarily due to thepresence of copyback DI genomes and correlated with their ability to self-anneal and form dsRNA. The level of IFNβ activation was found to beproportional to that of DI genome replication and to the ratio of DI to ND genomes during infection. Over-expression of the viral Vand C proteinswas as effective in blocking the copyback DI-induced activation of the IFNβ promoter as it was in reducing poly-I/C-induced activation, providingevidence that these DI infections activate IFNβ via dsRNA. Infection with an SeV stock that is highly contaminated with copyback DI genomes isthus a very particular way of potently activating IFNβ, presumably by providing plentiful dsRNA under conditions of reduced expression of viralproducts which block the host antiviral response.© 2006 Elsevier Inc. All rights reserved.

Keywords: Sendai virus; Defective-Interfering genome; Interferon beta; Innate immunity

Our understanding of how animal cells recognize and mountan innate antiviral response to intracellular RNA virusreplication has recently made great progress. Two DexD/Hbox helicases with CARD domains, RIG-I and Mda5/Helicard,were found to participate in the detection of cytoplasmicdsRNA (Andrejeva et al., 2004; Yoneyama et al., 2004). dsRNAis thought to be a common product of RNAvirus infections thatacts as a pathogen-associated molecular pattern (PAMP)responsible for initiating the innate antiviral response. RIG-Iand Mda5 initiate antiviral responses by coordinately activatingseveral transcription factors, including NF-κB and IRF-3, thatbind to the IFNβ promoter forming an enhanceosome thatactivates this primary host-response gene (McWhirter et al.,2005). Upon binding dsRNA, the CARD domains of thesehelicases are thought to be freed for interaction with thesynonymous domain of MAVS/VISA/cardif/IPS-1, a mitochon-drial protein which in turn is required for recruiting the kinasesthat activate these transcription factors (Xu et al., 2005; Meylan

⁎ Corresponding author.E-mail address: [email protected] (D. Kolakofsky).

0042-6822/$ - see front matter © 2006 Elsevier Inc. All rights reserved.doi:10.1016/j.virol.2006.03.022

et al., 2005; Seth et al., 2005; Kawai et al., 2005). Autocrineinteraction of secreted IFNβ with its cell surface receptor thencloses the “innate immunity loop”, leading to increased IFN-stimulated gene (ISG) products, such as other IFNs and theintracellular effectors of the antiviral state (Sen, 2001).

The role of Mda5 in dsRNA signaling to IFNβ wasuncovered because Mda5 binds to the paramyxovirus SV5 Vprotein (Andrejeva et al., 2004). This interaction, and that ofother paramyxovirus V proteins, blocks dsRNA signaling, andthis property of V maps to the highly conserved cys-rich domainat their C-termini (Poole et al., 2002; Andrejeva et al., 2004). Vproteins of different paramyxoviruses, however, are verydifferent at their N-terminal portions, which accounts for theirotherwise very different properties (Lamb and Kolakofsky,2001). Rubulavirus V proteins (e.g., SV5, PIV2) are associatedwith intracellular and virion nucleocapsids (NCs) and areimportant in promoting virus growth. PIV2 which cannotexpress the entire V protein is highly debilitated even in the mostpermissive cell culture (Nishio et al., 2005). Respirovirus (e.g.,Sendai virus (SeV)) V proteins, in contrast, are nonstructuralproteins, are not associated with NCs and their expression

Page 69: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

102 L. Strahle et al. / Virology 351 (2006) 101–111

inhibits genome replication (Curran et al., 1991). Moreover,although V expression is not required for replication in cellculture, SeVVexpression is essential for virulence in mice (Katoet al., 1997; Delenda et al., 1998). This requirement ispresumably due to its ability to block dsRNA-induced activationof the IFNβ promoter (Poole et al., 2002) as infection with SeVthat cannot specifically express the V protein strongly activatesIFNβ (Strahle et al., 2003). Finally, in contrast to rubulaviruses,respirovirus VmRNAs express a nested set of C proteins from anoverlapping ORF, and these C proteins also block the innateantiviral response, in several ways (e.g., by blocking IFNsignaling) (Gotoh et al., 1999; Garcin et al., 1999).

It is noteworthy that infection with wild-type SeV or SV5does not normally activate IFNβ. In contrast, infections withSV5 VΔC (which produces a C-terminally truncated V protein)or infections with SeV with mutations in either the leaderregion, two regions of the C protein or the V protein all lead toenhanced levels of IFNβ mRNA (He et al., 2002; Strahle et al.,2003). In the case of SeV, these viral genes also appear to beinvolved in preventing activation of inflammatory cytokinessuch as IL-8. For SeV, all the viral products that are not knownto play essential roles in the replication process itself (the VandC proteins, and leader and trailer RNAs) appear to function incountering the innate antiviral response. In the context of anormal, wild-type infection of cells in culture, the effect of thesevarious viral anti-host-response products is apparently sufficientto prevent, or severely limit, IFNβ activation.

The IFNβ promoter is normally activated in cells treatedwith dsRNA (poly I:poly C, or poly-I/C) or infected with virus.Sendai virus infection is often used in this respect, and virusstocks which strongly activate IFNβ are also availablecommercially. However, it has long been known that theremarkable ability of some SeV stocks to induce IFN secretionin macrophage and other cell lines is related to the presence ofdefective-interfering (DI) particles (Johnston, 1981; Poole et al.,2002), but the nature of the IFN-inducing agent in these stockswas not examined. Moreover, for VSV, another mononegavirus,DI particles containing “snap-back” DI genomes (see below)were found to be very potent inducers of IFN, even in theabsence of co-infecting nondefective (ND) helper virus (Marcusand Sekellick, 1977; Sekellick and Marcus, 1982). DI particlescontain deleted viral genomes which are generated spontane-ously as by-products of ND genome replication. DI genomeshave, sine qua non, gained the ability to successfully competewith their helper ND genomes for the viral replication substratesprovided by the latter; hence, they are also “interfering”(Perrault, 1981; Lazzarini et al., 1981). Because of theirreplicative advantage over ND genomes, DI genomes invariablyaccumulate in SeV stocks that are repeatedly passed in eggs,unless steps to prevent this accumulation are taken.

Nonsegmented negative-strand RNA viruses (NNV) DIgenomes can be of two types, internal deletion or copyback(Fig. 1). The replicative advantage of internal deletion DIgenomes over ND genomes is not well understood (Garcin etal., 1994), but that of copyback DI genomes is well studied.Copyback DI genomes have always replaced the weakergenomic replication promoter at the 3′ ends of their minus

strands with the stronger antigenomic promoter, and thus bothDI genomes and antigenomes initiate from the same strongantigenomic promoter. Paramyxovirus replication promoters arecontained within the 3′ terminal 91–96 nt of the genomic RNA(narrow boxes, Fig. 1), and all natural copyback DI genomeshave copied back 91 nt or more while carrying out this promoterexchange (see Fig. 1). Although these RNAs contain terminithat are perfectly complementary for ca. 100 nt, DI genomes arenormally present within nucleocapsids (NCs), where their RNAends are not free to anneal. However, when SDS is used togently dissociate the N protein from RNA, copyback DIgenomes (and antigenomes) rapidly form ssRNA circles withdsRNA panhandles, as seen in the EM and on biochemicalanalysis (Kolakofsky, 1976). Several copyback DI genomesfrom independent virus stocks were characterized initially. Theyall contained complementary termini of ca. 100 nt in length, andas expected, the rate at which they circularized on SDStreatment was inversely proportional to their length.

Similar copyback DI genomes with limited terminalcomplementarity are common for VSV. However, VSV, unlikeSeV, also generates an extreme form of copyback DI genomewhose sequences are complementary over their entire length ofca. 2 kb (snapback DIs) and which form long dsRNA “hairpins”(of ca. 1000 bp) rather than ss circles upon SDS treatment(Lazzarini et al., 1981; Perrault, 1981). It is these VSV snapbackDI genomes, like DI 011, that were reported to strongly induceIFN by themselves, in aged chick embryo fibroblasts (CEFs)and mouse L cells (Marcus and Gaccione, 1989; Marcus andSekellick, 1977). These reports have remained controversial,however, because this IFN induction was independent of co-infecting helper virus, whereas Youngner and colleagues foundthat it correlated with contaminating ND virus in L cells. Theselatter workers, moreover, were unable to find a correlationbetween the snapback content of their DIs and IFN induction(Frey et al., 1979). Sekellick and Marcus also reported thatsnapback DI induction of IFN was unaffected by heat treatmentthat would inactivate its RNA polymerase, or UV treatment thatwould prevent its genome from being copied, and concludedthat this IFN induction was due to a pre-existing molecule thatdid not require any synthetic events for its formation (Sekellickand Marcus, 1982; Marcus and Gaccione, 1989). Disassemblyof DI 011 NCs intracellularly would then appear to be the onlyexplanation for dsRNA formation. However, as the infection ofa single DI 011 particle per CEF was sufficient to induce aquantum (maximum) yield of IFN, the manner in which the DI011 NC is presumably disassembled so efficiently in vivo (topermit dsRNA formation) remains an enigma as NCs aregenerally very stable in vivo.

Given the growing appreciation that dsRNAmay be a commonproduct of RNAvirus infection that participates in the induction ofthe innate antiviral response, we have reexamined the requirementof SeV DI genomes for the activation of the IFNβ promoter.

Results

Three SeV stocks containing DI genomes (Figs. 1, 2a), aswell as a stock containing only ND genomes, were examined for

Page 70: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

Fig. 1. A schematic view of SeV RNA synthesis and DI genomes. The ND (−) genome is shown above, with its 3′ end genomic replication promoter (on the left) andthe complement of the antigenomic replication promoter (at the 5′ end), both contained within the terminal 96 nt, shown as thin boxes. The central protein codingregion, from the beginning of the N ORF to the end of the L ORF, is shown as thicker boxes; these regions are not drawn to scale. Only the N/P junction is shown, theremaining genes are lumped together as a P/V/C … L. The (+) RNAs transcribed from the ND genome (le RNA and the 6 mRNAs) are shown below the ND genome.The sole transcript of the ND-antigenome, tr RNA, is shown above. The lines below the ND genome indicate the progress of the replicase when generating internaldeletion (E307) or copyback (H4/ChR1) DI genomes. The structure of the DI (−) genomes is shown below; the dark shading on the left indicates the extent of theterminal complementarity, which is also indicated in brackets on the left. The inverted triangle indicates the yfp sequence used to tag H4. The arrows below the DIgenomes show the PCR amplification strategy used to clone the DI RNA of the Charles River SeV stock.

103L. Strahle et al. / Virology 351 (2006) 101–111

their ability to activate a luciferase reporter gene driven by theIFNβ promoter. The DI-H4 stock is composed predominantlyof a natural 1410 nt long copyback DI genome, whose terminiare perfectly complementary for 110 nt (Calain et al., 1992).The DI-S104 stock, like H4, was generated by passage in eggs,but this stock is composed of 5 major DI species. Only thesmallest DI of this stock has been cloned as DNA and found tobe an internal deletion DI of 1794 nt (called E307) (Engelhornet al., 1993). Their termini are complementary for only 12 nt,like those of ND genomes. However, Northern analysis with aleader/N gene probe, which anneals specifically to internaldeletion DI genomes, shows that this stock contains 3 internaldeletion as well as 2 copyback DI genomes (the latter of ca.2500 and 3500 nt in length) (Fig. 2a). DI-GP55, in contrast tothe others, was produced and rescued by recombinant means(Garcin et al., 1995) and is identical to E307 except that itcontains the 55 nt trailer region at the (−) DI genome 3′ end, inplace of the leader region. DI-GP55 is thus a copyback DIgenome whose termini are complementary for 55 nt (Fig. 1). Asmentioned above, copyback DI genomes with more extensiveterminal complementarity, such VSV DI 011, have not beenreported for SeV, and we have no stable SeV stocks that containonly internal deletion DI genomes.

Infection of 293T cells with all 3 DI stocks was found toactivate the IFNβ reporter, but to very different extents (Fig. 2c).

H4 was the most potent, followed by GP55, and S104 was theleast potent. In contrast, infection with ND genomes alone barelyactivated the reporter gene, even though the ND infectionaccumulated as much or more viral proteins than the DIinfections (Fig. 2b). In these and other experiments, there is oftenan inverse correlation between the accumulation of viral proteinsand the extent of IFNβ activation, as might be expected ifactivation is due to the presence of DI genomes. Moreimportantly, plaque purification of the H4 stock yields a viruspreparation that does not contain DI genomes and which doesnot activate the IFNβ promoter during infection (ND-H4, Fig.3). The ability of the DI-H4 stock to activate IFNβ is thus not dueto mutations within the ND genome, which could have arisenbecause of the presence of the DI genomes.

IFNβ activation requires modification of IRF-3, which ishyper-phosphorylated in response to viral infection, or dsRNA(Fitzgerald et al., 2003; Sharma et al., 2003). Activated IRF-3dimerizes andmigrates to the nucleus where it binds to the PRD Iand III elements of the IFNβ promoter, as part of anenhanceosome (Maniatis et al., 1998). The activation of theIFNβ promoter by SeV DI infection appears to require IRF-3since this activation is largely ablated by co-expression of adominant-negative form of IRF-3 (data not shown). When theextent of IRF-3 dimerization was examined, a significantfraction of the IRF-3 was found as dimers (on non-denaturing

Page 71: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

Fig. 2. Only DI infections activate IFNβ in 293T cells. (a) Northern blot of encapsidated (CsCl banded) RNAs from the various infections as indicated, using a (+)riboprobe from the end of the L gene (which detects all DI genomes) or a (+) riboprobe from the leader/N gene region, which detects only internal deletion DI genomes.The DI-H4 and GP-55 genomes do not anneal with the leader/N gene probe. (b) A Western blot of equal amounts of cell extracts (total proteins) of the variousinfections, using anti-P and anti-N antibodies. (c) 293T cells were transfected with a luciferase reporter plasmid under the control of the IFNβ promoter (and controlplasmid) for 24 h then infected (or not) with the various SeV stocks for 20 h. Cell extracts were prepared, and levels of the renilla and firefly luciferases weredetermined. The cumulative results of 3 experiments are shown. (d) Two different amounts (1× and 5×) of total cell proteins from extracts of the various infections wereseparated on nondenaturing gels andWestern blotted with anti-IRF-3. (e) Equal amounts of cell extracts of the various infections (total proteins) were separated on SDSgels andWestern blotted with anti-IRF-3. The right-hand lane shows uninfected cells that express transfected IRF-3 5D. The 5 isoforms, numbered 1 to 5, are indicated.

104 L. Strahle et al. / Virology 351 (2006) 101–111

gels) in extracts of all 3 DI infections, and the extent of dimerformation was roughly in proportion to the degree of IFNβactivation (Fig. 2d). In contrast, there was no evidence of dimersin the ND extract.We also examined the electrophoretic mobilityof IRF-3 on SDS-PAGE as this mobility is sensitive to IRF-3phosphorylation status (Hiscott et al., 2003; Hasegawa et al.,1992; Yoneyama et al., 2002). We could distinguish 5electrophoretic forms of IRF-3 in our extracts (Fig. 2e). Mock-infected extracts predominantly contained forms 2 and 3.Infection with ND SeV led to the strong appearance of fasterform 5, without loss of 2 and 3. In contrast, infection with DI-H4and GP55 led to the appearance of the slightly slower form 4,reduction in the intensity of forms 2 and 3, and the appearance ofthe slowest form 1, which co-migrates with phospho-mimeticIRF-3 5D that is constitutively active (Lin et al., 1999). IRF-3 is

thus being modified in response to the ND infection, but thesemodifications do not lead to dimerization or the activation of theIFNβ promoter. The presence of DI genomes during infectionsomehow causes IRF-3 to be modified differently, leading to itsdimerization and IFNβ promoter activation.

DI-H4-induced activation of IFNβ is proportional to DIgenome replication

The relative amounts of DI and ND genomes in cell extractscan be examined by Northern blotting (Figs. 2 and 3), but thisanalysis is linear only over a relatively narrow range and oftenunequally estimates RNAs that vary significantly in length(>10-fold in this case). RT/PCR is better suited to this task, butthis method cannot differentiate between DI-H4 and ND

Page 72: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

Fig. 4. Evolution of the DI-H4+yfp stock on passage in eggs. (a) The DI-H4+yfp genome was prepared from DNA by reverse genetics (Methods andmaterials) and rescued by superinfecting the BSR T7 culture with ND SeV(Methods and materials). The culture supernatant was inoculated into eggs, andthe resulting allantoic fluid was diluted 1 to 500 for reinoculation, for a total of 20times. The stocks from each passage level were assayed for their ability toactivate a GFP gene under the control of the IFNβ promoter. Only the resultsfrom passage levels 8 and 16 to 20 are shown, along with DI-H4 as positivecontrol and mock-infected as a negative control. (b) Total RNA was preparedfrom cells infected with passage levels 8 and 16 to 20 and examined by RT/PCRfor their levels of ND genome (N gene) and DI genome (YFP) relative to that ofGADPH mRNAs. Total RNA from a mock-infected culture served as thenegative control. (c) Equal amounts of extracts from cells infected with passagelevels 8 and 16 to 19 were examined by Western blotting with anti-N and anti-P/V/C and anti-actin as a loading control.

Fig. 3. Allantoic fluid stocks prepared from plaque-purified SeV do not activateIFNβ. The DI-H4 and ChR stocks were titered on LLC-MK2 cells in theabsence of serum. Single plaques from the highest positive dilution (10−7) werepicked, and 3/10 of each plaque was inoculated into hen's eggs that wereincubated at 33 °C for 3 days. The allantoic fluid stocks resulting from theplaque-purified virus (ca. 109 pfu/ml) were compared with the original stockscontaining the same amount of viral proteins (as determined by Coomassie bluestaining of SDS gels) for their ability to activate pIFNβ-GFP in 293T cells. ANorthern blot of encapsidated (CsCl banded) RNAs from equal amounts of ND-,H4-, and ChR1-infected cells, using a (+) riboprobe from the end of the L gene,is shown in the insert.

105L. Strahle et al. / Virology 351 (2006) 101–111

genomes unless the complementary termini are included in theamplification. However, the dsRNA panhandles of circular DIgenome RNAs are exceedingly stable and cannot be invaded byprimers extended by RTase even at elevated temperatures. Tocircumvent these technical problems, we prepared a taggedversion of the DI-H4 genome containing a 162 nt insertionfrom the YFP gene (Fig. 1), such that the DI and ND genomescould be independently and accurately estimated by RT/PCR,over a large range of values. A second reason for preparing DI-H4+yfp is that, although our H4 stocks are composedpredominantly of the 1410 nt long species cloned as DNA,overexposure of Northern blots shows that several other RNAsare present in much lower amounts and which have nototherwise been characterized. As we do not know whether allthe DI genomes in the H4 stock contribute equally to inducingIFNβ activation or whether activation is due to a particular (andperhaps uncharacterized) species, we cannot be sure that IFNβactivation is in fact due to the 1410 nt copyback DI genome.Recapitulation of these results with a tagged copy of the DI-H4genome would settle this issue as well.

DI-H4+yfp genomes were recovered from DNA in BSR T7cells that were subsequently co-infected with ND SeV. Stockscontaining this DI genome were then generated by multiplepassages in embyonated chicken eggs (Methods and materials),and each passage was tested for its ability to activate a GFPreporter gene under the control of the IFNβ promoter (pIFNβ-GFP) upon infection of 293T cells. Although some IFNβactivation above background appeared by passage 3, thisactivation increased slowly and erratically at first and eventuallyreached activation levels approximately half those of thereference H4 stock by passage 20 (Fig. 4a), During the laterpassages (16 to 20), the level of intracellular ND genomessteadily decreased (Fig. 4b), leading to a reduction in the levelsof viral proteins (Fig. 4c), whereas the levels of DI-H4+yfp

genomes steadily increased (Fig. 4b). The ability of SeV DIstocks to induce IFNβ activation thus correlates with therelative levels of DI genomes during infection. As DI and NDgenome NCs are relatively stable structures, these levels reflectthe rates that DI and ND genomes are synthesized duringinfection with the various passage levels. The evolution of theDI-H4+yfp stock towards IFNβ activation (during relativelyundiluted passage in eggs) thus correlates with the level of DIgenome synthesis during infection. The remarkable ability of

Page 73: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

106 L. Strahle et al. / Virology 351 (2006) 101–111

the DI-H4 stock to activate IFNβ can now also be traced to thepredominant 1410-nt-long copyback DI genome.

Examination of the UV inactivation kinetics of DI-inducedIFNβ activation, as compared to the UV inactivation kinetics ofND and DI genome replication, can provide broad informationon the nature of this IFNβ activation. DI-H4+yfp (P(passage)17, insert, Fig. 5) was irradiated with 256 nm UV light forvarious times (0.5 to 8 min) and used to infect 293T cellscontaining pIFNβ-GFP. Intracellular RNA was isolated at20 hpi, and the levels of ND genome RNA and DI-H4+yfpgenome RNAwere measured by RT/PCR. GFP expression wasmonitored by FACS (Fig. 5a). DI-H4+yfp (1572 nt) is 1/10 thelength of the ND genome (15,384 nt) and thus proportionatelyless sensitive to UV inactivation. This difference in the loss ofND and DI genome levels is most apparent at the shortest timesof UV irradiation and is lost at the higher doses, presumablybecause DI genome replication ultimately depends on NDgenomes to provide all the replication substrates (N, P and Lproteins). The reduction of GFP expression levels upon

Fig. 5. UV inactivation kinetics of DI-H4+yfp infections. (a) The allantoic fluidof DI-H4+yfp passage level 17 was irradiated with 254 nm light (110 μW/cm2)from 0.5 to 8min and then used to infect 293Tcells that had been transfected withpIFNβ-GFP. Total RNA was prepared at 24 hpi, and the levels of DI-H4+yfpgenome and ND genome were determined by RT/PCR. GFP expression levelswere monitored by FACS. The results are shown relative to RNA from cellsinfected with non-irradiated allantoic fluid. The cumulative results of 2experiments are shown. The error bars, always <20%, are not shown for clarity.(b) A Northern blot of encapsidated RNAs fromND, DI-H4 and DI-H4+yfp P17infections, using a (+) probe from the end of the L gene.

increasing UV irradiation parallels that of ND and DI genomesand most closely follows the loss of the DI genomes at thelowest doses of UV. More importantly, as the reduction of GFPexpression levels closely follows that of DI genomes over arange of 2 logs, IFNβ activation is clearly proportional to thelevel of copyback DI genome replication (in 293T cells) for agiven stock as well.

The manner in which SeV DI genomes presumably generatedsRNA that induces IFN is thus quite different from that ofVSV DI 011. SeV DI genomes not only require co-infectionwith ND helper virus, IFN induction here (293T cells) is strictlyproportional to the level of DI genome replication, in contrast toVSV snapback DI IFN induction (in aged CEFs) where viralRNA synthesis is not required.

The SeV stock of Charles Rivers Laboratory

As mentioned above, SeV stocks (Cantell strain) whoseinfection of cultured cells strongly activates IFNβ, are availablefrom Charles River Laboratory. A fresh allantoic fluid stock ofthis virus preparation was found to activate IFNβ to levelssimilar to those of DI-H4 (Fig. 3), and this stock was found tocontain a very small DI genome (of ∼600 nt) by Northernanalysis (insert, Fig. 3). When the ND virus of this preparationwas plaque purified on LLC-MK2 cells, allantoic fluid stocksprepared from the purified virus had lost the ability to activateIFNβ (ND-ChR1, Fig. 3). Thus, similar to DI-H4, the ability ofthe Charles River virus preparation to activate IFNβ appears tobe due to the presence of the DI genome(s), and not to mutationswithin its ND genome.

To determine the nature of this DI genome, we cloned theDI genome as DNA, as illustrated in Fig. 1 (small horizontalarrows are primers). We used one set of primers to amplifythe common right end of all DIs (arrows under GP55, Fig. 1)and 3 sets of primers that were specific to the left end ofeither internal deletion (arrows under E307, Fig. 1) orcopyback DIs (arrows under H4, Fig. 1). The common rightend primer set and the 3 copyback-specific left-end primersets all yielded a PCR product of the expected size, whereasthe 3 internal-deletion-specific left-end primers failed toproduce visible DNA (not shown). When these amplifiedDNA fragments were sequenced, DI-ChR1 was deduced to bea simple copyback DI genome of 546 nt (453 nt are co-linearwith the 5′ end of the ND (−) genome), with terminalcomplementarity over 93 nt. To our knowledge, this is thesmallest natural SeV DI genome described to date, and thisproperty may be related to its ability to activate IFNβ sostrongly.

The SeV V and/or C proteins inhibit DI-H4-induced IFNβactivation

Although our 293T cells produce IFN in response to DI-H4 infection, they do not respond to added IFN. 293T cellsare thus useful in studying IFNβ activation in isolationbecause the activation is not also driven by positive feedbackvia ISGs. However, to study the broader aspects of the

Page 74: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

107L. Strahle et al. / Virology 351 (2006) 101–111

DI-H4 infection, we used 2fTGH cells, which both produceIFN in response to infection and respond as well to thesecreted IFN. When 2fTGH cells containing a pIFNβ-lucreporter are treated with poly-I/C (either added to themedium or via transfection (*)) or IFNα, poly-I/C* and H4infection strongly activate IFNβ promoter, whereas IFNα hasno effect and poly-I/C treatment has little effect (IFNβ is notan ISG, and TLR3 may be poorly expressed in thesefibroblasts). Moreover, whereas poly-I/C* and IFNα clearlyincreased ISG levels (STAT1, RIG-I, ISG15 and ISG56), H4infection failed to increase these ISG levels above theuntreated control. The SeV V protein blocks poly-I/C-induced IFNβ activation (Poole et al., 2002) and presumablyshould also block that induced by DI-H4 infection. Wereasoned that, if DI-H4 infections generated abnormally largeamounts of dsRNA, the amount of V expressed from the NDgenome during DI infections might be insufficient to blockdsRNA signaling via RIG-I and Mda5. We therefore over-expressed the V and C proteins by plasmid transfection in2fTGH cells containing pIFNβ-luc and re-examined theeffects of the various treatments. As shown in Fig. 6a, over-expression of the V and C proteins was as effective inblocking the DI-H4-induced activation of the IFNβ promoteras it was in reducing the poly-I/C*-induced activation. Thisresult is consistent with the notion that DI-H4 infectioninduces IFNβ activation, at least in part, via dsRNA. Over-expression of the V and C proteins also partially blocked thepoly-I/C*- and IFNα-induced increase in ISG levels,including that of RIG-I.

To examine whether DI-H4 infection was indeed able toblock IFN signaling, we similarly treated 2fTGH cellscontaining a pIRSE-luc reporter and examined the effects onthe reporter gene and ISG protein levels (Fig. 6b). In contrastto IFNα and poly-I/C* treatment which activated thispromoter and increased ISG levels, DI-H4 infection did notactivate the ISRE reporter over the untreated control, and thelevels of STAT1, RIG-I, ISG15 and ISG56 were not increased

Fig. 6. (a) The effect of poly I/C, DI-H4 infection and IFNα treatment on IFNβand ISGs in 2fTGH cells, in the presence and absence of the SeV V and Cproteins. 2fTGH cells were transfected with a luciferase reporter plasmid underthe control of the IFNβ promoter (and the TK-renilla control plasmid) and pSeV-V/C that expresses V and C proteins from the same mRNA (or and emptyplasmid) for 24 h. Parallel cultures were then either treated with 100 μg/ml ofpoly I/C in MEM (poly-I/C), transfected with 1 μg of poly I/C (poly I/C*),infected with DI-H4 or treated with 1000 IU of IFNα. Cell extracts wereprepared 20 h later, and the levels of the renilla and firefly luciferase activitieswere determined. The cumulative results of 2 experiments are shown. Equalamounts of total protein of each extract were also examined for their levels ofSTAT1 (and actin), RIG-I, SeV P, V and C proteins and ISG-15 and ISG-56 byWestern blotting with specific antibodies (Methods and materials). (b) The effectof poly I/C, DI-H4 infection and IFNα treatment on ISGs in 2fTGH cells.2fTGH cells were transfected with a luciferase reporter plasmid under thecontrol of an ISRE promoter (and the TK-renilla control plasmid) for 24 h.Parallel cultures were then treated as in panel A. Cell extracts were prepared 20 hlater, and the levels of the renilla and firefly luciferase activities weredetermined. The cumulative results of 2 experiments are shown. Equal amountsof total protein of each extract were also examined for their levels of STAT1 (andactin), RIG-I and ISG-15 and ISG-56 by Western blotting.

in these extracts. Thus, there appears to be sufficient viralproteins expressed during DI-H4 infection to block IFNsignaling.

Page 75: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

108 L. Strahle et al. / Virology 351 (2006) 101–111

Discussion

dsRNA is thought to be a common product or PAMP of RNAvirus infections that initiates the innate antiviral response, in partby activating IFNβ. However, the source of this dsRNA ispresumably different for different viruses. (+) RNA virusgenomes contain highly conserved 2° and 3° structures at their5′ and 3′ ends that are essential for virus replication (Simmondset al., 2004), and these highly structured RNA regions bythemselves can initiate signaling to IFNβ upon binding to RIG-I(Sumpter et al., 2005). In contrast, NNV genomes are not knownto contain conserved 2° structures, and, moreover, NNVgenomes function in RNA synthesis not as free RNAs but asassembled NCs, in which the genome RNA cannot normallyanneal (Lamb and Kolakofsky, 2001). One possible source ofdsRNA during NDSeVreplication is the occasional extension ofthe trailer RNA beyond the trailer/L gene junction (see Fig. 1),producing run-on trailer RNAswhose 3′ sequences can anneal tothose of the L mRNA (Vidal and Kolakofsky, 1989). In a similarvein, the transcriptase which synthesizes the L mRNApresumably reads through the L gene-end site at a frequencyof ca. 5% (similar to other gene junctions; Le Mercier et al.,2002) and terminates at the genome 5′ end, thus providing read-through L transcripts that can anneal to trailer RNAs. Althoughthe NNV replication strategy appears to minimize dsRNApotential during intracellular replication, this strategy presum-ably cannot exclude the generation of small amounts of dsRNA.It is reasonable that the levels of V and C expressed during NDgenome replication are designed to counteract the small amountsof dsRNA generated. If so, the presence of significant amountsof copyback DI genomes during intracellular replication willcertainly change the nature of the SeV infection.

The most important new finding of this study is that not allSeV stocks that are heavily contaminated by DI genomes areequally able to activate IFNβ. The H4, GP55 and S104 stocks allcontain the H strain ND genome as helper and can be directlycompared. S104 infections accumulate more viral products thanH4 infections as the S104 DI genomes appear to interfere lesswith their helper virus replication than those of H4 (Fig. 2b). Atthe same time, S104 infections activated IFNβ 10 to 20-fold lessstrongly than H4 infections in multiple experiments (Fig. 2a anddata not shown). Besides the different extents to which S104 andH4 DI genomes interfere with ND genome expression and thusaffect the intracellular concentration of the viral VandC proteins,H4 stocks are composed exclusively of copyback DI genomes,whereas S104 stocks are composed predominantly (>70%) ofinternal deletion DI genomes. Moreover, the two copyback DIgenomes in this stock are both longer than DI-H4. DI-GP55 isalso longer than DI-H4, its termini are complementary for onlyhalf the length as H4 (55 nt), and DI-GP55 interferes with NDgenome expression less than DI-H4. Taken together, these datasuggest that the ability of SeV DI stocks to activate IFNβ isrelated both to (i) their ability to interfere with helper genomeexpression, which leads to lower levels of Vand C intracellularly,and (ii) their relative content of copyback DI genomes. The sizeof the DI genome may also play a role in this latter respect.However, it will be necessary to examine these properties of SeV

DI genomes more directly, e.g., by extending the terminalcomplementarity of DI-GP55 to 110 nt, or by altering the lengthof DI-H4+yfp, to be more certain of these conclusions.

Copyback DI genomes may be stronger activators of IFNβthan internal deletion DI genomes because they have a strongerpotential to form dsRNA. Copyback DI genomes are composedof equal amounts of genomes and antigenomes (rather than a10-fold excess of genomes), and their termini can self-annealintramolecularly as well. The question then remains of how thisdsRNA potential is expressed as the synthesis of DI genomes,like that of ND genomes, is thought to be coupled to theirassembly into NCs. The vast majority of these NCs, onceformed, are very stable in vivo and band in CsCl densitygradients as fully assembled NCs. However, it is not rare to findsmall amounts of DI-H4 genomes and antigenomes in extractsof infected cells which pellet through these gradients as freeRNAs (<5%, unpublished). It is not clear whether these non-assembled DI genomes were normally made as NCs whichsubsequently disassembled or were actually made de novowithout concurrent assembly with N protein, as reported forsome conditions of VSV DI genome replication in vitro(Chanda et al., 1980). Independent of how non-assembled DI-H4 genomes are presumably generated in vivo, their formationappears to be proportional to their synthesis (Fig. 4). Theirpresence would represent a vast increase in the dsRNA potentialof SeV DI vs. ND infections, under conditions where there isless V protein available to dampen dsRNA signaling to IFNβ(Fig. 4c). The relatively short lengths of the H4 and ChR1 DIgenomes may play a role in how frequently their non-assembledRNAs are formed in vivo, but this needs to be investigated.

There is one further aspect of SeV copyback DI infections thatshould be mentioned to explain their ability to induce IFN soefficiently, coupled with the remarkable fact that these DIgenomes with dsRNA potential are actually selected for onpassage in eggs (e.g., Fig. 4) (Le Mercier et al., 2002). We havepreviously prepared ambisense ND SeV in which an additionalmRNA was expressed from the 3′ end of the antigenome. Incontrast to copyback DI genomes, genomes and antigenomes ofthe ambisense SeV contain the weaker genomic promoter. Theseambisense SeV grew poorly in IFN-sensitive cultures and wererelatively IFN-sensitive. They were also highly unstable onpassage in eggs and reverted to virus that grows well even in IFN-pretreated cells that restrict vesicular stomatitis virus replication,i.e., the wild-type SeV phenotype. Since this reversion wasalways associated with a point mutation in the ambi-mRNA startsite that severely limited its expression, we concluded that theselection ofmutants unable to express ambi-mRNAon passage inchicken eggs was presumably due to increased levels of dsRNAduring infection (vRdRp read-through of the ambi-mRNA stopsite creates a capped transcript that can potentially extend theentire length of the antigenome, whereas extension of theuncapped trailer RNA (wt SeV) is limited by the poor processivityof its vRdRp). If ND ambisense SeV with dsRNA potential arestrongly selected against in eggs, then how are DI genomes withdsRNA potential positively selected under the same conditions?

There are two possible explanations for this conundrum.Firstly, the dsRNA potential of ambisense SeV is not associated

Page 76: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

109L. Strahle et al. / Virology 351 (2006) 101–111

with any selective advantage and a single point mutation in theambi-mRNA promoter will largely eliminate this potential. TheND genomes of SeV stocks containing copyback DI genomes,in contrast, cannot escape their DI genomes by simple mutation,and the dsRNA potential of copyback DI genomes is alwaysassociated with a strong selective advantage as copyback DIgenomes outcompete their ND genomes for the replicationsubstrates provided by the latter. Secondly, and perhaps moreimportantly, copyback DI infections are relatively non-cyto-pathic and often end as persistent infections (Roux et al., 1991).This is in part due to the ectopic expression of trailer RNA (inplace of leader RNA) from the copyback DI (−) genome (Garcinet al., 1998) (Fig. 1). Trailer RNA is known to interact withTIAR, a protein with many links to apoptosis, and thisinteraction is important in suppressing SeV-induced PCD(Iseni et al., 2002). The relative absence of leader RNAs duringcopyback DI infections may also contribute to this DIphenotype as mutations in the SeV leader region are associatedwith virulence in mice (Fujii et al., 2002), and the normalexpression of leader RNA appears to be required to preventIFNβ activation (Strahle et al., 2003). The ability of copybackDI infections to delay, and in many cases completely preventPCD, may compensate for the negative consequences ofincreased dsRNA during infection, which presumably selectsagainst SeV that express ambi-mRNAs.

In summary, infection with an SeV stock that is highlycontaminated with copyback DI genomes is a potent way ofactivating IFNβ. These DI infections presumably provideplentiful dsRNA, under conditions of reduced expression ofviral products which block the host response to dsRNA, andwith minimal cytopathic effects that lead to persistent infection.In contrast, infection with an SeV stock that is not contaminatedwith copyback DI genomes does not activate IFNβ and ishighly cytopathic. These are two very different virus infections,and they should not be confused when SeV stocks of unknowncomposition are used to activate IFNβ.

Methods and materials

Cells, viruses, and antibodies

BSR-T7 cells were grown in BHK-21 Medium (GlasgowMEM, Gibco) supplemented with 5% fetal calf serum (FCS) inthe presence of the relevant maintenance drug (G418 at 400 μg/ml). 2fTGH cells and 293T cells were grown in Dulbecco'smodified Eagle's medium supplemented with 10% fetal calfserum (FCS).

SeV stocks were grown in the allantoic cavities of 9-day-oldembryonated chicken eggs for 3 days at 33 °C. For ND stocks(109 pfu/ml), 0.1 ml of a 105 dilution (ca. 1000 pfu) wasinoculated per egg. In the case of DI stocks, 0.1 ml of a 102 to104 dilution was used. In all cases, the amount of viral proteinspresent in the resulting allantoic fluid was analyzed by sodiumdodecyl sulfate (SDS)-polyacrylamide gel electrophoresis andCoomassie blue staining of pelleted virus. Virus titers weredetermined by plaquing on LLC-MK2 cells. Anti-IRF-3 (SantaCruz), anti-actin (Chemicon), anti-Stat1 (C-terminus) (Trans-

duction Laboratories), anti-N-877 and anti-PCV, anti-ISG-15and anti-ISG-56 were provided by Dr. Ganes Sen from TheCleveland Clinic Institute. Anti-Rig-I was provided by TadaatsuImaizumi from Hirosaki University School of Medicine.

Plasmids, transient transfections, luciferase assay and FACS

pβ-IFN-fl-lucter, which contains the firefly luciferase geneunder the control of the human IFNβ promoter, is described inKing and Goodbourn (1994). The IFNα/β-responsive reporterplasmid, p(9–27)4tkD(239)lucter, referred to here as pISRE-fl-lucter, contains four tandem repeats of the IFN-inducible gene9–27 ISRE fused to the firefly luciferase gene (Didcock et al.,1999). pTK-rl-lucter used as a transfection standard contains theherpes simplex virus TK promoter region upstream of the renillaluciferase gene (Promega). pIRF-3ΔN, which expresses adominant negative form of IRF-3, and pIRF-3 5D, which isconstitutively active, were obtained from John Hiscott andPaula Pitha (Lin et al., 1998).

For transfections, 100,000 cells were plated in six-well plates20 h before transfection with 1 μg of pβ-IFN-fl-lucter or pISRE-fl-lucter, 0.3 μg of pTK-rl-lucter, with or without 1 μg of IRF-3ΔN, or 1 μg of EBS plasmid expressing SeV-V protein (Nishioet al., 2005), and Fugene (Roche) according to the manufac-turer's instructions. At 24 h post-transfection, the cells were (orwere not) infected with various Sendai virus stocks or treatedwith 100 μg of poly(I)–poly(C) (Sigma, St. Louis, MO) per ml ortransfected with 1 μg of poly(I)–poly(C) using Fugene. Twentyhours later, cells were harvested and assayed for firefly andrenilla luciferase activity (dual-luciferase reporter assay system;Promega). Relative expression levels were calculated bydividing the firefly luciferase values by those of renillaluciferase. pIFNβ-GFP, which expresses GFP under the controlof the IFNβ promoter, was constructed by cloning the IFNβpromoter region from pIFNβ-fl-lucter into pEGFP-N3 (BDBiosciences Clontech), between the AseI and HindIII sites. Fortransfections, 100,000 cells were plated in six-well plates 20 hbefore transfection with 1 μg of pIFNβ-GFP and CaPO4

(Stratagene) according to the manufacturer's instructions. At24 h post-transfection, the cells were (or were not) infected withSendai virus or different DI stocks. Twenty hours later, cells wereharvested and assayed for GFP expression by FACS analysis.

Preparation of DI-H4+yfp

100,000 BSR-T7 cells were plated in six-well plates 20 hbefore transfection with a mix containing 0.75 μg of pTM1-L,1.5 μg of pTM1-N, 1.5 μg of pTM1-P/Cstop (which does notexpress C proteins), 1 μg of the various pDI constructs andFugene. Six hours later, the transfection mix was discarded andreplaced with 2 ml of Glasgow MEM supplemented with 5%FCS. Twenty four hours post-transfection, the cells wereinfected with ND SeV. Forty eight hours post-infection, thecells were scraped into their medium and injected directly intothe allantoic cavity of 9-day embryonated chicken eggs. Threedays later, the allantoic fluids were harvested and injectedundiluted into eggs. For further passages, the virus stocks were

Page 77: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

110 L. Strahle et al. / Virology 351 (2006) 101–111

clarified by centrifugation (10 min at 3000 rpm) and diluted 1/500 before injection. The presence of viruses in the resultingstock was determined by pelleting allantoic fluids (100 μl)through a TNE (10 mM Tris–HCl [pH 8.0], 100 mM NaCl,1 mM EDTA)–25% glycerol cushion for 20 min at 14,000 rpmin an Eppendorf 5417C centrifuge. Virus pellets wereresuspended in sample buffer, and the proteins were separatedby sodium dodecyl sulfate–10% PAGE and stained withCoomassie brilliant blue, alongside an ND stock of known titer.

Analysis of encapsidated RNAs

Confluent 293T cells in 9 cm ∅ Petri dishes (2 × 107 cells)were infected with 10 pfu/cell of ND stocks, and an equivalentamount of viral protein for DI stocks. Two days post-infection,the cells were collected, and the intracellular viral nucleocapsids(NC) were purified by 20–40% (w/w) CsCl density gradientcentrifugation and pelleted. After treatment with SDS andproteinase K, the nucleocapsid RNAs were phenol-extractedand ethanol-precipitated. The resulting RNAs were character-ized by Taqman analysis using specifics oligonucleotides andTaqman probes and by Northern blotting using a biotinylatedriboprobe generated in vitro by T7 RNA polymerase transcrip-tion of plus strands complementary to nucleotides 13,397–14,850 of the (−) ND genome.

Reverse transcription and real-time PCR via Taqman

Total RNA was extracted using Trizol (Invitrogen). Twentymicrograms of total RNA was mixed with 0.5 μg RandomHexamers (Promega) and subjected to a Superscript reversetranscription (RT) reaction as described by the manufacturer(Gibco) in a total volume of 50 μl. Two microliters of eachcDNA was then combined with 1 μl of internal controlHuman GAPDH (Applied Biosystems), 11 μl MasterMix(Eurogentec), 20 pmol (each) of forward and reverse primersand 4.4 pmol of Taqman probe in a total volume of 22 μl.The following primers and probes (Eurogentec or Microsynth)were used: N gene: 5′-GCAATAACGGTGTCGATCACG-3′(Fwd); 5′-TGCCTGAGCCGATCGG-3′ (Rev); 5′-CGAAGAT-GACGATACCGCAGCAGTAGC-3′ (Probe). YFP gene:5′-CCGACAACCACTACCTGAGCTA-3′ (Fwd); 5′-GAACTC-CAGCAGGACCATGTG-3′ (Rev); 5′-AAAGACCCCAACGA-GAAGCGCGA-3′ (Probe). Real-time PCR was carried out in the7700 Sequence Detector (Applied Biosystems).

Acknowledgment

This work was supported by the Swiss National ScienceFund.

References

Andrejeva, J., Childs, K.S., Young, D.F., Carlos, T.S., Stock, N.,Goodbourn, S., Randall, R.E., 2004. The V proteins of paramyxovirusesbind the IFN-inducible RNA helicase, mda-5, and inhibit its activationof the IFN-beta promoter. Proc. Natl. Acad. Sci. U.S.A. 101,17264–17269.

Calain, P., Curran, J., Kolakofsky, D., Roux, L., 1992. Molecular cloning ofnatural paramyxovirus copy-back defective interfering RNAs and theirexpression from DNA. Virology 191, 62–71.

Chanda, P.K., Kang, C.Y., Banerjee, A.K., 1980. Synthesis in vitro of the full-length complement of defective-interfering particle RNA of vesicularstomatitis virus. Proc. Natl. Acad. Sci. U.S.A. 77, 3927–3931.

Curran, J., Boeck, R., Kolakofsky, D., 1991. The Sendai virus P gene expressesboth an essential protein and an inhibitor of RNA synthesis by shufflingmodules via mRNA editing. EMBO J. 10, 3079–3085.

Delenda, C., Taylor, G., Hausmann, S., Garcin, D., Kolakofsky, D., 1998. Sendaiviruses with altered P, V, and W protein expression. Virology 242, 327–337.

Didcock, L., Young, D.F., Goodbourn, S., Randall, R.E., 1999. Sendai virus andsimian virus 5 block activation of interferon-responsive genes: importancefor virus pathogenesis. J. Virol. JID-0113724 73, 3125–3133.

Engelhorn, M., Stricker, R., Roux, L., 1993. Molecular cloning andcharacterization of a Sendai virus internal deletion defective RNA. J. Gen.Virol. 74, 137–141.

Fitzgerald, K.A., McWhirter, S.M., Faia, K.L., Rowe, D.C., Latz, E.,Golenbock, D.T., Coyle, A.J., Liao, S.M., Maniatis, T., 2003. IKKepsilonand TBK1 are essential components of the IRF3 signaling pathway. Nat.Immunol. 4, 491–496.

Frey, T.K., Jones, E.V., Cardamone Jr., J.J., Youngner, J.S., 1979. Induction ofinterferon in L cells by defective-interfering (DI) particles of vesicularstomatitis virus: lack of correlation with content of [+/−] snapback RNA.Virology 99, 95–102.

Fujii, Y., Sakaguchi, T., Kiyotani, K., Huang, C., Fukuhara, N., Egi, Y., Yoshida,T., 2002. Involvement of the leader sequence in Sendai virus pathogenesisrevealed by recovery of a pathogenic field isolate from cDNA. J. Virol. 76,8540–8547.

Garcin, D., de Melo, M., Roux, L., Kolakofsky, D., Curran, J., 1994. Presence ofa truncated form of the Sendai virus P protein in a long-term persistentinfection: implications for the maintenance of the persistent state. Virology200.

Garcin, D., Pelet, T., Calain, P., Roux, L., Curran, J., Kolakofsky, D., 1995. Ahighly recombinogenic system for the recovery of infectious Sendaiparamyxovirus from cDNA: generation of a novel copy-back nondefectiveinterfering virus. EMBO J. 14, 6087–6094.

Garcin, D., Taylor, G., Tanebayashi, K., Compans, R., Kolakofsky, D., 1998.The short Sendai virus leader region controls induction of programmed celldeath. Virology 243, 340–353.

Garcin, D., Latorre, P., Kolakofsky, D., 1999. Sendai virus C proteins counteractthe interferon-mediated induction of an antiviral state. J. Virol. 73,6559–6565.

Gotoh, B., Takeuchi, K., Komatsu, T., Yokoo, J., Kimura, Y., Kurotani, A., Kato,A., Nagai, Y., 1999. Knockout of the Sendai virus C gene eliminates the viralability to prevent the interferon-alpha/beta-mediated responses. FEBS Lett.459, 205–210.

Hasegawa, H., Yoshida, M., Shiosaka, T., Fujita, S., Kobayashi, Y., 1992.Mutations in the envelope protein of Japanese encephalitis virus affect entryinto cultured cells and virulence in mice. Virology 191, 158–165.

He, B., Paterson, R.G., Stock, N., Durbin, J.E., Durbin, R.K., Goodbourn, S.,Randall, R.E., Lamb, R.A., 2002. Recovery of Paramyxovirus simian virus 5with a V protein lacking the conserved cysteine-rich domain: themultifunctional V protein blocks both interferon-beta induction andinterferon signaling. Virology 303, 15–32.

Hiscott, J., Grandvaux, N., Sharma, S., Tenoever, B.R., Servant, M.J., Lin, R.,2003. Convergence of the NF-kappaB and interferon signaling pathways inthe regulation of antiviral defense and apoptosis. Ann. N. Y. Acad. Sci. 1010,237–248.

Iseni, F., Garcin, D., Nishio, M., Kedersha, N., Anderson, P., Kolakofsky, D.,2002. Sendai virus trailer RNA binds TIAR, a cellular protein involved invirus-induced apoptosis. EMBO J. 21, 5141–5150.

Johnston, M.D., 1981. The characteristics required for a Sendai viruspreparation to induce high levels of interferon in human lymphoblastoidcells. J. Gen. Virol. 56, 175–184.

Kato, A., Kiyotani, K., Sakai, Y., Yoshida, T., Nagai, Y., 1997. Theparamyxovirus, Sendai virus, V protein encodes a luxury function requiredfor viral pathogenesis. EMBO J 16, 578–587.

Page 78: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

111L. Strahle et al. / Virology 351 (2006) 101–111

Kawai, T., Takahashi, K., Sato, S., Coban, C., Kumar, H., Kato, H., Ishii, K.J.,Takeuchi, O., Akira, S., 2005. IPS-1, an adaptor triggering RIG-I- andMda5-mediated type I interferon induction. Nat. Immunol. 6, 981–988.

King, P., Goodbourn, S., 1994. The beta-interferon promoter responds topriming through multiple independent regulatory elements. J. Biol. Chem.269, 30609–30615.

Kolakofsky, D., 1976. Isolation and characterization of Sendai virus DI-RNAs.Cell 8, 547–555.

Lamb, R.A., Kolakofsky, D., 2001. Paramyxoviridae: the viruses and theirreplication. In: Knipe, D.M., Howley, P.M. (Eds.), Fields Virology, vol. 1.Lippincott, Williams and Wilkins, pp. 1305–1340.

Lazzarini, R.A., Keene, J.D., Schubert, M., 1981. The origins of defectiveinterfering particles of the negative-strand RNA viruses. Cell 26, 145–154.

Le Mercier, P., Garcin, D., Hausmann, S., Kolakofsky, D., 2002. Ambisensesendai viruses are inherently unstable but are useful to study viral RNAsynthesis. J. Virol. 76, 5492–5502.

Lin, R., Heylbroeck, C., Pitha, P.M., Hiscott, J., 1998. Virus-dependentphosphorylation of the IRF-3 transcription factor regulates nucleartranslocation, transactivation potential, and proteasome-mediated degrada-tion. Mol. Cell. Biol. 18, 2986–2996.

Lin, R., Mamane, Y., Hiscott, J., 1999. Structural and functional analysis ofinterferon regulatory factor 3: localization of the transactivation andautoinhibitory domains. Mol. Cell. Biol. 19, 2465–2474.

Maniatis, T., Falvo, J.V., Kim, T.H., Kim, T.K., Lin, C.H., Parekh, B.S.,Wathelet, M.G., 1998. Structure and function of the interferon-betaenhanceosome. Cold Spring Harbor Symp. Quant. Biol. 63, 609–620.

Marcus, P.I., Gaccione, C., 1989. Interferon induction by viruses. XIX. Vesicularstomatitis virus-New Jersey: high multiplicity passages generate interferon-inducing defective-interfering particles. Virology 171, 630–633.

Marcus, P.I., Sekellick, M.J., 1977. Defective interfering particles withcovalently linked [+/−]RNA induce interferon. Nature 266, 815–819.

McWhirter, S.M., Tenoever, B.R., Maniatis, T., 2005. Connecting mitochondriaand innate immunity. Cell 122, 645–647.

Meylan, E., Curran, J., Hofmann, K., Moradpour, D., Binder, M., Bartens-chlager, R., Tschopp, J., 2005. Cardif is an adaptor protein in the RIG-Iantiviral pathway and is targeted by hepatitis C virus. Nature 437,1167–1172.

Nishio, M., Tsurudome, M., Ito, M., Garcin, D., Kolakofsky, D., Ito, Y., 2005.Identification of paramyxovirus V protein residues essential for STATprotein degradation and promotion of virus replication. J. Virol. 79,8591–8601.

Perrault, J., 1981. Origin and replication of defective interfering particles. Curr.Top. Microbiol. Immunol. 93, 151–207.

Poole, E., He, B., Lamb, R.A., Randall, R.E., Goodbourn, S., 2002. The Vproteins of Simian virus 5 and other paramyxoviruses inhibit induction ofinterferon-beta. Virology 303, 33–46.

Roux, L., Simon, A.E., Holland, J.J., 1991. Effects of defective interferingviruses on virus replication and pathogenesis. Advances in Virus Research,vol. 40. Academic Press Inc., pp. 181–211.

Sekellick, M.J., Marcus, P.I., 1982. Interferon induction by viruses. VIII.Vesicular stomatitis virus: [+/−]DI-011 particles induce interferon in theabsence of standard virions. Virology 117, 280–285.

Sen, G.C., 2001. Viruses and interferons. Annu. Rev. Microbiol. 55, 255–281.Seth, R.B., Sun, L., Ea, C.K., Chen, Z.J., 2005. Identification and

characterization of MAVS, a mitochondrial antiviral signaling protein thatactivates NF-kappaB and IRF 3. Cell 122, 669–682.

Sharma, S., Tenoever, B.R., Grandvaux, N., Zhou, G.P., Lin, R., Hiscott, J.,2003. Triggering the interferon antiviral response through an IKK-relatedpathway. Science 300, 1148–1151.

Simmonds, P., Tuplin, A., Evans, D.J., 2004. Detection of genome-scaleordered RNA structure (GORS) in genomes of positive-stranded RNAviruses: implications for virus evolution and host persistence. RNA 10,1337–1351.

Strahle, L., Garcin, D., Le Mercier, P., Schlaak, J.F., Kolakofsky, D., 2003.Sendai virus targets inflammatory responses, as well as the interferon-induced antiviral state, in a multifaceted manner. J. Virol. 77,7903–7913.

Sumpter Jr., R., Loo, Y.M., Foy, E., Li, K., Yoneyama, M., Fujita, T.,Lemon, S.M., Gale Jr., M., 2005. Regulating intracellular antiviraldefense and permissiveness to hepatitis C virus RNA replication througha cellular RNA helicase, RIG-I.J. Virology 79, 2689–2699.

Vidal, S., Kolakofsky, D., 1989. Modified model for the switch from Sendaivirus transcription to replication. J. Virol. 63, 1951–1958.

Xu, L.G., Wang, Y.Y., Han, K.J., Li, L.Y., Zhai, Z., Shu, H.B., 2005. VISA is anadapter protein required for virus-triggered IFN-beta signaling. Mol. Cell 19,727–740.

Yoneyama, M., Suhara, W., Fujita, T., 2002. Control of IRF-3 activation byphosphorylation. J. Interferon Cytokine Res. 22, 73–76.

Yoneyama, M., Kikuchi, M., Natsukawa, T., Shinobu, N., Imaizumi, T.,Miyagishi, M., Taira, K., Akira, S., Fujita, T., 2004. The RNA helicase RIG-I has an essential function in double-stranded RNA-induced innate antiviralresponses. Nat. Immunol. 5, 730–737.

Page 79: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

IV. PUBLICATION 3

"SENDAI VIRUS ACTIVATION OF THE IFNβ

PROMOTER REQUIRES RIG-I AND IS

INHIBITED BY THE VIRAL C PROTEINS"

Laura Strähle, Jean-Baptiste Marq, Albert Brini, Stéphane Hausmann,

Daniel Kolakofsky and Dominique Garcin

Accepted in JOURNAL OF VIROLOGY, August 2007

77

Page 80: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

INTRODUCTION TO PAPER THREE:

All viruses have evolved to evade the cellular innate immune system including the IFN

system by various mechanisms. Interestingly, for the Paramyxoviruses, the C and V accessory

proteins are likely to be responsible for this counteraction of the innate immune response.

Paper one showed that mutations within distinct regions of the V and C proteins or of the

leader (absence of leader or over-expression of the trailer) all increased the level of IFNβ

mRNAs. SeV V and C proteins as well as the region of the leader are thus important for

antagonizing IFN signaling. Paper two showed that the SeV stocks containing specifically

copyback DI genomes were responsible for the strong IFNβ activation. This phenomenon

upon DI infection seemed to be due 1) to the ability of the DI genomes (and antigenomes) to

self-anneal and form dsRNA, and 2) to the reduced production of the viral V and C proteins.

Innate immunity upon viral infection has been extensively studied these past few years. As

mentioned previously the two helicases RIG-I and Mda-5 were discovered to be important

components responding to cytoplasmic RNA. Although these helicases transmit an identical

signal leading to the activation of IRF-3, IRF-7 and NF-kB, they appear to target different

viral products. In addition to this, RIG-I was found to detect other viral products than dsRNA.

Indeed Sousa et al. showed that influenza A virus infection does not generate dsRNA and that

RIG-I could be activated by single-stranded viral genomic RNA bearing 5’triphosphates

(5’pppRNA) .

In this study, the contribution of RIG-I (and Mda-5) in the detection of SeV infection is

examined in mouse embryo fibroblasts (MEFs). Because 5’triphophorylated products have

become new potential targets of RIG-I, we decided to test whether SeV infections induced

IFNβ activation by producing pppRNAs as well as dsRNA. The involvement of both

helicases was also analysed. For this, we used polyI/C, in vitro pppRNA (leader and trailer

RNAs) and two different types of SeV infection: 1) SeV-DI-H4, containing copyback DI

genomes and overexpressing 5’ppptrailer RNAs, as well as small amount of unencapsidated

DI H4-genome RNA that can self anneal to form dsRNA panhandles with 5’ppp ends and 2)

SeV-GFP(+/-), a mixed stock of SeV capable of producing dsRNA with capped ends. For the

mixted stock, we produced two different SeV stocks: one of them carries a GPF sequence,

producing + mRNA that is translated in GFP expressing proteins, the other contains the

inverse GFP sequence. The coinfection of these viruses produces dsRNA from both mRNA

78

Page 81: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

GFP + and - , which enabled us to test more precisely the effect of dsRNA (annealed capped

mRNAs) on the activation of IFNβ (Fig. 20).

Genome (-)N P/C/V) M F HN L 5’3’

GFP mRNA (+)

GFPSeV-GFP(+) :

Protein GFP

Genome (-)N P/C/V) M F HN L 5’3’

GFP mRNA (-)

PFGSeV-GFP(-) :

No Protein GFP

Coinfection SeV-GFP(+) and SeV-GFP(-) : SeV-GFP(+/-) dsRNA formation (capped ends) IFNβ activation ???

GFP mRNA (+)GFP mRNA (-)

Little/No protein GPF

Figure 20: SeV-GFP(+) expresses the protein GFP from a transgene between the M and the F genes. SeV-

GFP(-) synthesises a mRNA containing the complement of the GFP ORF, but no GFP protein can be

expressed. Upon coinfection (= SeV-GFP(+/-)), both messengers (GFP+ and GFP-) are generated and can

form dsRNA with capped 5’ppp ends. These infections are tested for their induction of IFNβ activation.

The formation of dsRNA in the coinfection was tested by using the dsRNA-binding domain

of E3L (Vacinia virus) proteins. We also used a dominant-negative form of RIG-I to examine

whether the activation of the IFNβ upon RNA treatment or different SeV infections was

dependent on RIG-I.

Finally, since the viral C and V proteins are responsible for counteracting the host innate

immune response, we over-expressed the V or C proteins in infected (SeV-DI-H4 or SeV-

79

Page 82: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

GFP(+/-))(Fig.21), or transfected (dsRNA or pppRNA) cells and also used SeV infections

carrying mutation within the C or V genes (SeV-Cminus or SeV-Vminus ). We also tested

whether they could inhibit the RIG-I dependent-IFNβ activation. In order to see which region

was responsible for the inhibition of RIG-I, two different domains of the C protein (C1-23 and

C24-204) carrying different function were also compared.

IFNβ

HELICASE CARD CARD

RIG-I

a) SeV-GFP(+/-) coinfection

IFNβ

HELICASE CARD CARD

RIG-I

tr ppp 5’

b) SeV-DI –H4 infection

ppp 5’

Cytoplasm

GFP

Overexpressed C and V proteins

ppp 5’tr ppp 5’

dsRNA

? ?

Figure 21: Upon coinfection (SeV-GFP(+/-)) or upon SeV-DI-H4 infection, the cellular sensor RIG-I

presumably recognises dsRNA and 5’triphosphorylated RNAs and leads to IFNβ activation. The viral

RNA products that are detected by RIG-I upon SeV infection are still to be discovered: the trailer RNAs

carrying 5’ppp ends remain good candidates. In this study, we also tested whether the overexpression of

the viral C or V proteins could prevent the induction of IFNβ (via RIG-I) upon both SeV GFP (+/-) and

DI-H4 infections.

In this study, we demonstrated that SeV-GFP(+/-) and SeV-DI-H4 infections as well as

transfections of the trailer pppRNAs or dsRNA (polyI/C) induced IFNβ activation in MEFs

cells in a RIG-I dependent manner (not Mda-5). This suggests that RIG-I presumably detects

both the 5’triphosphorylated RNAs and dsRNA. Moreover, these data showed that in infected

or transfected MEF cells, the expression of SeV C protein had a much greater effect in

counteracting RIG-I dependent IFNβ activation than the SeV V protein; and that the C24-204

region was sufficient for this function.

80

Page 83: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

JOURNAL OF VIROLOGY, Nov. 2007, p. 12227–12237 Vol. 81, No. 220022-538X/07/$08.00�0 doi:10.1128/JVI.01300-07Copyright © 2007, American Society for Microbiology. All Rights Reserved.

Activation of the Beta Interferon Promoter by Unnatural Sendai VirusInfection Requires RIG-I and Is Inhibited by Viral C Proteins�

Laura Strahle, Jean-Baptiste Marq, Albert Brini, Stephane Hausmann,Daniel Kolakofsky,* and Dominique Garcin

Department of Microbiology and Molecular Medicine, University of Geneva School of Medicine,11 Ave. de Champel, CH1211 Geneva, Switzerland

Received 14 June 2007/Accepted 27 August 2007

As infection with wild-type (wt) Sendai virus (SeV) normally activates beta interferon (IFN-�) very poorly,two unnatural SeV infections were used to study virus-induced IFN-� activation in mouse embryonic fibro-blasts: (i) SeV-DI-H4, which is composed mostly of small, copyback defective interfering (DI) genomes andwhose infection overproduces short 5�-triphosphorylated trailer RNAs (pppRNAs) and underproduces viral Vand C proteins, and (ii) SeV-GFP(�/�), a coinfection that produces wt amounts of viral gene products but thatalso produces both green fluorescent protein (GFP) mRNA and its complement, which can form double-stranded RNA (dsRNA) with capped 5� ends. We found that (i) virus-induced signaling to IFN-� dependedpredominantly on RIG-I (as opposed to mda-5) for both SeV infections, i.e., that RIG-I senses both pppRNAsand dsRNA without 5�-triphosphorylated ends, and (ii) it is the viral C protein (as opposed to V) that isprimarily responsible for countering RIG-I-dependent signaling to IFN-�. Nondefective SeV that cannotspecifically express C proteins not only cannot prevent the effects of transfected poly(I-C) or pppRNAs on IFN-�activation but also synergistically enhances these effects. SeV-Vminus infection, in contrast, behaves mostly likewt SeV and counteracts the effects of transfected poly(I-C) or pppRNAs.

All viruses evade the cellular innate immune system in partby expressing gene products that interfere with the ability ofthe host cell to establish an antiviral state (6). In the case of theParamyxovirinae, this anti-host-defense activity is due mostly toviral C and V proteins (15, 27, 31). The C and V proteins areencoded by separate alternate open reading frames (ORFs),which both overlap that of the P protein. V and C are alsoreferred to as accessory gene products, as not all members ofthis virus subfamily express one or the other. More specifically,rubulavirus and avulavirus express V but do not express Cproteins, and human parainfluenza virus type 1 (PIV1), a res-pirovirus most closely related to Sendai virus (SeV), expressesC but does not express a V protein (16, 20).

Paramyxovirus V and C proteins antagonize interferon(IFN) signaling by various mechanisms, and they also targetthe production of type I IFN (15, 31). Beta IFN (IFN-�)production is one of the earliest events in the cellular innateimmune response, which leads to the establishment of an anti-viral state. IFN-� production requires the coordinated activa-tion of several transcription factors, including NF-�B and IRF3(15, 29). For intracellular RNA virus replication, the signalingpathway that leads to IRF3 activation starts with mda-5 andRIG-I, two cytoplasmic DExH/D-box helicases with N-termi-nal CARD domains. These helicases respond to double-stranded RNA (dsRNA) and, at least for RIG-I, to 5�-triphos-phorylated single-stranded RNA (ssRNA) (pppRNA), whichare generated in the cytoplasm during RNA virus replication

(9, 11, 25). Upon the detection of these viral RNAs, the CARDdomains of these helicases interact with IPS-1/Cardif/MAVS/VISA, which is present in the mitochondrial membrane, andthis CARD-CARD interaction is thought to lead to the re-cruitment and activation of TBK1, IKKε, and other IKK ki-nases that activate NF-�B and IRF3, thereby activating theIFN-� promoter (8). The production of these early IFNs ini-tiates autocrine and paracrine signal amplifications via theJak/Stat pathway to produce a generalized antiviral state andalso assists in the subsequent activation of adaptive immuneresponses.

The role of mda-5 in sensing RNA virus infection was un-covered because mda-5 was found to bind to the PIV5 Vprotein and other paramyxovirus V proteins, including SeV V.These V-protein–mda-5 interactions, moreover, preventedIFN-� activation in response to transfected poly(I-C) (1). Onthe other hand, other studies found that RIG-I and not mda-5acts as the sensor of paramyxovirus infection (13, 28). Thispaper provides evidence that for SeV infection of mouse em-bryonic fibroblasts (MEFs), it is the C protein (and not V) thatis primarily responsible for this effect and that C acts by coun-tering RIG-I (and not mda-5). Independent expression of Cwas found to inhibit RIG-I-dependent signaling to the IFN-�promoter induced by either pppRNAs or dsRNAs. Moreover,SeV that cannot specifically express C proteins was unable tocounteract poly(I-C)- or pppRNA-induced IFN-� activation,whereas SeV that cannot express V behaved mostly like wild-type (wt) SeV.

MATERIALS AND METHODS

Cells, viruses, and antibodies. MEFs were grown in Dulbecco’s modifiedEagle’s medium supplemented with 10% fetal calf serum.

SeV stocks were grown in the allantoic cavitiesof 9-day-old embryonatedchicken eggs for 3 days at 33°C. For nondefective stocks (109 PFU/ml), 0.1 ml of

* Corresponding author. Mailing address: Department of Microbi-ology and Molecular Medicine, University of Geneva School of Med-icine, 11 Ave. de Champel, CH1211 Geneva, Switzerland. Phone: 41-223795657. Fax: 41-223795702. E-mail: [email protected].

� Published ahead of print on 5 September 2007.

12227

at Bibliotheque F

aculte Medecine G

eneve on Novem

ber 9, 2007 jvi.asm

.orgD

ownloaded from

Page 84: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

12228

at Bibliotheque F

aculte Medecine G

eneve on Novem

ber 9, 2007 jvi.asm

.orgD

ownloaded from

Page 85: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

a 105 dilution (ca. 1,000 PFU) was inoculated per egg. In the case of DI stocks,0.1 ml of a 103 dilution was used. In all cases, the amount of viral proteins presentin the resulting allantoic fluid was analyzed by sodium dodecyl sulfate-poly-acrylamide gel electrophoresis and Coomassie blue staining of pelleted virus.Virus titers were determined by plaquing on LLC-MK2 cells.

SeV-GFP(�), which expresses green fluorescent protein (GFP) from a trans-gene between the M and F genes, and SeV-GFP(�) or SeV-RFP, which ex-presses antisense GFP mRNA or red fluorescent protein (RFP) (dsRED) fromsimilarly located transgenes, were prepared as previously described (31). DI-H4stocks were described previously (30).

Primary antibodies used included rabbit anti-RFP (AB3216; Chemicon); anti-actin monoclonal antibody (MAb) (1501; Chemicon); rabbit anti-GFP (632460;BD biosciences); rabbit anti-SeV-P/C/V (homemade); anti-hemagglutinin (HA)MAb (16B12; BABCO), anti-Flag MAb (F1804; Sigma), rabbit anti-mda-5 (J.Tschopp, Lausanne, Switzerland), and rabbit anti-RIG-I (T. Fujita, Kyoto,Japan).

Plasmids, transient transfections, infections, inductions, luciferase assay, andfluorescence-activated cell sorter analysis. EBS plasmids (3) expressed viral andfluorescent proteins and were constructed by standard methods; precise detailcan be obtained from the authors.

NS1 {residues 1 to 77 [NS1(1-77)]} (from Jacques Perrault) and E3L {residues100 to 190 [E3L(100-190)]} (from Bertram Jacobs), were HA tagged and clonedinto pEBS. Flag-tagged RIG-I, RIG-I-C, or RIG-�CARDS (dominant negative)and mouse mda-5 were obtained from Jurg Tshopp and Klaus Conzelmann.

p�-IFN-fl-lucter, which contains the firefly luciferase gene under the control ofthe human IFN-� promoter, was described previously (14). pTK-rl-lucter, usedas a transfection standard, contains the herpes simplex virus TK promoter regionupstream of the Renilla luciferase gene (Promega).

For transfections, 100,000 cells were plated into six-well plates 20 h beforetransfection with 1.5 �g of p�-IFN-fl-lucter; 0.5 �g of pTK-rl-lucter; 0.5 �g ofplasmids expressing RIG-I and MDA-5; 1.5 �g of plasmids expressing V (whoseC ORF is closed with a stop codon), C1-204 or C1-23-Tom-C24-204 (or C*),NS1(1-77), wt E3L, mutant E3L(100-190), or RIG-� proteins (as indicated); andTransIT-LT1 transfection reagent (Mirus). At 24 h posttransfection, the cellswere (or were not) infected with various SeV stocks or transfected with 5 �g ofpoly(I-C) using TransIT-LT1 transfection reagent. Twenty hours later, cells wereharvested and assayed for firefly and Renilla luciferase activity (dual-luciferasereporter assay system; Promega). Relative expression levels were calculated bydividing the firefly luciferase values by those of Renilla luciferase.

Immunoblotting. Cytoplasmic extracts were prepared using 0.5% NP-40buffer. Equal amounts of total proteins were separated by sodium dodecyl sul-fate-polyacrylamide gel electrophoresis and transferred onto Immobilon-P mem-branes by semidry transfer. The secondary antibodies used were alkaline phos-phatase-conjugated goat anti-rabbit (or mouse) immunoglobulin G (Bio-Rad).The immobilized proteins were detected by light-enhanced chemiluminescence(Pierce) and analyzed in a Bio-Rad light detector using Quantity One software.

In vitro synthesis of RNA, purification, and transfection. DNA for T7 RNApolymerase synthesis of model RNA1 was prepared by PCR using the followingpartially complementary primers: 5�-TAATACGACTCACTATA(ggg/gca)ACACACCACAACCAACCCACAAC-3� (forward) (start sites are in lowercase type)and 5�-GAAAGAAAGGTGTGGTGTTGGTGTGGTTGTTGTGGGTTGGTTGTGG-3� (reverse). In vitro transcription was performed on 100 pmol ofpurified PCR product using T7 MEGAshortcript from Ambion according to themanufacturer’s instructions. For RNA1 containing the unusual OHGCA startsite, RNA was initiated with the dinucleotide 5� OHGpC24 in a reaction withoutGTP. For RNA1 containing the usual pppGGG start site, part of the product wastreated with 20 units of calf intestinal phosphatase (Roche) for 30 min at 37°C

followed by proteinase K treatment (15 min at 37°C), phenol extraction, andethanol precipitation. The T7 transcripts were purified on NucAway Spin col-umns from Ambion (to remove unincorporated nucleotides). SeV trailer pppR-NAs were synthesized similarly by using specific PCR primers.

For RNA transfection, 1 �g (1�) to 3 �g (3�) of RNA was transfected intoMEF cells using TransMessenger transfection reagent (QIAGEN).

RT and real-time PCR via TaqMan. Confluent MEFs in 10-cm petri dishes(107 cells) were infected with 20 PFU/cell of SeV GFP(�), SeV GFP(�), or bothstocks. At 24 h postinfection (hpi), the cells were collected and lysed in 300 �l ofNP-40 lysis buffer. Cytoplasmic extracts were then centrifuged in a 20 to 40%(wt/wt) CsCl density gradient (16 h at 35,000 rpm at 12°C). The pellet RNAswere ethanol precipitated and resuspended in 50 �l of Tris-EDTA. Fifteenmicrograms of RNA was then mixed with 0.5 �g of the forward or the reverseGFP primer and subjected to a Superscript reverse transcription (RT) reaction,according to instructions provided by the manufacturer (Gibco), in a total vol-ume of 50 �l. Five microliters of each cDNA was then combined with 12 �lMasterMix (Eurogentec), 20 pmol (each) of forward and reverse primers, and 4.4pmol of TaqMan probe in a total volume of 25 �l. The following primers andprobes (Eurogentec or Microsynth) were used for RT and TaqMan analyses ofthe GFP gene: 5�-CCGACAACCACTACCTGAGCAC-3� (forward), 5�-GAACTCCAGCAGGACCATGTG-3� (reverse), and 5�-AAAGACCCCAACGAGAAGCGCGA-3� (probe). Real-time PCR was carried out in duplicates using a 7700sequence detector (Applied Biosystems).

RESULTS

Three ways to activate IFN-�. We have used three ways toinduce the activation of an IFN-� promoter expressing a lucif-erase reporter gene in MEFs (see Fig. 2A). The first is tosimply transfect a synthetic dsRNA, poly(I):poly(C) [poly(I-C)], into the cells. The second way is to infect the cells with anSeV stock that contains a well-characterized copyback DI ge-nome (H4) (30). The third way is to coinfect cells with SeV-GFP(�), which expresses a GFP transgene, and SeV-GFP(�),which expresses mRNA containing the complement of theGFP ORF, as recently described for vesicular stomatitis virus(VSV) (24). As shown in Fig. 1A, infection with increasingamounts of SeV-GFP(�) alone leads to increasing GFP ex-pression. Coinfection of 20 PFU/cell of SeV-GFP(�) withincreasing amounts of SeV-GFP(�) leads to the gradual de-crease of GFP expression (top). At 20 PFU/cell of SeV-GFP(�), there are roughly equal amounts of GFP and anti-GFP mRNAs intracellularly (by strand-specific quantitativeRT-PCR) (Fig. 1B and see Materials and Methods), and thereis a 90% loss of GFP expression (Fig. 1A, top). This loss ofGFP expression cannot be accounted for by the reduced levelof GFP mRNA (Fig. 1B). In contrast, coinfection with increas-ing amounts of SeV expressing RFP as a neutral control (SeV-RFP) has a reduced ability to interfere with GFP expression(Fig. 1A). More importantly, whereas infection with SeV-GFP(�) alone or its coinfection with SeV-RFP leads to little

FIG. 1. IFN-� activation induced by SeV-GFP(�/�) infections. (A) Parallel cultures of MEFs were first transfected with pIFN�-lucff andpTK-lucr and then infected with increasing amounts of either SeV-GFP(�) alone (which expresses a GFP mRNA from a transgene between theM and F genes) or 20 PFU/cell of SeV-GFP(�) plus increasing amounts of either SeV-GFP(�) (which expresses an anti-GFP mRNA from atransgene in the same location) or SeV-RFP (which expresses an RFP mRNA from a transgene in the same location), as indicated. GFP expressionwas monitored by fluorescence-activated cell sorter analysis at 20 hpi. Cell extracts were prepared at 20 hpi, and equal amounts were used todetermine luciferase activities (below). These transfections were carried out three times with independent virus stocks, with similar results.(B) Cytoplasmic extracts were centrifuged on CsCl density gradients to isolate nonencapsidated (pellet) RNAs. The levels of GFP and anti-GFPmRNAs in 15 �g of CsCl pellet RNA were determined using sense- and antisense-specific primers for RT, followed by quantitative PCR (TaqMan)(see Materials and Methods). (C) Parallel cultures of MEFs were first transfected with the luciferase reporter plasmids plus either an empty vector,one expressing wt E3L(100–190), or one expressing mutant E3L(100–190) (E3L-mut.) and then infected with increasing amounts of SeV-GFP(�)and SeV-GFP(�) as indicated. Cell extracts were prepared at 20 hpi, and equal amounts were used to determine luciferase activities. Equalamounts of cell extracts were also Western blotted using anti-N and anti-HA (below).

VOL. 81, 2007 ACTIVATION OF IFN-� PROMOTER BY SeV INFECTION 12229

at Bibliotheque F

aculte Medecine G

eneve on Novem

ber 9, 2007 jvi.asm

.orgD

ownloaded from

Page 86: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

12230

at Bibliotheque F

aculte Medecine G

eneve on Novem

ber 9, 2007 jvi.asm

.orgD

ownloaded from

Page 87: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

or no activation of IFN-� (Fig. 1A), coinfection with SeV-GFP(�) clearly activates the IFN-� promoter (Fig. 1A, bot-tom). This IFN-� activation is inhibited by the coexpression ofthe dsRNA-binding domain of the vaccinia virus E3L protein,whereas this activation is unaffected by a mutant form of E3Lcontaining two point mutations that eliminate the binding ofdsRNA (10) (Fig. 1C). Taken together, our results show thatonly SeV coinfections that can form GFP dsRNA induceIFN-� activation.

The two SeV infections that activate IFN-� differ from eachother in several respects. First, DI-H4 genomes are of thecopyback variety and contain the strong antigenomic replica-tion promoter at their 3� ends. DI-H4 genomes thus have astrong competitive advantage in replication over nondefective(ND) genomes, and this sometimes leads to less viral structuralproteins like N and P being present intracellularly (see, e.g., Pprotein in Fig. 3A), but sometimes, this difference is minimal(see, e.g., Fig. 2A). However, in either case, viral V and Cproteins are almost entirely absent in these DI-H4-infectedcells, whereas V and C are found at wild-type levels inGFP(�/�) infections (Fig. 2 and data not shown). Both ofthese viral proteins are thought to limit IFN-� activation dueto virus infection (15, 31). Second, the DI genome replicationpromoters, like those of the ND genomes, are always active inthe presence of viral polymerase, and short 5�-triphosphory-lated trailer RNAs (rather than full-length DI genomes) aretranscribed by a relatively nonprocessive polymerase, espe-cially when the N protein is limiting. Unlike genome synthesisthat is dependent on ongoing (N) protein synthesis, that oftrailer RNA actually increases when translation is blocked withcycloheximide (18). SeV trailer RNAs are known to specificallybind to TIAR, a cellular RNA binding protein of the ELAVfamily (4), and to prevent virus-induced apoptosis (7, 12).DI-H4 infections are expected to overproduce trailer RNAs,which may stimulate RIG-I (11, 25), similar to measles virusleader RNA (26). Lastly, DI-H4-infected cells also containsmall amounts of unencapsidated H4 genome RNA that canself-anneal in a concentration-independent manner to formdsRNA panhandles with 5�-triphosphorylated ends. SeV-GFP(�/�)-infected cells, on the other hand, can form dsRNAswith capped ends.

Relative contributions of mda-5 and RIG-I in sensing SeVinfections in MEFs. mda-5 signaling to IFN-� was discoveredbecause the PIV5 V protein was found to bind this helicase andthereby prevent poly(I-C)-induced IFN-� activation. Furtherwork showed that the V–mda-5 interaction is a general prop-erty of paramyxovirus V proteins, including that of SeV (1, 5).

Nevertheless, several groups have now found that SeV infec-tion is sensed by RIG-I (and not mda-5) (13). To determinewhether RIG-I was also responsible for signaling to the IFN-�promoter in our MEFs, we examined the effect of expressing adominant-negative form of RIG-I [RIG-I(�CARDs), whoseN-terminal CARD domains are deleted] (Fig. 2A). MEFs werefirst transfected with pIFN�-lucff, pTK-lucr, and plasmids ex-pressing either RIG-I(�CARDs) or an empty vector as a neg-ative control. After 24 h, the cells were either transfected withpoly(I-C) or infected with either SeV-DI-H4 or SeV-GFP(�/�),and luciferase levels were determined 24 h later. As shown inFig. 2A, both SeV infections strongly activated the IFN-� pro-moter, whereas transfected poly(I-C) had a more modest effectin this experiment. RIG-I(�CARDs) coexpression did not af-fect the levels of P, V, and C proteins found intracellularly(bottom), but this coexpression reduced IFN-� activation tobackground levels in all three cases.

To determine whether the loss of IFN-� activation by RIG-I(�CARDs) coexpression was due to its ability to also inhibitmda-5 signaling, e.g., by sequestering cytoplasmic viral RNAs,we examined whether RIG-I(�CARDs) could inhibit mda-5signaling. As mda-5 and RIG-I can be activated by simpleoverexpression, we examined the effect of RIG-I(�CARDs)expression on IFN-� activation due to the overexpression ofthese two helicases. As shown in Fig. 2B, IFN-� activationclearly occurred upon exogenous mda-5 or RIG-I expression.Moreover, whereas RIG-I(�CARDs) coexpression completelyinhibited activation due to exogenous RIG-I, RIG-I(�CARDs)coexpression had little effect in countering IFN-� activation dueto exogenous mda-5. In contrast, the coexpression of a dominant-negative form of mda-5 completely inhibited IFN-� activationdue to mda-5 overexpression (Fig. 2B). Taken together, theseresults suggest that IFN-� activation in our MEFs in response tothese SeV infections is predominantly, if not exclusively, due tothe action of RIG-I.

The coexpression of either SeV V or C proteins stronglyinhibited IFN-� activation due to RIG-I overexpression,whereas only the V protein strongly inhibited IFN-� activationdue to mda-5 overexpression (Fig. 2B). The finding that SeV Vinhibits RIG-I signaling as well as that of mda-5 is consistentwith data from Childs et al. (5), who reported that SeV V wasa possible exception to the rule that all V proteins inhibitedmda-5 but not RIG-I. They reported that SeV V did in factmodestly inhibit RIG-I (35%), whereas all other V proteinshad no effect.

SeV V and C inhibition of SeV-DI-H4 and SeV-GFP(�/�)induced IFN-� activation. We next examined the abilities of

FIG. 2. Relative contributions of mda-5 and RIG-I in sensing SeV infections in MEFs. (A) Parallel cultures of MEFs were first transfected withpIFN�-lucff, pTK-lucr, and plasmids expressing dominant-negative RIG-I(�CARDs) or an empty vector as a negative control. After 24 h, the cellswere either transfected with poly(I-C) or infected with either SeV-DI-H4 or SeV-GFP(�/�) (as indicated). Cytoplasmic extracts were preparedafter a further 20 h of incubation and used to determine firefly and Renilla luciferase levels and the relative levels of RIG-I(�CARDs) (anti-Flag)and viral proteins (anti-P/V/C) by Western blotting (bottom). All transfections were carried out in duplicate, and the range of values obtained isindicated by the error bars. Ctrl., control. (B) Parallel cultures of MEFs were transfected with pIFN�-lucff, pTK-lucr, and plasmids expressingFlag-tagged RIG-I or mda-5 or these helicases plus RIG-I(�CARDs), mda-5(�CARDs), SeV V (whose overlapping C ORF was closed by a stopcodon), SeV C, or an empty plasmid as a negative control, as indicated. Cytoplasmic extracts were prepared after 40 h of incubation and used todetermine firefly and Renilla luciferase levels. All transfections were carried out in duplicate, and the range of values obtained is indicated by errorbars. The relative levels of the Flag–RIG-I constructs were determined by Western blotting with anti-Flag, those of the Flag–mda-5 constructs weredetermined with anti-mda-5, and those of the viral V and C proteins were determined with anti-P/V/C serum (bottom). Vect., vector.

VOL. 81, 2007 ACTIVATION OF IFN-� PROMOTER BY SeV INFECTION 12231

at Bibliotheque F

aculte Medecine G

eneve on Novem

ber 9, 2007 jvi.asm

.orgD

ownloaded from

Page 88: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

the SeV V and C proteins to inhibit IFN-� activation inducedby SeV-DI-H4 and SeV-GFP(�/�) infections. As shown inFig. 3A, exogenous expression of the SeV V protein did notaffect the level of viral P, V, and C proteins in SeV infections,but it did reduce IFN-� activation due to DI-H4 infection (by60%). Remarkably, SeV V overexpression did not inhibitIFN-� activation due to SeV-GFP(�/�) infection. The coexpres-sion of exogenous SeV C protein (actually C1–23-Tom-C24–204,

which migrates just slightly slower than the viral P protein) sim-ilarly did not affect the level of viral P, V, and C proteins in SeVinfections. C overexpression, however, more strongly inhibitedIFN-� activation due to either SeV infection [DI-H4-inducedactivation was reduced by 90%, and GFP(�/�)-induced acti-vation was reduced by 75%]. Coexpression of the unmodified Cprotein produced similar results (Fig. 2B and data not shown).The ability of the GFP(�/�) infection to activate IFN-�, despite

FIG. 3. SeV V and C inhibition of IFN-� activation induced by SeV infections. (A) Parallel cultures of MEFs were transfected with pIFN�-lucff,pTK-lucr, and plasmids expressing the SeV V protein, the SeV C protein (actually C1–23-Tom-C24–204), or unmodified Tom as a negative control.After 24 h, the cells were infected with either SeV-DI-H4 or SeV-GFP(�/�) (as indicated). Cytoplasmic extracts were prepared after a further24 h of incubation and used to determine firefly and Renilla luciferase levels. The relative levels of viral P, V, and C proteins were determined byWestern blotting with anti-P/C/V serum (bottom). All transfections were carried out in duplicate, and the range of values obtained is indicated byerror bars. (B) Parallel cultures of MEFs were transfected with pIFN�-lucff, pTK-lucr, and plasmids expressing either Tom, RIG-I(�CARDs), IAVNS1(1–73), C1–23-Tom-C24–204 (C*), or V, as indicated. After 24 h, the cells were infected with increasing amounts of SeV-DI-H4 (1�, 2�, and4�). Cytoplasmic extracts were prepared after a further 24 h of incubation and used to determine luciferase levels.

12232 STRAHLE ET AL. J. VIROL.

at Bibliotheque F

aculte Medecine G

eneve on Novem

ber 9, 2007 jvi.asm

.orgD

ownloaded from

Page 89: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

normal levels of expression of the V and C proteins, is presumablydue to the early formation of GFP dsRNA. In this case, the SeVV protein is considerably less potent than C in preventing theresponse to this dsRNA.

As the DI-H4 infections accumulate so few V and C pro-teins, we compared the abilities of these proteins (expressedfrom plasmids) to inhibit IFN-� activation relative to RIG-I(�CARDs) and the dsRNA binding domain of the influenzaA virus (IAV) NS1(1–73), another viral protein that inhibitsRIG-I signaling (22, 25). As shown in Fig. 3B, the SeV Cprotein was as active as RIG-I(�CARDs) in combating anincreasing dose of DI-H4 infection and almost as active asNS1. Consistent with above-described results (Fig. 3A), theSeV V protein was less active than C but was still able to inhibitmost of the DI-H4-induced IFN-� activation.

The SeV C1–204 protein is composed of two domains: theN-terminal 23 amino acids (C1–23) which act as a plasma mem-brane (PM) targeting signal (19) and which is present in thelonger (C�/C) but not in the shorter (Y1/Y2) “C” proteins, andC24–204, or the Y1 protein, which acts as a protein interactiondomain. Whereas C24–204 (or Y1) is naturally expressed duringinfection, C1–23 is only found fused to Y1. In order to study thedifferent contributions of these two domains to C-protein func-tion, we have used tomato red fluorescent protein (Tom) inwhich C1–23 is fused to the N terminus of Tom and C24–204 isfused to its carboxy terminus as a carrier. The interposition ofTom between these two domains of C remarkably does notappear to affect any of the activities of C1–204 (19). MEFs weretransfected with the luciferase reporter plasmids along withplasmids expressing various Tom constructs as indicated (Fig.4). After 24 h, half of the cultures were infected with SeV-DI-H4, and luciferase levels were determined after a further 24 h.The expression of C1–23-Tom, which carries the wt PM anchor

and is localized at the cell surface, or P8P9-Tom, which carriesa mutant PM anchor and is distributed throughout the cyto-plasm (19), had little or no effect on the DI-H4-induced IFN-�activation. In contrast, both C1–23-Tom-C24–204 and P8P9-Tom-C24–204 reduced IFN-� activation to near-background levels.C24–204 alone (Tom-C24–204), moreover, was still quite active inthis respect (Fig. 4). Thus, the C24–204 or Y1 protein interac-tion domain appears to be responsible for inhibiting RIG-I-dependent IFN-� activation, and this inhibition is largelyindependent of whether C24–204 is localized at the PM.

SeV C protein inhibits IFN-� activation induced by trans-fected pppRNA. A general property of nonsegmented nega-tive-strand RNA viruses is that short, promoter-proximalpppRNAs (leader and trailer RNAs) are transcribed fromtheir replication promoters, especially when unassembled Nprotein is limiting (17, 18). The ability of SeV infections toinduce IFN is essentially due to the presence of DI genomesthat are present in their egg-grown stocks, especially those ofthe copyback variety (30). As mentioned above, copyback DIgenomes have a strong replicative advantage because they con-tain strong replication promoters at the DI genome and anti-genome 3� ends. Copyback DI genome replication thus gener-ates short trailer RNAs that are unmodified at either end andcan be considered as unstable, abortive replication products(see Discussion).

To examine whether trailer RNAs act as pathogen-associ-ated molecular patterns (PAMPs), we transfected trailer RNAmade by T7 RNA polymerase in vitro into our MEFs andmonitored the activation of IFN-�. As the ability of pppRNAsto induce IFN-� activation is not sequence dependent (11), wealso examined model RNAs that were initiated with GTP butthen treated with phosphatase or those initiated with the dinu-cleotide GpC rather than pppG (23). The transfections of all

FIG. 4. SeV C24–204 (or Y1) protein inhibits IFN-� activation induced by DI-H4 infection. Parallel cultures of MEFs were transfected withpIFN�-lucff, pTK-lucr, and plasmids expressing tomato constructs carrying either the wt (C1–23) or mutant (P8P9) C1–23 fused to their N termini,with and without C24–204 (or Y1) fused to their carboxy termini, C24–204 fused to the carboxy terminus alone (Tom-C24–204), or unmodified Tomas a negative control (ctrl.), as indicated. After 24 h, the cells were infected with SeV-DI-H4 (as indicated). Cytoplasmic extracts were preparedafter a further 24 h of incubation and used to determine luciferase levels. The relative levels of the various tomato constructs were determined byWestern blotting with anti-dsRED (bottom). All transfections were carried out in duplicate, and the range of values obtained is indicated by errorbars.

VOL. 81, 2007 ACTIVATION OF IFN-� PROMOTER BY SeV INFECTION 12233

at Bibliotheque F

aculte Medecine G

eneve on Novem

ber 9, 2007 jvi.asm

.orgD

ownloaded from

Page 90: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

FIG. 5. pppRNA-induced activation of IFN-�. (A) Parallel cultures of MEFs were transfected with pIFN�-lucff and pTK-lucr, and pRIG-I wasalso transfected in some cultures, as indicated. After 24 h, the cells were transfected for 3 h with increasing amounts (1 or 3 �g) of eitherpppGGG/RNA1, phosphatase-treated GGG/RNA1, pppGCA/RNA1, or OHGCA/RNA1, as indicated. Cytoplasmic extracts were prepared 18 hpost-RNA transfection and used to determine luciferase levels. (B) Parallel cultures of MEFs were transfected with pIFN�-lucff, pTK-lucr, andplasmids expressing Tom, RIG-I(�CARDs), IAV NS1(1–73), C1–23-Tom-C24–204 (C*), or V, as indicated. After 24 h, the cells were transfected withincreasing amounts (1 �g and 3 �g) of pppGGG/RNA1, as indicated. Cytoplasmic extracts were prepared after 3 h of RNA transfection and usedto determine luciferase levels. Rel., relative. (C) Same as above (B), except that the cells were transfected with 3 �g of ppptrailer RNA.

12234 STRAHLE ET AL. J. VIROL.

at Bibliotheque F

aculte Medecine G

eneve on Novem

ber 9, 2007 jvi.asm

.orgD

ownloaded from

Page 91: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

three 5�-triphosphorylated ssRNAs clearly led to IFN-� acti-vation, whereas both RNAs that contained 5�-hydroxyl endshad essentially lost their ability to activate IFN-� in paralleltransfections (Fig. 5A and C), confirming previously reportedresults (11, 25, 26). We then examined the ability of the SeV Cand V proteins to inhibit 5�-pppRNA-dependent activation ofIFN-� compared to RIG-I(�CARDs) and IAV NS1(1–73).MEFs were first transfected with plasmids expressing variousviral inhibitory proteins or an empty plasmid as a negativecontrol and then transfected with increasing amounts ofpppRNAs. IFN-� activation was monitored after a further 18 h.As shown in Fig. 5B, expression of the SeV C protein was asactive as RIG-I(�CARDs) in inhibiting IFN-� activation at allamounts of pppRNAs transfected although not quite as activeas NS1(1–73). Expression of SeV V was again the least inhib-itory; in fact, significant inhibition occurred only at the lowestamount of pppRNA. Thus, short 5�-triphosphorylated ssRNAssuch as trailer RNA are potent stimulators of IFN-� whentransfected into our MEFs, and expression of the SeV C pro-tein (but not the SeV V protein) can effectively inhibit thisstimulation (Fig. 5B and C).

Relative importance of C and V in inhibiting RIG-I-depen-dent signaling to IFN-�. Another way to investigate the rela-tive importance of the C and V proteins in inhibiting RIG-I-dependent signaling to IFN-� is to compare the relativeabilities of SeV infections that cannot specifically express the Cor V proteins to affect pppRNA- or poly(I-C)-induced IFN-�

activation. MEFs were therefore first infected with 20 PFU/mlof either wt SeV, SeV-Vminus (containing a stop codon in the VORF just downstream of the mRNA editing site, which pro-duces a W-like protein instead of V), or SeV-Cminus (contain-ing three stop codons in the C ORF downstream of the Y2initiation codon). The infected cells were then transfected (at24 hpi) with pIFN-�-luc plus either pppRNA, poly(I-C), or noRNA and then harvested after 18 h to determine reporter geneactivity. As shown in Fig. 6, these three SeVs replicate toclearly different levels in our highly IFN-competent MEFs(even though they replicate similarly in BSR T7, 293T, andVero cells), highlighting the essential functions that these ac-cessory proteins play in countering the innate immune re-sponse. Nevertheless, in the absence of transfected RNA, onlySeV-Cminus infection activates IFN-� to any extent or increasesRIG-I levels; RIG-I is an IFN-stimulated gene, and its levelreflects that of the antiviral state (Fig. 6, bottom). Transfec-tions of either pppRNA or poly(I-C) strongly activated thereporter gene and increased RIG-I levels. Prior infection witheither SeV wt or SeV-Vminus reduced transfected RNA stim-ulation of the reporter gene and prevented the increase inRIG-I levels (Fig. 6). SeV-Vminus was only slightly less effectivethan wt SeV in this respect. In sharp contrast, prior infectionwith SeV-Cminus not only did not prevent the increase in RIG-Ilevels but also acted synergistically with either pppRNA orpoly(I-C) transfection to increase reporter gene activity byincreasing the level of RIG-I. These results reinforce the view

FIG. 6. RNA-induced activation of IFN-� in cells infected with SeV that cannot express either V or C. Parallel cultures of MEFs were eithermock infected or infected with 20 PFU/ml of wt SeV, SeV-Vminus, or SeV-Cminus, as indicated. After 24 h, the cells were transfected with luciferasereporter plasmids and either pppGGG/RNA1, poly(I-C), or no RNA (untreated). Cytoplasmic extracts were prepared after 18 h of RNAtransfection, used to determine luciferase levels (above), and Western blotted to determine the levels of P, V, and C proteins as well as endogenousRIG-I and actin as a loading control. rSeV, recombinant SeV.

VOL. 81, 2007 ACTIVATION OF IFN-� PROMOTER BY SeV INFECTION 12235

at Bibliotheque F

aculte Medecine G

eneve on Novem

ber 9, 2007 jvi.asm

.orgD

ownloaded from

Page 92: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

that it is primarily the SeV C protein (and not V) that inhibitspppRNA- and dsRNA-induced signaling to the IFN-� pro-moter via RIG-I during SeV infection.

DISCUSSION

SeV has been one of the most extensively used model virusesto investigate IFN induction in infected cells. Most of this workhas used the commercially available Cantell strain of SeV,whose ability to induce IFN, like that of other SeV strains, isdue to the presence of DI genomes in egg-grown stocks. Non-defective SeVs that are plaque purified from these stocks,including that of the Cantell strain, do not induce IFN unlesscellular RIG-I levels are artificially increased (21, 30). Fornondefective SeV infection, the expression of the C and Vproteins is apparently sufficient to prevent IFN-� activationunder normal conditions. Measles virus infection, in con-trast, can apparently induce IFN in the absence of DI ge-nomes, and evidence that this induction is due to the syn-thesis of leader pppRNAs has recently been provided (26).Leader and trailer RNAs, which are unmodified at eitherend, are unstable in infected cells unless they are encapsi-dated with the N protein (presumably after their synthesis asfree RNAs) (2). Free leader and trailer RNAs are moreeasily detected in VSV infections, which synthesize largeramounts of viral RNAs over a shorter period of time. Fornondefective VSV infections, eight times as many trailerRNAs/antigenome template are found as leader RNAs/ge-nome template, consistent with the relative strengths oftheir respective replication promoters. For VSV copybackDI infections, there are 40 times as many trailer RNAs/template (nondefective antigenome plus DI genome) asleader RNAs/genome template, presumably reflecting theincreased strength of the copyback DI replication promot-ers. The VSV polymerase clearly has a strong preference forinitiating RNA synthesis at the 3� ends of copyback DIgenomes over both ND genomes and antigenomes (17).

We previously noted that not all SeV stocks that contain DIgenomes strongly induce IFN; this ability appears to be re-stricted to stocks containing relatively small copyback DI ge-nomes (the smaller the DI genomes, the more moles of endsare present for a given weight). The commercially availableCantell strain contains a copyback DI genome of only 546nucleotides in length, the smallest SeV DI genome describedto date (30). SeV copyback DI-H4 genomes (1,410 nucleo-tides) have the same strong replicative advantage as their VSVcounterparts because they also contain strong replication pro-moters at both their genome and antigenome 3� ends. Thus,SeV copyback DI infections presumably synthesize consider-ably more pppRNAs than standard virus infections. We alsopreviously noted that when cytoplasmic extracts of DI-H4-infected cells are centrifuged on CsCl density gradients, smallamounts of DI genome RNA are found in the pellet fraction(30). This indicates that this RNA is not encapsidated with theN protein and therefore forms dsRNA panhandles in a con-centration-independent manner. Thus, copyback DI-H4 infec-tions apparently produce considerably more of both knownPAMPs of RNA virus infection than do standard virus infec-tions. Coupled with their strongly reduced accumulation of the

viral C and V proteins, it is easy to see why these copyback DIinfections are such potent inducers of IFN-�.

All paramyxoviruses express either C or V proteins, andmany viruses express both. In viruses that express only C or V,we presume that either viral protein alone counteracts theinnate immune response of the host to aid virus replication.The C and V proteins, which bear no sequence similarity, likelytarget different key elements of the host response. Viruses thatexpress both C and V presumably have more diverse ways ofcountering innate immunity. In support of this notion, SeVinfections that cannot specifically express either the C or Vprotein contain increased levels of IFN-� and interleukin-8mRNAs relative to wt SeV infections (31), and the indepen-dent expression of the C or V protein will inhibit poly(I-C)- orNewcastle disease virus-dependent activation of IRF-3 (15).Previous work has identified mda-5 as being a key target ofparamyxovirus V proteins in countering the innate immuneresponse (1). This paper provides evidence that for SeV infec-tion of MEFs, it is the C protein (and not V) that is primarilyresponsible for this effect and that C acts by countering RIG-I-dependent signaling to IFN-�. For example, the independentexpression of either C or V inhibited IFN-� activation due toRIG-I overexpression (Fig. 2B). Also, both proteins inhibitedIFN-� activation due to DI-H4 infection, although C was al-ways more effective here than V (Fig. 3). However, only Cexpression effectively inhibited IFN-� activation due toGFP(�/�) infection (Fig. 3) or transfected poly(I-C) or pp-pRNAs (Fig. 5). Perhaps the strongest indication that the Cproteins are primarily responsible for countering the innateimmune response comes from experiments with SeV that can-not specifically express the C or V proteins. SeV-Cminus infec-tion not only cannot prevent the effects of transfectedpoly(I-C) or pppRNAs on IFN-� activation but also synergis-tically enhances these effects. SeV-Vminus infection, in contrast,behaves mostly like wt SeV infection and counteracts the ef-fects of transfected poly(I-C) or pppRNAs (Fig. 6).

Finally, we note that poly(I-C) (made with polynucleotidephosphorylase that generates 5� diphosphate ends and which istransfected into cells) and the presumed GFP dsRNA (that isdirectly generated in the cytoplasm via the viral transcriptaseand which contains capped 5� ends) both activate IFN-� viaRIG-I in MEFs. Thus, the ability of dsRNA to induce RIG-Isignaling does not depend on the manner in which it is intro-duced into this cell compartment, nor is it peculiar to thepresence of 5�-diphosphorylated ends that are not normallyfound in cells and could theoretically act as PAMPs. Moreover,in either case, the activation of IFN-� by these dsRNAs isinhibited by the SeV C protein and not V, presumably becausethis signaling passes through RIG-I and not mda-5. It appearsthat our MEFs contain insufficient amounts of mda-5 to senseSeV infection, as these MEFs respond well to the expression ofplasmid-derived mda-5 (Fig. 2B). This conclusion is also con-sistent with our finding that three different rubulavirus V pro-teins that are known to counteract poly(I-C)-induced mda-5signaling were unable to inhibit IFN-� activation in response toSeV-DI-H4 infection (data not shown). We expect that theSeV V protein will be more important in countering the innateimmune response in other cells in which mda-5 functions as aPAMP recognition receptor.

12236 STRAHLE ET AL. J. VIROL.

at Bibliotheque F

aculte Medecine G

eneve on Novem

ber 9, 2007 jvi.asm

.orgD

ownloaded from

Page 93: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

ACKNOWLEDGMENTS

We thank J. Tschopp (Lausanne), T. Fujita (Kyoto), J. Perrault (SanDiego), B. Jacobs (Arizona), K. Conzelmann (Munich), and R. Randall(St. Andrews) for sharing materials.

This work was supported by the Swiss National Science Fund.

REFERENCES

1. Andrejeva, J., K. S. Childs, D. F. Young, T. S. Carlos, N. Stock, S. Goodbourn,and R. E. Randall. 2004. The V proteins of paramyxoviruses bind the IFN-inducible RNA helicase, mda-5, and inhibit its activation of the IFN-beta pro-moter. Proc. Natl. Acad. Sci. USA 101:17264–17269.

2. Blumberg, B. M., M. Leppert, and D. Kolakofsky. 1981. Interaction of VSVleader RNA and nucleocapsid protein may control VSV genome replication.Cell 23:837–845.

3. Bontron, S., C. Ucla, B. Mach, and V. Steimle. 1997. Efficient repression ofendogenous major histocompatibility complex class II expression throughdominant negative CIITA mutants isolated by a functional selection strategy.Mol. Cell. Biol. 17:4249–4258.

4. Brennan, C. M., and J. A. Steitz. 2001. HuR and mRNA stability. Cell. Mol.Life Sci. 58:266–277.

5. Childs, K., N. Stock, C. Ross, J. Andrejeva, L. Hilton, M. Skinner, R.Randall, and S. Goodbourn. 2006. mda-5, but not RIG-I, is a common targetfor paramyxovirus V proteins. Virology 359:190–200.

6. Garcia-Sastre, A., and C. A. Biron. 2006. Type 1 interferons and the virus-host relationship: a lesson in detente. Science 312:879–882.

7. Garcin, D., G. Taylor, K. Tanebayashi, R. Compans, and D. Kolakofsky.1998. The short Sendai virus leader region controls induction of pro-grammed cell death. Virology 243:340–353.

8. Hiscott, J., R. Lin, P. Nakhaei, and S. Paz. 2006. MasterCARD: a pricelesslink to innate immunity. Trends Mol. Med. 12:53–56.

9. Hiscott, J., T. L. Nguyen, M. Arguello, P. Nakhaei, and S. Paz. 2006. Ma-nipulation of the nuclear factor-kappaB pathway and the innate immuneresponse by viruses. Oncogene 25:6844–6867.

10. Ho, C. K., and S. Shuman. 1996. Mutational analysis of the vaccinia virus E3protein defines amino acid residues involved in E3 binding to double-stranded RNA. J. Virol. 70:2611–2614.

11. Hornung, V., J. Ellegast, S. Kim, K. Brzozka, A. Jung, H. Kato, H. Poeck, S.Akira, K. K. Conzelmann, M. Schlee, S. Endres, and G. Hartmann. 2006.5�-Triphosphate RNA is the ligand for RIG-I. Science 314:994–997.

12. Iseni, F., D. Garcin, M. Nishio, N. Kedersha, P. Anderson, and D. Kolakofsky.2002. Sendai virus trailer RNA binds TIAR, a cellular protein involved invirus-induced apoptosis. EMBO J. 21:5141–5150.

13. Kato, H., O. Takeuchi, S. Sato, M. Yoneyama, M. Yamamoto, K. Matsui,S. Uematsu, A. Jung, T. Kawai, K. J. Ishii, O. Yamaguchi, K. Otsu, T.Tsujimura, C. S. Koh, C. Reis e Sousa, Y. Matsuura, T. Fujita, and S. Akira.2006. Differential roles of MDA5 and RIG-I helicases in the recognition ofRNA viruses. Nature 441:101–105.

14. King, P., and S. Goodbourn. 1994. The beta-interferon promoter responds topriming through multiple independent regulatory elements. J. Biol. Chem.269:30609–30615.

15. Komatsu, T., K. Takeuchi, J. Yokoo, and B. Gotoh. 2004. C and V proteins

of Sendai virus target signaling pathways leading to IRF-3 activation for thenegative regulation of interferon-beta production. Virology 325:137–148.

16. Lamb, R. A., and D. Kolakofsky. 2001. Paramyxoviridae: the viruses and theirreplication, p. 1305–1340. In D. M. Knipe and P. M. Howley (ed.), Fieldsvirology, 4th ed., vol. 1. Lippincott Williams & Wilkins, Philadelphia, PA.

17. Leppert, M., and D. Kolakofsky. 1980. Effect of defective interfering parti-cles on plus- and minus-strand leader RNAs in vesicular stomatitis virus-infected cells. J. Virol. 35:704–709.

18. Leppert, M., L. Rittenhouse, J. Perrault, D. F. Summers, and D. Kolakofsky.1979. Plus and minus strand leader RNAs in negative strand virus-infectedcells. Cell 18:735–747.

19. Marq, J. B., A. Brini, D. Kolakofsky, and D. Garcin. 2007. Targeting of theSendai virus C protein to the plasma membrane via a peptide-only mem-brane anchor. J. Virol. 81:3187–3197.

20. Matsuoka, Y., J. Curran, T. Pelet, D. Kolakofsky, R. Ray, and R. W. Compans.1991. The P gene of human parainfluenza virus type 1 encodes P and C proteinsbut not a cysteine-rich V protein. J. Virol. 65:3406–3410.

21. Meylan, E., J. Curran, K. Hofmann, D. Moradpour, M. Binder, R. Barten-schlager, and J. Tschopp. 2005. Cardif is an adaptor protein in the RIG-Iantiviral pathway and is targeted by hepatitis C virus. Nature 437:1167–1172.

22. Mibayashi, M., L. Martinez-Sobrido, Y. M. Loo, W. B. Cardenas, M. Gale,Jr., and A. Garcia-Sastre. 2007. Inhibition of retinoic acid-inducible geneI-mediated induction of beta interferon by the NS1 protein of influenza Avirus. J. Virol. 81:514–524.

23. Milligan, J. F., and O. C. Uhlenbeck. 1989. Synthesis of small RNAs using T7RNA polymerase. Methods Enzymol. 180:51–62.

24. Ostertag, D., T. M. Hoblitzell-Ostertag, and J. Perrault. 2007. Overproduc-tion of double-stranded RNA in vesicular stomatitis virus-infected cells ac-tivates a constitutive cell-type-specific antiviral response. J. Virol. 81:503–513.

25. Pichlmair, A., O. Schulz, C. P. Tan, T. I. Naslund, P. Liljestrom, F. Weber,and C. Reis e Sousa. 2006. RIG-I-mediated antiviral responses to single-stranded RNA bearing 5�-phosphates. Science 314:997–1001.

26. Plumet, S., F. Herschke, J. M. Bourhis, H. Valentin, S. Longhi, and D.Gerlier. 2007. Cytosolic 5�-triphosphate ended viral leader transcript of mea-sles virus as activator of the RIG I-mediated interferon response. PLoS ONE2:e279.

27. Poole, E., B. He, R. A. Lamb, R. E. Randall, and S. Goodbourn. 2002. TheV proteins of simian virus 5 and other paramyxoviruses inhibit induction ofinterferon-beta. Virology 303:33–46.

28. Rothenfusser, S., N. Goutagny, G. DiPerna, M. Gong, B. G. Monks, A.Schoenemeyer, M. Yamamoto, S. Akira, and K. A. Fitzgerald. 2005. TheRNA helicase Lgp2 inhibits TLR-independent sensing of viral replication byretinoic acid-inducible gene-I. J. Immunol. 175:5260–5268.

29. Stark, G. R., I. M. Kerr, B. R. Williams, R. H. Silverman, and R. D.Schreiber. 1998. How cells respond to interferons. Annu. Rev. Biochem.67:227–264.

30. Strahle, L., D. Garcin, and D. Kolakofsky. 2006. Sendai virus defective-interfering genomes and the activation of interferon-beta. Virology 351:101–111.

31. Strahle, L., D. Garcin, P. Le Mercier, J. F. Schlaak, and D. Kolakofsky. 2003.Sendai virus targets inflammatory responses, as well as the interferon-in-duced antiviral state, in a multifaceted manner. J. Virol. 77:7903–7913.

VOL. 81, 2007 ACTIVATION OF IFN-� PROMOTER BY SeV INFECTION 12237

at Bibliotheque F

aculte Medecine G

eneve on Novem

ber 9, 2007 jvi.asm

.orgD

ownloaded from

Page 94: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

V. ADDITIONAL DATA

“ THE IMPLICATION OF SEV NUCLEOCAPSID

IN THE INDUCTION OF THE IFN-β "

92

Page 95: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

Additional Data

The implication of SeV nucleocapsid in the induction of the IFN-β The success of the innate cellular defense to viral infection is dependent on the capacity of the

host to detect the presence of the invading pathogen. Upon infection, many cellular sensors

recognise different components of the virus and initiate in turn signal transduction cascades

(such as IRF-3) producing in the end cytokines, including IFNs. The RNA helicase RIG-1 has

been recently discovered to recognise viral RNA. RNA recognition leads to IRF-3 and NF-kB

activation and finally to the induction of type I IFNs and the antiviral state of the cell. To

circumvent the detection of their own RNA genome, the mononegalevirales possess the

nucleocapsid (NCs) that surrounds completely the RNA viral genomes (and antigenomes). It

is likely that their nucleocapsid never disassemble during genome expression. This protection

seems to be quite strong considerating that the RNA genomes (within the nucleocapsid) are

resistant to nuclease attack at any salt concentration and that the NC is very stable, as it

withstands the high salt and gravity forces of cesium chloride density gradient centrifugation

(Lamb and Kolakofsky, 1996). Further more, the N proteins are believed to be “sticky”, in the

way that they are tightly bound to the viral RNA genomes and can also encapsidate free RNA

independently of whether it comes from the virus or the cell.

It has been shown in 2002 that MeV nucleocapsid (NC) protein was the major component of

IRF-3 activation and triggered the induction of IFN (Tenoever et al., 2002). The ability to

activate IRF-3 during the course of infection has also been observed in other single stranded,

enveloped RNA viruses such as RSV, NDV, VSV and SeV (Casola et al., 2001; Servant et al.,

2001; Sundstrom et al., 2001). This suggests that the IRF-3 cascade could be involved in the

viral NC detection. Since NCs are the first viral elements that enter the cell, it is logical to

make the hypothesis that, like MeV, SeV NCs act as PAMPs and consequently induce

IFNβ activation upon SeV infection.

1) SeV N protein expressed alone does not activate IFNβ. When N is expressed alone

(in the absence of the viral P protein which forms a complex with N and prevent

“illegitimate” non-specific binding to cellular RNA), it is often found in high number

and aggregates together with non genome RNAs, which represent a bogus NCs. To

analyze the direct effect of the SeV N protein alone, MEF cells were first transfected

93

Page 96: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

with a plasmid containing a luciferase reporter gene under the control of the IFNβ

promoter and 24h later with a plasmid expressing SeV N protein (Fig. A). We have

examined the ability of the N protein to induce IFNβ activation after transfection and

compared the level of IFNβ activation with those of the copyback DI-H4 infection that

we used as a positive control. As shown in figure A, the expression of N protein alone

does not induce IFNβ activation. By contrast, DI-H4 infection activates stongly the

activation of the IFNβ in this experiment. These results show that under these

conditions, SeV viral N protein does not induce IFNβ activation.

0

10

20

30

40

50

60

70

80

ctl

SeV

-DI-H

4in

fect

ion

pEB

S_N

A.

Relative Luciferase

Activity

Figure A: Transfected SeV N proteins do not induce IFNβ activation. Parallel cultures of MEFs were first transfected with pIFNβ-lucff and pTK-lucr. After 24h, the cells were

either infected with SeV-DI-H4, or transfected with plasmids expressing SeV N protein (pEBS_N).

Cytoplasmic extracts were prepared after 20h of incubation, and used to determine firefly and renilla

luciferase levels.

2) Transfected encapsidated SeV (NCs) genomes induce IFNβ activation. To further

investigate the role of the NCs, 24 hours SeV infected cells were collected and the

NCs purified by CsCl centrifugation (cf. M&M). Two doses of purified SeV NCs (or

PolyI/C) were transfected into MEF cells and tested for their ability to activate IFNβ

by using the reporter plasmid in which the IFNβ promoter expresses a luciferase

reporter gene. Surprisingly, we observed that transfected purified NCs induce strongly

94

Page 97: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

IFNβ activation and that the transfection of 10μl of purified NCs (corresponding to

250000 infected cells) is almost as efficient as a DI-H4 infection. Moreover, we can

see that NCs transfection is more competent than the synthetic dsRNA (poly I/C)

treatment in activating IFNβ. These results suggest that induction of IFNβ requires the

presence of the NCs complex (fig. B).

0

0.1

0.2

0.3

0.4

0.5

0.6

0.7_

5ug

5ul

10ul

mock DI-H4 infection poly IC SeV NC

B.

Relative Luciferase

Activity

Figure B: Transfected NCs induce IFNβ activation.

Parallel cultures of MEFs were first transfected with pIFNβ-lucff and pTK-lucr. After 24h, the cells were

either infected with SeV-DI-H4, or transfected either with poly I/C (5ug) or purified SeV NCs (5-10ul)

isolated by CsCl density gradients centrifugation (cf.M&M). Cell extracts were prepared after further 20h

of incubation, and equal amounts were used to determine luciferase activities.

3) SeV C and V proteins expression decreases the IFNβ activation upon NCs

transfection. Since SeV C and V proteins are responsible of counteracting the host

innate immune response and because we have shown in paper three that IFNβ

activation upon DI-H4 infection was in part due to a downregulation of the viral C and

V proteins, we decided to over-express the V or C proteins in NCs-transfected MEF

cells. We observed that IFNβ activation was strongly reduced by C expression (4

folds), and more modestly by V expression (2 folds) (Fig C). Thus, the SeV V and C

proteins inhibit IFNβ activation induced by NCs transfection.

95

Page 98: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

0

50000

100000

150000

200000

- SeV-V

NCs - + - + - +

SeV-C Transfection -

Relative Luciferase

Activity

C.

Figure C: Over-expression of viral C and V proteins inhibits IFNβ activation induced by SeV NCs

transfection. Parallel cultures of MEFs were transfected with pIFNβ-lucff and pTK-lucr and plasmids

expressing the SeV V protein, or the SeV C protein. 24h post-transfection, the cells were transfected with

purified SeV NCs (5ul). Cytoplasmic extracts were prepared after further 20h of incubation, and used to

determine luciferase levels.

4) NCs from either SeV or DI-H4 infections induce IFNβ at the same level. Because

DI-H4 infection induces more strongly IFNβ activation compared to a ND SeV

infection, we decided to compare NCs coming from both infections and their ability to

induce IFNβ activation. Prior to transfection, the purified NCs were charged on a gel

and stained with Commassie Blue in order to compare the amount of N proteins from

each infection (SeV or DI-H4) (Fig D.). Because DI-H4 genome is 10 times smaller

than the ND genome (and thus contains 10 times more moles of ends), we asked

whether DI-H4 NCs could induce more IFNβ than a SeV NCs. Two different

concentrations of NCs were transfected (for both types of infection) into MEF cells

and IFNβ activation was monitored using the reporter plasmid carrying the IFNβ

promoter expressing the luciferase gene (Fig D). We observed that the level of IFNβ

activation is the same in both the SeV and the DI-H4 NCs transfections. These data

suggest that the IFNβ activation is not dependent on the number of moles of genome.

Furthermore, because the number of mole correlates with the number of 5’

96

Page 99: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

triphosphorylated extremities of the genome, we can suggest that the IFNβ activation

upon NCs transfection is not dependent on the number of 5’ppp ends of each genome.

To confirm this idea, we decided to remove the eventual triphosphates at the ends of

the genomes by treating the purified NCs with phosphatase (CIP) (cf.M&M). The NCs

were then transfected in MEF cells and the IFNβ activation was analyzed.

Interestingly, transfected NCs previously treated with CIP had no effect on the level of

IFNβ activation. This data suggest that 5’tri-phophorylated ends of the genomes

presumably do not act as a viral inducer of the IFNβ upon NCs transfection (Fig. E).

0

10

20

30

40

50

60

70

80

90

2,5ul 5ul 2,5ul 5ul

ctl NC SeV NC DI-H4

D.

NC SeV Z NC SeV H4

1ul 4ul 2ul 4ul

Relative Luciferase

Activity

Figure D: Transfections of purified NCs from SeV or DI-H4 infections induce IFNβ at the same level.

Parallel cultures of MEFs were first transfected with pIFNβ-lucff and pTK-lucr. After 24h, the cells were

transfected with the same amount of NCs (2.5-5ul) purified from either SeV or DI-H4 infections.

Cytoplasmic extracts were prepared after 20h of incubation, and used to determine luciferase levels. The

amount of viral N proteins after two CsCl gradient centrifugations was determined by SDS-PAGE and

Coomassie-blue staining and is represented in the upper panel.

5) RNase treatment shuts down the induction of IFNβ. In order to rule out the

possible RNA contamination of our purified NCs, we decided to treat the viral NCs

with the RNases T and A. After RNase treatment, the cells were transfected as usual

and challenged for the IFNβ activation. Unexpectedly, we can observe no more IFNβ

activation, suggesting that RNA products are presumably the ones responsible for the

strong induction of IFNβ upon NCs transfection (Fig. E).

97

Page 100: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

0

10

20

30

40

50

60

70

80

NCs - + + + CIP - - + - RNases - - - +

E.

NC SeV Z

Ctl CIP RNase A&T1

Relative Luciferase

Activity

Figure E: Effect of transfected NCs on the IFNβ activation after CIP and RNase treatment

Parallel cultures of MEFs were transfected with pIFNβ-lucff and pTK-lucr. 24h post-transfection, the cells

were transfected with purified SeV NCs (5ul) or with SeV NCs that were previously treated with CIP or

RNAse. Cytoplasmic extracts were prepared after further 20h of incubation, and used to determine

luciferase levels. The amount of viral N proteins after two CsCl gradient centrifugations was determined

by SDS-PAGE and Coomassie-blue staining and is represented in the upper panel.

CONCLUSION

We first wanted to determine whether viral NCs themselves could act as PAMPs. We found

that bona fide NCs (as opposed to N protein aggregates that form upon N expression in the

absence of viral infection) did indeed appear to activate IFNβ. This activation could be

inhibited by the viral C and V proteins, and did not appear to be influenced by the number of

5’ppp ends present in the preparation; and this for two reasons: first, the transfections of the

NCs from both the ND and DI-H4 did not show any difference in the level of IFNβ activation

and that phosphatase treatment had also no effect. Unexpectedly, we treated the NCs with

RNase as a control for the phosphatase treatment, and found that the IFNβ activation was shut

down. These results suggest that it is (cellular or viral) RNA trapped in the NCs that is

responsible for the IFNβ activation and not the NC itself.

98

Page 101: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

MATERIALS AND METHODS

Cells and viruses

Mouse embryo fibroblast cells (MEFs) were grown in Dulbecco's modified Eagle's medium

supplemented with 10% fetal calf serum.

SeV stocks were grown in the allantoic cavities of 9-days-old embryonated chicken eggs for 3

days at 33 °C. For nondefective stocks (109 pfu/ml), 0.1 ml of a 105 dilution (ca. 1000 pfu)

was inoculated per egg. In the case of DI stocks, 0.1 ml of a 103 dilution was used. In all

cases, the amount of viral proteins present in the resulting allantoic fluid was analyzed by

SDS-PAGE and Coomassie-blue staining of pelleted virus. Virus titers were determined by

plaquing on LLC-MK2 cells.

Plasmids, transient transfections, infections, luciferase assay

EBS plasmids expressing viral proteins were constructed by standard methods (Bontron et al.,

1997); the precise detail can be obtained from the authors.

pβ-IFN-fl-lucter, which contains the firefly luciferase gene under the control of the human

IFNβ promoter, is described in (King and Goodbourn, 1994). pTK-rl-lucter, used as a

transfection standard, contains the herpes simplex virus TK promoter region upstream of the

renilla luciferase gene (Promega).

For transfections, 100,000 cells were plated in six-well plates 20 h before transfection with

1.5 μg of pβ-IFN-fl-lucter, 0.5 μg of pTK-rl-lucter,1.5 μg of plasmids (EBS) expressing N. At

24 h post-transfection, the cells were (or not) infected with DI-H4 stocks or transfected with 5

μg of poly-I/C or with 5-10ul of purified SeV or DI-H4 NCs. (cf. M&M Purification and

analysis of nucleocapsid RNAs) using TransMessenger transfection reagent (QIAGEN).

20hrs later, cells were harvested and assayed for firefly and renilla luciferase activity (dual-

luciferase reporter assay system; Promega). Relative expression levels were calculated by

dividing the firefly luciferase values by those of renilla luciferase.

Purification of nucleocapsid RNAs and RNase/phosphatase treatments

Confluent MEFs in 10 cm Petri dishes (107 cells) were infected with 20 pfu/cell of SeV Z or

SeV-DI-H4. 24hpi, the cells were collected and lysed in 300 ul of NP40 lysis buffer.

Cytoplasmic extracts were then centrifuged in a 20–40% (w/w) CsCl density gradient

(16h/35,000 rpm/12°C) for purification. The bands containing the encapsidated RNAs

resulting from replication (SeV Z or SeV-DI-H4 genomes) were extracted, pelleted in TNE

99

Page 102: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

1X (45min/35000 rpm/4°C) and resuspended in 40ul TE 0.1% SDS, 10% glycerol. This step

was repeated twice in the case of the infections.

Encapsidated RNAs from the SeV infection were treated (or not) with 60U of calf intestine

alkaline phosphatase (CIP) in a buffer (10X) containing 5M Tris-HCl, 1mM EDTA (Roche),

or with 50U RNase T1 (Roche) and 1ug RNase A (Roche) for 30min at 37°C. CIP and

RNAse treatments were performed between the two CsCl density gradients.

100

Page 103: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

VI. GENERAL DISCUSSION

101

Page 104: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

General Discussion The ability of the host cell to fight viral infection requires recognition of the invading viral

pathogen as such and subsequent induction of signaling cascades that lead to the production

of innate and adaptive antiviral responses. IFNβ plays a crucial role in priming neighbouring

cells for possible infection and is also involved (with other IFN-stimulated genes) in

recruiting cells of the host immune system to the site of infection, providing a way to

eliminate infected cells. In the present study, we investigated the mechanisms used by the host

cell to recognize SeV infection, and those used by SeV to counteract the cellular innate

immune system.

By using different SeV stocks carrying mutations either in the promoter region or in the C

proteins, we demonstrated that the presence of the leader RNA transcript (or the

overexpression of the trailer) as well as the expression of the viral C and V proteins have an

important role in counteracting the IFNβ activation and the expression of the chemokine IL-8

(c.f. Paper 1). As opposed to the V protein and the leader RNA, a number of investigations

have been made on the SeV C protein. As mentioned in the first paper, this particular protein

is in charge of many functions during infection: (i) it stimulates viral RNA synthesis early in

infection (Latorre et al., 1998a); (ii) it inhibits viral RNA synthesis late in infection by binding

to the viral polymerase (Cadd et al., 1996b); (iii) it has a role in the virus particle budding,

which is facilitated by its binding to Alix, and in virion assembly, possibly by interaction with

the matrix (M) protein (Mottet et al., 1996; Sakaguchi et al., 2005); (iv) The C protein

interacts with Stat1 in two ways: counteracting IFN signaling and inducing Stat1 instability

(Garcin et al., 2003). (v) Finally, the C protein inhibits the IRF-3-dependent activation of

IFNβ as well as the activation of IL-8 expression (Strahle et al., 2003). These multiple

functions of SeV C protein during infection fit well with its property to interact with various

viral and cellular proteins, such as Stat1, Alix and Rig-I (unpublished). However, these

interactions remain to be closely analyzed. Additionally, the reason why IL-8 expression has

been only recently discovered could be explained by the fact that standard SeV induces very

little IL-8. Indeed IL-8 is only expressed upon infection of SeV bearing mutations in the C

and V proteins, and in the leader region. Moreover, it has been already reported that SeV and

MeV infections induce an other CC-chemokine RANTES via the activation of IRF-3 (Genin

et al., 2000; Tenoever et al., 2002). These data confirm the results suggesting that SeV targets

the inflammatory and adaptive immune responses (IL-6 and IL-8) as well as the IFN-induced

intracellular antiviral state (IFNβ and STAT1).

102

Page 105: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

SeV is commonly used by many laboratories for its capacity to strongly induce IFN. This

ability was long known to be associated with the presence of DI genomes in the SeV stock.

The third paper provides evidence that the strong induction of IFNβ activation upon SeV

infection (SeV stock containing DI genomes) is mainly due to the presence of copyback DI

genomes. Moreover, the level of IFNβ activation was found to be proportional to that of DI

genome replication. Apparently, the ability of DI-H4 to induce IFNβ is not only related to its

strong interference with the helper, which leads to lower levels of V and C proteins

intracellularly but also to the relative content of copyback DI genomes, which can presumably

form dsRNA. (cf. paper 2).

It is important to emphasize the difference between a standard SeV-WT infection from an

infection of SeV that contains DI genomes. In the case of a SeV-WT infection, the RNA

genomes and antigenomes are tightly assembled into NCs, meaning that they are never free to

anneal (like other NNV). Furthermore, Weber et al. have shown recently that no detectable

amounts of dsRNA produced by NNV have been found in infected cells (Weber et al., 2006).

This result can be explained by the fact that the amount of dsRNA produced by NNV are

below their detection limit, since one molecule of dsRNA per cell can be effective in

triggering an antiviral response. If it is the case, we can imagine two different ways for SeV-

WT (devoided of DI genomes) to produce dsRNA: 1) During SeV replication, the polymerase

ignores occasionally the stop signal at the end of the trailer template, which results in the

extension of the trailer beyond the trailer/L gene junction. This event will produce a long

trailer whose extended 3’sequences can anneal to those of the L gene (Vidal and Kolakofsky,

1989). 2) dsRNA can also be the result of a readthrough by the transcriptase of the L gene-end

site until the end of the genome (an event that can happen at 5% of the time at all gene

junctions). In this case the genome 5’end can anneal to the trailer RNAs (Le et al., 2002).

Finally, if we consider that there is a small amount of dsRNA that is generated in SeV

infection, it is evident that the presence of dsRNA will be dampen by the SeV C and V

proteins expressed during ND genome replication and that the expression of IFNβ will

consequently be blocked.

In the case of copyback DI infections, it is a different story. Because DI-H4 contains two

antigenomic promoters located at both the genomes and antigenomes ends, it has strong

replicative advantage over the standard virus. Consequently, a huge amount of DI genomes is

rapidly generated and the level of viral proteins (including the level of the nucleocapsid

protein) is also strongly attenuated. This rapid generation of DI genomes and the low level of

103

Page 106: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

NC proteins could explain why the genomes and antigenomes may not assemble into

nucleocapsids and would consequently form dsRNA. If DI-H4 genomes and antigenomes are

not encapsidated during infection, two forms of dsRNA can be found (in addition to those

mentioned for a SeV infection). First, the extremities of the DI genome (complementary on

110 nucleotides) can self anneal in a concentration independent manner and form dsRNA,

resulting in the shape of panhandles. Secondly, the genome and the antigenome can anneal

together, which remains unlikely since this would require the melting of intramolecular

secondary structure.

Recent evidence suggests that NNV infections could activate the innate immune

response by producing viral signatures other than dsRNA, namely 5’tri-phosphorylated

single-stranded RNA transcripts (5’pppRNAs). This discovery suggests that dsRNA may not

always be implicated in the IFNβ induction upon viral infection and that there is probably

more than one factor able to induce this primary cellular induction. Two cytoplasmic

helicases, namely RIG-I and Mda-5, have been recently found to respond to dsRNA and, at

least for RIG-I, to 5’pppRNAs. These RNAs are generated in the cytoplasm during RNA

virus replication. Upon detection of specific viral RNAs RIG-I and Mda-5 interact with Cardif

which is present in the mitochondrial membrane, and this interaction is thought to lead to the

recruitment and activation of the TBK1, IKKε and other IKK kinases that activate NF-kB and

IRF3, thereby activating the IFNβ promoter.

In the third paper, we investigated different potential inducers of IFNβ, including the

synthetic dsRNA (polyI/C) and 5’pppRNAs and the involvement of RIG-I in these inductions.

We showed evidences that i) transfection of poly-I/C and ii) a SeV coinfection that can

produce GFP dsRNA, as well as iii) transfections of small 5’pppRNAs can all induce IFNβ.

In all cases, the IFNβ induction was dependent on RIG-I. Not all 5’pppRNAs generated by

SeV can act as PAMPs: 1) SeV mRNAs carry 5’ ppp ends, but because they are capped, these

mRNAs cannot be potential targets. 2) SeV genomes and antigenomes also carry

triphosphorylated 5’ends, but because the RNA is tightly and entirely encapsidated, it cannot

be seen. 3) Finally, leader and trailer RNA transcript are the most susceptible to induce IFNβ,

because they are short unencapsidated 5’pppRNAs that are independent on on-going (N)

protein synthesis. For this reason, we suspect that, in the case of DI-H4 infection, there is a

real overproduction of pppRNAs (trailer) resulting in the induction of the IFNβ activation.

Alternatively, other viral structures may take the role of RNA as a danger signal for the host

cell. Indeed, it was previously shown that the NCs from VSV and MeV are capable of

104

Page 107: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

triggering IFN induction. It was interesting to observe in our experiments that NCs purified

from SeV infection could strongly induce IFNβ. However, RNase treatment revealed that it

was probably not the NCs that were responsible for this activation, but more likely RNA

products that have contaminated the CsCl banded NCs. Because the NC is the first viral

element that enters the cell, one can imagine that viruses have evolved to avoid its detection.

Thus, it is reasonable to think that NCs do not act as PAMPs. Concerning MeV, it was shown

recently that the NCs were not responsible for the IFNβ activation after all and that it was the

leader transcript of MeV that was responsible for the activation of IFNβ via the RIG-I

pathway (Helin et al., 2001; Plumet et al., 2007). This again comforts the idea that SeV leader

and trailer RNAs are more likely to be the main PAMPs responsible for triggering the

antiviral responses upon SeV infection.

Regarding the C and V proteins, we presented evidence that for SeV infection of

MEFs, it is the C protein (and not V) that is primarily responsible for this effect, and that C

acts by countering RIG-I dependent signaling to IFNβ. For example, independent expression

of either the C or the V proteins inhibited IFNβ activation due to RIG-I over-expression. In

addition to this, both proteins inhibited IFNβ activation due to DI-H4 infection, although C

was always more effective than V (in our experiments). However, only C expression

effectively inhibited IFNβ activation due to GFP(+/-) infection, or transfected poly-I/C or

pppRNAs (cf paper 3).

These new findings, suggesting that the 5’pppRNA is a PAMP specific to the NNV

infections, lead us to reconsider the way the cell could detect the presence of viruses.

However, no proof has been found concerning the role of 5’pppRNA products in the

activation of IFNβ in SeV infection. Further analyses of the precise RNAs involved in the

IFNβ activation need to be done. Additionally, more investigations are necessary to determine

how these PAMPs are detected and the mechanisms that drive cellular responses. Finally,

learning more about the protein targets of SeV proteins also appear important in order to

understand the details of the IFN suppressive activities of SeV and virus infection in general.

105

Page 108: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

VIII. Reference List

Ali,A. and Nayak,D.P. (2000). Assembly of Sendai virus: M protein interacts with F and HN proteins and with the cytoplasmic tail and transmembrane domain of F protein. Virology 276, 289-303.

Andrejeva,J., Childs,K.S., Young,D.F., Carlos,T.S., Stock,N., Goodbourn,S., and Randall,R.E. (2004). The V proteins of paramyxoviruses bind the IFN-inducible RNA helicase, mda-5, and inhibit its activation of the IFN-beta promoter. Proc. Natl. Acad. Sci. U. S. A 101, 17264-17269.

Baltimore,D. and Huang,A. (1975). Proceedings: Defective interfering particles. Neurology 25, 492.

Baron,M.D. and Barrett,T. (1997). Rescue of rinderpest virus from cloned cDNA. J Virol 71, 1265-1271.

Barrett,A.D. and Dimmock,N.J. (1986). Defective interfering viruses and infections of animals. Curr. Top. Microbiol. Immunol. 128, 55-84.

Basler,C.F. and Garcia-Sastre,A. (2007). Sensing RNA virus infections. Nat. Chem. Biol. 3, 20-21.

Basler,C.F., Mikulasova,A., Martinez-Sobrido,L., Paragas,J., Muhlberger,E., Bray,M., Klenk,H.D., Palese,P., and Garcia-Sastre,A. (2003). The Ebola virus VP35 protein inhibits activation of interferon regulatory factor 3. J Virol 77, 7945-7956.

Baudin,F., Bach,C., Cusack,S., and Ruigrok,R.W.H. (1994). Structure of influenza virus RNP. I. Influenza virus nucleoprotein melts secondary structure in panandle RNA and exposes the bases to the solvent. EMBO J. 13, 3158-3165.

Bontron,S., Ucla,C., Mach,B., and Steimle,V. (1997). Efficient repression of endogenous major histocompatibility complex class II expression through dominant negative CIITA mutants isolated by a functional selection strategy. Mol. Cell Biol. 17, 4249-4258.

Bowie,A.G. and Fitzgerald,K.A. (2007). RIG-I: tri-ing to discriminate between self and non-self RNA. Trends Immunol. 28, 147-150.

Brzozka,K., Finke,S., and Conzelmann,K.K. (2005). Identification of the rabies virus alpha/beta interferon antagonist: phosphoprotein P interferes with phosphorylation of interferon regulatory factor 3. J Virol 79, 7673-7681.

Buchholz,C.J., Spehner,D., Drillien,R., Neubert,W.J., and Homann,H.E. (1993). The conserved N-terminal region of Sendai virus nucleocapsid protein NP is required for nucleocapsid assembly. J Virol 67, 5803-5812.

Buchholz,U.J., Finke,S., and Conzelmann,K.K. (1999). Generation of bovine respiratory syncytial virus (BRSV) from cDNA: BRSV NS2 is not essential for virus replication in tissue

106

Page 109: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

culture, and the human RSV leader region acts as a functional BRSV genome promoter. J Virol 73, 251-259.

Cadd,T., Garcin,D., Tapparel,C., Itoh,M., Homma,M., Roux,L., Curran,J., and Kolakofsky,D. (1996b). The Sendai paramyxovirus accessory C proteins inhibit viral genome amplification in a promoter-specific fashion. J Virol 70, 5067-5074.

Cadd,T., Garcin,D., Tapparel,C., Itoh,M., Homma,M., Roux,L., Curran,J., and Kolakofsky,D. (1996a). The Sendai paramyxovirus accessory C proteins inhibit viral genome amplification in a promoter-specific fashion. J Virol 70, 5067-5074.

Calain,P., Curran,J., Kolakofsky,D., and Roux,L. (1992). Molecular cloning of natural paramyxovirus copy-back defective interfering RNAs and their expression from DNA. Virology 191, 62-71.

Calain,P. and Roux,L. (1993). The rule of six, a basic feature for efficient replication of Sendai virus defective interfering RNA. J Virol 67, 4822-4830.

Calain,P. and Roux,L. (1995). Functional characterisation of the genomic and antigenomic promoter of the Sendai virus. Virology 211, 163-173.

Casola,A., Burger,N., Liu,T., Jamaluddin,M., Brasier,A.R., and Garofalo,R.P. (2001). Oxidant tone regulates RANTES gene expression in airway epithelial cells infected with respiratory syncytial virus. Role in viral-induced interferon regulatory factor activation. J. Biol. Chem. 276, 19715-19722.

Chen,Z., Li,Y., and Krug,R.M. (1999). Influenza A virus NS1 protein targets poly(A)-binding protein II of the cellular 3'-end processing machinery. EMBO J. 18, 2273-2283.

Childs,K., Stock,N., Ross,C., Andrejeva,J., Hilton,L., Skinner,M., Randall,R., and Goodbourn,S. (2007). mda-5, but not RIG-I, is a common target for paramyxovirus V proteins. Virology 359, 190-200.

Choppin,P.W., Scheid,A., and Mountcastle,W.E. (1975). Proceedings: Paramyxoviruses, membranes, and persistent infections. Neurology 25, 494.

Collins,P.L., Mink,M.A., and Stec,D.S. (1991). Rescue of synthetic analogs of respiratory syncytial genomic RNA and effect of truncations and mutations on the expression of a foreign reporter gene. Proc. Natl. Acad. Sci. U. S. A. 88, 9663-9667.

Conzelmann,K.-K. and Schnell,M.J. (1994). Rescue of Synthetic Genomic RNA Analogs of Rabies Virus by Plasmid-Encoded Proteins. J Virol 68, 713-719.

Creagh,E.M. and O'Neill,L.A. (2006). TLRs, NLRs and RLRs: a trinity of pathogen sensors that co-operate in innate immunity. Trends Immunol. 27, 352-357.

Curran,J. and Kolakofsky,D. (1990). Sendai virus P gene produces multiple proteins from overlapping open reading frames. Enzyme 44, 244-249.

Curran,J. and Kolakofsky,D. (1988). Ribosomal initiation from an ACG codon in the Sendai virus P/C mRNA. EMBO J. 7, 245-251.

107

Page 110: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

Curran,J., Marq,J.-B., and Kolakofsky,D. (1995). An N-terminal domain of the Sendai paramyxovirus P protein actsw as a chaperone for the NP protein during the nascent chain assembly step of genome replication. J Virol 69, 849-855.

Curran,J.A. and Kolakofsky,D. (1991). Rescue of a Sendai virus DI genome by other parainfluenza viruses: implications for genome replication. Virology 182, 168-176.

Darnell,J.E., Jr., Kerr,I.M., and Stark,G.R. (1994). Jak-STAT pathways and transcriptional activation in response to IFNs and other extracellular signaling proteins. Science 264, 1415-1421.

Das,S.C. and Pattnaik,A.K. (2005). Role of the hypervariable hinge region of phosphoprotein P of vesicular stomatitis virus in viral RNA synthesis and assembly of infectious virus particles. J Virol 79, 8101-8112.

Decker,T., Stockinger,S., Karaghiosoff,M., Muller,M., and Kovarik,P. (2002). IFNs and STATs in innate immunity to microorganisms. J. Clin. Invest 109, 1271-1277.

Deshpande,K.L. and Portner,A. (1984). Structural and functional analysis of Sendai virus nucleocapsid protein NP with monoclonal antibodies. Virology 139, 32-42.

Donelan,N.R., Basler,C.F., and Garcia-Sastre,A. (2003). A recombinant influenza A virus expressing an RNA-binding-defective NS1 protein induces high levels of beta interferon and is attenuated in mice. J Virol 77, 13257-13266.

Enami,M. and Palese,P. (1991). High-efficiency formation of influenza virus transfectants. J Virol 65, 2711-2713.

Errington,W. and Emmerson,P.T. (1997). Assembly of recombinant Newcastle disease virus nucleocapsid protein into nucleocapsid-like structures is inhibited by the phosphoprotein. J. Gen. Virol. 78 ( Pt 9), 2335-2339.

Fischer,S.F., Ludwig,H., Holzapfel,J., Kvansakul,M., Chen,L., Huang,D.C., Sutter,G., Knese,M., and Hacker,G. (2006). Modified vaccinia virus Ankara protein F1L is a novel BH3-domain-binding protein and acts together with the early viral protein E3L to block virus-associated apoptosis. Cell Death. Differ. 13, 109-118.

Fitzgerald,K.A., Rowe,D.C., Barnes,B.J., Caffrey,D.R., Visintin,A., Latz,E., Monks,B., Pitha,P.M., and Golenbock,D.T. (2003). LPS-TLR4 signaling to IRF-3/7 and NF-kappaB involves the toll adapters TRAM and TRIF. J. Exp. Med. 198, 1043-1055.

Fortes,P., Beloso,A., and Ortin,J. (1994). Influenza virus NS1 protein inhibits pre-mRNA splicing and blocks mRNA nucleocytoplasmic transport. EMBO J. 13, 704-712.

Fouillot-Coriou,N. and Roux,L. (2000). Structure-function analysis of the Sendai virus F and HN cytoplasmic domain: different role for the two proteins in the production of virus particle. Virology 270, 464-475.

Fuerst,T.R., Niles,E.G., Studier,F.W., and Moss,B. (1986). Eukaryotic transient-expression system based on recombinant vaccinia virus that synthesizes bacteriophage T7 RNA polymerase. Proc. Natl. Acad. Sci. U. S. A 83, 8122-8126.

108

Page 111: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

Fujii,Y., Sakaguchi,T., Kiyotani,K., Huang,C., Fukuhara,N., Egi,Y., and Yoshida,T. (2002). Involvement of the leader sequence in Sendai virus pathogenesis revealed by recovery of a pathogenic field isolate from cDNA. J Virol 76, 8540-8547.

Garcin,D., Marq,J.B., Goodbourn,S., and Kolakofsky,D. (2003). The amino-terminal extensions of the longer Sendai virus C proteins modulate pY701-Stat1 and bulk Stat1 levels independently of interferon signaling. J Virol 77, 2321-2329.

Garcin,D., Marq,J.B., Strahle,L., Le,M.P., and Kolakofsky,D. (2002). All four Sendai Virus C proteins bind Stat1, but only the larger forms also induce its mono-ubiquitination and degradation. Virology 295, 256-265.

Garcin,D., Pelet,T., Calain,P., Roux,L., Curran,J., and Kolakofsky,D. (1995). A highly recombinogenic system for the recovery of infectious Sendai paramyxovirus from cDNA: generation of a novel copy-back nondefective interfering virus. EMBO J. 14, 6087-6094.

Gassen,U., Collins,F.M., Duprex,W.P., and Rima,B.K. (2000). Establishment of a rescue system for canine distemper virus. J Virol 74, 10737-10744.

Genin,P., Algarte,M., Roof,P., Lin,R., and Hiscott,J. (2000). Regulation of RANTES chemokine gene expression requires cooperativity between NF-kappa B and IFN-regulatory factor transcription factors. J. Immunol. 164, 5352-5361.

Gershon,P.D., Ahn,B.Y., Garfield,M., and Moss,B. (1991). Poly(A) polymerase and a dissociable polyadenylation stimulatory factor encoded by vaccinia virus. Cell 66, 1269-1278.

Gotoh,B., Takeuchi,K., Komatsu,T., and Yokoo,J. (2003). The STAT2 activation process is a crucial target of Sendai virus C protein for the blockade of alpha interferon signaling. J Virol 77, 3360-3370.

Gupta,K.C. and Patwardhan,S. (1988). ACG, the initiator codon for a Sendai virus protein. J. Biol. Chem. 263, 8553-8556.

Haller,O., Kochs,G., and Weber,F. (2006). The interferon response circuit: induction and suppression by pathogenic viruses. Virology 344, 119-130.

Haller,O., Staeheli,P., and Kochs,G. (2007). Interferon-induced Mx proteins in antiviral host defense. Biochimie 89, 812-818.

Hatada,E. and Fukuda,R. (1992). Binding of influenza A virus NS1 protein to dsRNA in vitro. J. Gen. Virol. 73 ( Pt 12), 3325-3329.

Hausmann,S., Garcin,D., Delenda,C., and Kolakofsky,D. (1999). The versatility of paramyxovirus RNA polymerase stuttering. J Virol 73, 5568-5576.

Helin,E., Vainionpaa,R., Hyypia,T., Julkunen,I., and Matikainen,S. (2001). Measles virus activates NF-kappa B and STAT transcription factors and production of IFN-alpha/beta and IL-6 in the human lung epithelial cell line A549. Virology 290, 1-10.

Hiscott,J., Pitha,P., Genin,P., Nguyen,H., Heylbroeck,C., Mamane,Y., Algarte,M., and Lin,R. (1999). Triggering the interferon response: the role of IRF-3 transcription factor. J. Interferon Cytokine Res. 19, 1-13.

109

Page 112: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

Holland,J.J., Grabau,E.A., Jones,C.L., and Semler,B.L. (1979). Evolution of multiple genome mutations during long-term persistent infection by vesicular stomatitis virus. Cell 16, 495-504.

Holland,J.J., Kennedy,I.T., Semler,B.L., Jones,C.L., Roux,L., and Grabau,E.A. (1980). Defective interfering RNA viruses and the host-cell response. In Comprehensive Virology, H.Fraenkel-Conrat and R.R.Wagner, eds. (New York and London: Plenum Press), pp. 137-192.

Horikami,S.M., Hector,R.E., Smallwood,S., and Moyer,S.A. (1997). The Sendai virus C protein binds the L polymerase protein to inhibit viral RNA synthesis. Virology 235, 261-270.

Horikami,S.M. and Moyer,S.A. (1982). Host range mutants of vesicular stomatitis virus defective in in vitro RNA methylation. Proc. Natl. Acad. Sci. U. S. A 79, 7694-7698.

Hornung,V., Ellegast,J., Kim,S., Brzozka,K., Jung,A., Kato,H., Poeck,H., Akira,S., Conzelmann,K.K., Schlee,M., Endres,S., and Hartmann,G. (2006). 5'-Triphosphate RNA Is the Ligand for RIG-I. Science.

Horodyski,F.M. and Holland,J.J. (1980). Viruses isolated from cells persistently infected with vesicular stomatitis virus show altered interactions with defective interfering particles. J Virol 36, 627-631.

Horvath,C.M. (2004). Weapons of STAT destruction. Interferon evasion by paramyxovirus V protein. Eur. J. Biochem. 271, 4621-4628.

Houben,K., Marion,D., Tarbouriech,N., Ruigrok,R.W., and Blanchard,L. (2007). INTERACTION OF THE C-TERMINAL DOMAINS OF SENDAI VIRUS N AND P PROTEINS: Comparison of polymerase-nucleocapsid interaction within the paramyxovirus family. J Virol.

Huang,A.S. (1977). Viral pathogenesis and molecular biology. Bacteriol. Rev. 41, 811-821.

Huang,A.S. and Baltimore,D. (1970). Defective viral particles and viral disease processes. Nature 226, 325-327.

Iseni,F., Barge,A., Baudin,F., Blondel,D., and Ruigrok,R.W. (1998). Characterization of rabies virus nucleocapsids and recombinant nucleocapsid-like structures. J Gen Virol 79, 2909-2919.

Iseni,F., Garcin,D., Nishio,M., Kedersha,N., Anderson,P., and Kolakofsky,D. (2002b). Sendai virus trailer RNA binds TIAR, a cellular protein involved in virus-induced apoptosis. EMBO J. 21, 5141-5150.

Iseni,F., Garcin,D., Nishio,M., Kedersha,N., Anderson,P., and Kolakofsky,D. (2002a). Sendai virus trailer RNA binds TIAR, a cellular protein involved in virus-induced apoptosis. EMBO J. 21, 5141-5150.

Ishida,N. and Homma,M. (1978). Sendai virus. Adv. Virus Res. 23, 349-383.

110

Page 113: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

Itoh,M., Isegawa,Y., Hotta,H., and Homma,M. (1997). Isolation of an avirulent mutant of Sendai virus with two amino acid mutations from a highly virulent field strain through adaptation to LLC-MK2 cells. J Gen Virol 78, 3207-3215.

Kang,D.C., Gopalkrishnan,R.V., Lin,L., Randolph,A., Valerie,K., Pestka,S., and Fisher,P.B. (2004). Expression analysis and genomic characterization of human melanoma differentiation associated gene-5, mda-5: a novel type I interferon-responsive apoptosis-inducing gene. Oncogene 23, 1789-1800.

Kato,H., Takeuchi,O., Sato,S., Yoneyama,M., Yamamoto,M., Matsui,K., Uematsu,S., Jung,A., Kawai,T., Ishii,K.J., Yamaguchi,O., Otsu,K., Tsujimura,T., Koh,C.S., Reis E Sousa, Matsuura,Y., Fujita,T., and Akira,S. (2006). Differential roles of MDA5 and RIG-I helicases in the recognition of RNA viruses. Nature 441, 101-105.

Katze,M.G., He,Y., and Gale,M., Jr. (2002). Viruses and interferon: a fight for supremacy. Nat. Rev. Immunol. 2, 675-687.

Kawai,T., Takahashi,K., Sato,S., Coban,C., Kumar,H., Kato,H., Ishii,K.J., Takeuchi,O., and Akira,S. (2005). IPS-1, an adaptor triggering RIG-I- and Mda5-mediated type I interferon induction. Nat. Immunol. 6, 981-988.

Kiley,M.P. and Wagner,R.R. (1972). Ribonucleic acid species of intracellular nucleocapsids and released virions of vesicular stomatitis virus. J Virol 10, 244-255.

King,P. and Goodbourn,S. (1994). The beta-interferon promoter responds to priming through multiple independent regulatory elements. J. Biol. Chem. 269, 30609-30615.

Kingston,R.L., Hamel,D.J., Gay,L.S., Dahlquist,F.W., and Matthews,B.W. (2004). Structural basis for the attachment of a paramyxoviral polymerase to its template. Proc. Natl. Acad. Sci. U. S. A 101, 8301-8306.

Kiyotani,K., Sakaguchi,T., Fujii,Y., and Yoshida,T. (2001). Attenuation of a field Sendai virus isolate through egg-passages is associated with an impediment of viral genome replication in mouse respiratory cells. Arch. Virol. 146, 893-908.

Kolakofsky,D. (1976). Isolation and characterization of Sendai virus DI-RNAs. Cell 8, 547-555.

Kolakofsky,D. (1979). Studies on the generation and amplification of Sendai virus defective-interfering genomes. Virology 93, 589-593.

Kolakofsky,D., Pelet,T., Garcin,D., Hausmann,S., Curran,J., and Roux,L. (1998). Paramyxovirus RNA synthesis and the requirement for hexamer genome length: the rule of six revisited. J Virol 72, 891-899.

Koromilas,A.E., Roy,S., Barber,G.N., Katze,M.G., and Sonenberg,N. (1992). Malignant transformation by a mutant of the IFN-inducible dsRNA-dependent protein kinase. Science 257, 1685-1689.

Krystal,M., Li,R., Lyles,D., Pavlakis,G., and Palese,P. (1986). Expression of the three influenza virus polymerase proteins in a single cell allows growth complementation of viral mutants. Proc. Natl. Acad. Sci. U. S. A 83, 2709-2713.

111

Page 114: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

Kurotani,A., Kiyotani,K., Kato,A., Shioda,T., Sakai,Y., Mizumoto,K., Yoshida,T., and Nagai,Y. (1998). Sendai virus C proteins are categorically nonessential gene products but silencing their expression severely impairs viral replication and pathogenesis. Genes Cells 3, 111-124.

Lamb,R.A. and Kolakofsky,D. (1996). Paramyxoviridae: The viruses and Their Replication. In Fields Virology, B.N.Fields, D.M.Knipe, and P.M.Howley, eds., pp. 1177-1204.

Lamb,R.A. and Kolakofsky,D. (2001). Paramyxoviridae: The viruses and Their Replication. In Fields Virology, D.M.Knipe and P.M.Howley, eds. Lippincott, Williams & Wilkins), pp. 1305-1340.

Lamb,R.A., Mahy,B.W.J., and Choppin,P.W. (1976). The synthesis of Sendai virus polypeptides in infected cells. Virology 69, 116-131.

Latorre,P., Cadd,T., Itoh,M., Curran,J., and Kolakofsky,D. (1998a). The various Sendai virus C proteins are not functionally equivalent, and exert both positive and negative effects on viral RNA accumulation during the course of infection. J Virol 72, 5984-5993.

Latorre,P., Kolakofsky,D., and Curran,J. (1998b). Sendai virus Y proteins are initiated by a ribosomal shunt. Mol Cell Biol 18, 5021-5031.

Le,M.P., Garcin,D., Garcia,E., and Kolakofsky,D. (2003). Competition between the Sendai virus N mRNA start site and the genome 3'-end promoter for viral RNA polymerase. J Virol 77, 9147-9155.

Le,M.P., Garcin,D., Hausmann,S., and Kolakofsky,D. (2002). Ambisense sendai viruses are inherently unstable but are useful to study viral RNA synthesis. J Virol 76, 5492-5502.

Lemaitre,B., Nicolas,E., Michaut,L., Reichhart,J.M., and Hoffmann,J.A. (1996). The dorsoventral regulatory gene cassette spatzle/Toll/cactus controls the potent antifungal response in Drosophila adults. Cell 86, 973-983.

Leppert,M., Kort,L., and Kolakofsky,D. (1977). Further characterization of Sendai virus DI-RNAs: a model for their generation. Cell 12, 539-552.

Li,S., Min,J.Y., Krug,R.M., and Sen,G.C. (2006). Binding of the influenza A virus NS1 protein to PKR mediates the inhibition of its activation by either PACT or double-stranded RNA. Virology 349, 13-21.

Luytjes,W., Krystal,M., Enami,M., Parvin,J.D., and Palese,P. (1989). Amplification, expression, and packaging of foreign gene by influenza virus. Cell 59, 1107-1113.

Lynch,S. and Kolakofsky,D. (1978). Ends of the RNA within Sendai virus defective interfering nucleocapsids are not free. J Virol 28, 584-589.

Mamane,Y., Heylbroeck,C., Genin,P., Algarte,M., Servant,M.J., LePage,C., DeLuca,C., Kwon,H., Lin,R., and Hiscott,J. (1999). Interferon regulatory factors: the next generation. Gene 237, 1-14.

112

Page 115: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

Marcus,P.I. and Gaccione,C. (1989). Interferon induction by viruses. XIX. Vesicular stomatitis virus--New Jersey: high multiplicity passages generate interferon-inducing, defective-interfering particles. Virology 171, 630-633.

Marq,J.B., Brini,A., Kolakofsky,D., and Garcin,D. (2007). Targeting of the Sendai virus C protein to the plasma membrane via a peptide-only membrane anchor. J Virol 81, 3187-3197.

Masters,P.S. and Banerjee,A.K. (1988). Resolution of multiple complexes of phosphoprotein NS with nucleocapsid protein N of vesicular stomatitis virus. J Virol 62, 2651-2657.

Medzhitov,R., Preston-Hurlburt,P., and Janeway,C.A., Jr. (1997). A human homologue of the Drosophila Toll protein signals activation of adaptive immunity. Nature 388, 394-397.

Meylan,E., Curran,J., Hofmann,K., Moradpour,D., Binder,M., Bartenschlager,R., and Tschopp,J. (2005). Cardif is an adaptor protein in the RIG-I antiviral pathway and is targeted by hepatitis C virus. Nature 437, 1167-1172.

Morrison,T.G. (2003). Structure and function of a paramyxovirus fusion protein. Biochim. Biophys. Acta 1614, 73-84.

Mottet,G., Mühlemann,A., Tapparel,C., Hoffmann,F., and Roux,L. (1996). A Sendai virus vector leading to the efficient expression of mutant M proteins interfering with virus particle budding. Virology 221, 159-171.

Murphy,S.K., Ito,Y., and Parks,G.D. (1998). A functional antigenomic promoter for the paramyxovirus simian virus 5 requires proper spacing between an essential internal segment and the 3' terminus. J Virol 72, 10-19.

Nimchuk,Z., Eulgem,T., Holt,B.F., III, and Dangl,J.L. (2003). Recognition and response in the plant immune system. Annu. Rev. Genet. 37, 579-609.

Nishio,M., Tsurudome,M., Ito,M., Kawano,M., Kusagawa,S., Komada,H., and Ito,Y. (1999). Mapping of domains on the human parainfluenza virus type 2 nucleocapsid protein (NP) required for NP-phosphoprotein or NP-NP interaction. J. Gen. Virol. 80 ( Pt 8), 2017-2022.

Novick,D., Cohen,B., and Rubinstein,M. (1994). The human interferon alpha/beta receptor: characterization and molecular cloning. Cell 77, 391-400.

Ogino,T., Kobayashi,M., Iwama,M., and Mizumoto,K. (2005). Sendai virus RNA-dependent RNA polymerase L protein catalyzes cap methylation of virus-specific mRNA. J. Biol. Chem. 280, 4429-4435.

Palosaari,H., Parisien,J.P., Rodriguez,J.J., Ulane,C.M., and Horvath,C.M. (2003). STAT protein interference and suppression of cytokine signal transduction by measles virus V protein. J Virol 77, 7635-7644.

Park,K.H., Huang,T., Correia,F.F., and Krystal,M. (1991). Rescue of a foreign gene by Sendai virus. Proc. Natl. Acad. Sci. U. S. A. 88, 5537-5541.

Pattnaik,A.K., Ball,L.A., LeGrone,A., and Wertz,G.W. (1995). The termini of VSV DI particles are sufficient to signal encapsidation, replication and budding to generate infectious particles. Virology 206, 760-764.

113

Page 116: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

Pattnaik,A.K. and Wertz,G.W. (1990). Replication and amplification of defective interfering particle RNAs of vesicular stomatitis virus in cells expressing viral proteins from vectors containing cloned cDNAs. J Virol 64, 2948-2957.

Pelet,T., Curran,J., and Kolakofsky,D. (1991). The P gene of bovine parainfluenza virus 3 expresses all three reading frames from a single mRNA editing site. EMBO J. 10, 443-448.

Pelet,T., Delenda,C., Gubbay,O., Garcin,D., and Kolakofsky,D. (1996). Partial characterization of a Sendai virus replication promoter and the rule of six. Virology 224, 405-414.

Perrault,J. and Leavitt,R.W. (1978). Inverted complementary terminal sequences in single-stranded RNAs and snap-back RNAs from vesicular stomatitis defective interfering particles. J. Gen. Virol. 38, 35-50.

Perrotta,A.T. and Been,M.D. (1990). The self-cleaving domain from the genomic RNA of hepatitis delta virus: sequence requirements and the effects of denaturant. Nucleic Acids Res. 18, 6821-6827.

Pestka,S., Langer,J.A., Zoon,K.C., and Samuel,C.E. (1987). Interferons and their actions. Annu. Rev. Biochem. 56, 727-777.

Pichlmair,A., Schulz,O., Tan,C.P., Naslund,T.I., Liljestrom,P., Weber,F., and Reis E Sousa (2006). RIG-I-Mediated Antiviral Responses to Single-Stranded RNA Bearing 5' Phosphates. Science.

Player,M.R. and Torrence,P.F. (1998). The 2-5A system: modulation of viral and cellular processes through acceleration of RNA degradation. Pharmacol. Ther. 78, 55-113.

Plumet,S., Herschke,F., Bourhis,J.M., Valentin,H., Longhi,S., and Gerlier,D. (2007). Cytosolic 5'-Triphosphate Ended Viral Leader Transcript of Measles Virus as Activator of the RIG I-Mediated Interferon Response. PLoS. ONE. 2, e279.

Poch,O., Blumberg,B.M., Bougueleret,L., and Tordo,N. (1990). Sequence comparison of five polymerases (L proteins) of unsegmented negative-strand RNA viruses: theoretical assignment of functional domains. J. Gen. Virol. 71 ( Pt 5), 1153-1162.

Qanungo,K.R., Shaji,D., Mathur,M., and Banerjee,A.K. (2004). Two RNA polymerase complexes from vesicular stomatitis virus-infected cells that carry out transcription and replication of genome RNA. Proc. Natl. Acad. Sci. U. S. A 101, 5952-5957.

Racaniello,V.R. and Baltimore,D. (1981). Cloned poliovirus complementary DNA is infectious in mammalian cells. Science 214, 916-919.

Radecke,F., Spielhofer,P., Schneider,H., Kaelin,K., Huber,M., Dötsch,C., Christiansen,G., and Billeter,M.A. (1995). Rescue of measles viruses from cloned DNA. EMBO J. 14, 5773-5784.

Reith,W. and Mach,B. (2001). The bare lymphocyte syndrome and the regulation of MHC expression. Annu. Rev. Immunol. 19, 331-373.

114

Page 117: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

Rodriguez,J.J., Wang,L.F., and Horvath,C.M. (2003). Hendra virus V protein inhibits interferon signaling by preventing STAT1 and STAT2 nuclear accumulation. J Virol 77, 11842-11845.

Rothenfusser,S., Goutagny,N., DiPerna,G., Gong,M., Monks,B.G., Schoenemeyer,A., Yamamoto,M., Akira,S., and Fitzgerald,K.A. (2005). The RNA helicase Lgp2 inhibits TLR-independent sensing of viral replication by retinoic acid-inducible gene-I. J. Immunol. 175, 5260-5268.

Roux,L., Simon,A.E., and Holland,J.J. (1991). Effects of defective interfering viruses on virus replication and pathogenesis. In Advances in Virus Research, Vol. 40, Academic Press Inc.), pp. 181-211.

Sakaguchi,T., Kato,A., Sugahara,F., Shimazu,Y., Inoue,M., Kiyotani,K., Nagai,Y., and Yoshida,T. (2005). AIP1/Alix is a binding partner of Sendai virus C protein and facilitates virus budding. J Virol 79, 8933-8941.

Sakaguchi,T., Kiyotani,K., Sakaki,M., Fujii,Y., and Yoshida,T. (1994b). A field isolate of Sendai virus: its high virulence to mice and genetic divergence form prototype strains. Arch. Virol. 135, 159-164.

Sakaguchi,T., Kiyotani,K., Sakaki,M., Fujii,Y., and Yoshida,T. (1994a). A field isolate of Sendai virus: its high virulence to mice and genetic divergence form prototype strains. Arch. Virol. 135, 159-164.

Sakaguchi,T., Kiyotani,K., Watanabe,H., Huang,C., Fukuhara,N., Fujii,Y., Shimazu,Y., Sugahara,F., Nagai,Y., and Yoshida,T. (2003). Masking of the contribution of V protein to Sendai virus pathogenesis in an infection model with a highly virulent field isolate. Virology 313, 581-587.

Samuel,C.E. (2007). Interferons, interferon receptors, signal transducer and transcriptional activators, and interferon regulatory factors. J. Biol. Chem. 282, 20045-20046.

Sato,M., Hata,N., Asagiri,M., Nakaya,T., Taniguchi,T., and Tanaka,N. (1998). Positive feedback regulation of type I IFN genes by the IFN-inducible transcription factor IRF-7. FEBS Lett. 441, 106-110.

Sato,M., Suemori,H., Hata,N., Asagiri,M., Ogasawara,K., Nakao,K., Nakaya,T., Katsuki,M., Noguchi,S., Tanaka,N., and Taniguchi,T. (2000). Distinct and essential roles of transcription factors IRF-3 and IRF-7 in response to viruses for IFN-alpha/beta gene induction. Immunity. 13, 539-548.

Schindler,C. (1999). Cytokines and JAK-STAT signaling. Exp. Cell Res. 253, 7-14.

Schindler,C. and Darnell,J.E., Jr. (1995). Transcriptional responses to polypeptide ligands: the JAK-STAT pathway. Annu. Rev. Biochem. 64, 621-651.

Schlaak,J.F., Hilkens,C.M., Costa-Pereira,A.P., Strobl,B., Aberger,F., Frischauf,A.M., and Kerr,I.M. (2002). Cell-type and donor-specific transcriptional responses to interferon-alpha. Use of customized gene arrays. J. Biol. Chem. 277, 49428-49437.

115

Page 118: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

Servant,M.J., ten,O.B., LePage,C., Conti,L., Gessani,S., Julkunen,I., Lin,R., and Hiscott,J. (2001). Identification of distinct signaling pathways leading to the phosphorylation of interferon regulatory factor 3. J. Biol. Chem. 276, 355-363.

Seth,R.B., Sun,L., Ea,C.K., and Chen,Z.J. (2005). Identification and characterization of MAVS, a mitochondrial antiviral signaling protein that activates NF-kappaB and IRF 3. Cell 122, 669-682.

Sharma,S., Tenoever,B.R., Grandvaux,N., Zhou,G.P., Lin,R., and Hiscott,J. (2003). Triggering the interferon antiviral response through an IKK-related pathway. Science 300, 1148-1151.

Shih,I.H. and Been,M.D. (2002). Catalytic strategies of the hepatitis delta virus ribozymes. Annu. Rev. Biochem. 71, 887-917.

Sidhu,M.S., Menonna,J.P., Cook,S.D., Dowling,P.C., and Udem,S.A. (1993). Canine distemper virus L gene: sequence and comparison with related viruses. Virology 193, 50-65.

Skiadopoulos,M.H., Surman,S.R., Riggs,J.M., Elkins,W.R., St,C.M., Nishio,M., Garcin,D., Kolakofsky,D., Collins,P.L., and Murphy,B.R. (2002). Sendai virus, a murine parainfluenza virus type 1, replicates to a level similar to human PIV1 in the upper and lower respiratory tract of African green monkeys and chimpanzees. Virology 297, 153-160.

Smallwood,S. and Moyer,S.A. (1993). Promoter analysis of the vesicular stomatitis virus RNA polymerase. Virology 192, 254-263.

Smallwood,S. and Moyer,S.A. (2004). The L polymerase protein of parainfluenza virus 3 forms an oligomer and can interact with the heterologous Sendai virus L, P and C proteins. Virology 318, 439-450.

Stark,G.R., Kerr,I.M., Williams,B.R., Silverman,R.H., and Schreiber,R.D. (1998). How cells respond to interferons. Annu. Rev. Biochem. 67, 227-264.

Strahle,L., Garcin,D., Le,M.P., Schlaak,J.F., and Kolakofsky,D. (2003). Sendai virus targets inflammatory responses, as well as the interferon-induced antiviral state, in a multifaceted manner. J Virol 77, 7903-7913.

Stricker,R., Mottet,G., and Roux,L. (1994). The Sendai virus matrix protein appears to be recruited in the cytoplasm by the viral nucleocapsid to function in viral assembly and budding. J. Gen. Virol. 75 ( Pt 5), 1031-1042.

Sundstrom,J.B., McMullan,L.K., Spiropoulou,C.F., Hooper,W.C., Ansari,A.A., Peters,C.J., and Rollin,P.E. (2001). Hantavirus infection induces the expression of RANTES and IP-10 without causing increased permeability in human lung microvascular endothelial cells. J Virol 75, 6070-6085.

Takimoto,T., Bousse,T., Coronel,E.C., Scroggs,R.A., and Portner,A. (1998). Cytoplasmic domain of sendai virus HN protein contains a specific sequence required for its incorporation into virions [In Process Citation]. J Virol 72, 9747-9754.

Takimoto,T., Murti,K.G., Bousse,T., Scroggs,R.A., and Portner,A. (2001). Role of matrix and fusion proteins in budding of Sendai virus. J Virol 75, 11384-11391.

116

Page 119: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

Takizawa,T., Ohashi,K., and Nakanishi,Y. (1996). Possible involvement of double-stranded RNA-activated protein kinase in cell death by influenza virus infection. J Virol 70, 8128-8132.

Tapparel,C., Hausmann,S., Pelet,T., Curran,J., Kolakofsky,D., and Roux,L. (1997). Inhibition of Sendai virus genome replication due to promoter-increased selectivity: a possible role for the accessory C proteins. J Virol 71, 9588-9599.

Tapparel,C., Maurice,D., and Roux,L. (1998). The activity of Sendai virus genomic and antigenomic promoters requires a second element past the leader template regions: a motif (GNNNNN)3 is essential for replication. J Virol 72, 3117-3128.

Tenoever,B.R., Servant,M.J., Grandvaux,N., Lin,R., and Hiscott,J. (2002). Recognition of the measles virus nucleocapsid as a mechanism of IRF-3 activation. J Virol 76, 3659-3669.

Thanos,D. and Maniatis,T. (1995). Virus induction of human IFN beta gene expression requires the assembly of an enhanceosome. Cell 83, 1091-1100.

Ting,J.P. and Davis,B.K. (2005). CATERPILLER: a novel gene family important in immunity, cell death, and diseases. Annu. Rev. Immunol. 23, 387-414.

Ulane,C.M., Rodriguez,J.J., Parisien,J.P., and Horvath,C.M. (2003). STAT3 ubiquitylation and degradation by mumps virus suppress cytokine and oncogene signaling. J Virol 77, 6385-6393.

Unterstab,G., Ludwig,S., Anton,A., Planz,O., Dauber,B., Krappmann,D., Heins,G., Ehrhardt,C., and Wolff,T. (2005). Viral targeting of the interferon-{beta}-inducing Traf family member-associated NF-{kappa}B activator (TANK)-binding kinase-1. Proc. Natl. Acad. Sci. U. S. A 102, 13640-13645.

Vidal,S., Curran,J., and Kolakofsky,D. (1992). Editing of the Sendai virus P/C mRNA by G insertion occurs during mRNA synthesis via a virus-encoded activity. Journal of Virology 64, 239-246.

Vidal,S. and Kolakofsky,D. (1989). Modified model for the switch from Sendai virus transcription to replication. J Virol 63, 1951-1958.

Vilcek,J.T. (1996). Cytokines in 1995. Cytokine Growth Factor Rev. 7, 103-106.

Vulliemoz,D., Cordey,S., Mottet-Osman,G., and Roux,L. (2005). Nature of a paramyxovirus replication promoter influences a nearby transcription signal. J. Gen. Virol. 86, 171-180.

Wang,X.-L., Itoh,M., Hotta,H., and Homma,M. (1994). A protease activation mutant, MVCES1, as a safe and potent live vaccine derived from currently prevailing Sendai virus. J Virol 68, 3369-3373.

Weber,F., Wagner,V., Rasmussen,S.B., Hartmann,R., and Paludan,S.R. (2006). Double-stranded RNA is produced by positive-strand RNA viruses and DNA viruses but not in detectable amounts by negative-strand RNA viruses. J Virol 80, 5059-5064.

Williams,B.R. (1999). PKR; a sentinel kinase for cellular stress. Oncogene 18, 6112-6120.

117

Page 120: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

Xu,L.G., Wang,Y.Y., Han,K.J., Li,L.Y., Zhai,Z., and Shu,H.B. (2005). VISA is an adapter protein required for virus-triggered IFN-beta signaling. Mol. Cell 19, 727-740.

Yamamoto,M. and Akirz,S. (2005). TIR domain-containing adaptors regulate TLR signaling pathways. Adv. Exp. Med. Biol. 560, 1-9.

Yokosawa,N., Yokota,S., Kubota,T., and Fujii,N. (2002). C-terminal region of STAT-1alpha is not necessary for its ubiquitination and degradation caused by mumps virus V protein. J Virol 76, 12683-12690.

Yoneda,M., Guillaume,V., Ikeda,F., Sakuma,Y., Sato,H., Wild,T.F., and Kai,C. (2006). Establishment of a Nipah virus rescue system. Proc. Natl. Acad. Sci. U. S. A 103, 16508-16513.

Yoneyama,M. and Fujita,T. (2007). Function of RIG-I-like receptors in antiviral innate immunity. J. Biol. Chem. 282, 15315-15318.

Yoneyama,M., Kikuchi,M., Matsumoto,K., Imaizumi,T., Miyagishi,M., Taira,K., Foy,E., Loo,Y.M., Gale,M., Jr., Akira,S., Yonehara,S., Kato,A., and Fujita,T. (2005). Shared and unique functions of the DExD/H-box helicases RIG-I, MDA5, and LGP2 in antiviral innate immunity. J. Immunol. 175, 2851-2858.

Yoshida,T., Nagai,Y., Maeno,K., Iinuma,M., Hamaguchi,M., Matsumoto,T., Nagayoshi,S., and Hoshino,M. (1979). Studies on the role of M protein in virus assembly using a ts mutant of HJV (Sendai virus). Virology 92, 139-154.

118

Page 121: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

JOURNAL OF VIROLOGY, July 2003, p. 7903–7913 Vol. 77, No. 140022-538X/03/$08.00�0 DOI: 10.1128/JVI.77.14.7903–7913.2003Copyright © 2003, American Society for Microbiology. All Rights Reserved.

Sendai Virus Targets Inflammatory Responses, as Well as theInterferon-Induced Antiviral State, in a Multifaceted Manner

Laura Strahle,1 Dominique Garcin,1 Philippe Le Mercier,1 Joerg F. Schlaak,2†and Daniel Kolakofsky1*

Department of Genetics and Microbiology, University of Geneva School of Medicine, CMU,CH1211 Geneva, Switzerland,1 and Imperial Cancer Research Fund,

London WC2A 3PX, United Kingdom2

Received 13 February 2003/Accepted 17 April 2003

We have used cDNA arrays to compare the activation of various cellular genes in response to infection withSendai viruses (SeV) that contain specific mutations. Three groups of cellular genes activated by mutant SeVinfection, but not by wild-type SeV, were identified in this way. While some of these genes are well known inter-feron (IFN)-stimulated genes, others, such as those for interleukin-6 (IL-6) and IL-8, are not directly inducedby IFN. The gene for beta IFN (IFN-�), which is critical for initiating an antiviral response, was also specifi-cally activated in mutant SeV infections. The SeV-induced activation of IFN-� was found to depend on IFN reg-ulatory factor 3, and the activation of all three cellular genes was independent of IFN signaling. Mutations thatdisrupt four distinct elements in the SeV genome (the leader RNA, two regions of the C protein, and the Vprotein) all lead to enhanced levels of IFN-� mRNA, and at least three of these viral genes also appear to beinvolved in preventing activation of IL-8. Our results suggest that SeV targets the inflammatory and adaptiveimmune responses as well as the IFN-induced intracellular antiviral state by using a multifaceted approach.

Alpha/beta interferons (IFN-�/�) are cytokines that act in apleiotropic manner to limit viral replication and spread (2, 57).In fibroblasts (e.g., the bronchial epithelial target of manyparamyxoviruses), the product of the single IFN-� gene isdirectly induced by viral infection, and IFN-� feeds back ontocells in an autocrine manner to induce multiple IFN-� genesand in a paracrine manner to prime neighboring cells for theirpossible infection (60). Since most viruses induce IFN-� tosome extent, intracellular double-stranded RNA (dsRNA)generated from the viral genome is traditionally assumed to bethe common signature of virus replication that sets the IFNsystem in motion (22, 32). dsRNA is thought to induce theformation of an enhanceosome at the IFN-� promoter thatincludes IFN regulatory factor 3 (IRF-3) and NF-�B (amongother transcription factors) (65). IFNs induce a cellular statethat is nonconducive for viral replication by signaling throughtheir cell surface receptor, leading to the phosphorylation ofcytoplasmic STAT proteins and their nuclear translocation.IFN-�/� responses are regulated primarily via IFN-stimulatedgene (ISG) factor 3, a heterotrimeric transcription factor com-posed of STAT1, STAT2, and IRF-9 (p48). ISG factor 3 bindsto a DNA element (IFN-stimulated response element) in thepromoters of ISGs and activates their expression (7).

The extravasation of neutrophils, eosinophils, basophils, andmononuclear cells is the salient feature of the innate responseto microorganisms in the lung. Localized and systemic pro- andanti-inflammatory cytokines thus also play an important role in

the outcome of viral infection and pathogenicity of this organ(58). The CC chemokine interleukin-8 (IL-8) is secreted fromepithelial surfaces in a polar fashion during infection withpathogenic bacteria such as Salmonella enterica serovar Typhi-murium and sets up a subepithelial chemotactic gradient di-recting neutrophils and other immune cells to the site of in-fection (27). In polarized epithelial monolayers, S. entericaserovar Typhimurium-induced IL-8 expression is controlled viathe activation of the mitogen-activated protein kinase cascadeand I�B� kinase, followed by NF-�B translocation to the nu-cleus and production of IL-8 mRNA. IL-8 secretion by primaryhuman monocytes in response to dengue virus infection is alsotightly linked to NF-�B activation (3). Sendai virus (SeV) in-fection of human embryonic kidney 293 cells induces theexpression of the CXC chemokine RANTES in an IRF-3-and NF-�B-dependent manner (23, 41). NF-�B, like IRF-3,is found in the cytoplasm of unstimulated cells, retained in acomplex with the inhibitory I�B proteins. Upon stimulationwith many inducers, including dsRNA and virus infection, I�Bis rapidly phosphorylated and degraded, resulting in NF-�Brelease and translocation to the nucleus (30, 33).

Given the importance of the host innate immune response tovirus infection, viruses have, during their coevolution withcells, developed strategies to regulate cytokine synthesis andaction. SeV, a model paramyxovirus and respiratory pathogenof mice, is known to use its C protein to evade the host inter-feron response by at least two mechanisms. (i) C binds STAT1,preventing its activation in response to IFN, and the carboxylpart of the C protein (i.e., residues 24 to 204, or the Y proteins)is sufficient for this activity. A phenylalanine at position 170 ofC is also critical for blocking STAT1 activation (18, 59). (ii) Calso targets STAT1 for degradation, and the amino-terminalresidues of the C proteins (residues 1 to 23, which are absent

* Corresponding author. Mailing address: Department of Geneticsand Microbiology, University of Geneva School of Medicine, CMU, 9Ave de Champel, CH1211 Geneva, Switzerland. Phone: 41 22 3795657. Fax: 41 22 379 5702. E-mail: [email protected].

† Present address: Department of Gastroenterology and Hepatol-ogy, University of Essen, 45122 Essen, Germany.

7903

Page 122: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

in the Y proteins) are essential for this activity (reference 17and references therein).

This paper reports that SeVs carrying specific mutations inthe C gene, in contrast to wild-type SeV (SeV-wt), activateIL-8 and IFN-� expression as well as that of several ISGs. Ourresults suggest that the products of virtually all of the viralaccessory genes (C and V proteins and leader RNA) act toprevent the expression of these cellular genes that are centralto the overall host antiviral response.

MATERIALS AND METHODS

Cells and viruses. 2C4 cells (39), 2fTGH cells (48), and their derived cell linesU3A (45) and U5A and U5A�IFNAR (43) were obtained from IM Kerr (Im-perial Cancer Research Fund, London, United Kingdom) and grown in Dulbec-co’s modified Eagle’s medium supplemented with 10% fetal bovine serum in thepresence of the relevant maintenance drug (hygromycin at 250 �g/ml or G418 at400 �g/ml). The generation of recombinant SeV (rSeV) expressing alternate Cand V (and P) proteins is described elsewhere (8, 19, 37, 38). All SeV stocks weregrown in the allantoic cavities of 10-day-old embryonated chicken eggs. Viruspresent in the allantoic fluid was analyzed by sodium dodecyl sulfate (SDS)-polyacrylamide gel electrophoresis and Coomassie blue staining after virus pel-leting. Virus titers were determined by plaquing on LLC-MK2 cells.

Virus infections. Cells were infected at a multiplicity of infection of 20 inDulbecco’s modified Eagle’s medium containing 10% fetal bovine serum. Afteran absorption period of 1 to 2 h, the inoculum was removed and replaced withfresh medium.

Plasmids, transient transfections, and luciferase assay. IRF-3 (54), IRF-3�N(41), and IRF-3-ribozyme (67) were obtained from John Hiscott and Paula Pitha.pDsRed2, expressing red fluorescent protein (RFP), was from Clontech. Thereporter plasmid with the firefly luciferase gene under the control of the humanIFN-� promoter was described by King and Goodbourn (35) and is referred tohere as p�-IFN-fl-lucter. pTK-rl-lucter, used as a transfection standard, containsthe herpes simplex virus thymidine kinase promoter region upstream of theRenilla luciferase gene (Promega). For transfections, 100,000 cells were plated insix-well plates 20 h before transfection with 1 �g of p�-IFN-fl-lucter, 0.3 �g ofpTK-rl-lucter, 1 �g of IRF-3-expressing plasmid, and 6.9 �l of Fugene (Roche)according to the manufacturer’s instructions. At 24 h posttransfection, the cellswere (or were not) infected with SeV recombinants or treated with 50 �g ofpoly(I)-poly(C) (Sigma, St. Louis, Mo.) per ml. Twenty hours later, cells wereharvested and assayed for firefly and Renilla luciferase activities (dual-luciferasereporter assay system; Promega). Relative expression levels were calculated bydividing the firefly luciferase values by those of Renilla luciferase.

RNA extraction and quantification. Total RNA was extracted with Trizolreagent (Invitrogen), and two dilutions were electrophoresed on agarose-HCHOgels. The gels were stained with ethidium bromide, and the intensities of the 18Sand 28S rRNA bands were quantified by using the ChemiDoc System (Bio-Rad)and One-D-scan software. All samples were diluted to a final concentration of1 �g/�l so that their subsequent transcription into DNA, if not quantitative,would be little influenced by this parameter.

RT and real-time PCR via TaqMan. Ten microliters of total RNA was mixedwith 0.5 �g of random hexamer primer (Promega) and subjected to a reversetranscription (RT) reaction with Superscript enzyme (Gibco), as described by themanufacturer, in a total volume of 50 �l. Two microliters of each cDNA was thencombined with 1 �l of internal control (either 20� ribosomal 18S or humanglyceraldehyde-3-phosphate dehydrogenase [GAPDH] [Applied Biosystems]),11 �l of MasterMix (Eurogentec), 20 pmol (each) of forward and reverse prim-ers, and 4.4 pmol of TaqMan probe in a total volume of 22 �l. The followingprimers and probes (Eurogentec or Microsynth) were used: for the IFN-� gene,5�-CAGCAATTTTCAGTGTCAGAAGCT-5� (forward), 5�-TCATCCTGTCCTTGAGGCAGT-3� (reverse), and 5�-CTGTGGCAATTGAATGGGAGGCTTGA-3� (probe); for the IL-8 gene, 5�-CGGTGGCTCTCTTGGCAG-3� (forward),5�-TTAGCACTCCTTGGCAAAACT-3� (reverse), and 5�-CTTCCTGATTTCTGCAGCTCTGTGTGAAGGT-3� (probe); for the IL-6 gene, 5�-TCCAGGAGCCCAGCTATGAA-3� (forward), 5�-CCCAGGGAGAAGGCAACTG-3� (re-verse), and 5�-TCCTTCTCCACAAGCGCCTTCGG-3� (probe); for the 6-16gene, 5�-CCTGCTGCTCTTCACTTGCA-3� (forward), 5�-AGCCGCTGTCCGAGCTC-3� (reverse), and 5�-TGGAGGCAGGTAAGAAAAAGTGCTGCTCGG-3� (probe); for the N gene of SeVZ, 5�-GCAATAACGGTGTCGATCACG-3�(forward), 5�-GATCCTAGATTCCTCCTACCCCA-3� (reverse), and 5�-CGAAGATGACGATACCGCAGCAGTAGC-3� (probe); and for the N gene of

SeVM, 5�-CGAAGAGGATGATGCCGC-3� (forward), 5�-GGGTCATGTATCCTAAATCCTCGT-3� (reverse), and 5�-CAGCAGCTGGGATGGGAGGAAT-3� (probe). Real-time PCR was carried out in a 7700 sequence detector(Applied Biosystems, Foster City, Calif.).

Generation of customized cDNA arrays. Macroarrays were prepared as de-scribed previously (55). 5� IMAGE clones 0.5 to 0.8 kb in length were chosen andobtained from the Human Genome Mapping Project (Hinxton, United King-dom), plated onto L agar plates and grown overnight at 37°C. Single colonieswere picked and propagated overnight in Luria-Bertani medium containing 50�g of ampicillin per ml. Bacterial lysates were generated by 1:10 dilution indistilled water. From these lysates, inserts were amplified by PCR as describedabove. After purification (QIAquick PCR purification kit; Qiagen, Crawley,United Kingdom), PCR products were sequenced (ABI Prism; Applied Biosys-tems). PCR-amplified cDNAs were transferred into 96-well plates and spottedmanually onto dry nylon membranes (Hybond N�; Amersham Pharmacia, LittleChalfont, United Kingdom) in triplicates by using 96-pin replicators (NalgeNunc, Naperville, Ill.; V&P Scientific, San Diego, Calif.). Membranes were airdried, denatured by alkaline treatment, and then neutralized. The membraneswere again air dried and UV cross-linked prior to the experiment.

Generation of labeled cDNA, hybridization, washing of membranes and anal-ysis. Radiolabeled cDNA was generated from 10 �g of total RNA by RT with400 U of reverse transcriptase (Superscript II; Gibco) in the presence of 30 �Ciof [�-33P]dCTP. After RT, residual RNA was hydrolyzed by alkaline treatmentat 70°C for 20 min. For removal of unincorporated nucleotides, the cDNA waspurified by using G-50 columns (Amersham Pharmacia) according to the instruc-tions of the manufacturer. Before hybridization to the arrays, the labeled cDNAwas mixed with 50 �g of COT-DNA (Gibco) and 10 �g of poly(A) DNA (Sigma),denatured at 95°C for 5 min, and hybridized for 1 h to minimize nonspecificbinding. The cDNA was then added directly to the membranes, which had beenprehybridized in 20 ml of hybridization buffer for at least 30 min. The membraneswere hybridized for 16 h at 65°C in hybridization bottles (Amersham Pharmacia)in a rotary hybridization oven. After hybridization, the hybridization buffer wasdiscarded and replaced by 150 ml of washing buffer. The membranes werewashed once in 2� SSC (1� SSC is 0.15 M NaCl plus 0.015 M sodium citrate)–0.1% SDS, twice in 0.2� SSC–0.1% SDS, and once in 0.1� SSC–0.1% SDS for20 min each at 65°C. The membranes were then exposed to phoshorimagescreens for 48 h and scanned with a phosphorimager (Storm; Molecular Dynam-ics, Little Chalfont, United Kingdom). For analysis, images were analyzed withImageQuant (Molecular Dynamics). Further data analysis was performed withExcel (Microsoft).

IL-8 assay. Levels of IL-8 in supernatants and in cell lysates were determinedby a sandwich enzyme-linked immunosorbent assay with paired monoclonalantibodies (Pierce) as described by the manufacturer. The IL-8 concentrationswere normalized by using total protein levels in the lysate.

RESULTS

In our experiments we have used two strains (or lineages) ofSeV whose history is relevant to studies of virulence. Thenatural host of SeV has not been clearly identified, and thisvirus is sometimes referred to as murine parainfluenza virustype I because it efficiently infects mice, causes disease, andspreads readily to uninfected animals. However, there is novirologic or serologic evidence of SeV in wild mouse popula-tions (29). There are two known lineages of SeV, Z/H/Fushimiand Ohita M/Hamanatsu (12, 31, 53, 62). The nucleotide se-quences within each lineage are 99% identical, and they are89% identical between lineages. Z/H/Fushimi comes from vi-ruses isolated in the early 1950s after an epidemic of pneumo-nitis of newborn infants in Sendai, Japan (29, 56). These SeVshave been passaged extensively in eggs in various labs since the1950s, and they are moderately virulent for mice (50% lethaldose [LD50] 103 to 104 PFU). All of the SeVZ strains usedin this study (including the wt) are recombinants.

Ohita M (SeVM) and Hamanatsu, in contrast, are highlyvirulent (LD50, 102), low-egg-passage (nonrecombinant) vi-ruses isolated from two completely separate, very severe epi-demics of animal houses in Japan. This lineage is presumably

7904 STRAHLE ET AL. J. VIROL.

Page 123: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

closer to the virus in its natural (unknown) host, and it is knownthat SeV passage in eggs attenuates its virulence in mice. SeVM

grew poorly in cell culture, and a clear-plaque variant emergedthat was avirulent (LD50, �105) and contained only two aminoacid substitutions, CF170S and LE2050A (31). When placed in therSeVZ background, the CF170S mutation was found to accountfor most or all of the loss of virulence (15). This virus isreferred to here as SeVM-CF170S. SeVM-CF170S appeared toinitiate the infection of mice normally, but the infection waslimited to the first day. This was the first evidence that the Cgene, like the SeV V gene (34), was involved in countering hostinnate defenses. Although SeVZ-wt is attenuated relative toSeVM-wt in laboratory mice, rSeVZ-wt still replicates as effi-ciently in the monkey and chimpanzee models of human re-spiratory disease as human parainfluenza virus type 1 (56), thevirus which is most closely related to SeV and which is endemicin children.

We have used a cDNA array designed to study the humancell response to IFN-�/� (55) to monitor the effects of variousSeV infections on host mRNA levels. Around 150 genes ofinterest were selected from the UniGene database. Thesegenes comprise known ISGs and genes of intrinsic interestwhich might or might not be induced by IFNs in different cellsystems. They include genes involved in cell proliferation, im-mune responses, and the responses to a variety of cytokines(see Table 1 of reference 55). We compared matched sets ofSeV carrying two different mutations in the C gene (SeVM-CF170S and SeVZ-C�10-15), whose products interact withSTAT1 in different ways (to interfere with IFN signaling and toinduce STAT1 instability, respectively) (Fig. 1D). We also usedmatched sets of SeV carrying mutations in the viral replicationpromoters (SeVZ-GP1-42 and -GP31-42) that prevent apopto-sis and lead to persistent infections (the numbers refer to thenucleotides of the genomic promoter that have been replacedwith the equivalent sequences of the antigenomic promoter)(16, 20). Promoter mutations are thought to act via mutantleader RNAs that are abundantly transcribed from the geno-mic replication promoter and which bind to cellular RNA-binding proteins that regulate mRNA fate (28).

Upon infection with SeVM-wt or SeVZ-wt, many of thesemRNA levels remain unchanged (Fig. 1A to C). This lack ofresponse is presumably due in part to active SeV countermea-sures that neutralize the cell’s antiviral response (24). Uponinfection with the mutant viruses, the mRNA levels of 15 of the150 genes examined were elevated, and three patterns of geneactivation were seen (all values are triplicates, and a twofolddifference is very significant [55]). One series of genes (group1, nine genes) (Fig. 1A) is activated by SeV-CF170S infectionalone; these mRNA levels are unchanged in SeV-C�10-15 andSeV-GP1/31-42 infections. The CF170S substitution inactivatesthe ability of all four C proteins (C�, C, Y1, and Y2) to stablybind STAT1 and to interdict IFN signaling (18, 59). Accordingto this view, any of the four C proteins may function to preventthese mRNA levels from increasing during SeV infection (Fig.1D) (14). The IL-6 gene is the sole representative of group 2;it is activated by SeV-C�10-15 as well as SeV-CF170S, but notby the promoter mutants or the wt viruses (Fig. 1B). Accordingto this view, a second function of the C gene, specific to theNH2-terminal 23 amino acids present only in the longer C

proteins, is also required for SeV to prevent IL-6 activation.The third group, consisting of five genes, is activated by SeV-C�10-15 and SeV-GP1-42 infections as well as SeV-CF170S, butnot by SeV-GP31-42 or SeV-wt infections (Fig. 1C). Appar-ently, a third function provided specifically by the first 30 nu-cleotides (nt) of the genomic promoter (or leader RNA) is alsorequired to prevent activation of genes such as that of IL-8(Fig. 1C). This third function is not the ability of mutant leaderRNA to bind TIAR, a host RNA-binding protein important forvirus-induced apoptosis, as this occurs with SeV-GP31-42 aswell (28). In summary, comparative analysis of host gene acti-vation with SeV with specific mutations has identified threegroups of cellular genes that respond differently to SeV infec-tion.

Real-time RT-PCR estimations of mRNA levels. IFN signal-ing through the JAK/STAT pathway activates many ISGs (suchas 6-16, PKR, etc.) that collectively contribute to the cellularantiviral response. SeVs that interdict IFN signaling wouldtherefore also prevent the activation of these ISGs. Whilemany of the genes activated by the mutant SeVs are wellknown ISGs, IL-6 and IL-8 are known to be non-ISGs; treat-ment of 2fTGH cells with 1,000 IU of IFN-� does not increaseIL-6 or IL-8 mRNA levels over those of the untreated control(55). We therefore continued our study of selected host genesactivated by SeV by real-time RT-PCR (TaqMan), a methodthat is more quantitative than DNA arrays. We first examinedIL-8 (a chemokine) and IFN-�, an early host response proteinwhose gene was absent in the DNA array. 2C4 cells (a 2fTGH-derived cell line) were infected with 20 PFU of the variousSeVs per cell, and the levels of various host mRNAs weredetermined, including that of GADPH as an internal control(see Materials and Methods). All four SeVs (SeVM-wt, CF170S,SeVZ-wt, and C�10-15) grow relatively well in 2C4 cells, asindicated by their accumulated N mRNAs (Fig. 2 and 3) or Nproteins (data not shown); however, the SeVM-wt infectionsaccumulated ca. 2-fold less N mRNA than the three other SeVinfections (Fig. 2C). The relative levels of virus replication arepresumably important in stimulating the host response, andSeVM-wt infections might therefore be expected to activateIL-8 and IFN-� less strongly than the other SeVs for thisreason alone. We found that IL-8 and IFN-� mRNA levelswere elevated �20-fold in SeVM-CF170S versus SeVM-C-wtinfections. A strong difference was also found between SeVZ-C�10-15 and SeVZ-C-wt infections, where N mRNAs had ac-cumulated identically (Fig. 2). Moreover, IL-8 and IFN-�mRNA levels were also elevated in other SeVM-CF170S versusSeVM-C-wt infections of 2C4 cells, where N mRNAs had ac-cumulated identically (Fig. 3). Thus, specific mutations in twodifferent regions of the SeV C proteins lead to increased acti-vation of IL-8 and IFN-�. In all cases, IL-8 and IFN-� mRNAlevels were more strongly increased by SeVM-CF170S than bySeVZ-C�10-15.

We also examined the effects of various SeV infections ofcells transfected with plasmids expressing IRF-3 (or RFP as aneutral control), and in some cases the cells were also treatedwith 50 �g of poly(I)-poly(C) (dsRNA) per ml. Elevated IRF-3levels should enhance the antiviral response of the cells to SeVinfection, and the combined treatment is thought to approxi-mate a virus infection in itself and should potentiate the anti-viral response. This appears to be so, as the levels of IL-6, IL-8,

VOL. 77, 2003 SENDAI VIRUS TARGETING OF INFLAMMATORY RESPONSES 7905

Page 124: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

FIG. 1. Comparison of host mRNA levels in 2fTGH cells infected with various SeVs. (A to C) Parallel cultures of 2fTGH cells were infectedwith 20 PFU of the various SeVs per cell. Total cytoplasmic RNA was prepared with Trizol at 24 hpi, and equal amounts (10 �g) were used asa template for oligo(dT)-primed [33P]cDNA synthesis. The [33P]cDNA was annealed to triplicate DNAs arrayed on nylon membranes, and the[33P]cDNA bound was quantitated in a PhosphorImager. The graphs show the fold increase in each mRNA relative to the mock control. (D)Schematic representation of the viral mutations and their effects on host gene activation. The C proteins are shown as two telescoping boxesrepresenting the longer (C� and C) and shorter (Y1 and Y2) C proteins, whose activities during infection, and the mutations investigated, areindicated. The promoter mutation GP42 is thought to exert its effect via mutant leader (Le) RNA. The presumed requirement for the various wtgenetic elements to prevent host gene activation is shown. The names of the mutant SeVs used are also indicated. Instab., instability; sig., signaling.

7906 STRAHLE ET AL. J. VIROL.

Page 125: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

and IFN-� mRNAs induced by SeV infection in general in-creased with increasing proresponse treatment (i.e., RFP,IRF-3, and IRF-3 plus dsRNA) (Fig. 3). IRF-3 overexpression(Fig. 3A to C, IRF-3 versus RFP) enhanced IFN-� and IL-6activation by SeV but did not enhance that of IL-8. The addi-tional dsRNA treatment had little effect on further enhancingIFN-� and IL-6 mRNA levels but strongly enhanced IL-8mRNA levels. These differences in the enhancement of IL-6and -8 and IFN-� activation upon treatment with IRF-3 with orwithout dsRNA presumably reflect different activation path-ways in response to SeV infection. It is possible that IL-8activation requires both IRF-3 and dsRNA, whereas IRF-3 issufficient for IL-6 and IFN-� activation.

SeV activation of IL-6, IL-8, and IFN-� is independent ofIFN signaling. 2fTGH human fibrosarcoma cells were chosenfor these experiments because sublines defective in specificcomponents of the IFN signaling system have been generatedfrom these cells by X irradiation (48). U5A cells, for example,are defective in the IFN-�/� receptor 2 chain, which is essentialfor IFN-�/� signaling, and these cells have been restored toIFN sensitivity by complementation with the IFNAR2 gene(U5A�IFNAR2 cells) (43). Even though IL-6 and -8 are notactivated upon simple IFN treatment of uninfected cells, IFNsecreted during SeV infection may act differently, as additionalsignaling pathways are being induced by the virus infection.Moreover, CF170S (in contrast to C�10-15) does not preventIFN signaling, and it is important to know whether this phe-notype is responsible for the activation of IL-6 and -8. Wetherefore examined the various SeV infections of U5A as wellU5A�IFNAR2 cells to determine whether activation of IL-6and IFN-� by SeV-CF170S and SeV-C�10-15 required IFNsignaling. We also examined the activation of the 6-16 gene, aknown ISG, as a positive control (10). As shown in Fig. 4, bothSeV-CF170S and SeV-C�10-15 activated IL-6, IL-8, and IFN-�in U5A cells relative to SeV-wt infection. The IFNAR2-com-

plemented cell line yielded similar results, except that theactivation of these genes was paradoxically reduced in U5A�IFNAR2 cells relative to U5A cells. In contrast to the case forIL-6, IL-8, and IFN-�, little or no activation of 6-16 occurredin SeV-C mutant-infected U5A cells, whereas a modest acti-vation was evident in U5A�IFNAR2 cells. Moreover, 6-16 wasthe only mRNA whose levels in U5A�IFNAR2 cells exceededthose in U5A cells. The activation of IL-6, IL-8, and IFN-�during SeV infection, in contrast to that of 6-16, is thus largelyindependent of IFN signaling.

STAT1-defective U3A cells. The SeV C proteins interactwith STAT1 in two ways (Fig. 1D). C and STAT1 form a stablecomplex in vitro and during SeV infection, and this complex isassociated with a loss of IFN signaling. These events areblocked by the CF170S mutation but not by C�10-15. Theshorter Y proteins are also active in this respect. The longer Cproteins alone also induce STAT1 instability, and in contrast totheir effects on IFN signaling, this effect does not require F170(Fig. 1D). To examine whether SeV-C mutant-induced activa-tion of IL-6, IL-8, and IFN-� requires STAT1, U3A cells,which are known to be defective for STAT1, were examined(45). However, we were unable to examine the companionU3A�STAT1 cells, as these cells were found to have lostSTAT1 expression. Moreover, attempts to recomplement U3Acells with STAT1 failed (data not shown).

The results of the U3A cell infections are shown in Fig. 5. Asbefore, IL-6 and -8 and IFN-� mRNA levels were all clearlyincreased in SeV-CF170S infections relative to SeV-wt infec-tions. However, in contrast to the case for U5A and 2fTGHcells, SeV-C�10-15 infections did not contain enhancedmRNA levels relative to SeV-wt infections. Given that STAT1is probably not the only gene that has been lost in U3A cellsthat have been X irradiated (which randomly destroys chro-mosomal DNA), we can conclude only that enhanced mRNAlevels due to SeV-C�10-15 infection require STAT1 and/or

FIG. 1—Continued.

VOL. 77, 2003 SENDAI VIRUS TARGETING OF INFLAMMATORY RESPONSES 7907

Page 126: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

another, unknown gene, whereas activation due to SeV-CF170S

requires neither STAT1 nor any other gene destroyed in U3Acells.

IL-8 gene activation and IL-8 secretion. IL-8 is not known toact intracellularly. We therefore examined whether the SeV-induced IL-8 gene activation in U5A/U5A�IFNAR2 cells also

led to increased IL-8 protein synthesis and secretion. WhenIL-8 protein levels in cytoplasmic extracts of the various SeV-infected cells were examined, they were found to roughly mir-ror the mRNA levels (Fig. 4E). However, when the culturesupernatants were examined, a somewhat different result wasfound (Fig. 4F). Whereas the increased IL-8 mRNA level ofSeV-C�10-15-infected U5A cells was accompanied by stronglyincreased IL-8 secretion, that of U5A�IFNAR2 cells led toonly a modest increase in IL-8 secretion. Moreover, the in-

FIG. 2. Effects of SeV C gene mutations on IFN-� and IL-8 mRNAlevels during infection. Parallel cultures of 2C4 cells were infected (ornot) in triplicate for 24 h with 20 PFU of the various SeVs per cell.Total cytoplasmic RNA was prepared from each culture, and the sameamount of RNA (ca. 1 �g) was transcribed into cDNA with randomhexadeoxynucleotides and murine leukemia virus reverse transcrip-tase. The relative amounts of IFN-� and IL-8 gene sequences, relativeto that of GADPH as an internal control, were determined by real-time PCR (see Materials and Methods). The average levels of themRNAs and their deviations in the triplicate infections are shown. Therelative SeV N mRNA levels of the SeVZ-C-wt and -C�10-15 infec-tions were also determined (SeVZ and SeVM are 10% different insequence, and their detection requires different primers and probes).

FIG. 3. Effects of IRF-3 and dsRNA treatment on SeV-inducedhost gene activation. Parallel cultures of 2C4 cells were transfectedwith pRFP or pIRF-3. The cultures were then infected (or not) with 20PFU of the various SeVs per cell at 20 h posttransfection, and some ofthe cultures were also treated with 50 �g of poly(I)-poly(C) per ml.The cells were harvested at 24 hpi. The relative amounts of IFN-�,IL-6, and IL-8 gene sequences present were determined as describedfor Fig. 2.

7908 STRAHLE ET AL. J. VIROL.

Page 127: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

creased IL-8 mRNA levels in either SeV-CF170S-infected cellline did not lead to clearly increased IL-8 secretion.

IL-8 expression can be controlled at both the transcriptionaland posttranscriptional levels. In polarized epithelial monolay-ers, S. enterica serovar Typhimurium-induced IL-8 secretionrequires not only the activation of NF-�B and production of

IL-8 mRNA but also the activation of the small, Rho familyGTPases Cdc42 and Rac1, which regulate endocytic proteintraffic from the Golgi network to the basolateral surface of thecell. In the absence of Cdc42 or Rac1 function, IL-8 mRNAlevels increase in response to bacterial infection but IL-8 is notsecreted (3), similar to the case for our SeV-CF170S-infected

FIG. 4. SeV infection of cells defective for the IFN receptor and their complemented pseudo-wt derivatives. (A to D) Parallel cultures of U5Aor U5A�IFNAR2 cells were infected (or not) in duplicate with 20 PFU of the various SeVs per cell for 24 h. The relative amounts of IFN-�, IL-6,IL-8, and 6-16 mRNAs present were determined as described for Fig. 2. (E and F) Equal samples of the culture supernatants (F) or cytoplasmicextracts of the cultures (E) were analyzed for IL-8 protein levels by enzyme-linked immunosorbent assay.

VOL. 77, 2003 SENDAI VIRUS TARGETING OF INFLAMMATORY RESPONSES 7909

Page 128: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

U5A cells. The requirement for Cdc42 and Rac1 activation,moreover, is cell type dependent (9). The CF170S and C�10-15mutations thus appear to affect IL-8 secretion differently.

SeV prevents IFN-� gene activation in several ways. Themanner in which IFN-� transcription is induced by virus infec-tion is well studied, and the activation of IRF-3 is central to thisprocess. IRF-3 is expressed constitutively and is found in thecytoplasm in an inactive, unphosphorylated state. Upon virusinfection or dsRNA treatment of cells, IRF-3 is phosphory-lated by an unknown kinase and translocates to the nucleus,

where, together with other transcription factors such as NF-�B(which is itself also directly activated by virus infection ordsRNA), it activates IFN-� transcription (41, 65, 68). Phos-phorylation of IRF-3 after viral infection is the first step in theactivation of a gene program that includes a positive feedbackloop of IFN-�/� and IRF family members (60). The resultsdescribed above suggest that the SeV C gene encodes func-tions that prevent virus-induced IFN-� transcription (directlyor indirectly). During the course of this work, it was reportedthat the SeV V protein, as well as the V proteins of therubulaviruses SV5 and hPIV2, also prevented IFN-� transcrip-tion (26, 50, 64).

Given that SeV appears to use two viral genes (C and V) toneutralize IFN-� expression, we have examined a broaderpanel of mutant rSeV infections for their relative activation ofthe IFN-� promoter compared to that of dsRNA treatment.Besides SeV-CF170S and C�10-15, we examined two promotermutants, SeV-AGP55, in which the first 55 nt of the antigeno-mic promoter is replaced with the equivalent leader sequencesof the genomic promoter (38). SeV-AGP55 transcribes leaderRNA from both promoters (and no trailer RNA). The con-verse SeV-GP48 has the first 48 nt of the genomic promoterreplaced with the equivalent trailer sequences, and SeV-GP48transcribes basically trailer RNA from both promoters (and noleader RNA; GP48 and GP1-42 are identical in this respect[data not shown]) (19, 20). Finally, we examined SeV-V�/W��, which contains a stop codon at the beginning of the Vopen reading frame (ORF), such that edited V mRNAs aretranslated into W-like proteins, and specifically no V protein isexpressed (8).

2fTGH cells were transfected with a reporter plasmid inwhich luciferase is controlled by the IFN-� promoter (pIFN�-luciferase). To determine whether virus-induced IFN-� tran-scription required IRF-3 activation, the cells were cotransfect-ed with either a dominant-negative mutant of IRF-3 (IRF-3�N),an anti-IRF-3 ribozyme, or an empty control plasmid (seeMaterials and Methods). The transfected cultures were theninfected (or not) with the various SeVs (or treated withdsRNA) 24 h later and were harvested at 20 h postinfection(hpi). As shown in Fig. 6, with the notable exception of SeV-AGP55, all of the mutant SeV infections induced the reportermore strongly than SeV-wt and as well as dsRNA treatment. Inall cases, the coexpression of IRF-3�N or an anti-IRF-3 ri-bozyme prevented the SeV-induced expression of the reporter.Thus, mutations in two regions of the C protein that carry outdifferent functions, as well as the loss of leader RNA expres-sion or the expression of the V protein, all lead to IFN-�promoter activation in an IRF-3-dependent manner. Overex-pression of the W protein cannot compensate for the lack of Vprotein, so the highly conserved Cys-rich carboxyl domain of Vis specifically required. Only the loss of trailer RNA expression(SeV-AGP55) did not result in IFN-� activation.

Leader and trailer RNAs, the promoter-proximal productsof viral RNA synthesis, are AU rich and are thought to bind tocellular RNA-binding proteins that bind AU-rich elements(28). In contrast to leader RNA, trailer RNA is expressedrelatively late in infection (upon antigenome accumulation)and would not be expected to counteract immediate-early re-sponse genes. However, many of the other viral products thatdo not have a dedicated essential role in the replication ma-

FIG. 5. SeV infection of cells defective for STAT1. Parallel cul-tures of U3A cells were infected (or not) in duplicate with 20 PFU ofthe various SeVs per cell for 24 h. The relative amounts of IFN-�, IL-6,and IL-8 mRNAs present were determined as described for Fig. 2.

7910 STRAHLE ET AL. J. VIROL.

Page 129: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

chinery per se (C and V proteins and leader RNA) appear tobe pressed into service to coordinately prevent IFN-� activa-tion.

DISCUSSION

We have used cDNA arrays to compare the activation ofvarious cellular genes in response to infection with SeVs thatcontain mutations in their C proteins or leader regions. Threegroups of cellular genes were identified in this way (Fig. 1). Ofthe mutant SeV examined by DNA arrays, only the leader(31-42) mutation failed to activate any of the cellular genes relativeto SeV-wt, and consistent with this failure, SeV-GP31-42 wasthe only one of the four mutant SeVs not to have lost virulencein mice (M. Itoh, unpublished data). The CF170S mutation isassociated with a ca. 5-log-unit increase in LD50 (31), and thismutation appears to be the most important in cell cultureinfections as well as in mice (15). This mutation activates IL-6,

IL-8, and IFN-� more strongly than C�10-15, and only thismutation activates all three groups of cellular genes that re-spond to SeV infection.

The IFN-� gene is both a primary response gene and anISG, and it plays a central role in initiating the IFN-inducedantiviral response. This is the first report that the SeV C pro-teins and leader RNA counteract the expression of this keyprimary response gene, and it confirms that the report of Pooleet al. (50) that the product of our hemagglutinin-tagged V geneis active in this respect. As expected, activation of IFN-� re-quired activation of IRF-3 (Fig. 6) and was independent of IFNsignaling (Fig. 4), but the details of how this occurs remain tobe elucidated. We have also provided evidence that the SeV Cproteins and leader RNA counteract the expression of thechemokine IL-8. Infections by other viruses, e.g., respiratorysyncytial virus (RSV) (44), dengue virus (3), hepatitis C virus(49), and human immunodeficiency virus type 1 (46, 52), areknown to induce IL-8 secretion, as do infections by bacteriaand parasites, including Mycobacterium tuberculosis (66). In arelated vein, cytomegalovirus, a large DNA virus, encodes achemokine receptor that may facilitate virus replication (13),and human herpesvirus 8/Kaposi’s sarcoma virus carries fourORFs whose products are related to chemokines (42). Viralmodulation of chemokine expression presumably representsone aspect of the continuous battle between viral parasites andantiviral, inflammatory, and immune responses of the host.SeV infection has been reported to induce the CXC chemo-kine RANTES via the activation of IRF-3 and NF-�B (23, 40).IL-8 may not have been noticed in these earlier studies, be-cause SeV-wt induces very little IL-8 (Fig. 2). Our resultssuggest that SeV targets the inflammatory and adaptive im-mune responses (IL-6 and IL-8) as well as the IFN-inducedintracellular antiviral state (IFN-� and STAT1). As IFN-� andIL-8 transcription both depend on NF-�B activation, SeV maytarget this key transcription factor as well.

The leader(1-42), C�10-15, CF170S, and V�/W�� mutationsappear to disrupt four distinct elements in the SeV program tocounteract the cellular antiviral response. The facts that theyall lead to enhanced levels of IFN-� mRNA and that at leastthree of them increase IL-8 mRNA levels suggest that SeVemploys a multifaceted approach to inhibit viral clearance byinflammatory cells as well as to prevent the IFN-induced an-tiviral state, sometimes using the same viral macromoleculesdue to its limited coding capacity. The best-studied example ofparamyxovirus-induced activation of IL-8 is that of RSV (21,69). The IL-8 promoter in A549 cells can be induced by RSVinfection in at least three distinct pathways: via tumor necrosisfactor alpha (which requires only an intact NF-�B bindingsite), directly by intracellular RSV replication (which also re-quires other transcription factor binding sites) (5), and via theinteraction of the viral F protein with Toll-like receptor 4 (inwhich IRF-3 plays an important role) (36). Measles virus Hprotein interaction with Toll-like receptor 2 also activates IL-8(1). If all three parallel cellular pathways for IL-8 expressionoperate during SeV infection of 2fTGH cells, several differentSeV products will be required to effectively prevent IL-8 acti-vation (Fig. 1D).

Rubulaviruses do not express C proteins, but their V pro-teins have recently been found to prevent IFN-� expression bypreventing the activation of IRF-3 and NF-�B, as well as in-

FIG. 6. Effect of SeV infection on IFN-� promoter reporter geneexpression. Parallel cultures of 2fTGH cells were transfected with amixture of pINF�-fl-lucter and pTK-rl-lucter along with pIRF-3-�N,pAnti-IRF-3-ribozyme, or an empty plasmid (Ctrl), as indicated inMaterials and Methods. Duplicate cultures were infected (or not) with20 PFU of the various SeVs per cell or treated with dsRNA andharvested at 20 hpi. The relative activities of the fl- and rl-luciferaseswere determined. Error bars indicate standard deviations.

VOL. 77, 2003 SENDAI VIRUS TARGETING OF INFLAMMATORY RESPONSES 7911

Page 130: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

ducing the degradation of STAT1 or STAT2 (26, 50). TheRubulavirus V proteins thus also counteract more than onearm of the innate antiviral response. The versatility of theseviral gene products continues to surprise us. The SeV C pro-teins have been more intensively studied than SeV V proteinor leader RNA. Like the influenza A virus NS1 protein (6, 63)and hepatitis C virus NS5A protein (49), the SeV C proteinsare pleiotropic polypeptides that have multiple activities dur-ing infection, presumably due to their interaction with variousviral and cellular proteins. Their multiple functions, deci-phered in large part via C gene mutations, include (i) stimu-lation of viral RNA synthesis early in infection (SeV-C�/C�

infections exhibit a 10-h delay in the accumulation of viralproducts) (37); (ii) inhibition of viral RNA synthesis in a pro-moter-specific manner late in infection, by interacting with theP4-L vRdRP (this selective inhibition may promote the switchfrom mRNA synthesis to genome replication and increase thefidelity of vRdRP promoter recognition) (4, 61); (iii) a role invirion assembly, possibly by interaction with the matrix (M)protein (SeV-4C� particles are poorly infectious and amor-phic) (25); (iv) interaction with STAT1 in two separate ways, toinhibit IFN signaling and to induce STAT1 instability (17); and(v) inhibition of the IRF-3-dependent activation of IFN-� andthe activation of IL-8 expression in an IFN signaling-indepen-dent manner (this work).

How C interacts specifically with all of its viral and cellularpartners remains an enigma and is reminiscent of acidic acti-vation domains of transcription factors that interact with mul-tiple partners. Acidic activation domains are “natively disor-dered” (11, 51), and this property apparently allows them tobind different surfaces with high specificity (multiple inducedfits) and limited stability. The NH2-terminal portion of themeasles virus P protein that contains the overlapping C proteinORF is, in fact, a recent example of such natively disorderedproteins, in accordance with the prediction of algorithms thatdetect unstructured regions (47). By using the same algorithms(PONDR), the SeV C protein is strongly predicted to be na-tively disordered, and this property is shared with the commonNH2-terminal portions of rubulavirus V, I, and P proteins (17,26). The C�10-15 deletion, moreover, is in a region of C withthe highest prediction of disorder. It will be of interest toexamine whether purified SeV C proteins are indeed nativelydisordered.

ACKNOWLEDGMENTS

We thank Ian Kerr (London, United Kingdom) for useful discus-sions and for numerous cell lines, Paula Pitha (Baltimore, Md.) andJohn Hiscott (Montreal, Canada) for the IRF-3 reagents, and theJerome Pugin lab (University of Geneva) for help with the IL-8 proteinassays.

ADDENDUM IN PROOF

Fujii et al. (Y. Fujii, T. Sakaguchi, K. Kiyotani, C. Huang, N.Fukuhara, Y. Egi, and T. Yoshida, J. Virol. 76:8540–8547,2002) have shown that mutations in the leader region specifi-cally attenuate virus virulence in mice.

REFERENCES

1. Bieback, K., E. Lien, I. M. Klagge, E. Avota, J. Schneider-Schaulies, W. P.Duprex, H. Wagner, C. J. Kirschning, V. ter Meulen, and S. Schneider-

Schaulies. 2002. Hemagglutinin protein of wild-type measles virus activatestoll-like receptor 2 signaling. J. Virol. 76:8729–8736.

2. Biron, C. A., and G. C. Sen. 2001. Interferons and other cytokines, p.321–349. In D. M. Knipe and P. M. Howley (ed.), Fields virology. Lippincott,Williams, and Wilkins, Philadelphia, Pa.

3. Bosch, I., K. Xhaja, L. Estevez, G. Raines, H. Melichar, R. V. Warke, M. V.Fournier, F. A. Ennis, and A. L. Rothman. 2002. Increased productionof interleukin-8 in primary human monocytes and in human epithelialand endothelial cell lines after dengue virus challenge. J. Virol. 76:5588–5597.

4. Cadd, T., D. Garcin, C. Tapparel, M. Itoh, M. Homma, L. Roux, J. Curran,and D. Kolakofsky. 1996. The Sendai paramyxovirus accessory C proteinsinhibit viral genome amplification in a promoter-specific fashion. J. Virol.70:5067–5074.

5. Casola, A., R. P. Garofalo, M. Jamaluddin, S. Vlahopoulos, and A. R.Brasier. 2000. Requirement of a novel upstream response element in respi-ratory syncytial virus-induced IL-8 gene expression. J. Immunol. 164:5944–5951.

6. Chen, Z., and R. M. Krug. 2000. Selective nuclear export of viral mRNAs ininfluenza-virus-infected cells. Trends Microbiol. 8:376–383.

7. Darnell, J. E. J. 1997. STATs and gene regulation. Science 277:1630–1635.8. Delenda, C., S. Hausmann, D. Garcin, and D. Kolakofsky. 1997. Normal

cellular replication of Sendai virus without the trans-frame, nonstructural Vprotein. Virology 228:55–62.

9. Denk, A., M. Goebeler, S. Schmid, I. Berberich, O. Ritz, D. Lindemann, S.Ludwig, and T. Wirth. 2001. Activation of NF-kappa B via the Ikappa Bkinase complex is both essential and sufficient for proinflammatory geneexpression in primary endothelial cells. J. Biol. Chem. 276:28451–28458.

10. Der, S. D., A. Zhou, B. R. Williams, and R. H. Silverman. 1998. Identificationof genes differentially regulated by interferon alpha, beta, or gamma usingoligonucleotide arrays. Proc. Natl. Acad. Sci. USA 95:15623–15628.

11. Feldmann, H., and H. D. Klenk. 1996. Marburg and Ebola viruses. Adv.Virus Res. 47:1–52.

12. Fujii, Y., K. Kiyotani, T. Yoshida, and T. Sakaguchi. 2001. Conserved andnon-conserved regions in the Sendai virus genome: evolution of a genepossessing overlapping reading frames. Virus Genes 22:47–52.

13. Gao, J. L., and P. M. Murphy. 1994. Human cytomegalovirus open readingframe US28 encodes a functional beta chemokine receptor. J. Biol. Chem.269:28539–28542.

14. Garcin, D., J. Curran, M. Itoh, and D. Kolakofsky. 2001. Longer and shorterforms of Sendai virus C proteins play different roles in modulating thecellular antiviral response. J. Virol. 75:6800–6807.

15. Garcin, D., M. Itoh, and D. Kolakofsky. 1997. A point mutation in the Sendaivirus accessory C proteins attenuates virulence for mice, but not virus growthin cell culture. Virology 238:424–431.

16. Garcin, D., P. Latorre, and D. Kolakofsky. 1999. Sendai virus C proteinscounteract the interferon-mediated induction of an antiviral state. J. Virol.73:6559–6565.

17. Garcin, D., J. B. Marq, S. Goodbourn, and D. Kolakofsky. 2003. The amino-terminal extensions of the longer Sendai virus C proteins modulate pY701-Stat1 and bulk Stat1 levels independently of interferon signaling. J. Virol.77:2321–2329.

18. Garcin, D., J. B. Marq, L. Strahle, P. Le Mercier, and D. Kolakofsky. 2002.All four Sendai virus C proteins bind Stat1, but only the larger forms alsoinduce its mono-ubiquitination and degradation. Virology 295:256–265.

19. Garcin, D., T. Pelet, P. Calain, L. Roux, J. Curran, and D. Kolakofsky. 1995.A highly recombinogenic system for the recovery of infectious Sendaiparamyxovirus from cDNA: generation of a novel copy-back nondefectiveinterfering virus. EMBO J. 14:6087–6094.

20. Garcin, D., G. Taylor, K. Tanebayashi, R. Compans, and D. Kolakofsky.1998. The short Sendai virus leader region controls induction of pro-grammed cell death. Virology 243:340–353.

21. Garofalo, R., M. Sabry, M. Jamaluddin, R. K. Yu, A. Casola, P. L. Ogra, andA. R. Brasier. 1996. Transcriptional activation of the interleukin-8 gene byrespiratory syncytial virus infection in alveolar epithelial cells: nuclear trans-location of the RelA transcription factor as a mechanism producing airwaymucosal inflammation. J. Virol. 70:8773–8781.

22. Geiss, G., G. Jin, J. Guo, R. Bumgarner, M. G. Katze, and G. C. Sen. 2001.A comprehensive view of regulation of gene expression by double-strandedRNA-mediated cell signaling. J. Biol. Chem. 276:30178–30182.

23. Genin, P., M. Algarte, P. Roof, R. Lin, and J. Hiscott. 2000. Regulation ofRANTES chemokine gene expression requires cooperativity between NF-kappa B and IFN-regulatory factor transcription factors. J. Immunol. 164:5352–5361.

24. Goodbourn, S., L. Didcock, and R. E. Randall. 2000. Interferons: cell sig-nalling, immune modulation, antiviral response and virus countermeasures.J. Gen. Virol. 81:2341–2364.

25. Hasan, M. K., A. Kato, M. Muranaka, R. Yamaguchi, Y. Sakai, I. Hatano, M.Tashiro, and Y. Nagai. 2000. Versatility of the accessory C proteins of Sendaivirus: contribution to virus assembly as an additional role. J. Virol. 74:5619–5628.

26. He, B., R. G. Paterson, N. Stock, J. E. Durbin, R. K. Durbin, S. Goodbourn,

7912 STRAHLE ET AL. J. VIROL.

Page 131: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

R. E. Randall, and R. A. Lamb. 2002. Recovery of paramyxovirus simianvirus 5 with a V protein lacking the conserved cysteine-rich domain: themultifunctional V protein blocks both interferon-beta induction and inter-feron signaling. Virology 303:15–32.

27. Hobert, M. E., K. A. Sands, R. J. Mrsny, and J. L. Madara. 2002. Cdc42and Rac1 regulate late events in Salmonella typhimurium-induced inter-leukin-8 secretion from polarized epithelial cells. J. Biol. Chem. 277:51025–51032.

28. Iseni, F., D. Garcin, M. Nishio, N. Kedersha, P. Anderson, and D. Kolakof-sky. 2002. Sendai virus trailer RNA binds TIAR, a cellular protein involvedin virus-induced apoptosis. EMBO J. 21:5141–5150.

29. Ishida, N., and M. Homma. 1978. Sendai virus. Adv. Virus Res. 23:349–383.30. Israel, A. 2000. The IKK complex: an integrator of all signals that activate

NF-kappaB? Trends Cell Biol. 10:129–133.31. Itoh, M., Y. Isegawa, H. Hotta, and M. Homma. 1997. Isolation of an

avirulent mutant of Sendai virus with two amino acid mutations from a highlyvirulent field strain through adaptation to LLC-MK2 cells. J. Gen Virol.78:3207–3215.

32. Jacobs, B. L., and J. O. Langland. 1996. When two strands are better thanone: the mediators and modulators of the cellular responses to double-stranded RNA. Virology 219:339–349.

33. Karin, M., and Y. Ben Neriah. 2000. Phosphorylation meets ubiquitination:the control of NF-�B activity. Annu. Rev. Immunol. 18:621–663.

34. Kato, A., K. Kiyotani, K. Sakai, T. Yoshida, and Y. Nagai. 1997. The para-myxovirus, Sendai virus, V protein encodes a luxury function required forviral pathogenesis. EMBO J. 16:578–685.

35. King, P., and S. Goodbourn. 1994. The beta-interferon promoter responds topriming through multiple independent regulatory elements. J. Biol. Chem.269:30609–30615.

36. Kurt-Jones, E. A., L. Popova, L. Kwinn, L. M. Haynes, L. P. Jones, R. A.Tripp, E. E. Walsh, M. W. Freeman, D. T. Golenbock, L. J. Anderson, andR. W. Finberg. 2000. Pattern recognition receptors TLR4 and CD14 mediateresponse to respiratory syncytial virus. Nat. Immunol. 1:398–401.

37. Latorre, P., T. Cadd, M. Itoh, J. Curran, and D. Kolakofsky. 1998. Thevarious Sendai virus C proteins are not functionally equivalent and exertboth positive and negative effects on viral RNA accumulation during thecourse of infection. J. Virol. 72:5984–5993.

38. Le Mercier, P., D. Garcin, S. Hausmann, and D. Kolakofsky. 2002. Am-bisense Sendai viruses are inherently unstable but are useful to study viralRNA synthesis. J. Virol. 76:5492–5502.

39. Lillemeier, B. F., M. Koster, and I. M. Kerr. 2001. STAT1 from the cellmembrane to the DNA. EMBO J. 20:2508–2517.

40. Lin, R., C. Heylbroeck, P. Genin, P. M. Pitha, and J. Hiscott. 1999. Essentialrole of interferon regulatory factor 3 in direct activation of RANTES che-mokine transcription. Mol. Cell. Biol. 19:959–966.

41. Lin, R., C. Heylbroeck, P. M. Pitha, and J. Hiscott. 1998. Virus-dependentphosphorylation of the IRF-3 transcription factor regulates nuclear translo-cation, transactivation potential, and proteasome-mediated degradation.Mol. Cell. Biol. 18:2986–2996.

42. Liu, C., Y. Okruzhnov, H. Li, and J. Nicholas. 2001. Human herpesvirus 8(HHV-8)-encoded cytokines induce expression of and autocrine signaling byvascular endothelial growth factor (VEGF) in HHV-8-infected primary-effusion lymphoma cell lines and mediate VEGF-independent antiapoptoticeffects. J. Virol. 75:10933–10940.

43. Lutfalla, G., S. J. Holland, E. Cinato, D. Monneron, J. Reboul, N. C. Rogers,J. M. Smith, G. R. Stark, K. Gardiner, K. E. Mogensen, et al. 1995. MutantU5A cells are complemented by an interferon-alpha beta receptor subunitgenerated by alternative processing of a new member of a cytokine receptorgene cluster. EMBO J. 14:5100–5108.

44. Mastronarde, J. G., B. He, M. M. Monick, N. Mukaida, K. Matsushima, andG. W. Hunninghake. 1996. Induction of interleukin (IL)-8 gene expressionby respiratory syncytial virus involves activation of nuclear factor (NF)-kappaB and NF-IL-6. J. Infect. Dis. 174:262–267.

45. Muller, M., C. Laxton, J. Briscoe, C. Schindler, T. Improta, J. E. Darnell,Jr., G. R. Stark, and I. M. Kerr. 1993. Complementation of a mutant cellline: central role of the 91 kDa polypeptide of ISGF3 in the interferon-alphaand -gamma signal transduction pathways. EMBO J. 12:4221–4228.

46. Ott, M., J. L. Lovett, L. Mueller, and E. Verdin. 1998. Superinduction of IL-8in T cells by HIV-1 Tat protein is mediated through NF-kappaB factors.J. Immunol. 160:2872–2880.

47. Palangat, M., and R. Landick. 2001. Roles of RNA:DNA hybrid stability,RNA structure, and active site conformation in pausing by human RNApolymerase II. J. Mol. Biol. 311:265–282.

48. Pellegrini, S., J. John, M. Shearer, I. M. Kerr, and G. R. Stark. 1989. Use of

a selectable marker regulated by alpha interferon to obtain mutations in thesignaling pathway. Mol. Cell. Biol. 9:4605–4612.

49. Polyak, S. J., K. S. A. Khabar, D. M. Paschal, H. J. Ezelle, G. Duverlie,G. N. Barber, D. E. Levy, N. Mukaida, and D. R. Gretch. 2001. HepatitisC virus nonstructural 5A protein induces interleukin-8, leading to partialinhibition of the interferon-induced antiviral response. J. Virol. 75:6095–6106.

50. Poole, E., B. He, R. A. Lamb, R. E. Randall, and S. Goodbourn. 2002. TheV proteins of simian virus 5 and other paramyxoviruses inhibit induction ofinterferon-beta. Virology 303:33–46.

51. Rochat, S., H. Komada, and D. Kolakofsky. 1992. Loss of V protein expres-sion in human parainfluenza virus type 1 is not a recent event. Virus Res.24:137–144.

52. Roux, P., C. Alfieri, M. Hrimech, E. A. Cohen, and J. E. Tanner. 2000.Activation of transcription factors NF-�B and NF-IL-6 by human immuno-deficiency virus type 1 protein R (Vpr) induces interleukin-8 expression.J. Virol. 74:4658–4665.

53. Sakaguchi, T., K. Kiyotani, M. Sakaki, Y. Fujii, and T. Yoshida. 1994. A fieldisolate of Sendai virus: its high virulence to mice and genetic divergence formprototype strains. Arch. Virol. 135:159–164.

54. Schafer, S. L., R. Lin, P. A. Moore, J. Hiscott, and P. M. Pitha. 1998.Regulation of type I interferon gene expression by interferon regulatoryfactor-3. J. Biol. Chem. 273:2714–2720.

55. Schlaak, J. F., C. M. Hilkens, A. P. Costa-Pereira, B. Strobl, F. Aberger,A. M. Frischauf, and I. M. Kerr. 2002. Cell-type and donor-specific tran-scriptional responses to interferon-alpha. Use of customized gene arrays.J. Biol. Chem. 277:49428–49437.

56. Skiadopoulos, M. H., S. R. Surman, J. M. Riggs, W. R. Elkins, M. St Claire,M. Nishio, D. Garcin, D. Kolakofsky, P. L. Collins, and B. R. Murphy. 2002.Sendai virus, a murine parainfluenza virus type 1, replicates to a level similarto human PIV1 in the upper and lower respiratory tract of African greenmonkeys and chimpanzees. Virology 297:153–160.

57. Stark, G. R., I. M. Kerr, B. R. Williams, R. H. Silverman, and R. D.Schreiber. 1998. How cells respond to interferons. Annu. Rev. Biochem.67:227–264.

58. Strieter, R. M., J. A. Belperio, and M. P. Keane. 2002. Cytokines in innatehost defense in the lung. J. Clin. Invest. 109:699–705.

59. Takeuchi, K., T. Komatsu, J. Yokoo, A. Kato, T. Shioda, Y. Nagai, and B.Gotoh. 2001. Sendai virus C protein physically associates with Stat1. GenesCells 6:545–557.

60. Taniguchi, T., K. Ogasawara, A. Takaoka, and N. Tanaka. 2001. IRF familyof transcription factors as regulators of host defense. Annu. Rev. Immunol.19:623–655.

61. Tapparel, C., S. Hausmann, T. Pelet, J. Curran, D. Kolakofsky, and L. Roux.1997. Inhibition of Sendai virus genome replication due to promoter-in-creased selectivity: a possible role for the accessory C proteins. J. Virol.71:9588–9599.

62. Wang, X.-L., M. Itoh, H. Hotta, and M. Homma. 1994. A protease activationmutant, MVCES1, as a safe and potent live vaccine derived from currentlyprevailing Sendai virus. J. Virol. 68:3369–3373.

63. Wang, X., M. Li, H. Zheng, T. Muster, P. Palese, A. A. Beg, and A. Garcia-Sastre. 2000. Influenza A virus NS1 protein prevents activation of NF-�Band induction of alpha/beta interferon. J. Virol. 74:11566–11573.

64. Wansley, E. K., and G. D. Parks. 2002. Naturally occurring substitutions inthe P/V gene convert the noncytopathic paramyxovirus simian virus 5 into avirus that induces alpha/beta interferon synthesis and cell death. J. Virol.76:10109–10121.

65. Wathelet, M. G., C. H. Lin, B. S. Parekh, L. V. Ronco, P. M. Howley, and T.Maniatis. 1998. Virus infection induces the assembly of coordinately acti-vated transcription factors on the IFN-beta enhancer in vivo. Mol. Cell1:507–518.

66. Yamada, Y., A. Nakamura, M. Hosoda, T. Kato, T. Asano, K. Tonegawa, andM. Itoh. 2001. Cytokines in pleural liquid for diagnosis of tuberculous pleu-risy. Respir. Med. 95:577–581.

67. Yeow, W. S., W. C. Au, W. J. Lowther, and P. M. Pitha. 2001. Downregula-tion of IRF-3 levels by ribozyme modulates the profile of IFNA subtypesexpressed in infected human cells. J. Virol. 75:3021–3027.

68. Yoneyama, M., W. Suhara, Y. Fukuhara, M. Fukuda, E. Nishida, and T.Fujita. 1998. Direct triggering of the type I interferon system by virus infec-tion: activation of a transcription factor complex containing IRF-3 and CBP/p300. EMBO J. 17:1087–1095.

69. Zhang, Y., B. A. Luxon, A. Casola, R. P. Garofalo, M. Jamaluddin, and A. R.Brasier. 2001. Expression of respiratory syncytial virus-induced chemokinegene networks in lower airway epithelial cells revealed by cDNA micro-arrays. J. Virol. 75:9044–9058.

VOL. 77, 2003 SENDAI VIRUS TARGETING OF INFLAMMATORY RESPONSES 7913

Page 132: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

6) 101–111www.elsevier.com/locate/yviro

Virology 351 (200

Sendai virus defective-interfering genomes and theactivation of interferon-beta

Laura Strahle, Dominique Garcin, Daniel Kolakofsky ⁎

Department of Microbiology and Molecular Medicine, University of Geneva School of Medicine, 11 Ave de Champel, CH1211 Geneva, Switzerland

Received 17 January 2006; returned to author for revision 23 February 2006; accepted 14 March 2006Available online 2 May 2006

Abstract

The ability of some Sendai virus stocks to strongly activate IFNβ has long been known to be associated with defective-interfering (DI)genomes. We have compared SeV stocks containing various copyback and internal deletion DI genomes (and those containing only nondefective(ND) genomes) for their ability to activate reporter genes driven by the IFNβ promoter. We found that this property was primarily due to thepresence of copyback DI genomes and correlated with their ability to self-anneal and form dsRNA. The level of IFNβ activation was found to beproportional to that of DI genome replication and to the ratio of DI to ND genomes during infection. Over-expression of the viral Vand C proteinswas as effective in blocking the copyback DI-induced activation of the IFNβ promoter as it was in reducing poly-I/C-induced activation, providingevidence that these DI infections activate IFNβ via dsRNA. Infection with an SeV stock that is highly contaminated with copyback DI genomes isthus a very particular way of potently activating IFNβ, presumably by providing plentiful dsRNA under conditions of reduced expression of viralproducts which block the host antiviral response.© 2006 Elsevier Inc. All rights reserved.

Keywords: Sendai virus; Defective-Interfering genome; Interferon beta; Innate immunity

Our understanding of how animal cells recognize and mountan innate antiviral response to intracellular RNA virusreplication has recently made great progress. Two DexD/Hbox helicases with CARD domains, RIG-I and Mda5/Helicard,were found to participate in the detection of cytoplasmicdsRNA (Andrejeva et al., 2004; Yoneyama et al., 2004). dsRNAis thought to be a common product of RNAvirus infections thatacts as a pathogen-associated molecular pattern (PAMP)responsible for initiating the innate antiviral response. RIG-Iand Mda5 initiate antiviral responses by coordinately activatingseveral transcription factors, including NF-κB and IRF-3, thatbind to the IFNβ promoter forming an enhanceosome thatactivates this primary host-response gene (McWhirter et al.,2005). Upon binding dsRNA, the CARD domains of thesehelicases are thought to be freed for interaction with thesynonymous domain of MAVS/VISA/cardif/IPS-1, a mitochon-drial protein which in turn is required for recruiting the kinasesthat activate these transcription factors (Xu et al., 2005; Meylan

⁎ Corresponding author.E-mail address: [email protected] (D. Kolakofsky).

0042-6822/$ - see front matter © 2006 Elsevier Inc. All rights reserved.doi:10.1016/j.virol.2006.03.022

et al., 2005; Seth et al., 2005; Kawai et al., 2005). Autocrineinteraction of secreted IFNβ with its cell surface receptor thencloses the “innate immunity loop”, leading to increased IFN-stimulated gene (ISG) products, such as other IFNs and theintracellular effectors of the antiviral state (Sen, 2001).

The role of Mda5 in dsRNA signaling to IFNβ wasuncovered because Mda5 binds to the paramyxovirus SV5 Vprotein (Andrejeva et al., 2004). This interaction, and that ofother paramyxovirus V proteins, blocks dsRNA signaling, andthis property of V maps to the highly conserved cys-rich domainat their C-termini (Poole et al., 2002; Andrejeva et al., 2004). Vproteins of different paramyxoviruses, however, are verydifferent at their N-terminal portions, which accounts for theirotherwise very different properties (Lamb and Kolakofsky,2001). Rubulavirus V proteins (e.g., SV5, PIV2) are associatedwith intracellular and virion nucleocapsids (NCs) and areimportant in promoting virus growth. PIV2 which cannotexpress the entire V protein is highly debilitated even in the mostpermissive cell culture (Nishio et al., 2005). Respirovirus (e.g.,Sendai virus (SeV)) V proteins, in contrast, are nonstructuralproteins, are not associated with NCs and their expression

Page 133: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

102 L. Strahle et al. / Virology 351 (2006) 101–111

inhibits genome replication (Curran et al., 1991). Moreover,although V expression is not required for replication in cellculture, SeVVexpression is essential for virulence in mice (Katoet al., 1997; Delenda et al., 1998). This requirement ispresumably due to its ability to block dsRNA-induced activationof the IFNβ promoter (Poole et al., 2002) as infection with SeVthat cannot specifically express the V protein strongly activatesIFNβ (Strahle et al., 2003). Finally, in contrast to rubulaviruses,respirovirus VmRNAs express a nested set of C proteins from anoverlapping ORF, and these C proteins also block the innateantiviral response, in several ways (e.g., by blocking IFNsignaling) (Gotoh et al., 1999; Garcin et al., 1999).

It is noteworthy that infection with wild-type SeV or SV5does not normally activate IFNβ. In contrast, infections withSV5 VΔC (which produces a C-terminally truncated V protein)or infections with SeV with mutations in either the leaderregion, two regions of the C protein or the V protein all lead toenhanced levels of IFNβ mRNA (He et al., 2002; Strahle et al.,2003). In the case of SeV, these viral genes also appear to beinvolved in preventing activation of inflammatory cytokinessuch as IL-8. For SeV, all the viral products that are not knownto play essential roles in the replication process itself (the VandC proteins, and leader and trailer RNAs) appear to function incountering the innate antiviral response. In the context of anormal, wild-type infection of cells in culture, the effect of thesevarious viral anti-host-response products is apparently sufficientto prevent, or severely limit, IFNβ activation.

The IFNβ promoter is normally activated in cells treatedwith dsRNA (poly I:poly C, or poly-I/C) or infected with virus.Sendai virus infection is often used in this respect, and virusstocks which strongly activate IFNβ are also availablecommercially. However, it has long been known that theremarkable ability of some SeV stocks to induce IFN secretionin macrophage and other cell lines is related to the presence ofdefective-interfering (DI) particles (Johnston, 1981; Poole et al.,2002), but the nature of the IFN-inducing agent in these stockswas not examined. Moreover, for VSV, another mononegavirus,DI particles containing “snap-back” DI genomes (see below)were found to be very potent inducers of IFN, even in theabsence of co-infecting nondefective (ND) helper virus (Marcusand Sekellick, 1977; Sekellick and Marcus, 1982). DI particlescontain deleted viral genomes which are generated spontane-ously as by-products of ND genome replication. DI genomeshave, sine qua non, gained the ability to successfully competewith their helper ND genomes for the viral replication substratesprovided by the latter; hence, they are also “interfering”(Perrault, 1981; Lazzarini et al., 1981). Because of theirreplicative advantage over ND genomes, DI genomes invariablyaccumulate in SeV stocks that are repeatedly passed in eggs,unless steps to prevent this accumulation are taken.

Nonsegmented negative-strand RNA viruses (NNV) DIgenomes can be of two types, internal deletion or copyback(Fig. 1). The replicative advantage of internal deletion DIgenomes over ND genomes is not well understood (Garcin etal., 1994), but that of copyback DI genomes is well studied.Copyback DI genomes have always replaced the weakergenomic replication promoter at the 3′ ends of their minus

strands with the stronger antigenomic promoter, and thus bothDI genomes and antigenomes initiate from the same strongantigenomic promoter. Paramyxovirus replication promoters arecontained within the 3′ terminal 91–96 nt of the genomic RNA(narrow boxes, Fig. 1), and all natural copyback DI genomeshave copied back 91 nt or more while carrying out this promoterexchange (see Fig. 1). Although these RNAs contain terminithat are perfectly complementary for ca. 100 nt, DI genomes arenormally present within nucleocapsids (NCs), where their RNAends are not free to anneal. However, when SDS is used togently dissociate the N protein from RNA, copyback DIgenomes (and antigenomes) rapidly form ssRNA circles withdsRNA panhandles, as seen in the EM and on biochemicalanalysis (Kolakofsky, 1976). Several copyback DI genomesfrom independent virus stocks were characterized initially. Theyall contained complementary termini of ca. 100 nt in length, andas expected, the rate at which they circularized on SDStreatment was inversely proportional to their length.

Similar copyback DI genomes with limited terminalcomplementarity are common for VSV. However, VSV, unlikeSeV, also generates an extreme form of copyback DI genomewhose sequences are complementary over their entire length ofca. 2 kb (snapback DIs) and which form long dsRNA “hairpins”(of ca. 1000 bp) rather than ss circles upon SDS treatment(Lazzarini et al., 1981; Perrault, 1981). It is these VSV snapbackDI genomes, like DI 011, that were reported to strongly induceIFN by themselves, in aged chick embryo fibroblasts (CEFs)and mouse L cells (Marcus and Gaccione, 1989; Marcus andSekellick, 1977). These reports have remained controversial,however, because this IFN induction was independent of co-infecting helper virus, whereas Youngner and colleagues foundthat it correlated with contaminating ND virus in L cells. Theselatter workers, moreover, were unable to find a correlationbetween the snapback content of their DIs and IFN induction(Frey et al., 1979). Sekellick and Marcus also reported thatsnapback DI induction of IFN was unaffected by heat treatmentthat would inactivate its RNA polymerase, or UV treatment thatwould prevent its genome from being copied, and concludedthat this IFN induction was due to a pre-existing molecule thatdid not require any synthetic events for its formation (Sekellickand Marcus, 1982; Marcus and Gaccione, 1989). Disassemblyof DI 011 NCs intracellularly would then appear to be the onlyexplanation for dsRNA formation. However, as the infection ofa single DI 011 particle per CEF was sufficient to induce aquantum (maximum) yield of IFN, the manner in which the DI011 NC is presumably disassembled so efficiently in vivo (topermit dsRNA formation) remains an enigma as NCs aregenerally very stable in vivo.

Given the growing appreciation that dsRNAmay be a commonproduct of RNAvirus infection that participates in the induction ofthe innate antiviral response, we have reexamined the requirementof SeV DI genomes for the activation of the IFNβ promoter.

Results

Three SeV stocks containing DI genomes (Figs. 1, 2a), aswell as a stock containing only ND genomes, were examined for

Page 134: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

Fig. 1. A schematic view of SeV RNA synthesis and DI genomes. The ND (−) genome is shown above, with its 3′ end genomic replication promoter (on the left) andthe complement of the antigenomic replication promoter (at the 5′ end), both contained within the terminal 96 nt, shown as thin boxes. The central protein codingregion, from the beginning of the N ORF to the end of the L ORF, is shown as thicker boxes; these regions are not drawn to scale. Only the N/P junction is shown, theremaining genes are lumped together as a P/V/C … L. The (+) RNAs transcribed from the ND genome (le RNA and the 6 mRNAs) are shown below the ND genome.The sole transcript of the ND-antigenome, tr RNA, is shown above. The lines below the ND genome indicate the progress of the replicase when generating internaldeletion (E307) or copyback (H4/ChR1) DI genomes. The structure of the DI (−) genomes is shown below; the dark shading on the left indicates the extent of theterminal complementarity, which is also indicated in brackets on the left. The inverted triangle indicates the yfp sequence used to tag H4. The arrows below the DIgenomes show the PCR amplification strategy used to clone the DI RNA of the Charles River SeV stock.

103L. Strahle et al. / Virology 351 (2006) 101–111

their ability to activate a luciferase reporter gene driven by theIFNβ promoter. The DI-H4 stock is composed predominantlyof a natural 1410 nt long copyback DI genome, whose terminiare perfectly complementary for 110 nt (Calain et al., 1992).The DI-S104 stock, like H4, was generated by passage in eggs,but this stock is composed of 5 major DI species. Only thesmallest DI of this stock has been cloned as DNA and found tobe an internal deletion DI of 1794 nt (called E307) (Engelhornet al., 1993). Their termini are complementary for only 12 nt,like those of ND genomes. However, Northern analysis with aleader/N gene probe, which anneals specifically to internaldeletion DI genomes, shows that this stock contains 3 internaldeletion as well as 2 copyback DI genomes (the latter of ca.2500 and 3500 nt in length) (Fig. 2a). DI-GP55, in contrast tothe others, was produced and rescued by recombinant means(Garcin et al., 1995) and is identical to E307 except that itcontains the 55 nt trailer region at the (−) DI genome 3′ end, inplace of the leader region. DI-GP55 is thus a copyback DIgenome whose termini are complementary for 55 nt (Fig. 1). Asmentioned above, copyback DI genomes with more extensiveterminal complementarity, such VSV DI 011, have not beenreported for SeV, and we have no stable SeV stocks that containonly internal deletion DI genomes.

Infection of 293T cells with all 3 DI stocks was found toactivate the IFNβ reporter, but to very different extents (Fig. 2c).

H4 was the most potent, followed by GP55, and S104 was theleast potent. In contrast, infection with ND genomes alone barelyactivated the reporter gene, even though the ND infectionaccumulated as much or more viral proteins than the DIinfections (Fig. 2b). In these and other experiments, there is oftenan inverse correlation between the accumulation of viral proteinsand the extent of IFNβ activation, as might be expected ifactivation is due to the presence of DI genomes. Moreimportantly, plaque purification of the H4 stock yields a viruspreparation that does not contain DI genomes and which doesnot activate the IFNβ promoter during infection (ND-H4, Fig.3). The ability of the DI-H4 stock to activate IFNβ is thus not dueto mutations within the ND genome, which could have arisenbecause of the presence of the DI genomes.

IFNβ activation requires modification of IRF-3, which ishyper-phosphorylated in response to viral infection, or dsRNA(Fitzgerald et al., 2003; Sharma et al., 2003). Activated IRF-3dimerizes andmigrates to the nucleus where it binds to the PRD Iand III elements of the IFNβ promoter, as part of anenhanceosome (Maniatis et al., 1998). The activation of theIFNβ promoter by SeV DI infection appears to require IRF-3since this activation is largely ablated by co-expression of adominant-negative form of IRF-3 (data not shown). When theextent of IRF-3 dimerization was examined, a significantfraction of the IRF-3 was found as dimers (on non-denaturing

Page 135: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

Fig. 2. Only DI infections activate IFNβ in 293T cells. (a) Northern blot of encapsidated (CsCl banded) RNAs from the various infections as indicated, using a (+)riboprobe from the end of the L gene (which detects all DI genomes) or a (+) riboprobe from the leader/N gene region, which detects only internal deletion DI genomes.The DI-H4 and GP-55 genomes do not anneal with the leader/N gene probe. (b) A Western blot of equal amounts of cell extracts (total proteins) of the variousinfections, using anti-P and anti-N antibodies. (c) 293T cells were transfected with a luciferase reporter plasmid under the control of the IFNβ promoter (and controlplasmid) for 24 h then infected (or not) with the various SeV stocks for 20 h. Cell extracts were prepared, and levels of the renilla and firefly luciferases weredetermined. The cumulative results of 3 experiments are shown. (d) Two different amounts (1× and 5×) of total cell proteins from extracts of the various infections wereseparated on nondenaturing gels andWestern blotted with anti-IRF-3. (e) Equal amounts of cell extracts of the various infections (total proteins) were separated on SDSgels andWestern blotted with anti-IRF-3. The right-hand lane shows uninfected cells that express transfected IRF-3 5D. The 5 isoforms, numbered 1 to 5, are indicated.

104 L. Strahle et al. / Virology 351 (2006) 101–111

gels) in extracts of all 3 DI infections, and the extent of dimerformation was roughly in proportion to the degree of IFNβactivation (Fig. 2d). In contrast, there was no evidence of dimersin the ND extract.We also examined the electrophoretic mobilityof IRF-3 on SDS-PAGE as this mobility is sensitive to IRF-3phosphorylation status (Hiscott et al., 2003; Hasegawa et al.,1992; Yoneyama et al., 2002). We could distinguish 5electrophoretic forms of IRF-3 in our extracts (Fig. 2e). Mock-infected extracts predominantly contained forms 2 and 3.Infection with ND SeV led to the strong appearance of fasterform 5, without loss of 2 and 3. In contrast, infection with DI-H4and GP55 led to the appearance of the slightly slower form 4,reduction in the intensity of forms 2 and 3, and the appearance ofthe slowest form 1, which co-migrates with phospho-mimeticIRF-3 5D that is constitutively active (Lin et al., 1999). IRF-3 is

thus being modified in response to the ND infection, but thesemodifications do not lead to dimerization or the activation of theIFNβ promoter. The presence of DI genomes during infectionsomehow causes IRF-3 to be modified differently, leading to itsdimerization and IFNβ promoter activation.

DI-H4-induced activation of IFNβ is proportional to DIgenome replication

The relative amounts of DI and ND genomes in cell extractscan be examined by Northern blotting (Figs. 2 and 3), but thisanalysis is linear only over a relatively narrow range and oftenunequally estimates RNAs that vary significantly in length(>10-fold in this case). RT/PCR is better suited to this task, butthis method cannot differentiate between DI-H4 and ND

Page 136: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

Fig. 4. Evolution of the DI-H4+yfp stock on passage in eggs. (a) The DI-H4+yfp genome was prepared from DNA by reverse genetics (Methods andmaterials) and rescued by superinfecting the BSR T7 culture with ND SeV(Methods and materials). The culture supernatant was inoculated into eggs, andthe resulting allantoic fluid was diluted 1 to 500 for reinoculation, for a total of 20times. The stocks from each passage level were assayed for their ability toactivate a GFP gene under the control of the IFNβ promoter. Only the resultsfrom passage levels 8 and 16 to 20 are shown, along with DI-H4 as positivecontrol and mock-infected as a negative control. (b) Total RNA was preparedfrom cells infected with passage levels 8 and 16 to 20 and examined by RT/PCRfor their levels of ND genome (N gene) and DI genome (YFP) relative to that ofGADPH mRNAs. Total RNA from a mock-infected culture served as thenegative control. (c) Equal amounts of extracts from cells infected with passagelevels 8 and 16 to 19 were examined by Western blotting with anti-N and anti-P/V/C and anti-actin as a loading control.

Fig. 3. Allantoic fluid stocks prepared from plaque-purified SeV do not activateIFNβ. The DI-H4 and ChR stocks were titered on LLC-MK2 cells in theabsence of serum. Single plaques from the highest positive dilution (10−7) werepicked, and 3/10 of each plaque was inoculated into hen's eggs that wereincubated at 33 °C for 3 days. The allantoic fluid stocks resulting from theplaque-purified virus (ca. 109 pfu/ml) were compared with the original stockscontaining the same amount of viral proteins (as determined by Coomassie bluestaining of SDS gels) for their ability to activate pIFNβ-GFP in 293T cells. ANorthern blot of encapsidated (CsCl banded) RNAs from equal amounts of ND-,H4-, and ChR1-infected cells, using a (+) riboprobe from the end of the L gene,is shown in the insert.

105L. Strahle et al. / Virology 351 (2006) 101–111

genomes unless the complementary termini are included in theamplification. However, the dsRNA panhandles of circular DIgenome RNAs are exceedingly stable and cannot be invaded byprimers extended by RTase even at elevated temperatures. Tocircumvent these technical problems, we prepared a taggedversion of the DI-H4 genome containing a 162 nt insertionfrom the YFP gene (Fig. 1), such that the DI and ND genomescould be independently and accurately estimated by RT/PCR,over a large range of values. A second reason for preparing DI-H4+yfp is that, although our H4 stocks are composedpredominantly of the 1410 nt long species cloned as DNA,overexposure of Northern blots shows that several other RNAsare present in much lower amounts and which have nototherwise been characterized. As we do not know whether allthe DI genomes in the H4 stock contribute equally to inducingIFNβ activation or whether activation is due to a particular (andperhaps uncharacterized) species, we cannot be sure that IFNβactivation is in fact due to the 1410 nt copyback DI genome.Recapitulation of these results with a tagged copy of the DI-H4genome would settle this issue as well.

DI-H4+yfp genomes were recovered from DNA in BSR T7cells that were subsequently co-infected with ND SeV. Stockscontaining this DI genome were then generated by multiplepassages in embyonated chicken eggs (Methods and materials),and each passage was tested for its ability to activate a GFPreporter gene under the control of the IFNβ promoter (pIFNβ-GFP) upon infection of 293T cells. Although some IFNβactivation above background appeared by passage 3, thisactivation increased slowly and erratically at first and eventuallyreached activation levels approximately half those of thereference H4 stock by passage 20 (Fig. 4a), During the laterpassages (16 to 20), the level of intracellular ND genomessteadily decreased (Fig. 4b), leading to a reduction in the levelsof viral proteins (Fig. 4c), whereas the levels of DI-H4+yfp

genomes steadily increased (Fig. 4b). The ability of SeV DIstocks to induce IFNβ activation thus correlates with therelative levels of DI genomes during infection. As DI and NDgenome NCs are relatively stable structures, these levels reflectthe rates that DI and ND genomes are synthesized duringinfection with the various passage levels. The evolution of theDI-H4+yfp stock towards IFNβ activation (during relativelyundiluted passage in eggs) thus correlates with the level of DIgenome synthesis during infection. The remarkable ability of

Page 137: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

106 L. Strahle et al. / Virology 351 (2006) 101–111

the DI-H4 stock to activate IFNβ can now also be traced to thepredominant 1410-nt-long copyback DI genome.

Examination of the UV inactivation kinetics of DI-inducedIFNβ activation, as compared to the UV inactivation kinetics ofND and DI genome replication, can provide broad informationon the nature of this IFNβ activation. DI-H4+yfp (P(passage)17, insert, Fig. 5) was irradiated with 256 nm UV light forvarious times (0.5 to 8 min) and used to infect 293T cellscontaining pIFNβ-GFP. Intracellular RNA was isolated at20 hpi, and the levels of ND genome RNA and DI-H4+yfpgenome RNAwere measured by RT/PCR. GFP expression wasmonitored by FACS (Fig. 5a). DI-H4+yfp (1572 nt) is 1/10 thelength of the ND genome (15,384 nt) and thus proportionatelyless sensitive to UV inactivation. This difference in the loss ofND and DI genome levels is most apparent at the shortest timesof UV irradiation and is lost at the higher doses, presumablybecause DI genome replication ultimately depends on NDgenomes to provide all the replication substrates (N, P and Lproteins). The reduction of GFP expression levels upon

Fig. 5. UV inactivation kinetics of DI-H4+yfp infections. (a) The allantoic fluidof DI-H4+yfp passage level 17 was irradiated with 254 nm light (110 μW/cm2)from 0.5 to 8min and then used to infect 293Tcells that had been transfected withpIFNβ-GFP. Total RNA was prepared at 24 hpi, and the levels of DI-H4+yfpgenome and ND genome were determined by RT/PCR. GFP expression levelswere monitored by FACS. The results are shown relative to RNA from cellsinfected with non-irradiated allantoic fluid. The cumulative results of 2experiments are shown. The error bars, always <20%, are not shown for clarity.(b) A Northern blot of encapsidated RNAs fromND, DI-H4 and DI-H4+yfp P17infections, using a (+) probe from the end of the L gene.

increasing UV irradiation parallels that of ND and DI genomesand most closely follows the loss of the DI genomes at thelowest doses of UV. More importantly, as the reduction of GFPexpression levels closely follows that of DI genomes over arange of 2 logs, IFNβ activation is clearly proportional to thelevel of copyback DI genome replication (in 293T cells) for agiven stock as well.

The manner in which SeV DI genomes presumably generatedsRNA that induces IFN is thus quite different from that ofVSV DI 011. SeV DI genomes not only require co-infectionwith ND helper virus, IFN induction here (293T cells) is strictlyproportional to the level of DI genome replication, in contrast toVSV snapback DI IFN induction (in aged CEFs) where viralRNA synthesis is not required.

The SeV stock of Charles Rivers Laboratory

As mentioned above, SeV stocks (Cantell strain) whoseinfection of cultured cells strongly activates IFNβ, are availablefrom Charles River Laboratory. A fresh allantoic fluid stock ofthis virus preparation was found to activate IFNβ to levelssimilar to those of DI-H4 (Fig. 3), and this stock was found tocontain a very small DI genome (of ∼600 nt) by Northernanalysis (insert, Fig. 3). When the ND virus of this preparationwas plaque purified on LLC-MK2 cells, allantoic fluid stocksprepared from the purified virus had lost the ability to activateIFNβ (ND-ChR1, Fig. 3). Thus, similar to DI-H4, the ability ofthe Charles River virus preparation to activate IFNβ appears tobe due to the presence of the DI genome(s), and not to mutationswithin its ND genome.

To determine the nature of this DI genome, we cloned theDI genome as DNA, as illustrated in Fig. 1 (small horizontalarrows are primers). We used one set of primers to amplifythe common right end of all DIs (arrows under GP55, Fig. 1)and 3 sets of primers that were specific to the left end ofeither internal deletion (arrows under E307, Fig. 1) orcopyback DIs (arrows under H4, Fig. 1). The common rightend primer set and the 3 copyback-specific left-end primersets all yielded a PCR product of the expected size, whereasthe 3 internal-deletion-specific left-end primers failed toproduce visible DNA (not shown). When these amplifiedDNA fragments were sequenced, DI-ChR1 was deduced to bea simple copyback DI genome of 546 nt (453 nt are co-linearwith the 5′ end of the ND (−) genome), with terminalcomplementarity over 93 nt. To our knowledge, this is thesmallest natural SeV DI genome described to date, and thisproperty may be related to its ability to activate IFNβ sostrongly.

The SeV V and/or C proteins inhibit DI-H4-induced IFNβactivation

Although our 293T cells produce IFN in response to DI-H4 infection, they do not respond to added IFN. 293T cellsare thus useful in studying IFNβ activation in isolationbecause the activation is not also driven by positive feedbackvia ISGs. However, to study the broader aspects of the

Page 138: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

107L. Strahle et al. / Virology 351 (2006) 101–111

DI-H4 infection, we used 2fTGH cells, which both produceIFN in response to infection and respond as well to thesecreted IFN. When 2fTGH cells containing a pIFNβ-lucreporter are treated with poly-I/C (either added to themedium or via transfection (*)) or IFNα, poly-I/C* and H4infection strongly activate IFNβ promoter, whereas IFNα hasno effect and poly-I/C treatment has little effect (IFNβ is notan ISG, and TLR3 may be poorly expressed in thesefibroblasts). Moreover, whereas poly-I/C* and IFNα clearlyincreased ISG levels (STAT1, RIG-I, ISG15 and ISG56), H4infection failed to increase these ISG levels above theuntreated control. The SeV V protein blocks poly-I/C-induced IFNβ activation (Poole et al., 2002) and presumablyshould also block that induced by DI-H4 infection. Wereasoned that, if DI-H4 infections generated abnormally largeamounts of dsRNA, the amount of V expressed from the NDgenome during DI infections might be insufficient to blockdsRNA signaling via RIG-I and Mda5. We therefore over-expressed the V and C proteins by plasmid transfection in2fTGH cells containing pIFNβ-luc and re-examined theeffects of the various treatments. As shown in Fig. 6a, over-expression of the V and C proteins was as effective inblocking the DI-H4-induced activation of the IFNβ promoteras it was in reducing the poly-I/C*-induced activation. Thisresult is consistent with the notion that DI-H4 infectioninduces IFNβ activation, at least in part, via dsRNA. Over-expression of the V and C proteins also partially blocked thepoly-I/C*- and IFNα-induced increase in ISG levels,including that of RIG-I.

To examine whether DI-H4 infection was indeed able toblock IFN signaling, we similarly treated 2fTGH cellscontaining a pIRSE-luc reporter and examined the effects onthe reporter gene and ISG protein levels (Fig. 6b). In contrastto IFNα and poly-I/C* treatment which activated thispromoter and increased ISG levels, DI-H4 infection did notactivate the ISRE reporter over the untreated control, and thelevels of STAT1, RIG-I, ISG15 and ISG56 were not increased

Fig. 6. (a) The effect of poly I/C, DI-H4 infection and IFNα treatment on IFNβand ISGs in 2fTGH cells, in the presence and absence of the SeV V and Cproteins. 2fTGH cells were transfected with a luciferase reporter plasmid underthe control of the IFNβ promoter (and the TK-renilla control plasmid) and pSeV-V/C that expresses V and C proteins from the same mRNA (or and emptyplasmid) for 24 h. Parallel cultures were then either treated with 100 μg/ml ofpoly I/C in MEM (poly-I/C), transfected with 1 μg of poly I/C (poly I/C*),infected with DI-H4 or treated with 1000 IU of IFNα. Cell extracts wereprepared 20 h later, and the levels of the renilla and firefly luciferase activitieswere determined. The cumulative results of 2 experiments are shown. Equalamounts of total protein of each extract were also examined for their levels ofSTAT1 (and actin), RIG-I, SeV P, V and C proteins and ISG-15 and ISG-56 byWestern blotting with specific antibodies (Methods and materials). (b) The effectof poly I/C, DI-H4 infection and IFNα treatment on ISGs in 2fTGH cells.2fTGH cells were transfected with a luciferase reporter plasmid under thecontrol of an ISRE promoter (and the TK-renilla control plasmid) for 24 h.Parallel cultures were then treated as in panel A. Cell extracts were prepared 20 hlater, and the levels of the renilla and firefly luciferase activities weredetermined. The cumulative results of 2 experiments are shown. Equal amountsof total protein of each extract were also examined for their levels of STAT1 (andactin), RIG-I and ISG-15 and ISG-56 by Western blotting.

in these extracts. Thus, there appears to be sufficient viralproteins expressed during DI-H4 infection to block IFNsignaling.

Page 139: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

108 L. Strahle et al. / Virology 351 (2006) 101–111

Discussion

dsRNA is thought to be a common product or PAMP of RNAvirus infections that initiates the innate antiviral response, in partby activating IFNβ. However, the source of this dsRNA ispresumably different for different viruses. (+) RNA virusgenomes contain highly conserved 2° and 3° structures at their5′ and 3′ ends that are essential for virus replication (Simmondset al., 2004), and these highly structured RNA regions bythemselves can initiate signaling to IFNβ upon binding to RIG-I(Sumpter et al., 2005). In contrast, NNV genomes are not knownto contain conserved 2° structures, and, moreover, NNVgenomes function in RNA synthesis not as free RNAs but asassembled NCs, in which the genome RNA cannot normallyanneal (Lamb and Kolakofsky, 2001). One possible source ofdsRNA during NDSeVreplication is the occasional extension ofthe trailer RNA beyond the trailer/L gene junction (see Fig. 1),producing run-on trailer RNAswhose 3′ sequences can anneal tothose of the L mRNA (Vidal and Kolakofsky, 1989). In a similarvein, the transcriptase which synthesizes the L mRNApresumably reads through the L gene-end site at a frequencyof ca. 5% (similar to other gene junctions; Le Mercier et al.,2002) and terminates at the genome 5′ end, thus providing read-through L transcripts that can anneal to trailer RNAs. Althoughthe NNV replication strategy appears to minimize dsRNApotential during intracellular replication, this strategy presum-ably cannot exclude the generation of small amounts of dsRNA.It is reasonable that the levels of V and C expressed during NDgenome replication are designed to counteract the small amountsof dsRNA generated. If so, the presence of significant amountsof copyback DI genomes during intracellular replication willcertainly change the nature of the SeV infection.

The most important new finding of this study is that not allSeV stocks that are heavily contaminated by DI genomes areequally able to activate IFNβ. The H4, GP55 and S104 stocks allcontain the H strain ND genome as helper and can be directlycompared. S104 infections accumulate more viral products thanH4 infections as the S104 DI genomes appear to interfere lesswith their helper virus replication than those of H4 (Fig. 2b). Atthe same time, S104 infections activated IFNβ 10 to 20-fold lessstrongly than H4 infections in multiple experiments (Fig. 2a anddata not shown). Besides the different extents to which S104 andH4 DI genomes interfere with ND genome expression and thusaffect the intracellular concentration of the viral VandC proteins,H4 stocks are composed exclusively of copyback DI genomes,whereas S104 stocks are composed predominantly (>70%) ofinternal deletion DI genomes. Moreover, the two copyback DIgenomes in this stock are both longer than DI-H4. DI-GP55 isalso longer than DI-H4, its termini are complementary for onlyhalf the length as H4 (55 nt), and DI-GP55 interferes with NDgenome expression less than DI-H4. Taken together, these datasuggest that the ability of SeV DI stocks to activate IFNβ isrelated both to (i) their ability to interfere with helper genomeexpression, which leads to lower levels of Vand C intracellularly,and (ii) their relative content of copyback DI genomes. The sizeof the DI genome may also play a role in this latter respect.However, it will be necessary to examine these properties of SeV

DI genomes more directly, e.g., by extending the terminalcomplementarity of DI-GP55 to 110 nt, or by altering the lengthof DI-H4+yfp, to be more certain of these conclusions.

Copyback DI genomes may be stronger activators of IFNβthan internal deletion DI genomes because they have a strongerpotential to form dsRNA. Copyback DI genomes are composedof equal amounts of genomes and antigenomes (rather than a10-fold excess of genomes), and their termini can self-annealintramolecularly as well. The question then remains of how thisdsRNA potential is expressed as the synthesis of DI genomes,like that of ND genomes, is thought to be coupled to theirassembly into NCs. The vast majority of these NCs, onceformed, are very stable in vivo and band in CsCl densitygradients as fully assembled NCs. However, it is not rare to findsmall amounts of DI-H4 genomes and antigenomes in extractsof infected cells which pellet through these gradients as freeRNAs (<5%, unpublished). It is not clear whether these non-assembled DI genomes were normally made as NCs whichsubsequently disassembled or were actually made de novowithout concurrent assembly with N protein, as reported forsome conditions of VSV DI genome replication in vitro(Chanda et al., 1980). Independent of how non-assembled DI-H4 genomes are presumably generated in vivo, their formationappears to be proportional to their synthesis (Fig. 4). Theirpresence would represent a vast increase in the dsRNA potentialof SeV DI vs. ND infections, under conditions where there isless V protein available to dampen dsRNA signaling to IFNβ(Fig. 4c). The relatively short lengths of the H4 and ChR1 DIgenomes may play a role in how frequently their non-assembledRNAs are formed in vivo, but this needs to be investigated.

There is one further aspect of SeV copyback DI infections thatshould be mentioned to explain their ability to induce IFN soefficiently, coupled with the remarkable fact that these DIgenomes with dsRNA potential are actually selected for onpassage in eggs (e.g., Fig. 4) (Le Mercier et al., 2002). We havepreviously prepared ambisense ND SeV in which an additionalmRNA was expressed from the 3′ end of the antigenome. Incontrast to copyback DI genomes, genomes and antigenomes ofthe ambisense SeV contain the weaker genomic promoter. Theseambisense SeV grew poorly in IFN-sensitive cultures and wererelatively IFN-sensitive. They were also highly unstable onpassage in eggs and reverted to virus that grows well even in IFN-pretreated cells that restrict vesicular stomatitis virus replication,i.e., the wild-type SeV phenotype. Since this reversion wasalways associated with a point mutation in the ambi-mRNA startsite that severely limited its expression, we concluded that theselection ofmutants unable to express ambi-mRNAon passage inchicken eggs was presumably due to increased levels of dsRNAduring infection (vRdRp read-through of the ambi-mRNA stopsite creates a capped transcript that can potentially extend theentire length of the antigenome, whereas extension of theuncapped trailer RNA (wt SeV) is limited by the poor processivityof its vRdRp). If ND ambisense SeV with dsRNA potential arestrongly selected against in eggs, then how are DI genomes withdsRNA potential positively selected under the same conditions?

There are two possible explanations for this conundrum.Firstly, the dsRNA potential of ambisense SeV is not associated

Page 140: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

109L. Strahle et al. / Virology 351 (2006) 101–111

with any selective advantage and a single point mutation in theambi-mRNA promoter will largely eliminate this potential. TheND genomes of SeV stocks containing copyback DI genomes,in contrast, cannot escape their DI genomes by simple mutation,and the dsRNA potential of copyback DI genomes is alwaysassociated with a strong selective advantage as copyback DIgenomes outcompete their ND genomes for the replicationsubstrates provided by the latter. Secondly, and perhaps moreimportantly, copyback DI infections are relatively non-cyto-pathic and often end as persistent infections (Roux et al., 1991).This is in part due to the ectopic expression of trailer RNA (inplace of leader RNA) from the copyback DI (−) genome (Garcinet al., 1998) (Fig. 1). Trailer RNA is known to interact withTIAR, a protein with many links to apoptosis, and thisinteraction is important in suppressing SeV-induced PCD(Iseni et al., 2002). The relative absence of leader RNAs duringcopyback DI infections may also contribute to this DIphenotype as mutations in the SeV leader region are associatedwith virulence in mice (Fujii et al., 2002), and the normalexpression of leader RNA appears to be required to preventIFNβ activation (Strahle et al., 2003). The ability of copybackDI infections to delay, and in many cases completely preventPCD, may compensate for the negative consequences ofincreased dsRNA during infection, which presumably selectsagainst SeV that express ambi-mRNAs.

In summary, infection with an SeV stock that is highlycontaminated with copyback DI genomes is a potent way ofactivating IFNβ. These DI infections presumably provideplentiful dsRNA, under conditions of reduced expression ofviral products which block the host response to dsRNA, andwith minimal cytopathic effects that lead to persistent infection.In contrast, infection with an SeV stock that is not contaminatedwith copyback DI genomes does not activate IFNβ and ishighly cytopathic. These are two very different virus infections,and they should not be confused when SeV stocks of unknowncomposition are used to activate IFNβ.

Methods and materials

Cells, viruses, and antibodies

BSR-T7 cells were grown in BHK-21 Medium (GlasgowMEM, Gibco) supplemented with 5% fetal calf serum (FCS) inthe presence of the relevant maintenance drug (G418 at 400 μg/ml). 2fTGH cells and 293T cells were grown in Dulbecco'smodified Eagle's medium supplemented with 10% fetal calfserum (FCS).

SeV stocks were grown in the allantoic cavities of 9-day-oldembryonated chicken eggs for 3 days at 33 °C. For ND stocks(109 pfu/ml), 0.1 ml of a 105 dilution (ca. 1000 pfu) wasinoculated per egg. In the case of DI stocks, 0.1 ml of a 102 to104 dilution was used. In all cases, the amount of viral proteinspresent in the resulting allantoic fluid was analyzed by sodiumdodecyl sulfate (SDS)-polyacrylamide gel electrophoresis andCoomassie blue staining of pelleted virus. Virus titers weredetermined by plaquing on LLC-MK2 cells. Anti-IRF-3 (SantaCruz), anti-actin (Chemicon), anti-Stat1 (C-terminus) (Trans-

duction Laboratories), anti-N-877 and anti-PCV, anti-ISG-15and anti-ISG-56 were provided by Dr. Ganes Sen from TheCleveland Clinic Institute. Anti-Rig-I was provided by TadaatsuImaizumi from Hirosaki University School of Medicine.

Plasmids, transient transfections, luciferase assay and FACS

pβ-IFN-fl-lucter, which contains the firefly luciferase geneunder the control of the human IFNβ promoter, is described inKing and Goodbourn (1994). The IFNα/β-responsive reporterplasmid, p(9–27)4tkD(239)lucter, referred to here as pISRE-fl-lucter, contains four tandem repeats of the IFN-inducible gene9–27 ISRE fused to the firefly luciferase gene (Didcock et al.,1999). pTK-rl-lucter used as a transfection standard contains theherpes simplex virus TK promoter region upstream of the renillaluciferase gene (Promega). pIRF-3ΔN, which expresses adominant negative form of IRF-3, and pIRF-3 5D, which isconstitutively active, were obtained from John Hiscott andPaula Pitha (Lin et al., 1998).

For transfections, 100,000 cells were plated in six-well plates20 h before transfection with 1 μg of pβ-IFN-fl-lucter or pISRE-fl-lucter, 0.3 μg of pTK-rl-lucter, with or without 1 μg of IRF-3ΔN, or 1 μg of EBS plasmid expressing SeV-V protein (Nishioet al., 2005), and Fugene (Roche) according to the manufac-turer's instructions. At 24 h post-transfection, the cells were (orwere not) infected with various Sendai virus stocks or treatedwith 100 μg of poly(I)–poly(C) (Sigma, St. Louis, MO) per ml ortransfected with 1 μg of poly(I)–poly(C) using Fugene. Twentyhours later, cells were harvested and assayed for firefly andrenilla luciferase activity (dual-luciferase reporter assay system;Promega). Relative expression levels were calculated bydividing the firefly luciferase values by those of renillaluciferase. pIFNβ-GFP, which expresses GFP under the controlof the IFNβ promoter, was constructed by cloning the IFNβpromoter region from pIFNβ-fl-lucter into pEGFP-N3 (BDBiosciences Clontech), between the AseI and HindIII sites. Fortransfections, 100,000 cells were plated in six-well plates 20 hbefore transfection with 1 μg of pIFNβ-GFP and CaPO4

(Stratagene) according to the manufacturer's instructions. At24 h post-transfection, the cells were (or were not) infected withSendai virus or different DI stocks. Twenty hours later, cells wereharvested and assayed for GFP expression by FACS analysis.

Preparation of DI-H4+yfp

100,000 BSR-T7 cells were plated in six-well plates 20 hbefore transfection with a mix containing 0.75 μg of pTM1-L,1.5 μg of pTM1-N, 1.5 μg of pTM1-P/Cstop (which does notexpress C proteins), 1 μg of the various pDI constructs andFugene. Six hours later, the transfection mix was discarded andreplaced with 2 ml of Glasgow MEM supplemented with 5%FCS. Twenty four hours post-transfection, the cells wereinfected with ND SeV. Forty eight hours post-infection, thecells were scraped into their medium and injected directly intothe allantoic cavity of 9-day embryonated chicken eggs. Threedays later, the allantoic fluids were harvested and injectedundiluted into eggs. For further passages, the virus stocks were

Page 141: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

110 L. Strahle et al. / Virology 351 (2006) 101–111

clarified by centrifugation (10 min at 3000 rpm) and diluted 1/500 before injection. The presence of viruses in the resultingstock was determined by pelleting allantoic fluids (100 μl)through a TNE (10 mM Tris–HCl [pH 8.0], 100 mM NaCl,1 mM EDTA)–25% glycerol cushion for 20 min at 14,000 rpmin an Eppendorf 5417C centrifuge. Virus pellets wereresuspended in sample buffer, and the proteins were separatedby sodium dodecyl sulfate–10% PAGE and stained withCoomassie brilliant blue, alongside an ND stock of known titer.

Analysis of encapsidated RNAs

Confluent 293T cells in 9 cm ∅ Petri dishes (2 × 107 cells)were infected with 10 pfu/cell of ND stocks, and an equivalentamount of viral protein for DI stocks. Two days post-infection,the cells were collected, and the intracellular viral nucleocapsids(NC) were purified by 20–40% (w/w) CsCl density gradientcentrifugation and pelleted. After treatment with SDS andproteinase K, the nucleocapsid RNAs were phenol-extractedand ethanol-precipitated. The resulting RNAs were character-ized by Taqman analysis using specifics oligonucleotides andTaqman probes and by Northern blotting using a biotinylatedriboprobe generated in vitro by T7 RNA polymerase transcrip-tion of plus strands complementary to nucleotides 13,397–14,850 of the (−) ND genome.

Reverse transcription and real-time PCR via Taqman

Total RNA was extracted using Trizol (Invitrogen). Twentymicrograms of total RNA was mixed with 0.5 μg RandomHexamers (Promega) and subjected to a Superscript reversetranscription (RT) reaction as described by the manufacturer(Gibco) in a total volume of 50 μl. Two microliters of eachcDNA was then combined with 1 μl of internal controlHuman GAPDH (Applied Biosystems), 11 μl MasterMix(Eurogentec), 20 pmol (each) of forward and reverse primersand 4.4 pmol of Taqman probe in a total volume of 22 μl.The following primers and probes (Eurogentec or Microsynth)were used: N gene: 5′-GCAATAACGGTGTCGATCACG-3′(Fwd); 5′-TGCCTGAGCCGATCGG-3′ (Rev); 5′-CGAAGAT-GACGATACCGCAGCAGTAGC-3′ (Probe). YFP gene:5′-CCGACAACCACTACCTGAGCTA-3′ (Fwd); 5′-GAACTC-CAGCAGGACCATGTG-3′ (Rev); 5′-AAAGACCCCAACGA-GAAGCGCGA-3′ (Probe). Real-time PCR was carried out in the7700 Sequence Detector (Applied Biosystems).

Acknowledgment

This work was supported by the Swiss National ScienceFund.

References

Andrejeva, J., Childs, K.S., Young, D.F., Carlos, T.S., Stock, N.,Goodbourn, S., Randall, R.E., 2004. The V proteins of paramyxovirusesbind the IFN-inducible RNA helicase, mda-5, and inhibit its activationof the IFN-beta promoter. Proc. Natl. Acad. Sci. U.S.A. 101,17264–17269.

Calain, P., Curran, J., Kolakofsky, D., Roux, L., 1992. Molecular cloning ofnatural paramyxovirus copy-back defective interfering RNAs and theirexpression from DNA. Virology 191, 62–71.

Chanda, P.K., Kang, C.Y., Banerjee, A.K., 1980. Synthesis in vitro of the full-length complement of defective-interfering particle RNA of vesicularstomatitis virus. Proc. Natl. Acad. Sci. U.S.A. 77, 3927–3931.

Curran, J., Boeck, R., Kolakofsky, D., 1991. The Sendai virus P gene expressesboth an essential protein and an inhibitor of RNA synthesis by shufflingmodules via mRNA editing. EMBO J. 10, 3079–3085.

Delenda, C., Taylor, G., Hausmann, S., Garcin, D., Kolakofsky, D., 1998. Sendaiviruses with altered P, V, and W protein expression. Virology 242, 327–337.

Didcock, L., Young, D.F., Goodbourn, S., Randall, R.E., 1999. Sendai virus andsimian virus 5 block activation of interferon-responsive genes: importancefor virus pathogenesis. J. Virol. JID-0113724 73, 3125–3133.

Engelhorn, M., Stricker, R., Roux, L., 1993. Molecular cloning andcharacterization of a Sendai virus internal deletion defective RNA. J. Gen.Virol. 74, 137–141.

Fitzgerald, K.A., McWhirter, S.M., Faia, K.L., Rowe, D.C., Latz, E.,Golenbock, D.T., Coyle, A.J., Liao, S.M., Maniatis, T., 2003. IKKepsilonand TBK1 are essential components of the IRF3 signaling pathway. Nat.Immunol. 4, 491–496.

Frey, T.K., Jones, E.V., Cardamone Jr., J.J., Youngner, J.S., 1979. Induction ofinterferon in L cells by defective-interfering (DI) particles of vesicularstomatitis virus: lack of correlation with content of [+/−] snapback RNA.Virology 99, 95–102.

Fujii, Y., Sakaguchi, T., Kiyotani, K., Huang, C., Fukuhara, N., Egi, Y., Yoshida,T., 2002. Involvement of the leader sequence in Sendai virus pathogenesisrevealed by recovery of a pathogenic field isolate from cDNA. J. Virol. 76,8540–8547.

Garcin, D., de Melo, M., Roux, L., Kolakofsky, D., Curran, J., 1994. Presence ofa truncated form of the Sendai virus P protein in a long-term persistentinfection: implications for the maintenance of the persistent state. Virology200.

Garcin, D., Pelet, T., Calain, P., Roux, L., Curran, J., Kolakofsky, D., 1995. Ahighly recombinogenic system for the recovery of infectious Sendaiparamyxovirus from cDNA: generation of a novel copy-back nondefectiveinterfering virus. EMBO J. 14, 6087–6094.

Garcin, D., Taylor, G., Tanebayashi, K., Compans, R., Kolakofsky, D., 1998.The short Sendai virus leader region controls induction of programmed celldeath. Virology 243, 340–353.

Garcin, D., Latorre, P., Kolakofsky, D., 1999. Sendai virus C proteins counteractthe interferon-mediated induction of an antiviral state. J. Virol. 73,6559–6565.

Gotoh, B., Takeuchi, K., Komatsu, T., Yokoo, J., Kimura, Y., Kurotani, A., Kato,A., Nagai, Y., 1999. Knockout of the Sendai virus C gene eliminates the viralability to prevent the interferon-alpha/beta-mediated responses. FEBS Lett.459, 205–210.

Hasegawa, H., Yoshida, M., Shiosaka, T., Fujita, S., Kobayashi, Y., 1992.Mutations in the envelope protein of Japanese encephalitis virus affect entryinto cultured cells and virulence in mice. Virology 191, 158–165.

He, B., Paterson, R.G., Stock, N., Durbin, J.E., Durbin, R.K., Goodbourn, S.,Randall, R.E., Lamb, R.A., 2002. Recovery of Paramyxovirus simian virus 5with a V protein lacking the conserved cysteine-rich domain: themultifunctional V protein blocks both interferon-beta induction andinterferon signaling. Virology 303, 15–32.

Hiscott, J., Grandvaux, N., Sharma, S., Tenoever, B.R., Servant, M.J., Lin, R.,2003. Convergence of the NF-kappaB and interferon signaling pathways inthe regulation of antiviral defense and apoptosis. Ann. N. Y. Acad. Sci. 1010,237–248.

Iseni, F., Garcin, D., Nishio, M., Kedersha, N., Anderson, P., Kolakofsky, D.,2002. Sendai virus trailer RNA binds TIAR, a cellular protein involved invirus-induced apoptosis. EMBO J. 21, 5141–5150.

Johnston, M.D., 1981. The characteristics required for a Sendai viruspreparation to induce high levels of interferon in human lymphoblastoidcells. J. Gen. Virol. 56, 175–184.

Kato, A., Kiyotani, K., Sakai, Y., Yoshida, T., Nagai, Y., 1997. Theparamyxovirus, Sendai virus, V protein encodes a luxury function requiredfor viral pathogenesis. EMBO J 16, 578–587.

Page 142: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

111L. Strahle et al. / Virology 351 (2006) 101–111

Kawai, T., Takahashi, K., Sato, S., Coban, C., Kumar, H., Kato, H., Ishii, K.J.,Takeuchi, O., Akira, S., 2005. IPS-1, an adaptor triggering RIG-I- andMda5-mediated type I interferon induction. Nat. Immunol. 6, 981–988.

King, P., Goodbourn, S., 1994. The beta-interferon promoter responds topriming through multiple independent regulatory elements. J. Biol. Chem.269, 30609–30615.

Kolakofsky, D., 1976. Isolation and characterization of Sendai virus DI-RNAs.Cell 8, 547–555.

Lamb, R.A., Kolakofsky, D., 2001. Paramyxoviridae: the viruses and theirreplication. In: Knipe, D.M., Howley, P.M. (Eds.), Fields Virology, vol. 1.Lippincott, Williams and Wilkins, pp. 1305–1340.

Lazzarini, R.A., Keene, J.D., Schubert, M., 1981. The origins of defectiveinterfering particles of the negative-strand RNA viruses. Cell 26, 145–154.

Le Mercier, P., Garcin, D., Hausmann, S., Kolakofsky, D., 2002. Ambisensesendai viruses are inherently unstable but are useful to study viral RNAsynthesis. J. Virol. 76, 5492–5502.

Lin, R., Heylbroeck, C., Pitha, P.M., Hiscott, J., 1998. Virus-dependentphosphorylation of the IRF-3 transcription factor regulates nucleartranslocation, transactivation potential, and proteasome-mediated degrada-tion. Mol. Cell. Biol. 18, 2986–2996.

Lin, R., Mamane, Y., Hiscott, J., 1999. Structural and functional analysis ofinterferon regulatory factor 3: localization of the transactivation andautoinhibitory domains. Mol. Cell. Biol. 19, 2465–2474.

Maniatis, T., Falvo, J.V., Kim, T.H., Kim, T.K., Lin, C.H., Parekh, B.S.,Wathelet, M.G., 1998. Structure and function of the interferon-betaenhanceosome. Cold Spring Harbor Symp. Quant. Biol. 63, 609–620.

Marcus, P.I., Gaccione, C., 1989. Interferon induction by viruses. XIX. Vesicularstomatitis virus-New Jersey: high multiplicity passages generate interferon-inducing defective-interfering particles. Virology 171, 630–633.

Marcus, P.I., Sekellick, M.J., 1977. Defective interfering particles withcovalently linked [+/−]RNA induce interferon. Nature 266, 815–819.

McWhirter, S.M., Tenoever, B.R., Maniatis, T., 2005. Connecting mitochondriaand innate immunity. Cell 122, 645–647.

Meylan, E., Curran, J., Hofmann, K., Moradpour, D., Binder, M., Bartens-chlager, R., Tschopp, J., 2005. Cardif is an adaptor protein in the RIG-Iantiviral pathway and is targeted by hepatitis C virus. Nature 437,1167–1172.

Nishio, M., Tsurudome, M., Ito, M., Garcin, D., Kolakofsky, D., Ito, Y., 2005.Identification of paramyxovirus V protein residues essential for STATprotein degradation and promotion of virus replication. J. Virol. 79,8591–8601.

Perrault, J., 1981. Origin and replication of defective interfering particles. Curr.Top. Microbiol. Immunol. 93, 151–207.

Poole, E., He, B., Lamb, R.A., Randall, R.E., Goodbourn, S., 2002. The Vproteins of Simian virus 5 and other paramyxoviruses inhibit induction ofinterferon-beta. Virology 303, 33–46.

Roux, L., Simon, A.E., Holland, J.J., 1991. Effects of defective interferingviruses on virus replication and pathogenesis. Advances in Virus Research,vol. 40. Academic Press Inc., pp. 181–211.

Sekellick, M.J., Marcus, P.I., 1982. Interferon induction by viruses. VIII.Vesicular stomatitis virus: [+/−]DI-011 particles induce interferon in theabsence of standard virions. Virology 117, 280–285.

Sen, G.C., 2001. Viruses and interferons. Annu. Rev. Microbiol. 55, 255–281.Seth, R.B., Sun, L., Ea, C.K., Chen, Z.J., 2005. Identification and

characterization of MAVS, a mitochondrial antiviral signaling protein thatactivates NF-kappaB and IRF 3. Cell 122, 669–682.

Sharma, S., Tenoever, B.R., Grandvaux, N., Zhou, G.P., Lin, R., Hiscott, J.,2003. Triggering the interferon antiviral response through an IKK-relatedpathway. Science 300, 1148–1151.

Simmonds, P., Tuplin, A., Evans, D.J., 2004. Detection of genome-scaleordered RNA structure (GORS) in genomes of positive-stranded RNAviruses: implications for virus evolution and host persistence. RNA 10,1337–1351.

Strahle, L., Garcin, D., Le Mercier, P., Schlaak, J.F., Kolakofsky, D., 2003.Sendai virus targets inflammatory responses, as well as the interferon-induced antiviral state, in a multifaceted manner. J. Virol. 77,7903–7913.

Sumpter Jr., R., Loo, Y.M., Foy, E., Li, K., Yoneyama, M., Fujita, T.,Lemon, S.M., Gale Jr., M., 2005. Regulating intracellular antiviraldefense and permissiveness to hepatitis C virus RNA replication througha cellular RNA helicase, RIG-I.J. Virology 79, 2689–2699.

Vidal, S., Kolakofsky, D., 1989. Modified model for the switch from Sendaivirus transcription to replication. J. Virol. 63, 1951–1958.

Xu, L.G., Wang, Y.Y., Han, K.J., Li, L.Y., Zhai, Z., Shu, H.B., 2005. VISA is anadapter protein required for virus-triggered IFN-beta signaling. Mol. Cell 19,727–740.

Yoneyama, M., Suhara, W., Fujita, T., 2002. Control of IRF-3 activation byphosphorylation. J. Interferon Cytokine Res. 22, 73–76.

Yoneyama, M., Kikuchi, M., Natsukawa, T., Shinobu, N., Imaizumi, T.,Miyagishi, M., Taira, K., Akira, S., Fujita, T., 2004. The RNA helicase RIG-I has an essential function in double-stranded RNA-induced innate antiviralresponses. Nat. Immunol. 5, 730–737.

Page 143: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

JOURNAL OF VIROLOGY, Nov. 2007, p. 12227–12237 Vol. 81, No. 220022-538X/07/$08.00�0 doi:10.1128/JVI.01300-07Copyright © 2007, American Society for Microbiology. All Rights Reserved.

Activation of the Beta Interferon Promoter by Unnatural Sendai VirusInfection Requires RIG-I and Is Inhibited by Viral C Proteins�

Laura Strahle, Jean-Baptiste Marq, Albert Brini, Stephane Hausmann,Daniel Kolakofsky,* and Dominique Garcin

Department of Microbiology and Molecular Medicine, University of Geneva School of Medicine,11 Ave. de Champel, CH1211 Geneva, Switzerland

Received 14 June 2007/Accepted 27 August 2007

As infection with wild-type (wt) Sendai virus (SeV) normally activates beta interferon (IFN-�) very poorly,two unnatural SeV infections were used to study virus-induced IFN-� activation in mouse embryonic fibro-blasts: (i) SeV-DI-H4, which is composed mostly of small, copyback defective interfering (DI) genomes andwhose infection overproduces short 5�-triphosphorylated trailer RNAs (pppRNAs) and underproduces viral Vand C proteins, and (ii) SeV-GFP(�/�), a coinfection that produces wt amounts of viral gene products but thatalso produces both green fluorescent protein (GFP) mRNA and its complement, which can form double-stranded RNA (dsRNA) with capped 5� ends. We found that (i) virus-induced signaling to IFN-� dependedpredominantly on RIG-I (as opposed to mda-5) for both SeV infections, i.e., that RIG-I senses both pppRNAsand dsRNA without 5�-triphosphorylated ends, and (ii) it is the viral C protein (as opposed to V) that isprimarily responsible for countering RIG-I-dependent signaling to IFN-�. Nondefective SeV that cannotspecifically express C proteins not only cannot prevent the effects of transfected poly(I-C) or pppRNAs on IFN-�activation but also synergistically enhances these effects. SeV-Vminus infection, in contrast, behaves mostly likewt SeV and counteracts the effects of transfected poly(I-C) or pppRNAs.

All viruses evade the cellular innate immune system in partby expressing gene products that interfere with the ability ofthe host cell to establish an antiviral state (6). In the case of theParamyxovirinae, this anti-host-defense activity is due mostly toviral C and V proteins (15, 27, 31). The C and V proteins areencoded by separate alternate open reading frames (ORFs),which both overlap that of the P protein. V and C are alsoreferred to as accessory gene products, as not all members ofthis virus subfamily express one or the other. More specifically,rubulavirus and avulavirus express V but do not express Cproteins, and human parainfluenza virus type 1 (PIV1), a res-pirovirus most closely related to Sendai virus (SeV), expressesC but does not express a V protein (16, 20).

Paramyxovirus V and C proteins antagonize interferon(IFN) signaling by various mechanisms, and they also targetthe production of type I IFN (15, 31). Beta IFN (IFN-�)production is one of the earliest events in the cellular innateimmune response, which leads to the establishment of an anti-viral state. IFN-� production requires the coordinated activa-tion of several transcription factors, including NF-�B and IRF3(15, 29). For intracellular RNA virus replication, the signalingpathway that leads to IRF3 activation starts with mda-5 andRIG-I, two cytoplasmic DExH/D-box helicases with N-termi-nal CARD domains. These helicases respond to double-stranded RNA (dsRNA) and, at least for RIG-I, to 5�-triphos-phorylated single-stranded RNA (ssRNA) (pppRNA), whichare generated in the cytoplasm during RNA virus replication

(9, 11, 25). Upon the detection of these viral RNAs, the CARDdomains of these helicases interact with IPS-1/Cardif/MAVS/VISA, which is present in the mitochondrial membrane, andthis CARD-CARD interaction is thought to lead to the re-cruitment and activation of TBK1, IKKε, and other IKK ki-nases that activate NF-�B and IRF3, thereby activating theIFN-� promoter (8). The production of these early IFNs ini-tiates autocrine and paracrine signal amplifications via theJak/Stat pathway to produce a generalized antiviral state andalso assists in the subsequent activation of adaptive immuneresponses.

The role of mda-5 in sensing RNA virus infection was un-covered because mda-5 was found to bind to the PIV5 Vprotein and other paramyxovirus V proteins, including SeV V.These V-protein–mda-5 interactions, moreover, preventedIFN-� activation in response to transfected poly(I-C) (1). Onthe other hand, other studies found that RIG-I and not mda-5acts as the sensor of paramyxovirus infection (13, 28). Thispaper provides evidence that for SeV infection of mouse em-bryonic fibroblasts (MEFs), it is the C protein (and not V) thatis primarily responsible for this effect and that C acts by coun-tering RIG-I (and not mda-5). Independent expression of Cwas found to inhibit RIG-I-dependent signaling to the IFN-�promoter induced by either pppRNAs or dsRNAs. Moreover,SeV that cannot specifically express C proteins was unable tocounteract poly(I-C)- or pppRNA-induced IFN-� activation,whereas SeV that cannot express V behaved mostly like wild-type (wt) SeV.

MATERIALS AND METHODS

Cells, viruses, and antibodies. MEFs were grown in Dulbecco’s modifiedEagle’s medium supplemented with 10% fetal calf serum.

SeV stocks were grown in the allantoic cavitiesof 9-day-old embryonatedchicken eggs for 3 days at 33°C. For nondefective stocks (109 PFU/ml), 0.1 ml of

* Corresponding author. Mailing address: Department of Microbi-ology and Molecular Medicine, University of Geneva School of Med-icine, 11 Ave. de Champel, CH1211 Geneva, Switzerland. Phone: 41-223795657. Fax: 41-223795702. E-mail: [email protected].

� Published ahead of print on 5 September 2007.

12227

at Bibliotheque F

aculte Medecine G

eneve on Novem

ber 9, 2007 jvi.asm

.orgD

ownloaded from

Page 144: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

12228

at Bibliotheque F

aculte Medecine G

eneve on Novem

ber 9, 2007 jvi.asm

.orgD

ownloaded from

Page 145: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

a 105 dilution (ca. 1,000 PFU) was inoculated per egg. In the case of DI stocks,0.1 ml of a 103 dilution was used. In all cases, the amount of viral proteins presentin the resulting allantoic fluid was analyzed by sodium dodecyl sulfate-poly-acrylamide gel electrophoresis and Coomassie blue staining of pelleted virus.Virus titers were determined by plaquing on LLC-MK2 cells.

SeV-GFP(�), which expresses green fluorescent protein (GFP) from a trans-gene between the M and F genes, and SeV-GFP(�) or SeV-RFP, which ex-presses antisense GFP mRNA or red fluorescent protein (RFP) (dsRED) fromsimilarly located transgenes, were prepared as previously described (31). DI-H4stocks were described previously (30).

Primary antibodies used included rabbit anti-RFP (AB3216; Chemicon); anti-actin monoclonal antibody (MAb) (1501; Chemicon); rabbit anti-GFP (632460;BD biosciences); rabbit anti-SeV-P/C/V (homemade); anti-hemagglutinin (HA)MAb (16B12; BABCO), anti-Flag MAb (F1804; Sigma), rabbit anti-mda-5 (J.Tschopp, Lausanne, Switzerland), and rabbit anti-RIG-I (T. Fujita, Kyoto,Japan).

Plasmids, transient transfections, infections, inductions, luciferase assay, andfluorescence-activated cell sorter analysis. EBS plasmids (3) expressed viral andfluorescent proteins and were constructed by standard methods; precise detailcan be obtained from the authors.

NS1 {residues 1 to 77 [NS1(1-77)]} (from Jacques Perrault) and E3L {residues100 to 190 [E3L(100-190)]} (from Bertram Jacobs), were HA tagged and clonedinto pEBS. Flag-tagged RIG-I, RIG-I-C, or RIG-�CARDS (dominant negative)and mouse mda-5 were obtained from Jurg Tshopp and Klaus Conzelmann.

p�-IFN-fl-lucter, which contains the firefly luciferase gene under the control ofthe human IFN-� promoter, was described previously (14). pTK-rl-lucter, usedas a transfection standard, contains the herpes simplex virus TK promoter regionupstream of the Renilla luciferase gene (Promega).

For transfections, 100,000 cells were plated into six-well plates 20 h beforetransfection with 1.5 �g of p�-IFN-fl-lucter; 0.5 �g of pTK-rl-lucter; 0.5 �g ofplasmids expressing RIG-I and MDA-5; 1.5 �g of plasmids expressing V (whoseC ORF is closed with a stop codon), C1-204 or C1-23-Tom-C24-204 (or C*),NS1(1-77), wt E3L, mutant E3L(100-190), or RIG-� proteins (as indicated); andTransIT-LT1 transfection reagent (Mirus). At 24 h posttransfection, the cellswere (or were not) infected with various SeV stocks or transfected with 5 �g ofpoly(I-C) using TransIT-LT1 transfection reagent. Twenty hours later, cells wereharvested and assayed for firefly and Renilla luciferase activity (dual-luciferasereporter assay system; Promega). Relative expression levels were calculated bydividing the firefly luciferase values by those of Renilla luciferase.

Immunoblotting. Cytoplasmic extracts were prepared using 0.5% NP-40buffer. Equal amounts of total proteins were separated by sodium dodecyl sul-fate-polyacrylamide gel electrophoresis and transferred onto Immobilon-P mem-branes by semidry transfer. The secondary antibodies used were alkaline phos-phatase-conjugated goat anti-rabbit (or mouse) immunoglobulin G (Bio-Rad).The immobilized proteins were detected by light-enhanced chemiluminescence(Pierce) and analyzed in a Bio-Rad light detector using Quantity One software.

In vitro synthesis of RNA, purification, and transfection. DNA for T7 RNApolymerase synthesis of model RNA1 was prepared by PCR using the followingpartially complementary primers: 5�-TAATACGACTCACTATA(ggg/gca)ACACACCACAACCAACCCACAAC-3� (forward) (start sites are in lowercase type)and 5�-GAAAGAAAGGTGTGGTGTTGGTGTGGTTGTTGTGGGTTGGTTGTGG-3� (reverse). In vitro transcription was performed on 100 pmol ofpurified PCR product using T7 MEGAshortcript from Ambion according to themanufacturer’s instructions. For RNA1 containing the unusual OHGCA startsite, RNA was initiated with the dinucleotide 5� OHGpC24 in a reaction withoutGTP. For RNA1 containing the usual pppGGG start site, part of the product wastreated with 20 units of calf intestinal phosphatase (Roche) for 30 min at 37°C

followed by proteinase K treatment (15 min at 37°C), phenol extraction, andethanol precipitation. The T7 transcripts were purified on NucAway Spin col-umns from Ambion (to remove unincorporated nucleotides). SeV trailer pppR-NAs were synthesized similarly by using specific PCR primers.

For RNA transfection, 1 �g (1�) to 3 �g (3�) of RNA was transfected intoMEF cells using TransMessenger transfection reagent (QIAGEN).

RT and real-time PCR via TaqMan. Confluent MEFs in 10-cm petri dishes(107 cells) were infected with 20 PFU/cell of SeV GFP(�), SeV GFP(�), or bothstocks. At 24 h postinfection (hpi), the cells were collected and lysed in 300 �l ofNP-40 lysis buffer. Cytoplasmic extracts were then centrifuged in a 20 to 40%(wt/wt) CsCl density gradient (16 h at 35,000 rpm at 12°C). The pellet RNAswere ethanol precipitated and resuspended in 50 �l of Tris-EDTA. Fifteenmicrograms of RNA was then mixed with 0.5 �g of the forward or the reverseGFP primer and subjected to a Superscript reverse transcription (RT) reaction,according to instructions provided by the manufacturer (Gibco), in a total vol-ume of 50 �l. Five microliters of each cDNA was then combined with 12 �lMasterMix (Eurogentec), 20 pmol (each) of forward and reverse primers, and 4.4pmol of TaqMan probe in a total volume of 25 �l. The following primers andprobes (Eurogentec or Microsynth) were used for RT and TaqMan analyses ofthe GFP gene: 5�-CCGACAACCACTACCTGAGCAC-3� (forward), 5�-GAACTCCAGCAGGACCATGTG-3� (reverse), and 5�-AAAGACCCCAACGAGAAGCGCGA-3� (probe). Real-time PCR was carried out in duplicates using a 7700sequence detector (Applied Biosystems).

RESULTS

Three ways to activate IFN-�. We have used three ways toinduce the activation of an IFN-� promoter expressing a lucif-erase reporter gene in MEFs (see Fig. 2A). The first is tosimply transfect a synthetic dsRNA, poly(I):poly(C) [poly(I-C)], into the cells. The second way is to infect the cells with anSeV stock that contains a well-characterized copyback DI ge-nome (H4) (30). The third way is to coinfect cells with SeV-GFP(�), which expresses a GFP transgene, and SeV-GFP(�),which expresses mRNA containing the complement of theGFP ORF, as recently described for vesicular stomatitis virus(VSV) (24). As shown in Fig. 1A, infection with increasingamounts of SeV-GFP(�) alone leads to increasing GFP ex-pression. Coinfection of 20 PFU/cell of SeV-GFP(�) withincreasing amounts of SeV-GFP(�) leads to the gradual de-crease of GFP expression (top). At 20 PFU/cell of SeV-GFP(�), there are roughly equal amounts of GFP and anti-GFP mRNAs intracellularly (by strand-specific quantitativeRT-PCR) (Fig. 1B and see Materials and Methods), and thereis a 90% loss of GFP expression (Fig. 1A, top). This loss ofGFP expression cannot be accounted for by the reduced levelof GFP mRNA (Fig. 1B). In contrast, coinfection with increas-ing amounts of SeV expressing RFP as a neutral control (SeV-RFP) has a reduced ability to interfere with GFP expression(Fig. 1A). More importantly, whereas infection with SeV-GFP(�) alone or its coinfection with SeV-RFP leads to little

FIG. 1. IFN-� activation induced by SeV-GFP(�/�) infections. (A) Parallel cultures of MEFs were first transfected with pIFN�-lucff andpTK-lucr and then infected with increasing amounts of either SeV-GFP(�) alone (which expresses a GFP mRNA from a transgene between theM and F genes) or 20 PFU/cell of SeV-GFP(�) plus increasing amounts of either SeV-GFP(�) (which expresses an anti-GFP mRNA from atransgene in the same location) or SeV-RFP (which expresses an RFP mRNA from a transgene in the same location), as indicated. GFP expressionwas monitored by fluorescence-activated cell sorter analysis at 20 hpi. Cell extracts were prepared at 20 hpi, and equal amounts were used todetermine luciferase activities (below). These transfections were carried out three times with independent virus stocks, with similar results.(B) Cytoplasmic extracts were centrifuged on CsCl density gradients to isolate nonencapsidated (pellet) RNAs. The levels of GFP and anti-GFPmRNAs in 15 �g of CsCl pellet RNA were determined using sense- and antisense-specific primers for RT, followed by quantitative PCR (TaqMan)(see Materials and Methods). (C) Parallel cultures of MEFs were first transfected with the luciferase reporter plasmids plus either an empty vector,one expressing wt E3L(100–190), or one expressing mutant E3L(100–190) (E3L-mut.) and then infected with increasing amounts of SeV-GFP(�)and SeV-GFP(�) as indicated. Cell extracts were prepared at 20 hpi, and equal amounts were used to determine luciferase activities. Equalamounts of cell extracts were also Western blotted using anti-N and anti-HA (below).

VOL. 81, 2007 ACTIVATION OF IFN-� PROMOTER BY SeV INFECTION 12229

at Bibliotheque F

aculte Medecine G

eneve on Novem

ber 9, 2007 jvi.asm

.orgD

ownloaded from

Page 146: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

12230

at Bibliotheque F

aculte Medecine G

eneve on Novem

ber 9, 2007 jvi.asm

.orgD

ownloaded from

Page 147: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

or no activation of IFN-� (Fig. 1A), coinfection with SeV-GFP(�) clearly activates the IFN-� promoter (Fig. 1A, bot-tom). This IFN-� activation is inhibited by the coexpression ofthe dsRNA-binding domain of the vaccinia virus E3L protein,whereas this activation is unaffected by a mutant form of E3Lcontaining two point mutations that eliminate the binding ofdsRNA (10) (Fig. 1C). Taken together, our results show thatonly SeV coinfections that can form GFP dsRNA induceIFN-� activation.

The two SeV infections that activate IFN-� differ from eachother in several respects. First, DI-H4 genomes are of thecopyback variety and contain the strong antigenomic replica-tion promoter at their 3� ends. DI-H4 genomes thus have astrong competitive advantage in replication over nondefective(ND) genomes, and this sometimes leads to less viral structuralproteins like N and P being present intracellularly (see, e.g., Pprotein in Fig. 3A), but sometimes, this difference is minimal(see, e.g., Fig. 2A). However, in either case, viral V and Cproteins are almost entirely absent in these DI-H4-infectedcells, whereas V and C are found at wild-type levels inGFP(�/�) infections (Fig. 2 and data not shown). Both ofthese viral proteins are thought to limit IFN-� activation dueto virus infection (15, 31). Second, the DI genome replicationpromoters, like those of the ND genomes, are always active inthe presence of viral polymerase, and short 5�-triphosphory-lated trailer RNAs (rather than full-length DI genomes) aretranscribed by a relatively nonprocessive polymerase, espe-cially when the N protein is limiting. Unlike genome synthesisthat is dependent on ongoing (N) protein synthesis, that oftrailer RNA actually increases when translation is blocked withcycloheximide (18). SeV trailer RNAs are known to specificallybind to TIAR, a cellular RNA binding protein of the ELAVfamily (4), and to prevent virus-induced apoptosis (7, 12).DI-H4 infections are expected to overproduce trailer RNAs,which may stimulate RIG-I (11, 25), similar to measles virusleader RNA (26). Lastly, DI-H4-infected cells also containsmall amounts of unencapsidated H4 genome RNA that canself-anneal in a concentration-independent manner to formdsRNA panhandles with 5�-triphosphorylated ends. SeV-GFP(�/�)-infected cells, on the other hand, can form dsRNAswith capped ends.

Relative contributions of mda-5 and RIG-I in sensing SeVinfections in MEFs. mda-5 signaling to IFN-� was discoveredbecause the PIV5 V protein was found to bind this helicase andthereby prevent poly(I-C)-induced IFN-� activation. Furtherwork showed that the V–mda-5 interaction is a general prop-erty of paramyxovirus V proteins, including that of SeV (1, 5).

Nevertheless, several groups have now found that SeV infec-tion is sensed by RIG-I (and not mda-5) (13). To determinewhether RIG-I was also responsible for signaling to the IFN-�promoter in our MEFs, we examined the effect of expressing adominant-negative form of RIG-I [RIG-I(�CARDs), whoseN-terminal CARD domains are deleted] (Fig. 2A). MEFs werefirst transfected with pIFN�-lucff, pTK-lucr, and plasmids ex-pressing either RIG-I(�CARDs) or an empty vector as a neg-ative control. After 24 h, the cells were either transfected withpoly(I-C) or infected with either SeV-DI-H4 or SeV-GFP(�/�),and luciferase levels were determined 24 h later. As shown inFig. 2A, both SeV infections strongly activated the IFN-� pro-moter, whereas transfected poly(I-C) had a more modest effectin this experiment. RIG-I(�CARDs) coexpression did not af-fect the levels of P, V, and C proteins found intracellularly(bottom), but this coexpression reduced IFN-� activation tobackground levels in all three cases.

To determine whether the loss of IFN-� activation by RIG-I(�CARDs) coexpression was due to its ability to also inhibitmda-5 signaling, e.g., by sequestering cytoplasmic viral RNAs,we examined whether RIG-I(�CARDs) could inhibit mda-5signaling. As mda-5 and RIG-I can be activated by simpleoverexpression, we examined the effect of RIG-I(�CARDs)expression on IFN-� activation due to the overexpression ofthese two helicases. As shown in Fig. 2B, IFN-� activationclearly occurred upon exogenous mda-5 or RIG-I expression.Moreover, whereas RIG-I(�CARDs) coexpression completelyinhibited activation due to exogenous RIG-I, RIG-I(�CARDs)coexpression had little effect in countering IFN-� activation dueto exogenous mda-5. In contrast, the coexpression of a dominant-negative form of mda-5 completely inhibited IFN-� activationdue to mda-5 overexpression (Fig. 2B). Taken together, theseresults suggest that IFN-� activation in our MEFs in response tothese SeV infections is predominantly, if not exclusively, due tothe action of RIG-I.

The coexpression of either SeV V or C proteins stronglyinhibited IFN-� activation due to RIG-I overexpression,whereas only the V protein strongly inhibited IFN-� activationdue to mda-5 overexpression (Fig. 2B). The finding that SeV Vinhibits RIG-I signaling as well as that of mda-5 is consistentwith data from Childs et al. (5), who reported that SeV V wasa possible exception to the rule that all V proteins inhibitedmda-5 but not RIG-I. They reported that SeV V did in factmodestly inhibit RIG-I (35%), whereas all other V proteinshad no effect.

SeV V and C inhibition of SeV-DI-H4 and SeV-GFP(�/�)induced IFN-� activation. We next examined the abilities of

FIG. 2. Relative contributions of mda-5 and RIG-I in sensing SeV infections in MEFs. (A) Parallel cultures of MEFs were first transfected withpIFN�-lucff, pTK-lucr, and plasmids expressing dominant-negative RIG-I(�CARDs) or an empty vector as a negative control. After 24 h, the cellswere either transfected with poly(I-C) or infected with either SeV-DI-H4 or SeV-GFP(�/�) (as indicated). Cytoplasmic extracts were preparedafter a further 20 h of incubation and used to determine firefly and Renilla luciferase levels and the relative levels of RIG-I(�CARDs) (anti-Flag)and viral proteins (anti-P/V/C) by Western blotting (bottom). All transfections were carried out in duplicate, and the range of values obtained isindicated by the error bars. Ctrl., control. (B) Parallel cultures of MEFs were transfected with pIFN�-lucff, pTK-lucr, and plasmids expressingFlag-tagged RIG-I or mda-5 or these helicases plus RIG-I(�CARDs), mda-5(�CARDs), SeV V (whose overlapping C ORF was closed by a stopcodon), SeV C, or an empty plasmid as a negative control, as indicated. Cytoplasmic extracts were prepared after 40 h of incubation and used todetermine firefly and Renilla luciferase levels. All transfections were carried out in duplicate, and the range of values obtained is indicated by errorbars. The relative levels of the Flag–RIG-I constructs were determined by Western blotting with anti-Flag, those of the Flag–mda-5 constructs weredetermined with anti-mda-5, and those of the viral V and C proteins were determined with anti-P/V/C serum (bottom). Vect., vector.

VOL. 81, 2007 ACTIVATION OF IFN-� PROMOTER BY SeV INFECTION 12231

at Bibliotheque F

aculte Medecine G

eneve on Novem

ber 9, 2007 jvi.asm

.orgD

ownloaded from

Page 148: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

the SeV V and C proteins to inhibit IFN-� activation inducedby SeV-DI-H4 and SeV-GFP(�/�) infections. As shown inFig. 3A, exogenous expression of the SeV V protein did notaffect the level of viral P, V, and C proteins in SeV infections,but it did reduce IFN-� activation due to DI-H4 infection (by60%). Remarkably, SeV V overexpression did not inhibitIFN-� activation due to SeV-GFP(�/�) infection. The coexpres-sion of exogenous SeV C protein (actually C1–23-Tom-C24–204,

which migrates just slightly slower than the viral P protein) sim-ilarly did not affect the level of viral P, V, and C proteins in SeVinfections. C overexpression, however, more strongly inhibitedIFN-� activation due to either SeV infection [DI-H4-inducedactivation was reduced by 90%, and GFP(�/�)-induced acti-vation was reduced by 75%]. Coexpression of the unmodified Cprotein produced similar results (Fig. 2B and data not shown).The ability of the GFP(�/�) infection to activate IFN-�, despite

FIG. 3. SeV V and C inhibition of IFN-� activation induced by SeV infections. (A) Parallel cultures of MEFs were transfected with pIFN�-lucff,pTK-lucr, and plasmids expressing the SeV V protein, the SeV C protein (actually C1–23-Tom-C24–204), or unmodified Tom as a negative control.After 24 h, the cells were infected with either SeV-DI-H4 or SeV-GFP(�/�) (as indicated). Cytoplasmic extracts were prepared after a further24 h of incubation and used to determine firefly and Renilla luciferase levels. The relative levels of viral P, V, and C proteins were determined byWestern blotting with anti-P/C/V serum (bottom). All transfections were carried out in duplicate, and the range of values obtained is indicated byerror bars. (B) Parallel cultures of MEFs were transfected with pIFN�-lucff, pTK-lucr, and plasmids expressing either Tom, RIG-I(�CARDs), IAVNS1(1–73), C1–23-Tom-C24–204 (C*), or V, as indicated. After 24 h, the cells were infected with increasing amounts of SeV-DI-H4 (1�, 2�, and4�). Cytoplasmic extracts were prepared after a further 24 h of incubation and used to determine luciferase levels.

12232 STRAHLE ET AL. J. VIROL.

at Bibliotheque F

aculte Medecine G

eneve on Novem

ber 9, 2007 jvi.asm

.orgD

ownloaded from

Page 149: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

normal levels of expression of the V and C proteins, is presumablydue to the early formation of GFP dsRNA. In this case, the SeVV protein is considerably less potent than C in preventing theresponse to this dsRNA.

As the DI-H4 infections accumulate so few V and C pro-teins, we compared the abilities of these proteins (expressedfrom plasmids) to inhibit IFN-� activation relative to RIG-I(�CARDs) and the dsRNA binding domain of the influenzaA virus (IAV) NS1(1–73), another viral protein that inhibitsRIG-I signaling (22, 25). As shown in Fig. 3B, the SeV Cprotein was as active as RIG-I(�CARDs) in combating anincreasing dose of DI-H4 infection and almost as active asNS1. Consistent with above-described results (Fig. 3A), theSeV V protein was less active than C but was still able to inhibitmost of the DI-H4-induced IFN-� activation.

The SeV C1–204 protein is composed of two domains: theN-terminal 23 amino acids (C1–23) which act as a plasma mem-brane (PM) targeting signal (19) and which is present in thelonger (C�/C) but not in the shorter (Y1/Y2) “C” proteins, andC24–204, or the Y1 protein, which acts as a protein interactiondomain. Whereas C24–204 (or Y1) is naturally expressed duringinfection, C1–23 is only found fused to Y1. In order to study thedifferent contributions of these two domains to C-protein func-tion, we have used tomato red fluorescent protein (Tom) inwhich C1–23 is fused to the N terminus of Tom and C24–204 isfused to its carboxy terminus as a carrier. The interposition ofTom between these two domains of C remarkably does notappear to affect any of the activities of C1–204 (19). MEFs weretransfected with the luciferase reporter plasmids along withplasmids expressing various Tom constructs as indicated (Fig.4). After 24 h, half of the cultures were infected with SeV-DI-H4, and luciferase levels were determined after a further 24 h.The expression of C1–23-Tom, which carries the wt PM anchor

and is localized at the cell surface, or P8P9-Tom, which carriesa mutant PM anchor and is distributed throughout the cyto-plasm (19), had little or no effect on the DI-H4-induced IFN-�activation. In contrast, both C1–23-Tom-C24–204 and P8P9-Tom-C24–204 reduced IFN-� activation to near-background levels.C24–204 alone (Tom-C24–204), moreover, was still quite active inthis respect (Fig. 4). Thus, the C24–204 or Y1 protein interac-tion domain appears to be responsible for inhibiting RIG-I-dependent IFN-� activation, and this inhibition is largelyindependent of whether C24–204 is localized at the PM.

SeV C protein inhibits IFN-� activation induced by trans-fected pppRNA. A general property of nonsegmented nega-tive-strand RNA viruses is that short, promoter-proximalpppRNAs (leader and trailer RNAs) are transcribed fromtheir replication promoters, especially when unassembled Nprotein is limiting (17, 18). The ability of SeV infections toinduce IFN is essentially due to the presence of DI genomesthat are present in their egg-grown stocks, especially those ofthe copyback variety (30). As mentioned above, copyback DIgenomes have a strong replicative advantage because they con-tain strong replication promoters at the DI genome and anti-genome 3� ends. Copyback DI genome replication thus gener-ates short trailer RNAs that are unmodified at either end andcan be considered as unstable, abortive replication products(see Discussion).

To examine whether trailer RNAs act as pathogen-associ-ated molecular patterns (PAMPs), we transfected trailer RNAmade by T7 RNA polymerase in vitro into our MEFs andmonitored the activation of IFN-�. As the ability of pppRNAsto induce IFN-� activation is not sequence dependent (11), wealso examined model RNAs that were initiated with GTP butthen treated with phosphatase or those initiated with the dinu-cleotide GpC rather than pppG (23). The transfections of all

FIG. 4. SeV C24–204 (or Y1) protein inhibits IFN-� activation induced by DI-H4 infection. Parallel cultures of MEFs were transfected withpIFN�-lucff, pTK-lucr, and plasmids expressing tomato constructs carrying either the wt (C1–23) or mutant (P8P9) C1–23 fused to their N termini,with and without C24–204 (or Y1) fused to their carboxy termini, C24–204 fused to the carboxy terminus alone (Tom-C24–204), or unmodified Tomas a negative control (ctrl.), as indicated. After 24 h, the cells were infected with SeV-DI-H4 (as indicated). Cytoplasmic extracts were preparedafter a further 24 h of incubation and used to determine luciferase levels. The relative levels of the various tomato constructs were determined byWestern blotting with anti-dsRED (bottom). All transfections were carried out in duplicate, and the range of values obtained is indicated by errorbars.

VOL. 81, 2007 ACTIVATION OF IFN-� PROMOTER BY SeV INFECTION 12233

at Bibliotheque F

aculte Medecine G

eneve on Novem

ber 9, 2007 jvi.asm

.orgD

ownloaded from

Page 150: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

FIG. 5. pppRNA-induced activation of IFN-�. (A) Parallel cultures of MEFs were transfected with pIFN�-lucff and pTK-lucr, and pRIG-I wasalso transfected in some cultures, as indicated. After 24 h, the cells were transfected for 3 h with increasing amounts (1 or 3 �g) of eitherpppGGG/RNA1, phosphatase-treated GGG/RNA1, pppGCA/RNA1, or OHGCA/RNA1, as indicated. Cytoplasmic extracts were prepared 18 hpost-RNA transfection and used to determine luciferase levels. (B) Parallel cultures of MEFs were transfected with pIFN�-lucff, pTK-lucr, andplasmids expressing Tom, RIG-I(�CARDs), IAV NS1(1–73), C1–23-Tom-C24–204 (C*), or V, as indicated. After 24 h, the cells were transfected withincreasing amounts (1 �g and 3 �g) of pppGGG/RNA1, as indicated. Cytoplasmic extracts were prepared after 3 h of RNA transfection and usedto determine luciferase levels. Rel., relative. (C) Same as above (B), except that the cells were transfected with 3 �g of ppptrailer RNA.

12234 STRAHLE ET AL. J. VIROL.

at Bibliotheque F

aculte Medecine G

eneve on Novem

ber 9, 2007 jvi.asm

.orgD

ownloaded from

Page 151: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

three 5�-triphosphorylated ssRNAs clearly led to IFN-� acti-vation, whereas both RNAs that contained 5�-hydroxyl endshad essentially lost their ability to activate IFN-� in paralleltransfections (Fig. 5A and C), confirming previously reportedresults (11, 25, 26). We then examined the ability of the SeV Cand V proteins to inhibit 5�-pppRNA-dependent activation ofIFN-� compared to RIG-I(�CARDs) and IAV NS1(1–73).MEFs were first transfected with plasmids expressing variousviral inhibitory proteins or an empty plasmid as a negativecontrol and then transfected with increasing amounts ofpppRNAs. IFN-� activation was monitored after a further 18 h.As shown in Fig. 5B, expression of the SeV C protein was asactive as RIG-I(�CARDs) in inhibiting IFN-� activation at allamounts of pppRNAs transfected although not quite as activeas NS1(1–73). Expression of SeV V was again the least inhib-itory; in fact, significant inhibition occurred only at the lowestamount of pppRNA. Thus, short 5�-triphosphorylated ssRNAssuch as trailer RNA are potent stimulators of IFN-� whentransfected into our MEFs, and expression of the SeV C pro-tein (but not the SeV V protein) can effectively inhibit thisstimulation (Fig. 5B and C).

Relative importance of C and V in inhibiting RIG-I-depen-dent signaling to IFN-�. Another way to investigate the rela-tive importance of the C and V proteins in inhibiting RIG-I-dependent signaling to IFN-� is to compare the relativeabilities of SeV infections that cannot specifically express the Cor V proteins to affect pppRNA- or poly(I-C)-induced IFN-�

activation. MEFs were therefore first infected with 20 PFU/mlof either wt SeV, SeV-Vminus (containing a stop codon in the VORF just downstream of the mRNA editing site, which pro-duces a W-like protein instead of V), or SeV-Cminus (contain-ing three stop codons in the C ORF downstream of the Y2initiation codon). The infected cells were then transfected (at24 hpi) with pIFN-�-luc plus either pppRNA, poly(I-C), or noRNA and then harvested after 18 h to determine reporter geneactivity. As shown in Fig. 6, these three SeVs replicate toclearly different levels in our highly IFN-competent MEFs(even though they replicate similarly in BSR T7, 293T, andVero cells), highlighting the essential functions that these ac-cessory proteins play in countering the innate immune re-sponse. Nevertheless, in the absence of transfected RNA, onlySeV-Cminus infection activates IFN-� to any extent or increasesRIG-I levels; RIG-I is an IFN-stimulated gene, and its levelreflects that of the antiviral state (Fig. 6, bottom). Transfec-tions of either pppRNA or poly(I-C) strongly activated thereporter gene and increased RIG-I levels. Prior infection witheither SeV wt or SeV-Vminus reduced transfected RNA stim-ulation of the reporter gene and prevented the increase inRIG-I levels (Fig. 6). SeV-Vminus was only slightly less effectivethan wt SeV in this respect. In sharp contrast, prior infectionwith SeV-Cminus not only did not prevent the increase in RIG-Ilevels but also acted synergistically with either pppRNA orpoly(I-C) transfection to increase reporter gene activity byincreasing the level of RIG-I. These results reinforce the view

FIG. 6. RNA-induced activation of IFN-� in cells infected with SeV that cannot express either V or C. Parallel cultures of MEFs were eithermock infected or infected with 20 PFU/ml of wt SeV, SeV-Vminus, or SeV-Cminus, as indicated. After 24 h, the cells were transfected with luciferasereporter plasmids and either pppGGG/RNA1, poly(I-C), or no RNA (untreated). Cytoplasmic extracts were prepared after 18 h of RNAtransfection, used to determine luciferase levels (above), and Western blotted to determine the levels of P, V, and C proteins as well as endogenousRIG-I and actin as a loading control. rSeV, recombinant SeV.

VOL. 81, 2007 ACTIVATION OF IFN-� PROMOTER BY SeV INFECTION 12235

at Bibliotheque F

aculte Medecine G

eneve on Novem

ber 9, 2007 jvi.asm

.orgD

ownloaded from

Page 152: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

that it is primarily the SeV C protein (and not V) that inhibitspppRNA- and dsRNA-induced signaling to the IFN-� pro-moter via RIG-I during SeV infection.

DISCUSSION

SeV has been one of the most extensively used model virusesto investigate IFN induction in infected cells. Most of this workhas used the commercially available Cantell strain of SeV,whose ability to induce IFN, like that of other SeV strains, isdue to the presence of DI genomes in egg-grown stocks. Non-defective SeVs that are plaque purified from these stocks,including that of the Cantell strain, do not induce IFN unlesscellular RIG-I levels are artificially increased (21, 30). Fornondefective SeV infection, the expression of the C and Vproteins is apparently sufficient to prevent IFN-� activationunder normal conditions. Measles virus infection, in con-trast, can apparently induce IFN in the absence of DI ge-nomes, and evidence that this induction is due to the syn-thesis of leader pppRNAs has recently been provided (26).Leader and trailer RNAs, which are unmodified at eitherend, are unstable in infected cells unless they are encapsi-dated with the N protein (presumably after their synthesis asfree RNAs) (2). Free leader and trailer RNAs are moreeasily detected in VSV infections, which synthesize largeramounts of viral RNAs over a shorter period of time. Fornondefective VSV infections, eight times as many trailerRNAs/antigenome template are found as leader RNAs/ge-nome template, consistent with the relative strengths oftheir respective replication promoters. For VSV copybackDI infections, there are 40 times as many trailer RNAs/template (nondefective antigenome plus DI genome) asleader RNAs/genome template, presumably reflecting theincreased strength of the copyback DI replication promot-ers. The VSV polymerase clearly has a strong preference forinitiating RNA synthesis at the 3� ends of copyback DIgenomes over both ND genomes and antigenomes (17).

We previously noted that not all SeV stocks that contain DIgenomes strongly induce IFN; this ability appears to be re-stricted to stocks containing relatively small copyback DI ge-nomes (the smaller the DI genomes, the more moles of endsare present for a given weight). The commercially availableCantell strain contains a copyback DI genome of only 546nucleotides in length, the smallest SeV DI genome describedto date (30). SeV copyback DI-H4 genomes (1,410 nucleo-tides) have the same strong replicative advantage as their VSVcounterparts because they also contain strong replication pro-moters at both their genome and antigenome 3� ends. Thus,SeV copyback DI infections presumably synthesize consider-ably more pppRNAs than standard virus infections. We alsopreviously noted that when cytoplasmic extracts of DI-H4-infected cells are centrifuged on CsCl density gradients, smallamounts of DI genome RNA are found in the pellet fraction(30). This indicates that this RNA is not encapsidated with theN protein and therefore forms dsRNA panhandles in a con-centration-independent manner. Thus, copyback DI-H4 infec-tions apparently produce considerably more of both knownPAMPs of RNA virus infection than do standard virus infec-tions. Coupled with their strongly reduced accumulation of the

viral C and V proteins, it is easy to see why these copyback DIinfections are such potent inducers of IFN-�.

All paramyxoviruses express either C or V proteins, andmany viruses express both. In viruses that express only C or V,we presume that either viral protein alone counteracts theinnate immune response of the host to aid virus replication.The C and V proteins, which bear no sequence similarity, likelytarget different key elements of the host response. Viruses thatexpress both C and V presumably have more diverse ways ofcountering innate immunity. In support of this notion, SeVinfections that cannot specifically express either the C or Vprotein contain increased levels of IFN-� and interleukin-8mRNAs relative to wt SeV infections (31), and the indepen-dent expression of the C or V protein will inhibit poly(I-C)- orNewcastle disease virus-dependent activation of IRF-3 (15).Previous work has identified mda-5 as being a key target ofparamyxovirus V proteins in countering the innate immuneresponse (1). This paper provides evidence that for SeV infec-tion of MEFs, it is the C protein (and not V) that is primarilyresponsible for this effect and that C acts by countering RIG-I-dependent signaling to IFN-�. For example, the independentexpression of either C or V inhibited IFN-� activation due toRIG-I overexpression (Fig. 2B). Also, both proteins inhibitedIFN-� activation due to DI-H4 infection, although C was al-ways more effective here than V (Fig. 3). However, only Cexpression effectively inhibited IFN-� activation due toGFP(�/�) infection (Fig. 3) or transfected poly(I-C) or pp-pRNAs (Fig. 5). Perhaps the strongest indication that the Cproteins are primarily responsible for countering the innateimmune response comes from experiments with SeV that can-not specifically express the C or V proteins. SeV-Cminus infec-tion not only cannot prevent the effects of transfectedpoly(I-C) or pppRNAs on IFN-� activation but also synergis-tically enhances these effects. SeV-Vminus infection, in contrast,behaves mostly like wt SeV infection and counteracts the ef-fects of transfected poly(I-C) or pppRNAs (Fig. 6).

Finally, we note that poly(I-C) (made with polynucleotidephosphorylase that generates 5� diphosphate ends and which istransfected into cells) and the presumed GFP dsRNA (that isdirectly generated in the cytoplasm via the viral transcriptaseand which contains capped 5� ends) both activate IFN-� viaRIG-I in MEFs. Thus, the ability of dsRNA to induce RIG-Isignaling does not depend on the manner in which it is intro-duced into this cell compartment, nor is it peculiar to thepresence of 5�-diphosphorylated ends that are not normallyfound in cells and could theoretically act as PAMPs. Moreover,in either case, the activation of IFN-� by these dsRNAs isinhibited by the SeV C protein and not V, presumably becausethis signaling passes through RIG-I and not mda-5. It appearsthat our MEFs contain insufficient amounts of mda-5 to senseSeV infection, as these MEFs respond well to the expression ofplasmid-derived mda-5 (Fig. 2B). This conclusion is also con-sistent with our finding that three different rubulavirus V pro-teins that are known to counteract poly(I-C)-induced mda-5signaling were unable to inhibit IFN-� activation in response toSeV-DI-H4 infection (data not shown). We expect that theSeV V protein will be more important in countering the innateimmune response in other cells in which mda-5 functions as aPAMP recognition receptor.

12236 STRAHLE ET AL. J. VIROL.

at Bibliotheque F

aculte Medecine G

eneve on Novem

ber 9, 2007 jvi.asm

.orgD

ownloaded from

Page 153: Sendai virus and the innate antiviral defence · protéines virales P et L forment la polymérase à ARN, qui est responsable de la transcription et de la réplication du virus. Le

ACKNOWLEDGMENTS

We thank J. Tschopp (Lausanne), T. Fujita (Kyoto), J. Perrault (SanDiego), B. Jacobs (Arizona), K. Conzelmann (Munich), and R. Randall(St. Andrews) for sharing materials.

This work was supported by the Swiss National Science Fund.

REFERENCES

1. Andrejeva, J., K. S. Childs, D. F. Young, T. S. Carlos, N. Stock, S. Goodbourn,and R. E. Randall. 2004. The V proteins of paramyxoviruses bind the IFN-inducible RNA helicase, mda-5, and inhibit its activation of the IFN-beta pro-moter. Proc. Natl. Acad. Sci. USA 101:17264–17269.

2. Blumberg, B. M., M. Leppert, and D. Kolakofsky. 1981. Interaction of VSVleader RNA and nucleocapsid protein may control VSV genome replication.Cell 23:837–845.

3. Bontron, S., C. Ucla, B. Mach, and V. Steimle. 1997. Efficient repression ofendogenous major histocompatibility complex class II expression throughdominant negative CIITA mutants isolated by a functional selection strategy.Mol. Cell. Biol. 17:4249–4258.

4. Brennan, C. M., and J. A. Steitz. 2001. HuR and mRNA stability. Cell. Mol.Life Sci. 58:266–277.

5. Childs, K., N. Stock, C. Ross, J. Andrejeva, L. Hilton, M. Skinner, R.Randall, and S. Goodbourn. 2006. mda-5, but not RIG-I, is a common targetfor paramyxovirus V proteins. Virology 359:190–200.

6. Garcia-Sastre, A., and C. A. Biron. 2006. Type 1 interferons and the virus-host relationship: a lesson in detente. Science 312:879–882.

7. Garcin, D., G. Taylor, K. Tanebayashi, R. Compans, and D. Kolakofsky.1998. The short Sendai virus leader region controls induction of pro-grammed cell death. Virology 243:340–353.

8. Hiscott, J., R. Lin, P. Nakhaei, and S. Paz. 2006. MasterCARD: a pricelesslink to innate immunity. Trends Mol. Med. 12:53–56.

9. Hiscott, J., T. L. Nguyen, M. Arguello, P. Nakhaei, and S. Paz. 2006. Ma-nipulation of the nuclear factor-kappaB pathway and the innate immuneresponse by viruses. Oncogene 25:6844–6867.

10. Ho, C. K., and S. Shuman. 1996. Mutational analysis of the vaccinia virus E3protein defines amino acid residues involved in E3 binding to double-stranded RNA. J. Virol. 70:2611–2614.

11. Hornung, V., J. Ellegast, S. Kim, K. Brzozka, A. Jung, H. Kato, H. Poeck, S.Akira, K. K. Conzelmann, M. Schlee, S. Endres, and G. Hartmann. 2006.5�-Triphosphate RNA is the ligand for RIG-I. Science 314:994–997.

12. Iseni, F., D. Garcin, M. Nishio, N. Kedersha, P. Anderson, and D. Kolakofsky.2002. Sendai virus trailer RNA binds TIAR, a cellular protein involved invirus-induced apoptosis. EMBO J. 21:5141–5150.

13. Kato, H., O. Takeuchi, S. Sato, M. Yoneyama, M. Yamamoto, K. Matsui,S. Uematsu, A. Jung, T. Kawai, K. J. Ishii, O. Yamaguchi, K. Otsu, T.Tsujimura, C. S. Koh, C. Reis e Sousa, Y. Matsuura, T. Fujita, and S. Akira.2006. Differential roles of MDA5 and RIG-I helicases in the recognition ofRNA viruses. Nature 441:101–105.

14. King, P., and S. Goodbourn. 1994. The beta-interferon promoter responds topriming through multiple independent regulatory elements. J. Biol. Chem.269:30609–30615.

15. Komatsu, T., K. Takeuchi, J. Yokoo, and B. Gotoh. 2004. C and V proteins

of Sendai virus target signaling pathways leading to IRF-3 activation for thenegative regulation of interferon-beta production. Virology 325:137–148.

16. Lamb, R. A., and D. Kolakofsky. 2001. Paramyxoviridae: the viruses and theirreplication, p. 1305–1340. In D. M. Knipe and P. M. Howley (ed.), Fieldsvirology, 4th ed., vol. 1. Lippincott Williams & Wilkins, Philadelphia, PA.

17. Leppert, M., and D. Kolakofsky. 1980. Effect of defective interfering parti-cles on plus- and minus-strand leader RNAs in vesicular stomatitis virus-infected cells. J. Virol. 35:704–709.

18. Leppert, M., L. Rittenhouse, J. Perrault, D. F. Summers, and D. Kolakofsky.1979. Plus and minus strand leader RNAs in negative strand virus-infectedcells. Cell 18:735–747.

19. Marq, J. B., A. Brini, D. Kolakofsky, and D. Garcin. 2007. Targeting of theSendai virus C protein to the plasma membrane via a peptide-only mem-brane anchor. J. Virol. 81:3187–3197.

20. Matsuoka, Y., J. Curran, T. Pelet, D. Kolakofsky, R. Ray, and R. W. Compans.1991. The P gene of human parainfluenza virus type 1 encodes P and C proteinsbut not a cysteine-rich V protein. J. Virol. 65:3406–3410.

21. Meylan, E., J. Curran, K. Hofmann, D. Moradpour, M. Binder, R. Barten-schlager, and J. Tschopp. 2005. Cardif is an adaptor protein in the RIG-Iantiviral pathway and is targeted by hepatitis C virus. Nature 437:1167–1172.

22. Mibayashi, M., L. Martinez-Sobrido, Y. M. Loo, W. B. Cardenas, M. Gale,Jr., and A. Garcia-Sastre. 2007. Inhibition of retinoic acid-inducible geneI-mediated induction of beta interferon by the NS1 protein of influenza Avirus. J. Virol. 81:514–524.

23. Milligan, J. F., and O. C. Uhlenbeck. 1989. Synthesis of small RNAs using T7RNA polymerase. Methods Enzymol. 180:51–62.

24. Ostertag, D., T. M. Hoblitzell-Ostertag, and J. Perrault. 2007. Overproduc-tion of double-stranded RNA in vesicular stomatitis virus-infected cells ac-tivates a constitutive cell-type-specific antiviral response. J. Virol. 81:503–513.

25. Pichlmair, A., O. Schulz, C. P. Tan, T. I. Naslund, P. Liljestrom, F. Weber,and C. Reis e Sousa. 2006. RIG-I-mediated antiviral responses to single-stranded RNA bearing 5�-phosphates. Science 314:997–1001.

26. Plumet, S., F. Herschke, J. M. Bourhis, H. Valentin, S. Longhi, and D.Gerlier. 2007. Cytosolic 5�-triphosphate ended viral leader transcript of mea-sles virus as activator of the RIG I-mediated interferon response. PLoS ONE2:e279.

27. Poole, E., B. He, R. A. Lamb, R. E. Randall, and S. Goodbourn. 2002. TheV proteins of simian virus 5 and other paramyxoviruses inhibit induction ofinterferon-beta. Virology 303:33–46.

28. Rothenfusser, S., N. Goutagny, G. DiPerna, M. Gong, B. G. Monks, A.Schoenemeyer, M. Yamamoto, S. Akira, and K. A. Fitzgerald. 2005. TheRNA helicase Lgp2 inhibits TLR-independent sensing of viral replication byretinoic acid-inducible gene-I. J. Immunol. 175:5260–5268.

29. Stark, G. R., I. M. Kerr, B. R. Williams, R. H. Silverman, and R. D.Schreiber. 1998. How cells respond to interferons. Annu. Rev. Biochem.67:227–264.

30. Strahle, L., D. Garcin, and D. Kolakofsky. 2006. Sendai virus defective-interfering genomes and the activation of interferon-beta. Virology 351:101–111.

31. Strahle, L., D. Garcin, P. Le Mercier, J. F. Schlaak, and D. Kolakofsky. 2003.Sendai virus targets inflammatory responses, as well as the interferon-in-duced antiviral state, in a multifaceted manner. J. Virol. 77:7903–7913.

VOL. 81, 2007 ACTIVATION OF IFN-� PROMOTER BY SeV INFECTION 12237

at Bibliotheque F

aculte Medecine G

eneve on Novem

ber 9, 2007 jvi.asm

.orgD

ownloaded from