Zurich Open Repository And Zora.uzh.ch Year: 2018

2y ago
36 Views
3 Downloads
3.30 MB
17 Pages
Last View : 16d ago
Last Download : 3m ago
Upload by : Giovanna Wyche
Transcription

Zurich Open Repository andArchiveUniversity of ZurichMain LibraryStrickhofstrasse 39CH-8057 Zurichwww.zora.uzh.chYear: 2018MxB is an interferon-induced restriction factor of human herpesvirusesCrameri, Michel; Bauer, Michael; Caduff, Nicole; Walker, Raphael; Steiner, Fiona; Franzoso, FrancescaD; Gujer, Cornelia; Boucke, Karin; Kucera, Talissa; Zbinden, Andrea; Münz, Christian; Fraefel, Cornel;Greber, Urs F; Pavlovic, JovanAbstract: The type I interferon (IFN) system plays an important role in controlling herpesvirus infections,but it is unclear which IFN-mediated effectors interfere with herpesvirus replication. Here we reportthat human myxovirus resistance protein B (MxB, also designated Mx2) is a potent human herpesvirusrestriction factor in the context of IFN. We demonstrate that ectopic MxB expression restricts a rangeof herpesviruses from the Alphaherpesvirinae and Gammaherpesvirinae, including herpes simplex virus1 and 2 (HSV-1 and HSV-2), and Kaposi’s sarcoma-associated herpesvirus (KSHV). MxB restrictionof HSV-1 and HSV-2 requires GTPase function, in contrast to restriction of lentiviruses. MxB inhibitsthe delivery of incoming HSV-1 DNA to the nucleus and the appearance of empty capsids, but not thecapsid delivery to the cytoplasm or tegument dissociation from the capsid. Our study identifies MxB asa potent pan-herpesvirus restriction factor which blocks the uncoating of viral DNA from the incomingviral capsid.DOI: https://doi.org/10.1038/s41467-018-04379-2Posted at the Zurich Open Repository and Archive, University of ZurichZORA URL: https://doi.org/10.5167/uzh-151624Published VersionOriginally published at:Crameri, Michel; Bauer, Michael; Caduff, Nicole; Walker, Raphael; Steiner, Fiona; Franzoso, FrancescaD; Gujer, Cornelia; Boucke, Karin; Kucera, Talissa; Zbinden, Andrea; Münz, Christian; Fraefel, Cornel; Greber, Urs F; Pavlovic, Jovan (2018). MxB is an interferon-induced restriction factor of humanherpesviruses. Nature Communications, 9(1):1980.DOI: https://doi.org/10.1038/s41467-018-04379-2

ARTICLEDOI: 10.1038/s41467-018-04379-2OPENMxB is an interferon-induced restriction factor ofhuman herpesviruses1234567890():,;Michel Crameri1,2, Michael Bauer 2,3, Nicole Caduff2,4, Raphael Walker1, Fiona Steiner1,2,Francesca D. Franzoso5,6, Cornelia Gujer4, Karin Boucke3, Talissa Kucera1, Andrea Zbinden1, Christian Münz4,Cornel Fraefel5, Urs F. Greber 3 & Jovan Pavlovic1The type I interferon (IFN) system plays an important role in controlling herpesvirus infections, but it is unclear which IFN-mediated effectors interfere with herpesvirus replication.Here we report that human myxovirus resistance protein B (MxB, also designated Mx2) is apotent human herpesvirus restriction factor in the context of IFN. We demonstrate thatectopic MxB expression restricts a range of herpesviruses from the Alphaherpesvirinae andGammaherpesvirinae, including herpes simplex virus 1 and 2 (HSV-1 and HSV-2), and Kaposi’ssarcoma-associated herpesvirus (KSHV). MxB restriction of HSV-1 and HSV-2 requiresGTPase function, in contrast to restriction of lentiviruses. MxB inhibits the delivery ofincoming HSV-1 DNA to the nucleus and the appearance of empty capsids, but not the capsiddelivery to the cytoplasm or tegument dissociation from the capsid. Our study identifies MxBas a potent pan-herpesvirus restriction factor which blocks the uncoating of viral DNA fromthe incoming viral capsid.1 Institute of Medical Virology, University of Zurich, Winterthurerstrasse 190, 8057 Zürich, Switzerland. 2 Life Science Zurich Graduate School,Winterthurerstrasse 190, 8057 Zürich, Switzerland. 3 Institute of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057 Zürich,Switzerland. 4 Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, 8057 Zürich, Switzerland. 5 Institute of Virology,University of Zurich, Winterthurerstrasse 266a, 8057 Zürich, Switzerland. 6 Department of Neurosurgery and Neuropathology, University Hospital Zurich,Frauenklinikstrasse 10, 8091 Zürich, Switzerland. These authors contributed equally: Cornel Fraefel, Urs F. Greber. Correspondence and requests for materialsshould be addressed to J.P. (email: pavlovic.jovan@virology.uzh.ch)NATURE COMMUNICATIONS (2018)9:1980 DOI: 10.1038/s41467-018-04379-2 www.nature.com/naturecommunications1

ARTICLENATURE COMMUNICATIONS DOI: 10.1038/s41467-018-04379-2Viruses are ubiquitous and affect all forms of life. Inhumans, livestock and plants, they cause disease byrecurrent or chronic infections, and have had a majorimpact on human evolution. For example, viruses influenced theemergence and maintenance of elaborate host defence systems,such as innate and adaptive immunity1–4. In the course of aninfection, viruses elicit danger signals, and trigger cell-basedinnate immunity owing to viral components, including virions,viral proteins and nucleic acids, and cell damage5,6. A majorbranch of mammalian innate immunity against viruses is theinterferon (IFN) system7. This system comprises type I, II and IIIIFN, and involves hundreds of IFN-stimulated genes (ISGs). ISGexpression can lead to the so-called restriction factors whichdirectly inhibit a process essential to the production of virusprogeny, or ISG expression can act indirectly by building up anelaborate antiviral network8.Human myxovirus resistance protein B (MxB) has recentlybeen identified as an IFN-induced restriction factor of humanimmunodeficiency virus type 1 (HIV-1) and other primatelentiviruses9–11. MxB is a member of the dynamin-like largeGTPases and is closely related to MxA, an ISG product withbroad antiviral activity against a multitude of RNA viruses andsome DNA viruses but not against herpes simplex virus 1 (HSV1) or HIV-19,12,13. At the subcellular level, MxB can be found inthe cytoplasm, on the cytoplasmic face of nuclear membranesand in the nucleus10,12,14. Under certain conditions, MxB hasbeen shown to bind to HIV-1 capsid-like recombinant assemblies via its amino-terminal domain15–17. This has been taken asevidence that MxB prevents uncoating of proviral DNA fromincoming HIV-1 capsids. The antivirally active form of MxB islikely an antiparallel dimer and the formation of higher-orderoligomeric structures does not appear to be required to exert fullantiviral function15,18. However, binding of MxB to HIV-1capsids is clearly not sufficient for the observed antiviral activity,since MxB retains binding capacity to capsid mutants that areresistant to the antiviral effect of MxB15. Intriguingly, the GTPhydrolysing function of MxB appears to be dispensable for theantiviral activity against HIV-19,10,19, which stands in contrastto the antiviral activity of MxA that requires GTPasefunction13,20. Antiviral activity of MxB is likely not restricted toHIV-1, as indicated by a recent analysis of MX2 (the geneencoding MxB) evolution21. In their study, Mitchell et al.21found that amino acid residues, which are important for antilentivirus activity, have not evolved under diversifying selectionin primates, indicating that MxB functions beyond lentivirusrestriction. We therefore evaluated whether other human viruses, which replicate in the nucleus, are restricted by MxB andfocused on HSV-1 and human adenovirus C serotype 5 (HAdVC5).HAdV-C5 is a widespread human pathogen of the respiratorytracts and is life-threatening in immunosuppressed individuals22.Its replication is suppressed by IFN and can lead to persistentinfection with low levels of virus production23. HAdV-C5 entersepithelial cells by clathrin-dependent and clathrin-independent,dynamin-2-dependent endocytosis24. It escapes from nonacidified early endosomes, is transported by dynein-dependentand microtubule-dependent transport to the nucleus, binds anduncoats at the nuclear pore complex (NPC) and imports adouble-stranded linear DNA genome in complex with viralproteins into the nucleus25–30. Adenovirus intercepts IFNrestriction by its immediate-early protein E1A, which inhibits theE3 ubiquitin ligase hBre1, and thereby results in transcriptionalsuppression of ISGs31,32.Human herpesviruses are prevalent in humans, and causedisease ranging from subclinical manifestations to encephalitisand cancer, particularly in immunocompromised individuals.2NATURE COMMUNICATIONS (2018)9:1980Members of each subfamily Alphaherpesvirinae, Betaherpesvirinae and Gammaherpesvirinae establish lifelong persistence bylatent infections. The virions contain a double-stranded DNAgenome wrapped in an icosahedral capsid surrounded by a proteinaceous layer referred to as tegument, and a lipid envelope thatharbours the glycoproteins required for entry into host cells.Entry can either occur through fusion of the viral envelope withthe plasma membrane or by endocytosis33,34. Upon entry, thetegument gradually dissociates and the capsid is transportedalong microtubules to the nuclear envelope. Injection of the viralgenomic DNA into the nucleus occurs at the NPC in a processthat involves tegument and capsid proteins as well as cellularnuclear import factors and nucleoporins (reviewed in Refs. 35–38).Nuclear entry is followed by transcription of immediate-earlygenes including RL2, encoding the infected cell protein 0 (ICP0),which in turn drives HSV-1 early and late gene expression and isrequired for efficient replication.Incoming herpesviruses trigger a pronounced innate immuneresponse including the production of type I IFN39,40. Althoughthe recognition of herpesviruses by pattern recognition receptorsmediating IFN synthesis has been studied extensively (reviewed inRefs. 41–43), little is known about the IFN-induced effectormechanisms that result in the inhibition of virus replication.Several ISGs with antiviral function such as the dsRNAdependent protein kinase R (PKR), viperin, tetherin, zinc-fingerantiviral protein and 2′–5′ oligoadenylate synthetase (OAS) havebeen implicated in restricting herpesviruses, but their contribution remains to be elucidated (reviewed in Refs. 44,45).Here we report that MxB efficiently blocks herpesvirus infection at an early post-entry step, which precludes the nucleartranslocation of the incoming viral DNA.ResultsInterferon-induced MxB inhibits HSV-1 replication. To assessthe impact of MxB expression on HSV-1 infection, we transfectedT98G glioblastoma cells known to express high levels of MxB inresponse to type I IFN46 (see also Supplementary Fig. 1a) withnon-targeting control small interfering RNA (siRNA) (siNT) andMX2-targeting siRNAs (siMxB 3'-untranslated region (UTR),siMxB #1). MxB was expressed in IFN-α2-treated cells transfectedwith control siRNA, where it accumulated at the nuclear envelopeas well as in the cytoplasm, as previously described10,47. Transfection with MX2-specific siRNAs prevented the accumulation ofMxB to detectable levels (Fig. 1a–c). Infection of siNT-transfectedT98G cells with two distinct HSV-1 strains (MacIntyre and F)yielded viral titres of approximately 5 107 TCID50/ml. In orderto exclude non-specific effects of siRNA transfection, relativetitres are shown (Fig. 1b, c). Pretreatment of T98G cells with 500IU/ml IFN-α2 reduced viral titres of both strains approximately1000-fold. Remarkably, in T98G cells transfected with siRNAsspecific for MX2, the observed IFN-mediated inhibition of HSV-1infection was partially released, resulting in approximately 10fold increased viral titres (Fig. 1b, c). Increased titres in cellstransfected with MX2-specific siRNAs were reflected in markedlyenhanced levels of HSV-1 virion protein 16 (VP16) when compared to cells transfected with control siRNA (Fig. 1b, c).We next monitored the influence of MxB expression on HSV-1growth in an infection kinetics experiment by employing a livecell fluorescence imaging system and a recombinant HSV-1 strainencoding green fluorescent protein (GFP, strain C1248). Weinfected T98G cells with C12 virus following transfection withnon-targeting or MX2-specific siRNAs and stimulation with IFNα2. In the absence of IFN-α2 stimulation, the C12 strain grewrapidly, reaching maximal GFP signal (40–56 arbitrary units (a.u.)) between 60 and 72 h post infection (p.i.), irrespective of the DOI: 10.1038/s41467-018-04379-2 www.nature.com/naturecommunications

ARTICLENATURE COMMUNICATIONS DOI: 10.1038/s41467-018-04379-2was gradually released and GFP signals reached between 27% and45% of the IFN-α2-untreated controls (Fig. 1d). Hence, MxBcontributes significantly to the IFN-mediated inhibition ofHSV-1 infection. As a control, we performed an analogoussiRNA used (Fig. 1d and Supplementary Fig. 1b). By contrast, inIFN-α2-stimulated cells transfected with non-targeting siRNA,HSV-1 grew slowly, reaching approximately 6 a.u. at 72 h p.i. Incells pretreated with MX2-specific siRNAs, however, the blockadNormalised GFP signal intensity (a.u.)–IFN–α2MxBHSV-1 C12siMxB UTRsiNTMxBDNA504030siNTsiMxB UTRsiMxB #1–IFN–α2siNTsiMxB UTRsiMxB #1 IFN–α2Mock infection20100081624324048h post infection IFN–α25664**** **** IFN–α2siNTRelative area under the curve1.0siMxB UTR72 IFN–α20.80.60.40.20.0siNT siMxB siMxB siNT siMxB siMxB MockUTR#1UTR#1bcHSV-1 MacIntyreHSV-1 F**** ****1001001010Relative viral titre (%)Relative viral titre (%)**** ****110.010.0175100 IFN–α20.10.1kDa 100–IFN–α2siNT siMxB siMxB siNT siMxB siMxB#1UTRUTR#1 IFN-α2MxB0.00.00.0100.00.00.0% .06.824.515.9% MxAVP1635NATURE COMMUNICATIONS (2018)9:1980% VP16GAPDHkDa 100siNT siMxB siMxB siNT siMxB siMxB#1UTRUTR#1 IFN-α2MxB751000.00.00.0100.00.00.0% MxB75750.00.00.0100.0104.295.1% MxA5540100.0100.0100.03.720.937.7MxAVP1635 DOI: 10.1038/s41467-018-04379-2 www.nature.com/naturecommunications% VP16GAPDH3

ARTICLENATURE COMMUNICATIONS DOI: 10.1038/s41467-018-04379-2experiment using two siRNAs targeting the coding region of PKR,an ISG with known anti-HSV-1 activity44. The observed PKRsiRNA-dependent release of HSV-1 restriction resulted in22–26% of viral growth compared to the untreated controls(Supplementary Figs. 1c, d and 7d). We therefore concluded thatin this setting, the potential of MxB to restrict HSV-1 iscomparable to PKR.Overexpression of MxB inhibits herpesvirus growth. We nexttested whether ectopic expression of MxB in the absence of IFNα2 stimulation would restrict HSV-1 and other members of theHerpesviridae family. For this purpose, we generated severalstably transduced A549 and Vero cell clones constitutivelyexpressing glutathione S-transferase M1 (GST, control) or MxBby means of lentiviral vectors. In MxB-expressing A549 cells,ectopic MxB was found at the nuclear membrane and in apunctate pattern in the cytoplasm (Fig. 2a). A similar phenotypewas observed in Vero cells stably overexpressing MxB (Supplementary Fig. 2c). Transfection with siRNAs against the codingregion of MX2 (siMxB #1, siMxB #2) reduced the level of MxBexpression to about 30% or less of the control (siNT), whereas ansiRNA against the 3′-UTR of the endogenous MX2 mRNA(siMxB UTR) not present in the overexpressed gene had noinhibitory effect (Fig. 2b and Supplementary Fig. 2d). Controlinfection of A549-GST and A549-MxB cell clones with the HIV1-based luciferase reporter virus NL-Luc49 and influenza A virus(IAV) confirmed that MxB restricts replication of HIV-1 but notIAV9–12 (Supplementary Fig. 5b and Fig. 2c). Vesicular stomatitisvirus (VSV) was previously reported to be inhibited by MxB50. Inour cell culture system, we observed a fivefold reduction of VSVinfection in cells expressing MxB as compared to GST (Fig. 2d).Further, we tested whether MxB would inhibit growth of humanHAdV-C5. We observed a 2.4-fold reduction of HAdV-C5 titresin multi-round infections of A549-MxB cells (Fig. 2e), and aninhibition of GFP expression in single-round infections usingGFP or late viral protein expression as a readout (SupplementaryFig. 5c). However, transfection of siRNA against MX2 did notrestore HAdV-C5 reporter gene expression, suggesting that theeffects of MxB on HAdV-C5 infection were indirect or unspecific(Supplementary Fig. 5d). In contrast, we found that MxBexpression had a pronounced negative effect on several representatives of the Alphaherpesvirinae subfamily. In our experiments, we used stocks of three different HSV-1 strains(MacIntyre, F or C12) grown under identical conditions. Whenwe challenged A549 cells expressing MxB with these three strains,viral titres were reduced about 75-fold as compared to A549-GSTcells (Fig. 2f–h). Similarly, the commonly used HSV-2 strain Gshowed a reduction of approximately 50-fold (Fig. 2i). Transfection of an siRNA against the 3′-UTR of endogenous MX2mRNA had no effect on HSV growth. On the contrary, transfection with siRNAs against the coding region of MX2 largelyrestored titres of all tested HSV-1 strains and HSV-2 (Fig. 2f–i).Cell lysates prepared at the time of virus supernatant collectionrevealed that HSV late protein VP16 was expressed at considerably lower levels in A549-MxB cells compared to controlcells, yet VP16 expression was fully restored upon MX2 silencing(Supplementary Fig. 7b). Importantly, siRNA transfection did notimpact herpesvirus titres or VP16 expression in A549-GST cells,and the effect of the individual siRNAs on cell viability in thedifferent cell lines was negligible, thereby excluding unspecificeffects of the different siRNAs used in this system (Supplementary Fig. 7a). In order to rule out possible effects of individual cellclones on HSV titres, two additional, independently generatedA549-MxB cell clones (MxB 2 and MxB 3) were tested forrestriction of HSV-1 strain C12 and HSV-2. Both additionalA549-MxB clones inhibited HSV-1 and HSV-2 to the same extentas the original A549-MxB clone, and this effect was again revertedwith MX2-specific siRNAs (Supplementary Fig. 2a, b). The factthat the rescue of the HSV titres was not always complete(Fig. 2f–i, Supplementary Fig. 2b) may be due to incompletesilencing of MX2 expression in A549-MxB cells at the time ofinfection (Fig. 2b and Supplementary Fig. 7e). In Vero cells, theMxB-mediated inhibition of HSV-1 replication was readilymeasurable but less pronounced as compared to A549 cells(Supplementary Fig. 2e). We then tested whether Kaposi’ssarcoma-associated herpesvirus (KSHV), a representative of theGammaherpesvirinae subfamily, was restricted by MxB in A549cells. Here, we made use of a recombinant KSHV that expressesGFP upon virus entry into the host cell nucleus and red fluorescent protein (RFP) upon lytic reactivation51. While KSHV wasdetected in 79–88% of A549 cells expressing GST, only 27–31% ofA549 cells expressing MxB showed a detectable level of KSHVinfection (Fig. 3a). Pretreatment of A549-MxB cells with MX2specific siRNAs siMxB #1 and siMxB #2 resulted in a strongrescue of KSHV infectivity (58–62% in A549-MxB cells comparedto 70–87% in A549-GST cells, Fig. 3a). Moreover, we employedimmunofluorescence assays to assess the accumulation of latencyassociated nuclear antigen (LANA), one of the three main KSHVproteins expressed during persistent latent infection. EndogenousLANA staining is widely used to detect KSHV infection, as KSHVestablishes latency as a default program in various systemsin vitro and in vivo, including epithelial cells51–54. In A549-GSTFig. 1 Interferon-induced MxB inhibits HSV-1 replication. a T98G human glioblastoma cells were transfected with non-targeting siRNA (siNT) or siRNAtargeting endogenous MX2 (siMxB UTR). At 30 h post transfection, cells were mock-stimulated or stimulated with human IFN-α2 (1000 IU/ml) for 18 h.MxB protein expression and intracellular localisation was assessed by immunostaining. Nuclei were stained with Hoechst 33342. Scale bar, 20 µm. b, cT98G cells were transfected with non-targeting siRNA (siNT) or two different siRNAs targeting endogenous MX2 (siMxB UTR, siMxB #1). At 30 h posttransfection, cells were mock-stimulated or stimulated with human IFN-α2 (500 IU/ml). At 48 h post transfection, cells were infected with HSV-1 strainMacIntyre (b) or F (c

Zurich Open Repository and Archive University of Zurich Main Library Strickhofstrasse 39 CH-8057 Zurich www.zora.

Related Documents:

11 University of Zurich , Institute of Molecular Life Sciences, Neuroscience Center Zurich and Center for Integrative Human Physiology, Winterthurerstrasse 190, CH-8057 Zurich , Switzerland 22 University Hospital Zurich , Department of Neurology, Frauenklinikstrass

William Miller, Zora Hurston and the Chinaberry Tree. New York: Lee & Low Books, 1994. Recounts a story from Hurston’s childhood. Grades K-4. A.P. Porter, Jump at de Sun: The Story of Zora Neale Hurston. Minneapolis: Carolrhoda Books, 1992. Describes the life and times of Hurston. Grades 4-6. Della Yannuzzi, Zora Neale Hurston: Southern .

Zurich Management Liability Insurance About Zurich The insurer of this product is Zurich Australian Insurance Limited (ZAIL), ABN 13 000 296 640, AFS Licence Number 232507, a subsidiary of Zurich Financial Services Australia Limited (ZFSA). In this document, ZAIL may also be expressed as 'Zurich', 'we', 'us' or 'our'.

solaris repository description Local\ copy\ of\ the\ Oracle\ Solaris\ 11.1\ repository solaris repository legal-uris solaris repository mirrors solaris repository name Oracle\ Solaris\ 11.1\ Package\ Repository solaris repository origins solaris repository

Creating, Restoring, and Configuring the Informatica Repository 78 Starting the Informatica Repository Server 78 Creating or Restoring the Informatica Repository 79 Dropping the Informatica Repository (Optional) 81 Registering the Informatica Repository Server in Repository Server Administration Console 81 Pointing to the Informatica Repository 82

About Zurich The insurer of this product is Zurich Australian Insurance Limited (ZAIL), ABN 13 000 296 640, AFS Licence Number 232507. In this document, ZAIL may also be expressed as 'Zurich', 'we', 'us' or 'our'. ZAIL is part of the Zurich Insurance Group, a leading multi-line insurer that serves its customers in global and local

Korea Sin Moo Hapkido 8038 Zürich www.hapkido.ch KSC Kantonsschulsportclub Wiedikon, Bas 8003 Zürich www.kscw.ch KTV Albisrieden 8048 Zürich Kunstturnerinnen Zürich - Oerlikon 8051 Zürich www.kutu.ch Küsnachter Schwimmverein 8700 Küsnacht ZH www.kuesnachtersv.ch Kyokushinkai Karat

Boris Fausto Frontmatter Moreinformation. xiv Preface the tendency to consider Brazilian history as an evolutionary trend characterized by constant progress. This is a simplistic point of view that events in recent years have belied. On the other hand, I have also rejected the point of view that emphasizes inertia – that suggests, for example, that problems caused by political patronage, by .