<<

336

Viral invasion and host defense: strategies and counter-strategies James C Carrington∗ and Steven A Whitham

The outcome of infection of plants by is determined pendent RNA polymerase activity and other associated by the net effects of compatibility functions and defense activities (e.g. helicase, capping, priming) in close asso- responses. Recent advances reveal that viruses have the ciation with cytoplasmic surfaces of membranes [2]. The capacity to modulate host compatibility and defense functions docking of replication complexes to membranes appears by a variety of mechanisms. to be a point of intimate interaction between and host cell. In several cases, complexes associate specifically with endoplasmic reticulum (ER)-derived membranes Addresses or vesicles through specific membrane-binding functions Institute of Biological Chemistry, Washington State University, Pullman, WA 99164-6340, USA of replication proteins [3,4]. The basis for membrane ∗e-mail: [email protected] specificity, however, remains obscure. The participation of cellular factors, such as host proteins or protein complexes, Current Opinion in Plant Biology 1998, 1:336–341 has been a point of speculation for many years, with http://biomednet.com/elecref/1369526600100336 only limited direct evidence to support functional roles  Current Biology Ltd ISSN 1369-5266 of host factors [2]. This may change in the near future through the clever use of yeast as an adopted host for Abbreviations brome mosaic virus (BMV) to analyze RNA replication; BMV brome mosaic virus CaMV cauliflower mosaic virus Paul Ahlquist and colleagues have developed selectable ER endoplasmic reticulum and counter-selectable BMV RNA-based replicons for HC-Pro helper component-proteinase yeast [5•,6,7]. For example, BMV replicons expressing HR hypersensitive response URA3 allow ura3 yeast to grow in the absence of uracil mab maintenance of BMV functions MP movement protein and thus provide a positive selection for cells with PSBMV pea seedborne mosaic active BMV replication. Conversely, URA3 confers a lethal PVX potato virus X phenotype in the presence of 5-fluoroortic acid, and thus TBRV tomato black ring provides a selection for yeast cells that lose the ability TEV tobacco etch virus TMV tobacco mosaic virus to support BMV replication. The characteristics of BMV VIGS viral induced gene silencing replicon amplification are remarkably similar in yeast cells and plant cells, providing a rationale for applying yeast genetics to identify host factors that influence Introduction replication. The MAB1, MAB2 and MAB3 (Maintenance of When discussing susceptibility or resistance of plants to BMV functions) loci contribute distinct functions to BMV viruses, it is convenient to consider ‘susceptibility factors’ genomic RNA replication, subgenomic RNA synthesis and ‘resistance factors’ as separate entities governing and accumulation of BMV-encoded replication proteins •• infection. It can be argued, however, that the distinction [8 ]. Recessive mutations at each locus condition a between susceptibility and resistance to viruses may not temperature-sensitive yeast growth phenotype, suggesting be so straightforward. We are particularly intrigued by that the wild-type gene products are necessary for the the possibility that viruses can be active promoters of normal cellular function. Dissection of the roles of the susceptibility or suppressors of defense. In this review, MAB gene products in viral RNA replication will represent we will consider selected aspects of susceptibility, such a significant milestone in understanding compatibility as virus–host interactions that govern virus genome during virus–host interactions. replication and intercellular movement, and a limited discussion of defense responses. We will also summarize DNA-containing viruses, such as the geminiviruses, repli- recent findings, as well as our views, on how viruses cate in the host plant nucleus where the cellular DNA may modulate host functions to ensure their own efficient replication apparatus is employed. The geminiviruses replication and movement. use host DNA polymerase and associated factors in conjunction with a virus-encoded replication protein Virus–host interactions: susceptibility factors (termed RepA or AL1). The RepA/AL1 protein provides A plant host is considered fully susceptible to a given virus several key activities that integrate viral and host functions if the virus can successfully complete three processes: controlling early gene expression, late gene expression and genome replication, cell-to-cell movement (local) and DNA replication [9,10]. This protein binds to the stem long-distance (vasculature-dependent) movement [1]. For structure at the replication origin and nicks the DNA prior most RNA-genome viruses, RNA replication is catalyzed to initiation of rolling-circle DNA replication [9,11–13]. by a core set of virus-encoded enzymes with RNA-de- Like many animal-infecting DNA viruses that use the Viral invasion and host defense: strategies and counter-strategies Carrington and Whitham 337

cellular DNA replication apparatus, the geminiviruses face therefore, must overcome the lack of DNA replication a serious problem; they infect terminally differentiated, factors in G0 cells. Two significant breakthroughs have resting state (G0) cells, but these cells lack factors required shed light on possible mechanisms whereby geminiviruses for DNA replication ([14], and references therein). As solve this problem. First, geminiviruses induce expression detailed below, geminiviruses likely solve this problem of at least some of the components necessary for DNA through modulating factors that normally control the cell synthesis upon infection. Proliferating cell nuclear antigen cycle. (PCNA), a DNA polymerase accessory factor normally found only in S-phase cells, is induced in quiescent Virus-encoded susceptibility factors controlling cell-to- mesophyll cells by infection with tomato golden mosaic cell and long-distance movement are called ‘movement geminivirus (TGMV) [14]. Using AL1 transgenic plants, proteins’ (MPs) and have been reviewed extensively induction of PCNA was shown to require only the TGMV in recent years [1,15,16]. Movement proteins facilitate AL1 gene. Second, the AL1/RepA protein physically movement through interactions with the viral genome or interacts with host-encoded retinoblastoma-like tumor assembled virus particle, intracellular transport pathways suppressor proteins (pRbs) [28••,29–31]. Wheat dwarf and structures, and plasmodesmata. For geminiviruses, geminivirus mutants with substitutions affecting the prob- two MPs are required for cell-to-cell movement. One able pRb-like protein binding site of Rep exhibit defects (BR1) is necessary to transport viral ssDNA genomes in viral DNA synthesis [29], supporting the concept between the nucleus and the cytoplasm [17,18]. The that this interaction is necessary. The well-characterized BR1 protein, therefore, is a nuclear shuttle protein with mammalian pRB protein serves as key G1 checkpoint nuclear import and export signals. The other (BL1) regulator, preventing completion of G1 and entry into interacts with ER-derived membranes and plasmodesmata S-phase [32]. Phosphorylation by cyclin-dependent ki- to facilitate transit of viral DNA to adjacent plant cells, nases relieves the cell cycle-inhibitory activity of pRb and possibly by modifying the plasmodesmata structure and allows progression into S-phase [32]. Plants, like mammals, transport properties [19,20]. Tobacco mosaic virus (TMV) likely use one or more pRb-like proteins as checkpoint MP interacts with viral RNA and modifies plasmodesmata inhibitors of the cell cycle. It is reasonable to suggest, [21–23]. The TMV MP was shown to interact with therefore, that geminivirus Rep protein interacts with, microtubules in infected or transfected cells [24,25], and it and inactivates (or at least diverts), pRb-like protein in was proposed that this might enable intracellular transport infected cells. This would relieve the transcriptional block of movement complexes from sites of genome replication to production of S-phase-specific mRNAs and provide to plasmodesmata. Recent data, however, suggest that a pool of enzymes and factors necessary for viral DNA the MP–microtubule association may occur relatively late replication. Thus, plant geminiviruses join the ranks in the infection process, possibly as part of a pathway of animal DNA tumor-inducing viruses, such as SV40 to dissipate high local concentrations or to degrade MP and adenovirus, which encode proteins that affect the [26]. Early during the infection process, TMV MP, like cell cycling apparatus through interactions with tumor the geminivirus BL1 [19], associates with ER-derived suppressor proteins [33]. membranes and plasmodesmata [26]. Might the association between the ER and diverse MPs from different viruses A novel defense response against viruses be telling us something important? Recall that plasmodes- Defense responses involving dominant resistance (R) mata contain an extension of the ER, the desmotubule, genes, hypersensitive cell death and induction of systemic which is threaded through the central channel [16,27]. acquired resistance have been reviewed nicely by others Quite possibly, transport complex movement through [34•,35•,36,37] and will not be covered here. Rather, we plasmodesmata requires specific interactions first with will focus on a novel defense response — post-transcrip- cortical ER and then with the desmotubule component in tional gene silencing — that has only recently been linked the channel. to natural forms of virus resistance.

Induction of host compatibility factors by Post-transcriptional gene silencing is a remarkable pro- viruses cess that has been reviewed extensively [38–41]. The Do viruses have the ability to promote their own infections hallmarks of post-transcriptional silencing of nuclear-de- by stimulation of host susceptibility factors or structures rived sequences include, among other features, high needed for genome replication, genome expression or transcription levels of the silenced gene but relatively low movement? Given that the number of cellular factors steady-state cytoplasmic levels of the transcript. Silencing shown to play definitive roles in promoting virus infection occurs through a homology-dependent mechanism and is small, there are relatively few examples to ponder. The functions in trans to suppress identical or closely related geminiviruses, however, provide an excellent case to argue sequences. Evidence suggests that silencing is due that viruses have the potential to modulate susceptibility. to activation of a sequence-specific RNA degradation apparatus, perhaps by a mechanism involving synthesis of As mentioned above, geminiviruses are dependent on complementary nucleic acids. Silencing of a nuclear-de- the host DNA replication apparatus for replication, and, rived sequence can be triggered by either nuclear genes 338 Plant–microbe interactions

or by cytoplasmic, replicating nucleic acids containing transient depletion of many, but not all, classes of host homologous sequences. Furthermore, a silenced state in cell mRNAs in pea embryos [50,51]. The host mRNA a plant can be generated systemically through transport of modulation occurs specifically in cells supporting virus a mobile signal [42••,43••]. replication, and exhibits some of the characteristics of the heat shock response, such as induction of HSP70 and Whereas it has been known for some time that viral polyubiquitin mRNA expression. Although the bases for sequences can induce silencing of an endogenous nuclear suppression and subsequent recovery of transcript levels gene or a transgene [44,45], it was only recently demon- are not clear, the consequences of inhibition of host cell strated that viruses can trigger silencing in the apparent mRNA accumulation on induction of defense responses absence of homologous nuclear sequences. Infection are potentially far-reaching. As most types of known of Nicotiana clevelandii by tomato blackring nepovirus induced defense responses require host gene expression (TBRV) strain W22 results in an initial symptomatic phase [34•], inhibition of mRNA accumulation or translation may in which the virus moves systemically, followed by a recov- severely limit the extent or timing of a defense program. ery state in which new tissue developing post-inoculation is asymptomatic and largely devoid of virus [46••]. Further, Additional lines of evidence suggest that can silencing of the viral sequence is active in the recovered modulate host defense responses, and that modulation tissue, conferring effective resistance against W22 and a can condition enhanced accumulation and movement of closely related strain. Prolonged infection of Brassica napus potyviruses as well as heterologous viruses. Typically, by cauliflower mosaic virus (CaMV), a pararetrovirus, also in single-infections by a non-potyvirus, virus genome induces post-transcriptional gene silencing independent of amplification at the single-cell level shuts off after an homologous transgenes [47••]. These findings are highly exponential amplification phase within 12–24 hours of significant as they indicate that homology-dependent gene inoculation [52]. Tobacco etch virus (TEV), in contrast, silencing may be a general antiviral response. The logic accumulates at a slower initial rate but for considerably for such a response is impeccable: extreme resistance can longer periods of time (e.g., 72 hours post-inoculation; be elicited after infection by a broad range of infecting [53]). TEV mutants with defects in helper component- viruses using a general, adaptive silencing apparatus. An proteinase (HC-Pro) exhibit a premature amplification important unresolved issue, however, is the extent to shut-off phenotype by 24 hours post-inoculation and which viruses, other than those mentioned, elicit silencing are restricted in long-distance movement [54•]. These upon infection. Most compatible virus–host combinations observations suggest that wild-type HC-Pro may function do not show the recovery phenotypes as do TBRV and to suppress one or more induced defense responses. CaMV. It is possible, however, that silencing is triggered Significantly, when heterologous viruses are co-inoculated transiently to limit the extent of virus replication in with TEV or other potyviruses, or when heterologous infected tissue, thus lessening the extent of disease, viruses infect plants or cells in the presence of functional but not in time to limit spread systemically. Silencing HC-Pro, the heterologous viruses exhibit an enhanced as a general antivirus response in both compatible and genome accumulation phenotype [55,56,57•]. In the case incompatible virus–host combinations deserves careful of infections of potato virus X (PVX) expressing HC-Pro attention. or an HC-Pro polyprotein precursor, the enhancement of PVX is actually the result of continued amplification Do viruses fight back? beyond the point at which amplification normally shuts off If one examines the way animal viruses deal with innate [57•]. A plausible explanation for all of these results is that and adaptive immune system defenses, it becomes clear potyviruses, through a key activity of HC-Pro, suppress that these agents are masters of the counter-defensive induction of host defense responses that would normally strategy. For example, poxviruses encode several types result in early amplification shut-off and restricted ac- of cytokine receptor mimic proteins [48]. These atten- cumulation (Figure 1). The antiviral defense responses uate the inflammatory and immune responses normally could have broad-spectrum activity against a range of triggered by virus-infected cells. Also, numerous animal diverse viruses, or could be induced to have specificity viruses encode inhibitors of apoptosis proteins (IAPs), against many different viruses. The mechanistic details preventing normal immune signaling that occurs through of this system are not yet resolved, although recently activation of cell death pathways [49]. obtained evidence is consistent with HC-Pro mediating a suppression of homology-dependent gene silencing (KD Do plant viruses deploy counter-defensive strategies? Kasschau and JC Carrington, unpublished data). Recent findings offer suggestions that certain plant viruses are able to interdict defense mechanisms or alter host Finally, the potential influence of viruses on signal cell metabolism for their own benefit. In a series of transduction pathways controlling a wide range of cellular elegant experiments examining host cell transcript and activities is just now being realized. Some of the best protein levels in situ in tissue across an advancing ongoing work in this area deals with the virulence-attenu- infection front, Andy Maule and colleagues demonstrated ating of the blight fungus, Cryphonectria that pea seedborne mosaic potyvirus (PSBMV) induces parasitica. Donald Nuss and co-workers have shown that Viral invasion and host defense: strategies and counter-strategies Carrington and Whitham 339

Figure 1

ER HC-Pro

Accumulation of Local VIGS or Systemic acquired viral RNAs other induced silencing or other defense responses systemic responses?

Viral RNA Amplification shut-off replication complex or viral RNA degradation Current Opinion in Plant Biology

Modulation of antiviral defense responses by potyvirus TEV HC-Pro. The effects of HC-Pro on accumulation and movement of TEV and heterologous viruses [54•,55,56,57•] suggest that HC-Pro protein may suppress local and systemic defense responses. The defense responses affected by HC-Pro may include virus-induced gene silencing (VIGS), induced (non-HR) reactions, systemic acquired silencing or other systemic responses. The dotted line indicates that there is currently no direct experimental evidence suggesting that HC-Pro does interfere with a systemic host defense response. The viral replication complex is comprised of virus-encoded and possibly host-encoded proteins (represented by gray balls), which synthesize viral RNAs in association with endoplasmic reticulum (ER) derived membranes. hypovirus infection suppresses a G-protein signaling cas- (HR- and non-HR-dependent) virus–host combinations cade through down-regulation of the G-protein a-subunit, will reveal loci necessary for recognition and response CPG-1 [58–60]. This G-protein signaling pathway nega- signaling during defense. Clearly, the successes in recovery tively regulates cAMP levels. Significantly, virus infection, of highly informative mutants with defects in the sig- transgenic co-suppression of CPG-1, and drug-induced naling pathways for defense against bacterial and fungal elevation of cAMP levels each result in effects consistently pathogens sets an encouraging precedence [61]. Second, associated with hypovirulence of the fungus, including loss-of-susceptibility mutants using normally compatible reduced asexual sporulation, reduced pigmentation and virus–host combinations will reveal loci necessary for altered colony morphology. The ramifications of induction genome replication and movement. Besides the yeast mab of a hypovirulent state by hypoviruses are not immediately mutants with defects in supporting BMV RNA replication, clear, although it is possible that an ecological advantage an Arabidopsis mutant (tom1) has been recovered with is afforded. As sexual reproduction of hypovirus-containing a defect in supporting TMV replication [62]. Recovery C. parasitica strains is suppressed, genetic recombination in of large numbers of susceptibility mutants, however, a hypovirulent population should be limited. This would may require particular finesse, as the cellular factors promote proliferation of uniform anastomosis compatibility and structure needed to support virus infection almost groups, which in turn would promote distribution of the certainly are critical for normal cellular function. We virus by its normal transmission route — hyphal fusion. believe that the necessary mutants are attainable, however, While the influence of hypovirus infection on signaling in through development of novel virus–host genetic systems C. parasitica is now becoming clear, the effects of plant that are amenable to high-throughput selections and viruses on G-protein or other signaling cascades has yet to screens using mass inoculation procedures and selectable be established. The tools are available, however, to address or screenable viruses. this issue in detail. Acknowledgements Conclusions and future prospects Work in the author’s laboratory was supported by grants from the National Institute of Health (GM18529 to SA Whitham; AI27832 to JC Carrington) Although we have witnessed some significant develop- and the United States Department of Agriculture (95-37303-1867). ments recently, we suggest that the most exciting days in the quest to understand compatibility and incompatibility References and recommended reading functions governing virus infection are still ahead of us. Papers of particular interest, published within the annual period of review, A major limitation at this point is the paucity of useful have been highlighted as: host mutants that exhibit altered susceptibility phenotypes • of special interest and that can serve as starting points for isolation of •• of outstanding interest relevant genes. Two types of host mutants should be pursued aggressively. First, gain-of-susceptibility, or 1. Carrington JC, Kasschau KD, Mahajan SK, Schaad MC: Cell-to- cell and long-distance movement of viruses in plants. Plant loss-of-resistance, mutants using normally incompatible Cell 1996, 8:1669-1681. 340 Plant–microbe interactions

2. Lai MMC: Cellular factors in the transcription and replication 20. Noueiry AO, Lucas WJ, Gilbertson RL: Two proteins of a plant of viral RNA genomes: a parallel to DNA-dependent RNA DNA virus coordinate nuclear and plasmodesmal transport. transcription. Virology 1998, 244:1-12. Cell 1994, 76:925-932. 3. Schaad MC, Jensen P, Carrington JC: Formation of plant 21. Citovsky V, Knorr D, Schuster G, Zambryski P: The P30 RNA virus replication complexes on membranes: role of an movement protein of tobacco mosaic virus is a single-strand endoplasmic reticulum-targeted viral protein. EMBO J 1997, nucleic acid binding protein. Cell 1990, 60:637-647. 16:4049-4059. 22. Wolf S, Deom CM, Beachy RN, Lucas WJ: Movement protein of 4. Restrepo-Hartwig MA, Ahlquist P: Brome mosaic virus helicase- tobacco mosaic virus modifies plasmodesmata size exclusion and polymerase-like proteins colocalize on the endoplasmic limit. Science 1989, 246:377-379. reticulum at sites of viral RNA synthesis. J Virol 1996, 70:8908- 23. Wolf S, Deom CM, Beachy R, Lucas WJ: Plasmodesmatal 8916. function is probed using transgenic tobacco plants that express a virus movement protein. Plant Cell 1991, 3:593-604. 5. Ishikawa M, Janda M, Krol MA, Ahlquist P: In vivo DNA • expression of functional brome mosaic virus RNA replicons 24. Heinlein M, Epel BL, Padgett HS, Beachy RN: Interaction of in Saccharomyces cerevisiae. J Virol 1997, 71:7781-7790. movement proteins with the plant cytoskeleton. This paper describes the development and use of selectable BMV replicons Science 1995, 270:1983-1985. in yeast. These replicons have wide utility in genetic selections for yeast 25. McLean BG, Zupan J, Zambryski PC: Tobacco mosaic virus mutants with altered capacity to support replication. movement protein associates with the cytoskeleton in tobacco 6. Janda M, Ahlquist P: RNA-dependent replication, transcription, cells. Plant Cell 1995, 7:2101-2114. and persistence of brome mosaic virus RNA replicons in 26. Heinlein M, Hal S, Padgett J, Gens S, Pickard BG, Casper SJ, S. cerevisiae. Cell 1993, 72:961-970. Epel BL, Beachy RN: Changing patterns of localization of TMV movement protein and replicase to endoplasmic reticulum and Formation of brome 7. Quadt R, Ishikawa M, Janda M, Ahlquist P: microtubules during infection. 10 mosaic virus RNA-dependent RNA polymerase in yeast Plant Cell 1998, :in press. requires coexpression of viral proteins and viral RNA. Proc Natl 27. McLean BG, Hempel FD, Zambryski PC: Plant intercellular Acad Sci USA 1995, 92:4892-4896. communication via plasmodesmata. Plant Cell 1997, 9:1043- 1054. 8. Ishikawa M, Diez J, Restrepo-Hartwig M, Ahlquist P: Yeast •• mutations in multiple complementation groups inhibit brome 28. Ach RA, Durfee T, Miller AB, Taranto P, Hanley-Bowdoin L, mosaic virus RNA replication and transcription and perturb •• Zambryski PC, Gruissem W: RRB1 and RRB2 encode maize regulated expression of the viral polymerase-like gene. Proc retinoblastoma-related proteins that interact with a plant D- Natl Acad Sci USA 1997, 94:13810-13815. type cyclin and geminivirus replication protein. Mol Cell Biol The yeast mab mutants, which exhibit altered abilities to support BMV repli- 1997, 17:5077-5086. con amplification, are described. Understanding the roles of the MAB gene This paper is the latest in a series including [29–31] which establishes that products will provide key insights the critical virus–host interactions required plants encode pRB homologues. The mammalian pRB protein serves as a for eukaryotic cells to support viral RNA replication. critical negative regulator of the cell cycle by inhibiting progression through G1 and into S-phase. These papers also demonstrate convincingly that plant 9. Eagle PA, Orozco BM, Hanley-Bowdoin L: A DNA sequence pRB proteins interact directly with geminivirus AL1/RepA protein. Interac- required for geminivirus replication also mediates tion between AL1/RepA and pRB protein may alter pRB function, thereby transcriptional regulation. Plant Cell 1994, 6:1157-1170. inducing activation of events necessary to produce S-phase-specific nuclear factors required for geminivirus DNA replication. 10. Sunter G, Hartitz MD, Bisaro DM: Tomato golden mosaic virus leftward gene expression: autoregulation of geminivirus 29. Xie Q, Suarez-Lopez P, Gutierrez C: Identification and analysis replication protein. Virology 1993, 195:275-280. of a retinoblastoma binding motif in the replication protein of a plant DNA virus: requirement for efficient viral DNA replication. 11. Fontes EPB, Eagle PA, Sipe PA, Luckow VA, Hanley-Bowdoin L: EMBO J 1995, 14:4073-4082. Interaction between a geminivirus replication protein and origin DNA is essential for viral replication. J Biol Chem 1994, 30. Xie Q, Sanz-Burgos AP, Hannon GJ, Gutierrez C: Plant cells 269:8459-8465. contain a novel member of the retinoblastoma family of growth regulatory proteins. EMBO J 1996, 15:4900-4908. Rep 12. Desbiez C, David C, Mettouchi A, Laufs J, Gronenborn B: 31. Collin S, Fernandez-Lobato M, Gooding PS, Mullineaux PM, protein of tomato yellow leaf curl geminivirus has an ATPase Fenoll C: The two nonstructural proteins from wheat dwarf activity required for viral DNA replication. Proc Natl Acad Sci virus involved in viral gene expression and replication are 92 USA 1995, :5640-5644. retinoblastoma-binding proteins. Virology 1996, 219:324-329. 13. Hanson SF, Hoogstraten RA, Ahlquist P, Gilbertson RL, Russell 32. Weinberg RA: The retinoblastoma protein and cell cycle DR, Maxwell DP: Mutational analysis of a putative NTP-binding control. Cell 1995, 81:323-330. domain in the replication-associated protein (AC1) of bean golden mosaic geminivirus. Virology 1995, 211:1-9. 33. Vousden KH: Regulation of the cell cycle by viral oncoproteins. Semin Cancer Biol 1995, 6:109-116. 14. Nagar S, Pedersen TJ, Carrick KM, Hanley-Bowdoin L, 34. Baker B, Zambryski P, Staskawicz B, Dinesh-Kumar SP: Signaling Robertson D: A geminivirus induces expression of a host DNA • in plant–microbe interactions. Science 1997, 276:726-733. synthesis protein in terminally differentiated plant cells. Plant Recent highlights of research into plant–microbe interactions are summa- Cell 1995, 7:705-719. rized in this excellent review. 15. Nelson RS, van Bel AJE: The mystery of virus trafficking into, 35. Hammond-Kosack KE, Jones JDG: Plant disease resistance through and out of vascular tissue. Prog Botany 1998, 59:476- • genes. Annu Rev Plant Physiol Plant Mol Biol 1997, 48:575-607. 533. This is a fine review on structure, function and evolution of dominant resis- 16. Lucas WJ: Plasmodesmata: intercellular channels for tance genes. macromolecular transport in plants. Curr Opin Cell Biol 1995, 36. Dangl JL, Dietrich RA, Richberg MH: Death don’t have no mercy: 7:673-680. cell death programs in plant-microbe interactions. Plant Cell 1996, 8:1793-1807. 17. Sanderfoot AA, Ingram DJ, Lazarowitz SG: A viral movement protein as a nuclear shuttle. The geminivirus BR1 movement 37. Ryals JA, Neuenschwander UH, Willits MG, Molina A, Steiner protein contains domains essential for interaction with BL1 HY, Hunt MD: Systemic acquired resistance. Plant Cell 1996, and nuclear localization. Plant Physiol 1996, 110:23-33. 8:1809-1819. 18. Pascal E, Sanderfoot AA, Ward BM, Medville R, Turgeon R, 38. Depicker A, Van Montagu M: Post-transcriptional gene silencing Lazarowitz SG: The geminivirus BR1 movement protein binds in plants. Curr Opin Cell Biol 1997, 9:373-382. single-stranded DNA and localizes to the cell nucleus. Plant 39. Kumpatla SP, Chandrasekharan MB, Iyer LM, Li G, Hall TC: Cell 1994, 6:995-1006. Genome intruder scanning and modulation systems and transgene silencing. Trends Plant Sci 1998, 3:97-104. 19. Ward BM, Medville R, Lazarowitz SG, Turgeon R: The geminivirus BL1 movement protein is associated with endoplasmic 40. Baulcombe DC, English JJ: Ectopic pairing of homologous DNA reticulum-derived tubules in developing phloem cells. J Virol and post-transcriptional gene silencing in transgenic plants. 1997, 71:3726-3733. Curr Opin Biotechnol 1996, 7:173-180. Viral invasion and host defense: strategies and counter-strategies Carrington and Whitham 341

41. Baulcombe DC: RNA as a target and an initiator of post- 52. Aoki S, Takebe I: Replication of tobacco mosaic virus RNA in transcriptional gene silencing in transgenic plants. Plant Mol tobacco mesophyll protoplasts inoculated in vitro. Virology Biol 1996, 32:79-88. 1975, 65:343-354. 42. Voinnet O, Baulcombe DC: Systemic signalling in gene 53. Li XH, Carrington JC: Complementation of tobacco etch •• silencing. Nature 1997, 389:553-555. potyvirus mutants by active RNA polymerase expressed in This seminal paper demonstrates that post-transcriptional gene silencing can transgenic cells. Proc Natl Acad Sci USA 1995, 92:457-461. be induced in tissues by a systemic signal derived from another part of the plant. 54. Kasschau KD, Cronin S, Carrington JC: Genome amplification • and long-distance movement functions associated with the 43. Palauqui JC, Elmayan T, Pollien JM, Vaucheret H: Systemic central domain of tobacco etch potyvirus helper component- •• acquired silencing: transgene-specific post-transcriptional proteinase. Virology 1997, 228:251-262. silencing is transmitted by grafting from silenced stocks to Using a series of alanine-scanning mutants, the role of HC-Pro as an en- non-silenced scions. EMBO J 1997, 16:4738-4745. hancer of genome amplification and long-distance movement was investi- This seminal paper demonstrates that post-transcriptional gene silencing can gated. It was proposed that HC-Pro may serve as a suppressor of one or be induced in tissues by a systemic signal derived from another part of the more host defense responses. plant. 55. Vance VB: Replication of potato virus X RNA is altered in 44. Kumagai MH, Donson J, Della-Cioppa G, Harvey D, Hanley K, coinfections with potato virus Y. Virology 1991, 182:486-494. Grill LK: Cytoplasmic inhibition of carotenoid biosynthesis with virus-derived RNA. Proc Natl Acad Sci USA 1995, 92:1679- 56. Shi XM, Miller H, Verchot J, Carrington JC, Vance VB: Mutational 1683. analysis of the potyviral sequence that mediates potato virus X/potyviral synergistic disease. Virology 1996, 231:35-42. 45. Goodwin J, Chapman K, Swaney S, Parks TD, Wernsman EA, Dougherty WG: Genetic and biochemical dissection of 57. Pruss G, Ge X, Shi XM, Carrington JC, Vance VB: Plant viral transgenic RNA-mediated virus resistance. Plant Cell 1996, • synergism: the potyviral genome encodes a broad-range 8:95-105. pathogenicity enhancer that transactivates replication of heterologous viruses. Plant Cell 1997, 9:859-868. 46. Ratcliff F, Harrison BD, Baulcombe DC: A similarity between viral Using transgenic plants and recombinant viruses, the tobacco etch virus HC- •• defence and gene silencing in plants. Science 1997, 276:1558- Pro protein was shown to stimulate accumulation of, and increase disease 1560. severity induced by, several heterologous viruses. In this paper, and in [54•], These two papers ([46••] and [47••]) show that virus-induced gene silencing HC-Pro was proposed to serve as a suppressor of one or more defense can be established independent of homologous nuclear sequences. This responses, possibly including virus-induced gene silencing. implies that potentially any virus could induce a silenced state upon infection. Plants may use silencing as an anti-viral defense mechanism to limit the 58. Chen B, Gao S, Choi GH, Nuss DL: Extensive alteration of extent of infection. fungal gene transcript accumulation and elevation of G- 47. Al-Kaff NS, Covey SN, Kreike MM, Page AM, Pinder R, Dale PJ: protein-regulated cAMP levels by a virulence-attenuating •• Transcriptional and post-transcriptional plant gene silencing in hypovirus. Proc Natl Acad Sci USA 1996, 93:7996-8000. response to a pathogen. Science 1998, 279:2113-2115. 59. Choi GH, Chen B, Nuss DL: Virus-mediated or transgenic •• See annotation for [46 ]. suppression of a G-protein alpha subunit and attenuation of 48. Ploegh HL: Viral strategies of immune evasion. Science 1998, fungal virulence. Proc Natl Acad Sci USA 1995, 92:305-309. 280:248-253. 60. Kasahara S, Nuss DL: Targeted disruption of a fungal G-protein 49. Gillet G, Brun G: Viral inhibition of apotosis. Trends Microbiol subunit gene results in increased vegetative growth but 1996, 4:312-317. reduced virulence. Mol Plant Microbe Interact 1997, 10:984-993. 50. Wang D, Maule AJ: Inhibition of host gene expression 61. Glazebrook J, Rogers EE, Ausubel FM: Use of Arabidopsis for associated with plant virus replication. Science 1995, 267:229- genetic dissection of plant defense responses. Annu Rev Genet 231. 1997, 31:547-569. 51. Aranda MA, Escaler M, Wang D, Maule AJ: Induction of HSP70 62. Ishikawa M, Naito S, Ohno T: Effects of the tom1 mutation of and polyubiquitin expression associated with plant virus Arabidopsis thaliana on the multiplication of tobacco mosaic replication. Proc Natl Acad Sci USA 1996, 93:15289-15293. virus RNA in protoplasts. J Virol 1993, 67:5328-5338.