Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 148
Filter
1.
PLoS Pathog ; 20(3): e1012085, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38484009

ABSTRACT

Elaborate viral replication organelles (VROs) are formed to support positive-strand RNA virus replication in infected cells. VRO formation requires subversion of intracellular membranes by viral replication proteins. Here, we showed that the key ATG8f autophagy protein and NBR1 selective autophagy receptor were co-opted by Tomato bushy stunt virus (TBSV) and the closely-related carnation Italian ringspot virus. Knockdown of ATG8f or NBR1 in plants led to reduced tombusvirus replication, suggesting pro-viral function for selective autophagy. BiFC and proximity-labeling experiments showed that the TBSV p33 replication protein interacted with ATG8f and NBR1 to recruit them to VROs. In addition, we observed that several core autophagy proteins, such as ATG1a, ATG4, ATG5, ATG101 and the plant-specific SH3P2 autophagy adaptor proteins were also re-localized to TBSV VROs, suggesting that TBSV hijacks the autophagy machinery in plant cells. We demonstrated that subversion of autophagy components facilitated the recruitment of VPS34 PI3 kinase and enrichment of phospholipids, such as phosphatidylethanolamine and PI3P phosphoinositide in the VRO membranes. Hijacking of autophagy components into TBSV VROs led to inhibition of autophagic flux. We also found that a fraction of the subverted ATG8f and NBR1 was sequestered in biomolecular condensates associated with VROs. We propose that the VRO-associated condensates trap those autophagy proteins, taking them away from the autophagy pathway. Overall, tombusviruses hijack selective autophagy to provide phospholipid-rich membranes for replication and to regulate the antiviral autophagic flux.


Subject(s)
Tombusvirus , Tombusvirus/physiology , Saccharomyces cerevisiae/genetics , Intracellular Membranes/metabolism , Virus Replication/physiology , Phospholipids/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Autophagy , Organelles/metabolism , RNA, Viral/genetics
2.
J Virol ; 96(12): e0016821, 2022 06 22.
Article in English | MEDLINE | ID: mdl-35638821

ABSTRACT

Positive-strand RNA viruses build large viral replication organelles (VROs) with the help of coopted host factors. Previous works on tomato bushy stunt virus (TBSV) showed that the p33 replication protein subverts the actin cytoskeleton by sequestering the actin depolymerization factor, cofilin, to reduce actin filament disassembly and stabilize the actin filaments. Then, TBSV utilizes the stable actin filaments as "trafficking highways" to deliver proviral host factors into the protective VROs. In this work, we show that the cellular intrinsic restriction factors (CIRFs) also use the actin network to reach VROs and inhibit viral replication. Disruption of the actin filaments by expression of the Legionella RavK protease inhibited the recruitment of plant CIRFs, including the CypA-like Roc1 and Roc2 cyclophilins, and the antiviral DDX17-like RH30 DEAD box helicase into VROs. Conversely, temperature-sensitive actin and cofilin mutant yeasts with stabilized actin filaments reduced the levels of copurified CIRFs, including cyclophilins Cpr1, CypA, Cyp40-like Cpr7, cochaperones Sgt2, the Hop-like Sti1, and the RH30 helicase in viral replicase preparations. Dependence of the recruitment of both proviral and antiviral host factors into VROs on the actin network suggests that there is a race going on between TBSV and its host to exploit the actin network and ultimately to gain the upper hand during infection. We propose that, in the highly susceptible plants, tombusviruses efficiently subvert the actin network for rapid delivery of proviral host factors into VROs and ultimately overcome host restriction factors via winning the recruitment race and overwhelming cellular defenses. IMPORTANCE Replication of positive-strand RNA viruses is affected by the recruitment of host components, which provide either proviral or antiviral functions during virus invasion of infected cells. The delivery of these host factors into the viral replication organelles (VROs), which represent the sites of viral RNA replication, depends on the cellular actin network. Using TBSV, we uncover a race between the virus and its host with the actin network as the central player. We find that in susceptible plants, tombusviruses exploit the actin network for rapid delivery of proviral host factors into VROs and ultimately overcome host restriction factors. In summary, this work demonstrates that the actin network plays a major role in determining the outcome of viral infections in plants.


Subject(s)
Actins , Antiviral Restriction Factors , Organelle Biogenesis , Tombusvirus , Virus Replication , Actin Depolymerizing Factors/metabolism , Actins/metabolism , Carrier Proteins/metabolism , Cyclophilins/metabolism , DNA Viruses/genetics , RNA, Viral/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/virology , Saccharomyces cerevisiae Proteins , Tombusvirus/genetics , Tombusvirus/physiology , Viral Proteins/metabolism
3.
J Virol ; 95(21): e0107621, 2021 10 13.
Article in English | MEDLINE | ID: mdl-34406861

ABSTRACT

Positive-strand RNA viruses induce the biogenesis of unique membranous organelles called viral replication organelles (VROs), which perform virus replication in infected cells. Tombusviruses have been shown to rewire cellular trafficking and metabolic pathways, remodel host membranes, and recruit multiple host factors to support viral replication. In this work, we demonstrate that tomato bushy stunt virus (TBSV) and the closely related carnation Italian ringspot virus (CIRV) usurp Rab7 small GTPase to facilitate building VROs in the surrogate host yeast and in plants. Depletion of Rab7 small GTPase, which is needed for late endosome and retromer biogenesis, strongly inhibits TBSV and CIRV replication in yeast and in planta. The viral p33 replication protein interacts with Rab7 small GTPase, which results in the relocalization of Rab7 into the large VROs. Similar to the depletion of Rab7, the deletion of either MON1 or CCZ1 heterodimeric GEFs (guanine nucleotide exchange factors) of Rab7 inhibited TBSV RNA replication in yeast. This suggests that the activated Rab7 has proviral functions. We show that the proviral function of Rab7 is to facilitate the recruitment of the retromer complex and the endosomal sorting nexin-BAR proteins into VROs. We demonstrate that TBSV p33-driven retargeting of Rab7 into VROs results in the delivery of several retromer cargos with proviral functions. These proteins include lipid enzymes, such as Vps34 PI3K (phosphatidylinositol 3-kinase), PI4Kα-like Stt4 phosphatidylinositol 4-kinase, and Psd2 phosphatidylserine decarboxylase. In summary, based on these and previous findings, we propose that subversion of Rab7 into VROs allows tombusviruses to reroute endocytic and recycling trafficking to support virus replication. IMPORTANCE The replication of positive-strand RNA viruses depends on the biogenesis of viral replication organelles (VROs). However, the formation of membranous VROs is not well understood yet. Using tombusviruses and the model host yeast, we discovered that the endosomal Rab7 small GTPase is critical for the formation of VROs. Interaction between Rab7 and the TBSV p33 replication protein leads to the recruitment of Rab7 into VROs. TBSV-driven usurping of Rab7 has proviral functions through facilitating the delivery of the co-opted retromer complex, sorting nexin-BAR proteins, and lipid enzymes into VROs to create an optimal milieu for virus replication. These results open up the possibility that controlling cellular Rab7 activities in infected cells could be a target for new antiviral strategies.


Subject(s)
Nicotiana/virology , Organelles/virology , Saccharomyces cerevisiae/virology , Tombusvirus/physiology , Viral Proteins/metabolism , Virus Replication , rab GTP-Binding Proteins/physiology , 1-Phosphatidylinositol 4-Kinase/metabolism , Endosomes/metabolism , Gene Knockdown Techniques , Guanine Nucleotide Exchange Factors/physiology , Host Microbial Interactions , Organelles/metabolism , Plant Diseases/virology , Protein Binding , Protein Transport , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/physiology , Sorting Nexins/metabolism
4.
Virology ; 563: 1-19, 2021 11.
Article in English | MEDLINE | ID: mdl-34399236

ABSTRACT

To further our understanding of the pro-viral roles of the host cytosolic heat shock protein 70 (Hsp70) family, we chose the conserved Arabidopsis thaliana Hsp70-2 and the unique Erd2 (early response to dehydration 2), which contain Hsp70 domains. Based on in vitro studies with purified components, we show that AtHsp70-2 and AtErd2 perform pro-viral functions equivalent to that of the yeast Ssa1 Hsp70. These functions include activation of the tombusvirus RdRp, and stimulation of replicase assembly. Yeast-based complementation studies demonstrate that AtHsp70-2 or AtErd2 are present in the purified tombusvirus replicase. RNA silencing and over-expression studies in Nicotiana benthamiana suggest that both Hsp70-2 and Erd2 are co-opted by tomato bushy stunt virus (TBSV). Moreover, we used allosteric inhibitors of Hsp70s to inhibit replication of TBSV and related plant viruses in plants. Altogether, interfering with the functions of the co-opted Hsp70s could be an effective antiviral approach against tombusviruses in plants.


Subject(s)
Arabidopsis/metabolism , HSP70 Heat-Shock Proteins/metabolism , Tombusvirus/physiology , Virus Replication/physiology , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Benzothiazoles/pharmacology , Gene Expression Regulation, Plant/immunology , Gene Expression Regulation, Viral/drug effects , Gene Expression Regulation, Viral/physiology , Gene Knockdown Techniques , Genetic Complementation Test , HSP70 Heat-Shock Proteins/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Plant Leaves/metabolism , Plant Leaves/virology , Pyridinium Compounds/pharmacology , RNA, Viral/physiology , Nicotiana/metabolism , Nicotiana/virology , Two-Hybrid System Techniques , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication/drug effects
5.
PLoS Pathog ; 17(6): e1009680, 2021 06.
Article in English | MEDLINE | ID: mdl-34161398

ABSTRACT

Positive-strand (+)RNA viruses take advantage of the host cells by subverting a long list of host protein factors and transport vesicles and cellular organelles to build membranous viral replication organelles (VROs) that support robust RNA replication. How RNA viruses accomplish major recruitment tasks of a large number of cellular proteins are intensively studied. In case of tomato bushy stunt virus (TBSV), a single viral replication protein, named p33, carries out most of the recruitment duties. Yet, it is currently unknown how the viral p33 replication protein, which is membrane associated, is capable of the rapid and efficient recruitment of numerous cytosolic host proteins to facilitate the formation of large VROs. In this paper, we show that, TBSV p33 molecules do not recruit each cytosolic host factor one-by-one into VROs, but p33 targets a cytosolic protein interaction hub, namely Rpn11, which interacts with numerous other cytosolic proteins. The highly conserved Rpn11, called POH1 in humans, is the metalloprotease subunit of the proteasome, which couples deubiquitination and degradation of proteasome substrates. However, TBSV takes advantage of a noncanonical function of Rpn11 by exploiting Rpn11's interaction with highly abundant cytosolic proteins and the actin network. We provide supporting evidence that the co-opted Rpn11 in coordination with the subverted actin network is used for delivering cytosolic proteins, such as glycolytic and fermentation enzymes, which are readily subverted into VROs to produce ATP locally in support of VRO formation, viral replicase complex assembly and viral RNA replication. Using several approaches, including knockdown of Rpn11 level, sequestering Rpn11 from the cytosol into the nucleus in plants or temperature-sensitive mutation in Rpn11 in yeast, we show the inhibition of recruitment of glycolytic and fermentation enzymes into VROs. The Rpn11-assisted recruitment of the cytosolic enzymes by p33, however, also requires the combined and coordinated role of the subverted actin network. Accordingly, stabilization of the actin filaments by expression of the Legionella VipA effector in yeast and plant, or via a mutation of ACT1 in yeast resulted in more efficient and rapid recruitment of Rpn11 and the selected glycolytic and fermentation enzymes into VROs. On the contrary, destruction of the actin filaments via expression of the Legionella RavK effector led to poor recruitment of Rpn11 and glycolytic and fermentation enzymes. Finally, we confirmed the key roles of Rpn11 and the actin filaments in situ ATP production within TBSV VROs via using a FRET-based ATP-biosensor. The novel emerging theme is that TBSV targets Rpn11 cytosolic protein interaction hub driven by the p33 replication protein and aided by the subverted actin filaments to deliver several co-opted cytosolic pro-viral factors for robust replication within VROs.


Subject(s)
Actin Cytoskeleton/metabolism , Endopeptidases/metabolism , Host-Pathogen Interactions/physiology , Tombusvirus/physiology , Virus Replication/physiology , Cytosol/metabolism , Proteasome Endopeptidase Complex/metabolism
6.
Curr Opin Virol ; 48: 30-41, 2021 06.
Article in English | MEDLINE | ID: mdl-33845410

ABSTRACT

Positive-strand RNA viruses depend on intensive manipulation of subcellular organelles and membranes to create unique viral replication organelles (VROs), which represent the sites of robust virus replication. The host endomembrane-based protein-trafficking and vesicle-trafficking pathways are specifically targeted by many (+)RNA viruses to take advantage of their rich resources. We summarize the critical roles of co-opted endoplasmic reticulum subdomains and associated host proteins and COPII vesicles play in tombusvirus replication. We also present the surprising contribution of the early endosome and the retromer tubular transport carriers to VRO biogenesis. The central player is tomato bushy stunt virus (TBSV), which provides an outstanding system based on the identification of a complex network of interactions with the host cells. We present the emerging theme on how TBSV uses tethering and membrane-shaping proteins and lipid modifying enzymes to build the sophisticated VRO membranes with unique lipid composition.


Subject(s)
Host-Pathogen Interactions/physiology , Organelles/virology , Tombusvirus/physiology , Virus Replication/physiology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/virology , Genes, Viral/genetics , Host-Pathogen Interactions/genetics , Lipid Metabolism , Lipids , Magnoliopsida/virology , RNA Viruses , Tombusvirus/genetics , Virus Replication/genetics
7.
Virology ; 559: 15-29, 2021 07.
Article in English | MEDLINE | ID: mdl-33799077

ABSTRACT

Positive-strand RNA viruses build viral replication organelles (VROs) with the help of co-opted host factors. The energy requirement of intensive viral replication processes is less understood. Previous studies on tomato bushy stunt virus (TBSV) showed that tombusviruses hijack two ATP-producing glycolytic enzymes to produce ATP locally within VROs. In this work, we performed a cDNA library screen with Arabidopsis thaliana proteins and the TBSV p33 replication protein. The p33 - plant interactome contained highly conserved glycolytic proteins. We find that the glycolytic Hxk2 hexokinase, Eno2 phosphopyruvate hydratase and Fba1 fructose 1,6-bisphosphate aldolase are critical for TBSV replication in yeast or in a cell-free replicase reconstitution assay. The recruitment of Fba1 is important for the local production of ATP within VROs. Altogether, our data support the model that TBSV recruits and compartmentalizes possibly most members of the glycolytic pathway. This might allow TBSV to avoid competition with the host for ATP.


Subject(s)
Adenosine Triphosphate/metabolism , Glycolysis , Nicotiana/enzymology , Tombusvirus/physiology , Virus Replication/physiology , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Host Microbial Interactions , Nicotiana/metabolism , Nicotiana/virology , Tombusvirus/genetics , Tombusvirus/metabolism
8.
PLoS Pathog ; 17(3): e1009423, 2021 03.
Article in English | MEDLINE | ID: mdl-33725015

ABSTRACT

Plus-stranded RNA viruses have limited coding capacity and have to co-opt numerous pro-viral host factors to support their replication. Many of the co-opted host factors support the biogenesis of the viral replication compartments and the formation of viral replicase complexes on subverted subcellular membrane surfaces. Tomato bushy stunt virus (TBSV) exploits peroxisomal membranes, whereas the closely-related carnation Italian ringspot virus (CIRV) hijacks the outer membranes of mitochondria. How these organellar membranes can be recruited into pro-viral roles is not completely understood. Here, we show that the highly conserved Fis1 mitochondrial fission protein is co-opted by both TBSV and CIRV via direct interactions with the p33/p36 replication proteins. Deletion of FIS1 in yeast or knockdown of the homologous Fis1 in plants inhibits tombusvirus replication. Instead of the canonical function in mitochondrial fission and peroxisome division, the tethering function of Fis1 is exploited by tombusviruses to facilitate the subversion of membrane contact site (MCS) proteins and peroxisomal/mitochondrial membranes for the biogenesis of the replication compartment. We propose that the dynamic interactions of Fis1 with MCS proteins, such as the ER resident VAP tethering proteins, Sac1 PI4P phosphatase and the cytosolic OSBP-like oxysterol-binding proteins, promote the formation and facilitate the stabilization of virus-induced vMCSs, which enrich sterols within the replication compartment. We show that this novel function of Fis1 is exploited by tombusviruses to build nuclease-insensitive viral replication compartment.


Subject(s)
Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Tombusvirus/physiology , Virus Replication/physiology , Saccharomyces cerevisiae/virology , Nicotiana/virology
9.
Virology ; 554: 106-119, 2021 02.
Article in English | MEDLINE | ID: mdl-33418272

ABSTRACT

Cucumber necrosis virus (CNV) is a (+)ssRNA virus that elicits spreading local and systemic necrosis in Nicotiana benthamiana. We previously showed that the CNV coat protein (CP) arm functions as a chloroplast transit peptide that targets a CP fragment containing the S and P domains to chloroplasts during infection. Here we show that several CP arm mutants that inefficiently target chloroplasts, along with a mutant that lacks the S and P domains, show an early onset of more localized necrosis along with protracted induction of pathogenesis related protein (PR1a). Agroinfiltrated CNV CP is shown to interfere with CNV p33 and Tomato bushy stunt virus p19 induced necrosis. Additionally, we provide evidence that a CP mutant that does not detectably enter the chloroplast stroma induces relatively higher levels of several plant defense-related genes compared to WT CNV. Together, our data suggest that targeting of CNV CP to the chloroplast stroma interferes with chloroplast-mediated plant defense.


Subject(s)
Capsid Proteins/metabolism , Chloroplasts/metabolism , Plant Necrosis and Chlorosis/virology , Tombusvirus/physiology , Capsid Proteins/chemistry , Capsid Proteins/genetics , Genes, Plant , Mutant Proteins/metabolism , N-Acetylneuraminic Acid/metabolism , Plant Immunity/genetics , Plant Necrosis and Chlorosis/immunology , Plant Proteins/genetics , Plant Proteins/metabolism , Protein Domains , Signal Transduction , Nicotiana/genetics , Nicotiana/immunology , Nicotiana/virology , Tombusvirus/genetics , Up-Regulation , Viral Proteins/metabolism
10.
PLoS Pathog ; 16(12): e1009120, 2020 12.
Article in English | MEDLINE | ID: mdl-33370420

ABSTRACT

Positive-strand RNA viruses replicate in host cells by forming large viral replication organelles, which harbor numerous membrane-bound viral replicase complexes (VRCs). In spite of its essential role in viral replication, the biogenesis of the VRCs is not fully understood. The authors identified critical roles of cellular membrane-shaping proteins and PI(3)P (phosphatidylinositol 3-phosphate) phosphoinositide, a minor lipid with key functions in endosomal vesicle trafficking and autophagosome biogenesis, in VRC formation for tomato bushy stunt virus (TBSV). The authors show that TBSV co-opts the endosomal SNX-BAR (sorting nexin with Bin/Amphiphysin/Rvs- BAR domain) proteins, which bind to PI(3)P and have membrane-reshaping function during retromer tubular vesicle formation, directly into the VRCs to boost progeny viral RNA synthesis. We find that the viral replication protein-guided recruitment and pro-viral function of the SNX-BAR proteins depends on enrichment of PI(3)P at the site of viral replication. Depletion of SNX-BAR proteins or PI(3)P renders the viral double-stranded (ds)RNA replication intermediate RNAi-sensitive within the VRCs in the surrogate host yeast and in planta and ribonuclease-sensitive in cell-free replicase reconstitution assays in yeast cell extracts or giant unilamellar vesicles (GUVs). Based on our results, we propose that PI(3)P and the co-opted SNX-BAR proteins are coordinately exploited by tombusviruses to promote VRC formation and to play structural roles and stabilize the VRCs during viral replication. Altogether, the interplay between the co-opted SNX-BAR membrane-shaping proteins, PI(3)P and the viral replication proteins leads to stable VRCs, which provide the essential protection of the viral RNAs against the host antiviral responses.


Subject(s)
Phosphatidylinositol Phosphates/metabolism , Sorting Nexins/metabolism , Tombusvirus/physiology , Viral Replicase Complex Proteins/metabolism , Arabidopsis/metabolism , Arabidopsis/virology , Cells, Cultured , Host-Pathogen Interactions/genetics , Organisms, Genetically Modified , Phosphatidylinositols/metabolism , Protein Interaction Domains and Motifs , RNA, Viral/genetics , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/metabolism , RNA-Dependent RNA Polymerase/physiology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/virology , Sorting Nexins/chemistry , Sorting Nexins/physiology , Nicotiana/metabolism , Nicotiana/virology , Tombusvirus/genetics , Tombusvirus/metabolism , Viral Replicase Complex Proteins/physiology , Virus Replication/genetics
11.
Virus Res ; 286: 198048, 2020 09.
Article in English | MEDLINE | ID: mdl-32522536

ABSTRACT

Gentian virus A (GeVA), a novel tombusvirus isolated from Japanese gentian, has shown only a limited ability to infect Japanese gentians under experimental conditions. In this study, temperature was found to affect the efficient multiplication of GeVA in Japanese gentians. GeVA efficiently multiplied in inoculated leaves of gentians at 18 °C but not at 23 °C. This low-temperature (18 °C)-preferred GeVA multiplication was specifically observed in Japanese gentians and Arabidopsis thaliana but not in other experimental plants, including Nicotiana benthamiana. In A. thaliana, visible defense responses, including pathogenesis-related protein 1 expression, were not detected at 23 °C. Furthermore, several A. thaliana mutants, including those defective in RNA silencing, with altered plant immunities did not allow GeVA to multiply to detectable levels at 23 °C. Taken together, these data suggest that unique interaction between GeVA and gentians/A. thaliana, which is independent of RNA silencing, may underlie the low-temperature-preferred multiplication of GeVA.


Subject(s)
Cold Temperature , Gentiana/virology , Host Microbial Interactions , Tombusvirus/physiology , Virus Replication , Arabidopsis/virology , Plant Leaves/virology , RNA, Viral/metabolism , Nicotiana/virology , Tombusvirus/genetics , Tombusvirus/pathogenicity
12.
Viruses ; 12(1)2020 01 03.
Article in English | MEDLINE | ID: mdl-31947719

ABSTRACT

Recent discoveries on virus-driven hijacking and compartmentalization of the cellular glycolytic and fermentation pathways to support robust virus replication put the spotlight on the energy requirement of viral processes. The active recruitment of glycolytic enzymes in combination with fermentation enzymes by the viral replication proteins emphasizes the advantages of producing ATP locally within viral replication structures. This leads to a paradigm shift in our understanding of how viruses take over host metabolism to support the virus's energy needs during the replication process. This review highlights our current understanding of how a small plant virus, Tomato bushy stunt virus, exploits a conserved energy-generating cellular pathway during viral replication. The emerging picture is that viruses not only rewire cellular metabolic pathways to obtain the necessary resources from the infected cells but the fast replicating viruses might have to actively hijack and compartmentalize the energy-producing enzymes to provide a readily available source of ATP for viral replication process.


Subject(s)
Adenosine Triphosphate/metabolism , Energy Metabolism , Tombusvirus/physiology , Virus Replication , Aerobiosis , Fermentation , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , Glycolysis , Host-Pathogen Interactions , Neoplasms/metabolism , RNA, Viral/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/virology
13.
PLoS Pathog ; 15(5): e1007771, 2019 05.
Article in English | MEDLINE | ID: mdl-31136641

ABSTRACT

Positive-stranded RNA viruses replicate inside cells and depend on many co-opted cellular factors to complete their infection cycles. To combat viruses, the hosts use conserved restriction factors, such as DEAD-box RNA helicases, which can function as viral RNA sensors or as effectors by blocking RNA virus replication. In this paper, we have established that the plant DDX17-like RH30 DEAD-box helicase conducts strong inhibitory function on tombusvirus replication when expressed in plants and yeast surrogate host. The helicase function of RH30 was required for restriction of tomato bushy stunt virus (TBSV) replication. Knock-down of RH30 levels in Nicotiana benthamiana led to increased TBSV accumulation and RH30 knockout lines of Arabidopsis supported higher level accumulation of turnip crinkle virus. We show that RH30 DEAD-box helicase interacts with p33 and p92pol replication proteins of TBSV, which facilitates targeting of RH30 from the nucleus to the large TBSV replication compartment consisting of aggregated peroxisomes. Enrichment of RH30 in the nucleus via fusion with a nuclear retention signal at the expense of the cytosolic pool of RH30 prevented the re-localization of RH30 into the replication compartment and canceled out the antiviral effect of RH30. In vitro replicase reconstitution assay was used to demonstrate that RH30 helicase blocks the assembly of viral replicase complex, the activation of the RNA-dependent RNA polymerase function of p92pol and binding of p33 replication protein to critical cis-acting element in the TBSV RNA. Altogether, these results firmly establish that the plant DDX17-like RH30 DEAD-box helicase is a potent, effector-type, restriction factor of tombusviruses and related viruses. The discovery of the antiviral role of RH30 DEAD-box helicase illustrates the likely ancient roles of RNA helicases in plant innate immunity.


Subject(s)
Antiviral Agents/pharmacology , DEAD-box RNA Helicases/pharmacology , Nicotiana/virology , Plant Proteins/metabolism , Tombusvirus/drug effects , Viral Proteins/metabolism , Virus Replication/drug effects , Arabidopsis/metabolism , Arabidopsis/virology , Host-Pathogen Interactions , Plant Diseases/virology , Plant Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Nicotiana/drug effects , Nicotiana/metabolism , Tombusvirus/physiology , Viral Proteins/genetics , Virus Assembly/drug effects
14.
PLoS Pathog ; 14(5): e1007028, 2018 05.
Article in English | MEDLINE | ID: mdl-29746582

ABSTRACT

Positive-strand RNA viruses assemble numerous membrane-bound viral replicase complexes within large replication compartments to support their replication in infected cells. Yet the detailed mechanism of how given subcellular compartments are subverted by viruses is incompletely understood. Although, Tomato bushy stunt virus (TBSV) uses peroxisomal membranes for replication, in this paper, we show evidence that the ER-resident SNARE (soluble NSF attachment protein receptor) proteins play critical roles in the formation of active replicase complexes in yeast model host and in plants. Depletion of the syntaxin 18-like Ufe1 and Use1, which are components of the ER SNARE complex in the ERAS (ER arrival site) subdomain, in yeast resulted in greatly reduced tombusvirus accumulation. Over-expression of a dominant-negative mutant of either the yeast Ufe1 or the orthologous plant Syp81 syntaxin greatly interferes with tombusvirus replication in yeast and plants, thus further supporting the role of this host protein in tombusvirus replication. Moreover, tombusvirus RNA replication was low in cell-free extracts from yeast with repressed Ufe1 or Use1 expression. We also present evidence for the mislocalization of the tombusviral p33 replication protein to the ER membrane in Ufe1p-depleted yeast cells. The viral p33 replication protein interacts with both Ufe1p and Use1p and co-opts them into the TBSV replication compartment in yeast and plant cells. The co-opted Ufe1 affects the virus-driven membrane contact site formation, sterol-enrichment at replication sites, recruitment of several pro-viral host factors and subversion of the Rab5-positive PE-rich endosomes needed for robust TBSV replication. In summary, we demonstrate a critical role for Ufe1 and Use1 SNARE proteins in TBSV replication and propose that the pro-viral functions of Ufe1 and Use1 are to serve as assembly hubs for the formation of the extensive TBSV replication compartments in cells. Altogether, these findings point clearly at the ERAS subdomain of ER as a critical site for the biogenesis of the TBSV replication compartment.


Subject(s)
SNARE Proteins/metabolism , SNARE Proteins/physiology , Tombusvirus/physiology , DNA Replication , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/physiology , Endosomes/metabolism , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/physiology , Mitochondrial Membranes/metabolism , Qa-SNARE Proteins/metabolism , Qa-SNARE Proteins/physiology , Qc-SNARE Proteins/metabolism , Qc-SNARE Proteins/physiology , RNA, Viral/genetics , RNA-Dependent RNA Polymerase/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/physiology , Tombusvirus/genetics , Tombusvirus/metabolism , Tombusvirus/pathogenicity , Viral Proteins/genetics , Virus Replication/physiology
15.
Virology ; 519: 207-222, 2018 06.
Article in English | MEDLINE | ID: mdl-29734044

ABSTRACT

Similar to other (+)RNA viruses, tomato bushy stunt virus (TBSV) utilizes metabolites, lipids, membranes, and co-opted host factors during replication. The coordination of cell metabolism and growth with environmental cues is performed by the target of rapamycin (TOR) kinase in eukaryotic cells. In this paper, we find that TBSV replication partially inhibits TOR activity, likely due to recruitment of glycolytic enzymes to the viral replication compartment, which results in reduced ATP levels in the cytosol. Complete inhibition of TOR activity with rapamycin in yeast or AZD8055 inhibitor in plants reduces tombusvirus replication. We find that high glucose concentration, which stimulates TOR activity, enhanced tombusvirus replication in yeast. Depletion of yeast Sch9 or plant S6K1 kinase, a downstream effector of TOR, also inhibited tombusvirus replication in yeast and plant or the assembly of the viral replicase in vitro. Altogether, the TOR pathway is crucial for TBSV to replicate efficiently in hosts.


Subject(s)
Mechanistic Target of Rapamycin Complex 1/metabolism , Nicotiana/virology , RNA, Viral/biosynthesis , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/virology , Tombusvirus/genetics , Transcription Factors/metabolism , Virus Replication , Adenosine Triphosphate/metabolism , Glycolysis , Host-Pathogen Interactions , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Morpholines/pharmacology , RNA-Dependent RNA Polymerase/metabolism , Ribonucleotides/biosynthesis , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Sirolimus/pharmacology , Nicotiana/drug effects , Nicotiana/metabolism , Tombusvirus/physiology , Transcription Factors/antagonists & inhibitors
16.
J Biol Chem ; 293(24): 9486-9495, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29700118

ABSTRACT

Unlike the mRNAs of their eukaryotic hosts, many RNAs of viruses lack a 5' m7GpppN cap and the 3' polyadenosine tail, and yet they are translated efficiently. Plant RNA viruses, in particular, have complex structures within their mRNA UTRs that allow them to bypass some cellular translation control steps. In the 3' UTR of maize necrotic streak virus (MNeSV), an I-shaped RNA structure (ISS) has been shown to bind eukaryotic initiation factor (eIF)4F and to mediate viral translation initiation. A 5'-3' RNA "kissing-loop" interaction is required for optimal translation. However, the details of how the 3' ISS mediates translation initiation are not well understood. Here, we studied the binding of the 3' ISS with eIFs. The eIF4A-eIF4B complex was found to increase binding affinity of eIF4F with the 3' ISS by 4-fold (from KD = 173 ± 34 nm to KD = 48 ± 11 nm). Pre-steady-state analysis indicated that the eIF4A-eIF4B complex increased the RNA association rate and decreased the dissociation rate in an ATP-independent manner. Furthermore, our findings suggest that eIF4F could promote binding of the 3' ISS with the MNeSV 5'UTR, enhancing the long-distance kissing-loop interaction. However, the association of the 5'UTR with the 3' ISS-eIF4F complex did not increase 40S ribosomal subunit binding affinity. These quantitative results suggest a stepwise model in which the first committed step is eIF4F binding to the 3' ISS, followed by an interaction with the 5'UTR and subsequent 40S ribosomal subunit binding.


Subject(s)
Eukaryotic Initiation Factor-4F/metabolism , Plant Diseases/virology , Plant Proteins/metabolism , RNA, Messenger/metabolism , RNA, Viral/metabolism , Tombusvirus/physiology , Triticum/virology , 3' Untranslated Regions , Entropy , Eukaryotic Initiation Factors/metabolism , Nucleic Acid Conformation , Protein Biosynthesis , RNA, Messenger/chemistry , RNA, Viral/chemistry , Ribosome Subunits, Small, Eukaryotic/metabolism , Ribosome Subunits, Small, Eukaryotic/virology , Thermodynamics , Triticum/metabolism
17.
Virus Genes ; 54(3): 368-375, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29480423

ABSTRACT

Non-structural protein 1 (NS1) of influenza A virus is a multifunctional dimeric protein that contains a conserved N-terminal RNA binding domain. Studies have shown that NS1 suppresses RNA silencing and the NS1 proteins encoded by different influenza A virus strains exhibit differential RNA silencing suppression activities. In this study, we showed that the NS1 protein from avian influenza virus (AIV) H9N2 suppressed systemic RNA silencing induced by sense RNA or dsRNA. It resulted in more severe Potato virus X symptom, but could not reverse established systemic green fluorescent protein silencing in Nicotiana benthamiana. In addition, its systemic silencing suppression activity was much weaker than that of p19. The local silencing suppression activity of AIV H9N2 NS1 was most powerful at 7 dpi and was even stronger than that of p19. And the inhibition ability to RNA silencing of NS1 is stronger than that of p19 in human cells. Collectively, these results indicate that AIV H9N2 NS1 is an effective RNA silencing suppressor that likely targets downstream step(s) of dsRNA formation at an early stage in RNA silencing. Although NS1 and p19 both bind siRNA, their suppression mechanisms seem to differ because of differences in their suppression activities at various times post-infiltration and because p19 can reverse established systemic RNA silencing, but NS1 cannot.


Subject(s)
Influenza A Virus, H9N2 Subtype/physiology , RNA Interference , Tombusvirus/physiology , Viral Nonstructural Proteins/physiology , Agrobacterium/genetics , DNA, Viral , Green Fluorescent Proteins/genetics , Influenza A Virus, H9N2 Subtype/genetics , Plants, Genetically Modified , RNA, Double-Stranded , Sequence Analysis, DNA , Nicotiana , Viral Nonstructural Proteins/genetics , Viral Proteins/physiology
18.
PLoS Pathog ; 13(7): e1006520, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28759634

ABSTRACT

Reconstituted antiviral defense pathway in surrogate host yeast is used as an intracellular probe to further our understanding of virus-host interactions and the role of co-opted host factors in formation of membrane-bound viral replicase complexes in protection of the viral RNA against ribonucleases. The inhibitory effect of the RNA interference (RNAi) machinery of S. castellii, which only consists of the two-component DCR1 and AGO1 genes, was measured against tomato bushy stunt virus (TBSV) in wild type and mutant yeasts. We show that deletion of the co-opted ESCRT-I (endosomal sorting complexes required for transport I) or ESCRT-III factors makes TBSV replication more sensitive to the RNAi machinery in yeast. Moreover, the lack of these pro-viral cellular factors in cell-free extracts (CFEs) used for in vitro assembly of the TBSV replicase results in destruction of dsRNA replication intermediate by a ribonuclease at the 60 min time point when the CFE from wt yeast has provided protection for dsRNA. In addition, we demonstrate that co-opted oxysterol-binding proteins and membrane contact sites, which are involved in enrichment of sterols within the tombusvirus replication compartment, are required for protection of viral dsRNA. We also show that phosphatidylethanolamine level influences the formation of RNAi-resistant replication compartment. In the absence of peroxisomes in pex3Δ yeast, TBSV subverts the ER membranes, which provide as good protection for TBSV dsRNA against RNAi or ribonucleases as the peroxisomal membranes in wt yeast. Altogether, these results demonstrate that co-opted protein factors and usurped lipids are exploited by tombusviruses to build protective subcellular environment against the RNAi machinery and possibly other cellular ribonucleases.


Subject(s)
Endosomal Sorting Complexes Required for Transport/metabolism , RNA Interference , RNA, Double-Stranded/genetics , RNA, Viral/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/virology , Tombusvirus/genetics , Endosomal Sorting Complexes Required for Transport/genetics , RNA, Double-Stranded/metabolism , RNA, Viral/metabolism , Saccharomyces/genetics , Saccharomyces/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Tombusvirus/physiology , Virus Replication
19.
Curr Opin Virol ; 26: 132-140, 2017 10.
Article in English | MEDLINE | ID: mdl-28843111

ABSTRACT

Plant RNA viruses are widespread pathogens that need to interact intricately with their hosts to co-opt numerous cellular factors to facilitate their replication. Currently, there are only a limited number of plant resistance genes against a limited number of viruses. To develop novel antiviral approaches, the interaction network between the given virus and the host cell could be targeted. Yeast (Saccharomyces cerevisiae) has been developed as a surrogate host for tomato bushy stunt virus (TBSV), allowing systematic genome-wide screens to identify both susceptibility and restriction factors for TBSV. Importantly, pro-viral or antiviral functions of several of the characterized yeast proteins have been validated in plant hosts. This paper describes how yeast susceptibility and restriction factors of TBSV could be used as antiviral approaches. The gained knowledge on host factors could lead to novel, inducible, broad-range, and durable antiviral tools against plant viruses.


Subject(s)
Host-Pathogen Interactions , Plant Diseases/immunology , Plant Diseases/virology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/virology , Tombusvirus/immunology , Tombusvirus/physiology , Models, Theoretical
20.
J Virol ; 91(7)2017 04 01.
Article in English | MEDLINE | ID: mdl-28100609

ABSTRACT

Membranous structures derived from various organelles are important for replication of plus-stranded RNA viruses. Although the important roles of co-opted host proteins in RNA virus replication have been appreciated for a decade, the equally important functions of cellular lipids in virus replication have been gaining full attention only recently. Previous work with Tomato bushy stunt tombusvirus (TBSV) in model host yeast has revealed essential roles for phosphatidylethanolamine and sterols in viral replication. To further our understanding of the role of sterols in tombusvirus replication, in this work we showed that the TBSV p33 and p92 replication proteins could bind to sterols in vitro The sterol binding by p33 is supported by cholesterol recognition/interaction amino acid consensus (CRAC) and CARC-like sequences within the two transmembrane domains of p33. Mutagenesis of the critical Y amino acids within the CRAC and CARC sequences blocked TBSV replication in yeast and plant cells. We also showed the enrichment of sterols in the detergent-resistant membrane (DRM) fractions obtained from yeast and plant cells replicating TBSV. The DRMs could support viral RNA synthesis on both the endogenous and exogenous templates. A lipidomic approach showed the lack of enhancement of sterol levels in yeast and plant cells replicating TBSV. The data support the notion that the TBSV replication proteins are associated with sterol-rich detergent-resistant membranes in yeast and plant cells. Together, the results obtained in this study and the previously published results support the local enrichment of sterols around the viral replication proteins that is critical for TBSV replication.IMPORTANCE One intriguing aspect of viral infections is their dependence on efficient subcellular assembly platforms serving replication, virion assembly, or virus egress via budding out of infected cells. These assembly platforms might involve sterol-rich membrane microdomains, which are heterogeneous and highly dynamic nanoscale structures usurped by various viruses. Here, we demonstrate that TBSV p33 and p92 replication proteins can bind to sterol in vitro Mutagenesis analysis of p33 within the CRAC and CARC sequences involved in sterol binding shows the important connection between the abilities of p33 to bind to sterol and to support TBSV replication in yeast and plant cells. Together, the results further strengthen the model that cellular sterols are essential as proviral lipids during viral replication.


Subject(s)
Cholesterol/chemistry , Protoplasts/virology , Saccharomyces cerevisiae/virology , Tombusvirus/physiology , Viral Proteins/chemistry , Amino Acid Sequence , Binding Sites , Cholesterol/physiology , Membrane Microdomains/metabolism , Protein Binding , Nicotiana/virology , Viral Proteins/physiology , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...