Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
New Phytol ; 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38515267

ABSTRACT

Positive-strand RNA viruses co-opt organellar membranes for biogenesis of viral replication organelles (VROs). Tombusviruses also co-opt pro-viral cytosolic proteins to VROs. It is currently not known what type of molecular organization keeps co-opted proteins sequestered within membranous VROs. In this study, we employed tomato bushy stunt virus (TBSV) and carnation Italian ringspot virus (CIRV) - Nicotiana benthamiana pathosystems to identify biomolecular condensate formation in VROs. We show that TBSV p33 and the CIRV p36 replication proteins sequester glycolytic and fermentation enzymes in unique condensate substructures associated with membranous VROs. We find that p33 and p36 form droplets in vitro driven by intrinsically disordered region. The replication protein organizes partitioning of co-opted host proteins into droplets. VRO-associated condensates are critical for local adenosine triphosphate production to support energy for virus replication. We find that co-opted endoplasmic reticulum membranes and actin filaments form meshworks within and around VRO condensates, contributing to unique composition and structure. We propose that p33/p36 organize liquid-liquid phase separation of co-opted concentrated host proteins in condensate substructures within membranous VROs. Overall, we demonstrate that subverted membranes and condensate substructures co-exist and are critical for VRO functions. The replication proteins induce and connect the two substructures within VROs.

2.
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
3.
J Vis Exp ; (197)2023 07 21.
Article in English | MEDLINE | ID: mdl-37590538

ABSTRACT

Cerebral thrombosis, a blood clot in a cerebral artery or vein, is the most common type of cerebral infarction. The study of the cell components of cerebral blood clots is important for diagnosis, treatment, and prognosis. However, the current approaches to studying the cell components of the clots are mainly based on in situ staining, which is unsuitable for the comprehensive study of the cell components because cells are tightly wrapped in the clots. Previous studies have successfully isolated a fibrinolytic enzyme (sFE) from Sipunculus nudus, which can degrade the cross-linked fibrin directly, releasing the cell components. This study established a comprehensive method based on the sFE to study the cell components of cerebral thrombus. This protocol includes clot dissolving, cell releasing, cell staining, and routine blood examination. According to this method, the cell components could be studied quantitatively and qualitatively. The representative results of experiments using this method are shown.


Subject(s)
Thrombosis , Humans , Cerebral Infarction , Fibrin , Staining and Labeling , Veins
4.
J Med Virol ; 95(1): e28129, 2023 01.
Article in English | MEDLINE | ID: mdl-36068190

ABSTRACT

CD7 and CD57 are related to the differentiation and functional stages of CD8+ T cells. However, the role of their combined presence in CD8+ T cells in patients with chronic hepatitis B virus (HBV) infection, especially those with end-stage liver disease, remains unclear. Blood samples from healthy volunteers and patients with chronic hepatitis B were analyzed via Luminex assay and ELISA to measure plasma cytokine levels. Further, recombinant IL-22 was used to stimulate peripheral blood mononuclear cells from healthy volunteers, and the frequency of CD3+ CD4- CD7+ CD57- T cells and apoptosis rates were investigated via flow cytometry. Patients with end-stage liver disease, particularly those with acute to chronic liver failure, showed decreased CD3+ CD4- CD7+ CD57- T cell frequency. Furthermore, the prevalence of CD3+ CD4- CD7+ CD57- T cells was negatively correlated with disease severity, prognosis, and complications (ascites). We also observed that IL-22 promoted apoptosis and brought about a decrease in the number of CD3+ CD4- CD7+ CD57- T cells in a dose-dependent manner. CD3+ CD4- CD7+ CD57- T cells displayed a B and T lymphocyte attenuator (BTLA)high CD25high CD127high immunosuppressive phenotype and showed low interferon-γ, tumor necrosis factor-α, granzyme A, and perforin expression levels. The present findings will elucidate the pathogenesis of HBV-related end-stage liver disease and aid the identification of novel drug targets.


Subject(s)
End Stage Liver Disease , Hepatitis B, Chronic , Humans , Hepatitis B virus , Hepatitis B, Chronic/complications , CD8-Positive T-Lymphocytes , Leukocytes, Mononuclear , T-Lymphocyte Subsets , Disease Progression
5.
Virology ; 572: 1-16, 2022 07.
Article in English | MEDLINE | ID: mdl-35533414

ABSTRACT

Positive-strand RNA viruses induce the biogenesis of viral replication organelles (VROs), which support viral replication in infected cells. VRO formation requires viral replication proteins, co-opted host factors and intracellular membranes. Here, we show that the conserved Atg11 autophagy scaffold protein is co-opted by Tomato bushy stunt virus (TBSV) via direct interactions with the viral replication proteins. Deletion of ATG11 in yeast or knockdown of the homologous Atg11 in plants led to reduced tombusvirus replication, thus indicating pro-viral function for Atg11. Based on co-purification, BiFC and proximity-labeling experiments, we find that Atg11 is co-opted to stabilize virus-induced membrane contact sites (vMCS) within VROs. We propose that the tethering and scaffold function of Atg11 is critical in vMCSs for lipid enrichment. Absence of Atg11 interferes with sterols enrichment in VROs, rendering VROs RNAi-sensitive. Altogether, the expanding roles of co-opted host proteins with tethering functions suggest that the tombusvirus VROs are elaborate structures.


Subject(s)
Saccharomyces cerevisiae Proteins , Tombusvirus , Autophagy , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/metabolism , Host-Pathogen Interactions/genetics , RNA, Viral/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Tombusvirus/genetics , Vesicular Transport Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication/genetics
6.
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
7.
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
8.
J Virol ; 94(12)2020 06 01.
Article in English | MEDLINE | ID: mdl-32269127

ABSTRACT

Positive-strand RNA [(+)RNA] viruses assemble numerous membrane-bound viral replicase complexes (VRCs) with the help of viral replication proteins and co-opted host proteins within large viral replication compartments in the cytosol of infected cells. In this study, we found that deletion or depletion of Sac1 phosphatidylinositol 4-phosphate [PI(4)P] phosphatase reduced tomato bushy stunt virus (TBSV) replication in yeast (Saccharomyces cerevisiae) and plants. We demonstrate a critical role for Sac1 in TBSV replicase assembly in a cell-free replicase reconstitution assay. The effect of Sac1 seems to be direct, based on its interaction with the TBSV p33 replication protein, its copurification with the tombusvirus replicase, and its presence in the virus-induced membrane contact sites and within the TBSV replication compartment. The proviral functions of Sac1 include manipulation of lipid composition, sterol enrichment within the VRCs, and recruitment of additional host factors into VRCs. Depletion of Sac1 inhibited the recruitment of Rab5 GTPase-positive endosomes and enrichment of phosphatidylethanolamine in the viral replication compartment. We propose that Sac1 might be a component of the assembly hub for VRCs, likely in collaboration with the co-opted the syntaxin18-like Ufe1 SNARE protein within the TBSV replication compartments. This work also led to demonstration of the enrichment of PI(4)P phosphoinositide within the replication compartment. Reduction in the PI(4)P level due to chemical inhibition in plant protoplasts; depletion of two PI(4)P kinases, Stt4p and Pik1p; or sequestration of free PI(4)P via expression of a PI(4)P-binding protein in yeast strongly inhibited TBSV replication. Altogether, Sac1 and PI(4)P play important proviral roles during TBSV replication.IMPORTANCE Replication of positive-strand RNA viruses depends on recruitment of host components into viral replication compartments or organelles. Using TBSV, we uncovered the critical roles of Sac1 PI(4)P phosphatase and its substrate, PI(4)P phosphoinositide, in promoting viral replication. Both Sac1 and PI(4)P are recruited to the site of viral replication to facilitate the assembly of the viral replicase complexes, which perform viral RNA replication. We found that Sac1 affects the recruitment of other host factors and enrichment of phosphatidylethanolamine and sterol lipids within the subverted host membranes to promote optimal viral replication. In summary, this work demonstrates the novel functions of Sac1 and PI(4)P in TBSV replication in the model host yeast and in plants.


Subject(s)
Host-Pathogen Interactions/genetics , Phosphatidylinositol Phosphates/metabolism , Phosphoric Monoester Hydrolases/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Tombusvirus/genetics , Virus Replication/genetics , 1-Phosphatidylinositol 4-Kinase/genetics , 1-Phosphatidylinositol 4-Kinase/metabolism , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis/virology , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Endosomes/metabolism , Gene Expression Regulation , Phosphatidylethanolamines/genetics , Phosphatidylethanolamines/metabolism , Phosphoric Monoester Hydrolases/deficiency , Phosphoric Monoester Hydrolases/metabolism , Plant Cells/metabolism , Plant Cells/virology , Plant Leaves/genetics , Plant Leaves/metabolism , Plant Leaves/virology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protoplasts/metabolism , Qa-SNARE Proteins/genetics , Qa-SNARE Proteins/metabolism , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/virology , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction , Sterols/metabolism , Nicotiana/genetics , Nicotiana/metabolism , Nicotiana/virology , Tombusvirus/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , rab5 GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/metabolism
9.
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
10.
PLoS Pathog ; 15(10): e1008092, 2019 10.
Article in English | MEDLINE | ID: mdl-31648290

ABSTRACT

The viral replication proteins of plus-stranded RNA viruses orchestrate the biogenesis of the large viral replication compartments, including the numerous viral replicase complexes, which represent the sites of viral RNA replication. The formation and operation of these virus-driven structures require subversion of numerous cellular proteins, membrane deformation, membrane proliferation, changes in lipid composition of the hijacked cellular membranes and intensive viral RNA synthesis. These virus-driven processes require plentiful ATP and molecular building blocks produced at the sites of replication or delivered there. To obtain the necessary resources from the infected cells, tomato bushy stunt virus (TBSV) rewires cellular metabolic pathways by co-opting aerobic glycolytic enzymes to produce ATP molecules within the replication compartment and enhance virus production. However, aerobic glycolysis requires the replenishing of the NAD+ pool. In this paper, we demonstrate the efficient recruitment of pyruvate decarboxylase (Pdc1) and alcohol dehydrogenase (Adh1) fermentation enzymes into the viral replication compartment. Depletion of Pdc1 in combination with deletion of the homologous PDC5 in yeast or knockdown of Pdc1 and Adh1 in plants reduced the efficiency of tombusvirus replication. Complementation approach revealed that the enzymatically functional Pdc1 is required to support tombusvirus replication. Measurements with an ATP biosensor revealed that both Pdc1 and Adh1 enzymes are required for efficient generation of ATP within the viral replication compartment. In vitro reconstitution experiments with the viral replicase show the pro-viral function of Pdc1 during the assembly of the viral replicase and the activation of the viral p92 RdRp, both of which require the co-opted ATP-driven Hsp70 protein chaperone. We propose that compartmentalization of the co-opted fermentation pathway in the tombusviral replication compartment benefits the virus by allowing for the rapid production of ATP locally, including replenishing of the regulatory NAD+ pool by the fermentation pathway. The compartmentalized production of NAD+ and ATP facilitates their efficient use by the co-opted ATP-dependent host factors to support robust tombusvirus replication. We propose that compartmentalization of the fermentation pathway gives an evolutionary advantage for tombusviruses to replicate rapidly to speed ahead of antiviral responses of the hosts and to outcompete other pathogenic viruses. We also show the dependence of turnip crinkle virus, bamboo mosaic virus, tobacco mosaic virus and the insect-infecting Flock House virus on the fermentation pathway, suggesting that a broad range of viruses might induce this pathway to support rapid replication.


Subject(s)
Alcohol Dehydrogenase/metabolism , Pyruvate Decarboxylase/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/virology , Tombusvirus/growth & development , Virus Replication/physiology , Adenosine Triphosphate/biosynthesis , Fermentation/physiology , Glycolysis/physiology , HSP70 Heat-Shock Proteins/metabolism , Host-Pathogen Interactions/physiology , NAD/metabolism , RNA, Viral/biosynthesis , Saccharomyces cerevisiae/metabolism , Nicotiana/virology , Tombusvirus/genetics , Virus Replication/genetics
11.
Virol Sin ; 34(3): 295-305, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30868360

ABSTRACT

Banana bunchy top virus (BBTV) poses a serious danger to banana crops worldwide. BBTV-encoded protein B4 is a determinant of pathogenicity. However, the relevant molecular mechanisms underlying its effects remain unknown. In this study, we found that a functional peptide could be liberated from protein B4, likely via proteolytic processing. Site-directed mutagenesis indicated that the functional processing of protein B4 is required for its pathogenic effects, including dwarfism and sterility, in plants. The released protein fragment targets host proteins, such as the large subunit of RuBisCO (RbcL) and elongation factor 2 (EF2), involved in protein synthesis. Therefore, the peptide released from B4 (also a precursor) may act as a non-canonical modifier to influence host-pathogen interactions involving BBTV and plants.


Subject(s)
Babuvirus/pathogenicity , Musa/virology , Peptides/metabolism , Plant Diseases/virology , Plant Viral Movement Proteins/metabolism , DNA, Viral , Host-Pathogen Interactions , Peptides/genetics , Plant Viral Movement Proteins/genetics , Plants, Genetically Modified/virology , Nicotiana/genetics , Nicotiana/virology , Virulence
12.
Arch Virol ; 163(5): 1317-1323, 2018 May.
Article in English | MEDLINE | ID: mdl-29392491

ABSTRACT

The nonstructural protein pc6 encoded by rice grassy stunt virus (RGSV) plays a significant role in viral cell-to-cell movement, presumably by transport through plasmodesmata (PD). We confirmed the association of pc6 with PD, and also elucidated the mechanisms of protein targeting to PD. Several inhibitor treatments showed conclusively that pc6 is targeted to PD via the ER-to-Golgi secretory system and actin filaments. In addition, VIII-1 myosin was also found to be involved in pc6 PD targeting. Deletion mutants demonstrated that C-terminal amino acid residues 209-229 (transmembrane domain 2; TM2) are essential for pc6 to move through PD.


Subject(s)
Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Plant Viral Movement Proteins/metabolism , Plasmodesmata/virology , Tenuivirus/metabolism , Viral Nonstructural Proteins/metabolism , Actin Cytoskeleton/metabolism , Myosins/metabolism , Plant Diseases/virology , Protein Transport , Secretory Pathway , Sequence Deletion , Tenuivirus/chemistry , Tenuivirus/genetics , Nicotiana/virology , Viral Nonstructural Proteins/genetics
13.
Arch Virol ; 162(5): 1335-1339, 2017 May.
Article in English | MEDLINE | ID: mdl-28050737

ABSTRACT

Bamboo mosaic virus (BaMV) is a well-characterized virus and a model of virus-host interaction in plants. Here, we identified naturally occurring BaMV isolates from Fujian Province, China and furthermore describe a naturally occurring BaMV coinfection in bamboo (Bambusa xiashanensis) plants. Two different types of BaMV were identified, represented by isolates BaMV-XSNZHA7 (X7) and BaMV-XSNZHA10 (X10). The phylogenetic relationships between X7- and X10-like isolates and published BaMV isolates were determined based on genomic RNA and amino acid sequences. Three clusters were identified, indicating that BaMV is highly diverse. The in planta viral replication kinetics were determined for X7 and X10 in single infections and in an X7/X10 coinfection. The peak viral load during coinfection was significantly greater than that during single infection with either virus and contained a slightly higher proportion of X10 virus than X7, suggesting that X10-like viruses may have a fitness advantage when compared to X7-like viruses.


Subject(s)
Bambusa/virology , Plant Diseases/virology , Potexvirus/classification , Potexvirus/genetics , RNA, Viral/genetics , Amino Acid Sequence/genetics , Base Sequence , China , Coinfection/virology , Host-Pathogen Interactions , Phylogeny , Potexvirus/isolation & purification , Sequence Analysis, RNA , Viral Load
14.
Arch Virol ; 162(2): 505-510, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27743256

ABSTRACT

We characterised the virus-derived small interfering RNAs (vsiRNA) of bamboo mosaic virus (Ba-vsiRNAs) and its associated satellite RNA (satRNA)-derived siRNAs (satsiRNAs) in a bamboo plant (Dendrocalamus latiflorus) by deep sequencing. Ba-vsiRNAs and satsiRNAs of 21-22 nt in length, with both (+) and (-) polarity, predominated. The 5'-terminal base of Ba-vsiRNA was biased towards A, whereas a bias towards C/U was observed in sense satsiRNAs, and towards A in antisense satsiRNAs. A large set of bamboo genes were identified as potential targets of Ba-vsiRNAs and satsiRNAs, revealing RNA silencing-based virus-host interactions in plants. Moreover, we isolated and characterised new isolates of bamboo mosaic virus (BaMV; 6,350 nt) and BaMV-associated satRNA (satBaMV; 834 nt), designated BaMV-MAZSL1 and satBaMV-MAZSL1, respectively.


Subject(s)
Bambusa/virology , Genes, Plant , Potexvirus/genetics , RNA, Satellite/genetics , RNA, Small Interfering/genetics , RNA, Viral/genetics , High-Throughput Nucleotide Sequencing , Host-Pathogen Interactions , Open Reading Frames , Plant Diseases/virology , Potexvirus/classification , Potexvirus/isolation & purification , RNA Interference
15.
Arch Virol ; 161(4): 1091-4, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26923929

ABSTRACT

The complete genome sequences of three isolates of bamboo mosaic virus (BaMV) from mainland China were determined and compared to those of BaMV isolates from Taiwan. Sequence analysis showed that isolate BaMV-JXYBZ1 from Fuzhou shares 98 % nucleotide sequence identity with BaMV-YTHSL14 from nucleotides 2586 to 6306, and more than 94 % nucleotide sequence identity with BaMV-MUZHUBZ2 in other regions. Recombination and phylogenetic analyses indicate that BaMV-JXYBZ1 is a recombinant with one recombination breakpoint. To our knowledge, this is the first report of a BaMV recombinant worldwide.


Subject(s)
Plant Diseases/virology , Poaceae/virology , Potexvirus/genetics , Reassortant Viruses , Bambusa/virology , China , Phylogeny , Potexvirus/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...