Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.625
Filter
1.
Nat Commun ; 15(1): 6817, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39122673

ABSTRACT

Arboviruses can be paternally transmitted by male insects to offspring for long-term persistence, but the mechanism remains largely unknown. Here, we use a model system of a destructive rice reovirus and its leafhopper vector to find that insect ribosome-rescuer Pelo-Hbs1 complex expressed on the sperm surface mediates paternal arbovirus transmission. This occurs through targeting virus-containing tubules constituted by viral nonstructural protein Pns11 to sperm surface via Pns11-Pelo interaction. Tubule assembly is dependent on Hsp70 activity, while Pelo-Hbs1 complex inhibits tubule assembly via suppressing Hsp70 activity. However, virus-activated ubiquitin ligase E3 mediates Pelo ubiquitinated degradation, synergistically causing Hbs1 degradation. Importantly, Pns11 effectively competes with Pelo for binding to E3, thus antagonizing E3-mediated Pelo-Hbs1 degradation. These processes cause a slight reduction of Pelo-Hbs1 complex in infected testes, promoting effective tubule assembly. Our findings provide insight into how insect sperm-specific Pelo-Hbs1 complex is modulated to promote paternal virus transmission without disrupting sperm function.


Subject(s)
Hemiptera , Insect Proteins , Spermatozoa , Animals , Male , Spermatozoa/metabolism , Spermatozoa/virology , Hemiptera/virology , Hemiptera/metabolism , Insect Proteins/metabolism , Insect Proteins/genetics , Arboviruses , HSP70 Heat-Shock Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Reoviridae/physiology , Insect Vectors/virology , Insect Vectors/metabolism , Ribosomes/metabolism , Arbovirus Infections/transmission , Arbovirus Infections/metabolism , Arbovirus Infections/virology
2.
Nat Commun ; 15(1): 6918, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39134555

ABSTRACT

Salivary proteins of insect herbivores can suppress plant defenses, but the roles of many remain elusive. One such protein is glyceraldehyde-3-phosphate dehydrogenase (GAPDH) from the saliva of the Recilia dorsalis (RdGAPDH) leafhopper, which is known to transmit rice gall dwarf virus (RGDV). Here we show that RdGAPDH was loaded into exosomes and released from salivary glands into the rice phloem through an exosomal pathway as R. dorsalis fed. In infected salivary glands of R. dorsalis, the virus upregulated the accumulation and subsequent release of exosomal RdGAPDH into the phloem. Once released, RdGAPDH consumed H2O2 in rice plants owing to its -SH groups reacting with H2O2. This reduction in H2O2 of rice plant facilitated R. dorsalis feeding and consequently promoted RGDV transmission. However, overoxidation of RdGAPDH could cause potential irreversible cytotoxicity to rice plants. In response, rice launched emergency defense by utilizing glutathione to S-glutathionylate the oxidization products of RdGAPDH. This process counteracts the potential cellular damage from RdGAPDH overoxidation, helping plant to maintain a normal phenotype. Additionally, salivary GAPDHs from other hemipterans vectors similarly suppressed H2O2 burst in plants. We propose a strategy by which plant viruses exploit insect salivary proteins to modulate plant defenses, thus enabling sustainable insect feeding and facilitating viral transmission.


Subject(s)
Hemiptera , Hydrogen Peroxide , Oryza , Plant Diseases , Saliva , Animals , Hemiptera/virology , Hydrogen Peroxide/metabolism , Oryza/virology , Oryza/metabolism , Plant Diseases/virology , Saliva/metabolism , Saliva/virology , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Salivary Glands/virology , Salivary Glands/metabolism , Insect Proteins/metabolism , Insect Proteins/genetics , Insect Vectors/virology , Phloem/virology , Phloem/metabolism , Reoviridae/physiology , Glutathione/metabolism , Salivary Proteins and Peptides/metabolism , Plant Viruses/physiology , Plant Defense Against Herbivory
3.
J Virol ; 98(9): e0102824, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39194247

ABSTRACT

Grass carp reovirus (GCRV) is the most virulent pathogen in the genus Aquareovirus, belonging to the family Spinareoviridae. Members of the Spinareoviridae family are known to replicate and assemble in cytoplasmic inclusion bodies termed viroplasms; however, the detailed mechanism underlying GCRV viroplasm formation and its specific roles in virus infection remains largely unknown. Here, we demonstrate that GCRV viroplasms form through liquid-liquid phase separation (LLPS) of the nonstructural protein NS80 and elucidate the specific role of LLPS during reovirus infection and immune evasion. We observe that viroplasms coalesce within the cytoplasm of GCRV-infected cells. Immunofluorescence and transmission electron microscopy indicate that GCRV viroplasms are membraneless structures. Live-cell imaging and fluorescence recovery after photobleaching assay reveal that GCRV viroplasms exhibit liquid-like properties and are highly dynamic structures undergoing fusion and fission. Furthermore, by using a reagent to inhibit the LLPS process and constructing an NS80 mutant defective in LLPS, we confirm that the liquid-like properties of viroplasms are essential for recruiting viral dsRNA, viral RdRp, and viral proteins to participate in viral genome replication and virion assembly, as well as for sequestering host antiviral factors for immune evasion. Collectively, our findings provide detailed insights into reovirus viroplasm formation and reveal the specific functions of LLPS during virus infection and immune evasion, identifying potential targets for the prevention and control of this virus. IMPORTANCE: Grass carp reovirus (GCRV) poses a significant threat to the aquaculture industry, particularly in China, where grass carp is a vital commercial fish species. However, detailed information regarding how GCRV viroplasms form and their specific roles in GCRV infection remains largely unknown. We discovered that GCRV viroplasms exhibit liquid-like properties and are formed through a physico-chemical biological phenomenon known as liquid-liquid phase separation (LLPS), primarily driven by the nonstructural protein NS80. Furthermore, we confirmed that the liquid-like properties of viroplasms are essential for virus replication, assembly, and immune evasion. Our study not only contributes to a deeper understanding of GCRV infection but also sheds light on broader aspects of viroplasm biology. Given that viroplasms are a universal feature of reovirus infection, inhibiting LLPS and then blocking viroplasms formation may serve as a potential pan-reovirus inhibition strategy.


Subject(s)
Carps , Immune Evasion , Reoviridae Infections , Reoviridae , Viral Nonstructural Proteins , Virus Replication , Reoviridae/genetics , Reoviridae/physiology , Animals , Viral Nonstructural Proteins/metabolism , Viral Nonstructural Proteins/genetics , Carps/virology , Reoviridae Infections/virology , Inclusion Bodies, Viral/metabolism , Fish Diseases/virology , Fish Diseases/immunology , Cytoplasm/virology , Cytoplasm/metabolism , Genome, Viral , Cell Line , RNA, Viral/genetics , Phase Separation
4.
Virology ; 598: 110196, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39098183

ABSTRACT

Reovirus (Reo) has shown promising potential in specifically killing tumor cells, and offering new possibilities for ovarian cancer (OC) treatment. However, neutralizing antibodies in the ascites from OC patients greatly limit the further application of Reo. In this study, we employed cationic liposomes (Lipo) to deliver Reo, significantly enhancing its ability to enter OC cells and its effectiveness in killing these cells under ascitic conditions. Pre-treatment with the MßCD inhibitor notably decreased Reo-mediated tumor cell death, indicating that Lipo primarily enables Reo's cellular uptake through caveolin-mediated endocytosis. Our results demonstrate that Lipo effectively facilitates the entry of Reo into the cytoplasm and triggers cell apoptosis. The above findings provide a new strategy to overcome the obstacle of neutralizing antibodies in the clinical application of Reo.


Subject(s)
Antibodies, Neutralizing , Liposomes , Ovarian Neoplasms , Reoviridae , Female , Humans , Ovarian Neoplasms/immunology , Antibodies, Neutralizing/immunology , Reoviridae/immunology , Reoviridae/physiology , Cell Line, Tumor , Oncolytic Virotherapy/methods , Apoptosis , Animals , Cations , Oncolytic Viruses/immunology , Mice
5.
J Invertebr Pathol ; 206: 108170, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39173824

ABSTRACT

The silkworm holds pivotal economic importance, serving not only as a primary source of silk but also as a prominent model organism in scientific research. Nonetheless, silkworm farming remains vulnerable to diverse factors, with viral infections posing the gravest threat to the sericulture industry. Among these, the Bombyx mori cytoplasmic polyhedrosis virus (BmCPV), a member of the Reoviridae family and the cytoplasmic polyhedrosis virus genus, emerges as a significant pathogen in silkworm production. BmCPV infection primarily induces midgut sepsis in silkworms, spreads rapidly, and can inflict substantial economic losses on sericulture production. Presently, effective strategies for preventing and treating BmCPV infections are lacking. Long non-coding RNA (lncRNA) constitutes a class of RNA molecules with transcripts exceeding 200 nt, playing a crucial role in mediating the interplay between pathogens and host cells. Investigation through high-throughput technology has unveiled that BmCPV infection markedly upregulates the expression of Linc20486. This observation suggests potential involvement of Linc20486 in regulating virus replication. Indeed, as anticipated, knockdown of Linc20486 in cells profoundly impedes BmCPV replication, whereas overexpression significantly enhances virus propagation. To probe into the mechanism underlying Linc20486's impact on virus replication, its effects on autophagy, innate immunity, and RNAi-related pathways were scrutinized. The findings revealed that Linc20486 exerts significant influence on the expression of RNAi pathway-related genes, such as Dicer1, Dicer2 and AGO2. This discovery holds promise for unveiling novel avenues to comprehend and combat BmCPV infections in silkworms.


Subject(s)
Bombyx , RNA, Long Noncoding , Reoviridae , Virus Replication , Animals , Bombyx/virology , Reoviridae/physiology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Ribonuclease III/genetics , Ribonuclease III/metabolism , Insect Proteins/genetics , Insect Proteins/metabolism
6.
Fish Shellfish Immunol ; 153: 109861, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39216711

ABSTRACT

This study explored the key molecules and signal pathways in the pathogenesis of grass carp reovirus (GCRV). Using immunoprecipitation mass spectrometry and Co-IP validation, the protein CiANXA4 was identified which interacts indirectly with CiLGP2. CiANXA4 encodes 321 amino acids, including 4 ANX domains. To explore the role of CiANXA4 in the anti-GCRV immune response, we used overexpression and siRNA knockdown in cells. The results showed that overexpression of the CiANXA4 gene significantly increased the mRNA content of vp2 and vp7 in GCRV-infected cells, and the virus titer greatly increased. Knockdown of CiANXA4 significantly inhibited the mRNA levels of vp2 and vp7, and the protein levels of viral protein VP7 also significantly decreased. This suggests that CiANXA4 promotes viral proliferation. Further, we demonstrate that the ANX3 and ANX4 domains are key domains that limit CiANXA4 function by constructing domain-deletion mutants. Finally, we investigated the relationship between CiLGP2 and CiANXA4. RT-PCR and Western blot results showed that CiLGP2 mRNA and protein expression levels were not affected by CiANXA4 overexpression. In contrast, overexpression of CiLGP2 resulted in significant reductions in CiANXA4 mRNA and protein levels. This suggests that the function of CiANXA4 is restricted by CiLGP2, and CiANXA4 is a downstream molecule of CiLGP2. These results reveal that CiANXA4 plays a critical role in the anti-GCRV innate immune response of grass carp, and provides new targets and strategies to develop antiviral drugs and improve disease resistance in grass carp.


Subject(s)
Carps , Fish Diseases , Fish Proteins , Immunity, Innate , Reoviridae Infections , Reoviridae , Animals , Fish Diseases/immunology , Fish Diseases/virology , Carps/genetics , Carps/immunology , Reoviridae Infections/immunology , Reoviridae Infections/veterinary , Reoviridae Infections/virology , Fish Proteins/genetics , Fish Proteins/immunology , Fish Proteins/chemistry , Reoviridae/physiology , Immunity, Innate/genetics , Gene Expression Regulation/immunology , Virus Replication
7.
New Phytol ; 243(6): 2368-2384, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39075808

ABSTRACT

Catalase (CAT) is the main reactive oxygen species (ROS)-scavenging enzyme in plants and insects. However, it remains elusive whether and how insect saliva CAT suppresses ROS-mediated plant defense, thereby promoting initial virus transmission by insect vectors. Here, we investigated how leafhopper Recilia dorsalis catalase (RdCAT) was secreted from insect salivary glands into rice phloem, and how it was perceived by rice chaperone NO CATALASE ACTIVITY1 (OsNCA1) to scavenge excessive H2O2 during insect-to-plant virus transmission. We found that the interaction of OsNCA1 with RdCAT activated its enzymatic activity to decompose H2O2 in rice plants during leafhopper feeding. However, initial insect feeding did not significantly change rice CATs transcripts. Knockout of OsNCA1 in transgenic lines decreased leafhopper feeding-activated CAT activity and caused higher H2O2 accumulation. A devastating rice reovirus activated RdCAT expression and promoted the cosecretion of virions and RdCAT into leafhopper salivary cavities and ultimately into the phloem. Virus-mediated increase of RdCAT secretion suppressed excessive H2O2, thereby promoting host attractiveness to insect vectors and initial virus transmission. Our findings provide insights into how insect saliva CAT is secreted and perceived by plant chaperones to suppress the early H2O2 burst during insect feeding, thereby facilitating viral transmission.


Subject(s)
Catalase , Hemiptera , Hydrogen Peroxide , Insect Vectors , Oryza , Saliva , Animals , Hydrogen Peroxide/metabolism , Hemiptera/virology , Hemiptera/physiology , Saliva/virology , Saliva/enzymology , Catalase/metabolism , Catalase/genetics , Insect Vectors/virology , Oryza/virology , Oryza/genetics , Oryza/enzymology , Reoviridae/physiology , Plant Diseases/virology , Phloem/virology
8.
Virology ; 598: 110171, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39018682

ABSTRACT

In addition to chemotherapy, oncolytic viruses are an efficient treatment for acute myeloid leukemia (AML). Like other oncolytic viruses, the anti-tumor efficacy of reovirus when administered intravenously is reduced due to the presence of neutralizing antibodies. In this study, we evaluated the role of exosomes in human umbilical cord-derived mesenchymal stem cells (UC-MSCs) to deliver reovirus to AML cells. We show that UC-MSCs loaded with reovirus can deliver reovirus to tumor cells without cellular contact. We further demonstrate that the exosome inhibitor, GW4869, inhibits the release of exosomes as well as inhibited the transfer of reovirus from UC-MSCs to tumor cells. Mechanistically, we show that exosomes derived from reovirus-infected UC-MSCs (MSCREO-EXOs) have a tumor lysis effect and transmit reovirus to tumor cells mainly through clathrin-mediated endocytosis (CME) and macropinocytosis. In addition, we demonstrate the feasibility of using MSC-derived exosomes (MSC-EXOs) as a reovirus carrier to exert an anti-tumor effect on AML cells. Collectively, our data indicate that UC-MSCs transfer reovirus to AML cells via exosome release and prompt further study of MSC-EXOs as a potential reovirus carrier to treat AML.


Subject(s)
Exosomes , Leukemia, Myeloid, Acute , Mesenchymal Stem Cells , Oncolytic Virotherapy , Oncolytic Viruses , Umbilical Cord , Humans , Exosomes/metabolism , Mesenchymal Stem Cells/virology , Mesenchymal Stem Cells/metabolism , Leukemia, Myeloid, Acute/therapy , Umbilical Cord/cytology , Oncolytic Viruses/physiology , Oncolytic Virotherapy/methods , Cell Line, Tumor , Reoviridae/physiology , Aniline Compounds/pharmacology , Endocytosis , Benzylidene Compounds
9.
BMC Genomics ; 25(1): 736, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39080552

ABSTRACT

Rice black-streaked dwarf virus (RBSDV) is an etiological agent of a destructive disease infecting some economically important crops from the Gramineae family in Asia. While RBSDV causes high yield losses, genetic characteristics of replicative viral populations have not been investigated within different host plants and insect vectors. Herein, eleven publicly available RNA-Seq datasets from Chinese RBSDV-infected rice, maize, and viruliferous planthopper (Laodelphax striatellus) were obtained from the NCBI database. The patterns of SNP and RNA expression profiles of expected RBSDV populations were analyzed by CLC Workbench 20 and Geneious Prime software. These analyses discovered 2,646 mutations with codon changes in RBSDV whole transcriptome and forty-seven co-mutated hotspots with high variant frequency within the crucial regions of S5-1, S5-2, S6, S7-1, S7-2, S9, and S10 open reading frames (ORFs) which are responsible for some virulence and host range functions. Moreover, three joint mutations are located on the three-dimensional protein of P9-1. The infected RBSDV-susceptible rice cultivar KTWYJ3 and indigenous planthopper datasets showed more co-mutated hotspot numbers than others. Our analyses showed the expression patterns of viral genomic fragments varied depending on the host type. Unlike planthopper, S5-1, S2, S6, and S9-1 ORFs, respectively had the greatest read numbers in host plants; and S5-2, S9-2, and S7-2 were expressed in the lowest level. These findings underscore virus/host complexes are effective in the genetic variations and gene expression profiles of plant viruses. Our analysis revealed no evidence of recombination events. Interestingly, the negative selection was observed at 12 RBSDV ORFs, except for position 1015 in the P1 protein, where a positive selection was detected. The research highlights the potential of SRA datasets for analysis of the virus cycle and enhances our understanding of RBSDV's genetic diversity and host specificity.


Subject(s)
Insect Vectors , Oryza , Plant Diseases , Plant Viruses , Animals , Oryza/virology , Oryza/genetics , Insect Vectors/virology , Insect Vectors/genetics , Plant Viruses/genetics , Plant Diseases/virology , Plant Diseases/genetics , Hemiptera/virology , Hemiptera/genetics , Genetic Variation , RNA-Seq , Transcriptome , Reoviridae/genetics , Zea mays/virology , Zea mays/genetics , Polymorphism, Single Nucleotide , Mutation , Gene Expression Profiling , Open Reading Frames/genetics
10.
Front Immunol ; 15: 1419321, 2024.
Article in English | MEDLINE | ID: mdl-39081319

ABSTRACT

Similar to other RNA viruses, grass carp reovirus, the causative agent of the hemorrhagic disease, replicates in cytoplasmic viral inclusion bodies (VIBs), orchestrated by host proteins and lipids. The host pathways that facilitate the formation and function of GCRV VIBs are poorly understood. This work demonstrates that GCRV manipulates grass carp oxysterol binding protein 1 (named as gcOSBP1) and vesicle-associated membrane protein-associated protein A/B (named as gcVAP-A/B), 3 components of cholesterol transport pathway, to generate VIBs. By siRNA-mediated knockdown, we demonstrate that gcOSBP1 is an essential host factor for GCRV replication. We reveal that the nonstructural proteins NS80 and NS38 of GCRV interact with gcOSBP1, and that the gcOSBP1 is recruited by NS38 and NS80 for promoting the generation of VIBs. gcOSBP1 increases the expression of gcVAP-A/B and promotes the accumulation of intracellular cholesterol. gcOSBP1 also interacts with gcVAP-A/B for forming gcOSBP1-gcVAP-A/B complexes, which contribute to enhance the accumulation of intracellular cholesterol and gcOSBP1-mediated generation of VIBs. Inhibiting cholesterol accumulation by lovastatin can completely abolish the effects of gcOSBP1 and/or gcVAP-A/B in promoting GCRV infection, suggesting that cholesterol accumulation is vital for gcOSBP1- and/or gcVAP-A/B-mediated GCRV replication. Thus, our results, which highlight that gcOSBP1 functions in the replication of GCRV via its interaction with essential viral proteins for forming VIBs and with host gcVAP-A/B, provide key molecular targets for obtaining anti-hemorrhagic disease grass carp via gene editing technology.


Subject(s)
Carps , Cholesterol , Inclusion Bodies, Viral , Receptors, Steroid , Reoviridae , Virus Replication , Animals , Reoviridae/physiology , Carps/virology , Carps/metabolism , Inclusion Bodies, Viral/metabolism , Cholesterol/metabolism , Receptors, Steroid/metabolism , Fish Diseases/virology , Fish Diseases/metabolism , Fish Diseases/immunology , Host-Pathogen Interactions , Reoviridae Infections/veterinary , Reoviridae Infections/metabolism , Reoviridae Infections/virology , Fish Proteins/metabolism , Fish Proteins/genetics , Viral Nonstructural Proteins/metabolism , Viral Nonstructural Proteins/genetics
11.
BMC Genomics ; 25(1): 715, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39048939

ABSTRACT

BF/C2 is a crucial molecule in the coagulation complement cascade pathway and plays a significant role in the immune response of grass carp through the classical, alternative, and lectin pathways during GCRV infection. In vivo experiments demonstrated that the mRNA expression levels of BF/C2 (A, B) in grass carp positively correlated with GCRV viral replication at various stages of infection. Excessive inflammation leading to death coincided with peak levels of BF/C2 (A, B) mRNA expression and GCRV viral replication. Correspondingly, BF/C2 (A, B) recombinant protein, CIK cells and GCRV co-incubation experiments yielded similar findings. Therefore, 3 h (incubation period) and 9 h (death period) were selected as critical points for this study. Transcriptome sequencing analysis revealed significant differences in the expression of BF/C2A and BF/C2B during different stages of CIK infection with GCRV and compared to the blank control group (PBS). Specifically, the BF/C2A_3 and BF/C2A_9 groups exhibited 2729 and 2228 differentially expressed genes (DEGs), respectively, with 1436 upregulated and 1293 downregulated in the former, and 1324 upregulated and 904 downregulated in the latter. The BF/C2B_3 and BF/C2B_9 groups showed 2303 and 1547 DEGs, respectively, with 1368 upregulated and 935 downregulated in the former, and 818 upregulated and 729 downregulated in the latter. KEGG functional enrichment analysis of these DEGs identified shared pathways between BF/C2A and PBS groups at 3 and 9 h, including the C-type lectin receptor signaling pathway, protein processing in the endoplasmic reticulum, Toll-like receptor signaling pathway, Salmonella infection, apoptosis, tight junction, and adipocytokine signaling pathway. Additionally, the BF/C2B groups at 3 and 9 h shared pathways related to protein processing in the endoplasmic reticulum, glycolysis/gluconeogenesis, and biosynthesis of amino acids. The mRNA levels of these DEGs were validated in cellular models, confirming consistency with the sequencing results. In addition, the mRNA expression levels of these candidate genes (mapk1, il1b, rela, nfkbiab, akt3a, hyou1, hsp90b1, dnajc3a et al.) in the head kidney, kidney, liver and spleen of grass carp immune tissue were significantly different from those of the control group by BF/C2 (A, B) protein injection in vivo. These candidate genes play an important role in the response of BF/C2 (A, B) to GCRV infection and it also further confirmed that BF/C2 (A, B) of grass carp plays an important role in coping with GCRV infection.


Subject(s)
Carps , Fish Diseases , Fish Proteins , Reoviridae Infections , Reoviridae , Animals , Carps/genetics , Carps/virology , Carps/immunology , Fish Diseases/virology , Fish Diseases/immunology , Fish Diseases/genetics , Reoviridae Infections/veterinary , Reoviridae Infections/immunology , Reoviridae Infections/genetics , Reoviridae Infections/virology , Fish Proteins/genetics , Fish Proteins/metabolism , Reoviridae/physiology , Gene Expression Profiling , Transcriptome , Virus Replication , Gene Expression Regulation
12.
Plant J ; 119(5): 2484-2499, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39007841

ABSTRACT

Pathogen infection induces massive reprogramming of host primary metabolism. Lipid and fatty acid (FA) metabolism is generally disrupted by pathogens and co-opted for their proliferation. Lipid droplets (LDs) that play important roles in regulating cellular lipid metabolism are utilized by a variety of pathogens in mammalian cells. However, the function of LDs during pathogenic infection in plants remains unknown. We show here that infection by rice black streaked dwarf virus (RBSDV) affects the lipid metabolism of maize, which causes elevated accumulation of C18 polyunsaturated fatty acids (PUFAs) leading to viral proliferation and symptom development. The overexpression of one of the two novel LD-associated proteins (LDAPs) of maize (ZmLDAP1 and ZmLDAP2) induces LD clustering. The core capsid protein P8 of RBSDV interacts with ZmLDAP2 and prevents its degradation through the ubiquitin-proteasome system mediated by a UBX domain-containing protein, PUX10. In addition, silencing of ZmLDAP2 downregulates the expression of FA desaturase genes in maize, leading to a decrease in C18 PUFAs levels and suppression of RBSDV accumulation. Our findings reveal that plant virus may recruit LDAP to regulate cellular FA metabolism to promote viral multiplication and infection. These results expand the knowledge of LD functions and viral infection mechanisms in plants.


Subject(s)
Fatty Acids , Plant Diseases , Plant Proteins , Virus Replication , Zea mays , Zea mays/virology , Zea mays/metabolism , Zea mays/genetics , Plant Proteins/metabolism , Plant Proteins/genetics , Plant Diseases/virology , Fatty Acids/metabolism , Lipid Metabolism , Lipid Droplet Associated Proteins/metabolism , Lipid Droplet Associated Proteins/genetics , Lipid Droplets/metabolism , Lipid Droplets/virology , Plant Viruses/physiology , Gene Expression Regulation, Plant , Reoviridae/physiology
13.
Viruses ; 16(6)2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38932213

ABSTRACT

The mode and outcome of fish-virus interactions are influenced by many abiotic factors, among which water temperature is especially important in poikilothermic fish. Rare minnow Gobiocypris rarus is a eurythermal small cyprinid fish that is sensitive to infection with genotype II grass carp reovirus (GCRV). HSP70, a conservative and key player in heat shock response, is previously identified as an induced pro-viral factor during GCRV infection in vitro. Here, rare minnow was subjected to heat shock treatment (HST), 1 h treatment at 32 °C followed by reverting to a normal temperature of 24 °C, and subsequently challenged with GCRV-II at a dosage of 1 × LD50. The effect of HST on GCRV virulence in vivo was evaluated by calculating virus-associated mortality and viral load in both dead and survival fish. The results revealed that HST enhanced the mortality of rare minnow infected with GCRV; the fact that viral loads in the tissue samples of HST-treated fish were significantly higher than those in samples of the control group at 6, 8 d p.i. reflected a faster infection process due to HST. Quantitative gene expression analysis was further employed to show that the expression levels of Hsp70 in intestine and liver tissues from the HST group declined faster than muscle tissue after HST. HST W/O GCRV challenge upregulated proinflammatory cytokines such as MyD88 and Nf-κB, which was in consistence with the inflammation observed in histopathological analysis. This study shed light on the complexity of the interaction between fish abiotic and biotic stress response, which suggested that HST, an abiotic stress, could enhance the virulence of GCRV in Gobiocypris rarus that involved modulating the gene expression of host heat shock, as well as a pro-inflammatory response.


Subject(s)
Cyprinidae , Fish Diseases , Reoviridae Infections , Reoviridae , Animals , Fish Diseases/virology , Reoviridae/pathogenicity , Reoviridae/genetics , Reoviridae/physiology , Virulence , Reoviridae Infections/virology , Reoviridae Infections/veterinary , Cyprinidae/virology , Viral Load , Carps/virology , Heat-Shock Response , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Hot Temperature
14.
Int J Mol Sci ; 25(12)2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38928143

ABSTRACT

Grass Carp Reovirus (GCRV) and Aeromonas hydrophila (Ah) are the causative agents of haemorrhagic disease in grass carp. This study aimed to investigate the molecular mechanisms and immune responses at the miRNA, mRNA, and protein levels in grass carp kidney cells (CIK) infected by Grass Carp Reovirus (GCRV, NV) and Aeromonas hydrophilus (Bacteria, NB) to gain insight into their pathogenesis. Within 48 h of infection with Grass Carp Reovirus (GCRV), 99 differentially expressed microRNA (DEMs), 2132 differentially expressed genes (DEGs), and 627 differentially expressed proteins (DEPs) were identified by sequencing; a total of 92 DEMs, 3162 DEGs, and 712 DEPs were identified within 48 h of infection with Aeromonas hydrophila. It is worth noting that most of the DEGs in the NV group were primarily involved in cellular processes, while most of the DEGs in the NB group were associated with metabolic pathways based on KEGG enrichment analysis. This study revealed that the mechanism of a grass carp haemorrhage caused by GCRV infection differs from that caused by the Aeromonas hydrophila infection. An important miRNA-mRNA-protein regulatory network was established based on comprehensive transcriptome and proteome analysis. Furthermore, 14 DEGs and 6 DEMs were randomly selected for the verification of RNA/small RNA-seq data by RT-qPCR. Our study not only contributes to the understanding of the pathogenesis of grass carp CIK cells infected with GCRV and Aeromonas hydrophila, but also serves as a significant reference value for other aquatic animal haemorrhagic diseases.


Subject(s)
Aeromonas hydrophila , Carps , MicroRNAs , RNA, Messenger , Reoviridae , Transcriptome , Animals , Carps/genetics , Carps/microbiology , Carps/virology , Carps/immunology , MicroRNAs/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reoviridae/physiology , Proteomics/methods , Fish Diseases/microbiology , Fish Diseases/immunology , Fish Diseases/virology , Fish Diseases/genetics , Gene Expression Profiling , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/veterinary , Gram-Negative Bacterial Infections/microbiology , Gram-Negative Bacterial Infections/genetics , Cell Line , Reoviridae Infections/veterinary , Reoviridae Infections/immunology , Reoviridae Infections/genetics , Gene Regulatory Networks
15.
Fish Shellfish Immunol ; 151: 109712, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38901682

ABSTRACT

The grass carp (Ctenopharyngodon idella) constitutes a significant economic resource within the aquaculture sector of our nation, yet it has been chronically afflicted by the Grass Carp Reovirus (GCRV) disease. The complement system, a vital component of fish's innate immunity, plays a crucial role in combating viral infections. This research investigates the potential role of MASP1, a key molecule in the lectin pathway of the complement system, in the GCRV infection in grass carp. An analysis of the molecular characteristics of MASP1 in grass carp revealed that its identity and similarity percentages range from 35.10 to 91.00 % and 35.30-91.00 %, respectively, in comparison to other species. Phylogenetically, MASP1 in C. idella aligns closely with species such as Danio rerio, Cyprinus carpio, and Carassius carassius, exhibiting chromosomal collinearity with the zebrafish. Subsequent tissue analysis in both healthy and GCRV-infected grass carp indicated that MASP1's basal expression was predominantly in the liver. Post-GCRV infection, MASP1 expression in various tissues exhibited temporal variations: peaking in the liver on day 5, spleen on day 7, and kidney on day 14. Furthermore, employing Complement Component 3 (C3) as a benchmark for complement system activation, it was observed that MASP1 could activate and cleave C3 to C3b. MASP1 also demonstrated an inhibitory effect on GCRV replication (compared with the control group, VP2 and VP7 decreased by 6.82-fold and 4.37-fold) and enhanced the expression of antiviral genes, namely IRF3, IRF7 and IFN1 (compared with the control group, increased 2.25-fold, 45.38-fold and 22.37-fold, respectively). In vivo protein injection experiments substantiated MASP1's influence on the relative mRNA expression levels of C3 in various tissues and its protein expression in serum. This study also verified that C3 could modulate the expression of antiviral genes such as IFN1 and IRF3.


Subject(s)
Carps , Fish Diseases , Fish Proteins , Immunity, Innate , Mannose-Binding Protein-Associated Serine Proteases , Phylogeny , Reoviridae Infections , Reoviridae , Animals , Reoviridae Infections/immunology , Reoviridae Infections/veterinary , Fish Diseases/immunology , Fish Diseases/virology , Carps/immunology , Carps/genetics , Reoviridae/physiology , Fish Proteins/genetics , Fish Proteins/immunology , Mannose-Binding Protein-Associated Serine Proteases/genetics , Mannose-Binding Protein-Associated Serine Proteases/immunology , Immunity, Innate/genetics , Gene Expression Regulation/immunology , Gene Expression Profiling/veterinary , Complement System Proteins/immunology , Complement System Proteins/genetics , Amino Acid Sequence , Sequence Alignment/veterinary
16.
Fish Shellfish Immunol ; 151: 109730, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38942250

ABSTRACT

RLR helicases RIG-I and MDA5, which are known as pattern recognition receptors to sense cytoplasmic viral RNAs and trigger antiviral immune responses, are DExD/H-box helicases. In teleost, whether and how non-RLR helicases regulate RLR helicases to affect viral infection remains unclear. Here, we report that the non-RLR helicase DHX40 from grass carp (namely gcDHX40) is a negative regulator of grass carp reovirus (GCRV) infection and RLR-mediated type I IFN production. GcDHX40 was a cytoplasmic protein. Ectopic expression of gcDHX40 facilitated GCRV replication and suppressed type I IFN production induced by GCRV infection and by those genes involved the RLR antiviral signaling pathway. Mechanistically, gcDHX40 promoted the generation of viral inclusion bodies (VIBs) by interacting with the NS38 protein of GCRV. Additionally, gcDHX40 interacted with RLR helicase, and impaired the formation of RLR-MAVS functional complexes. Taken together, our results indicate that gcDHX40 is a novel important proviral host factor involving in promoting the generation of GCRV VIBs and inhibiting the production of RLR-mediated type I IFNs.


Subject(s)
Carps , DEAD-box RNA Helicases , Fish Diseases , Fish Proteins , Immunity, Innate , Reoviridae Infections , Reoviridae , Viral Nonstructural Proteins , Animals , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/immunology , Viral Nonstructural Proteins/metabolism , Fish Diseases/immunology , Fish Diseases/virology , Fish Proteins/genetics , Fish Proteins/immunology , Carps/immunology , Carps/genetics , Reoviridae Infections/veterinary , Reoviridae Infections/immunology , Reoviridae/physiology , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/immunology , DEAD-box RNA Helicases/metabolism , Immunity, Innate/genetics , RNA Helicases/genetics , RNA Helicases/metabolism , RNA Helicases/immunology , Gene Expression Regulation/immunology
17.
ACS Infect Dis ; 10(8): 2728-2740, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-38873897

ABSTRACT

Nonenveloped viruses employ unique entry mechanisms to breach and infect host cells. Understanding these mechanisms is crucial for developing antiviral strategies. Prevailing perspective suggests that nonenveloped viruses release membrane pore-forming peptides to breach host membranes. However, the precise involvement of the viral capsid in this entry remains elusive. Our study presents direct observations elucidating the dynamically distinctive steps through which metastable reovirus capsids disrupt host lipid membranes as they uncoat into partially hydrophobic intermediate particles. Using both live cells and model membrane systems, our key finding is that reovirus capsids actively deform and permeabilize lipid membranes in a cholesterol-dependent process. Unlike membrane pore-forming peptides, these metastable viral capsids induce more extensive membrane perturbations, including budding, bridging between adjacent membranes, and complete rupture. Notably, cholesterol enhances subviral particle adsorption, resulting in the formation of pores equivalent to the capsid size. This cholesterol dependence is attributed to the lipid condensing effect, particularly prominent at an intermediate cholesterol level. Furthermore, our results reveal a positive correlation between membrane disruption extent and efficiency of viral variants in establishing infection. This study unveils the crucial role of capsid-lipid interaction in nonenveloped virus entry, providing new insights into how cholesterol homeostasis influences virus infection dynamics.


Subject(s)
Capsid , Cell Membrane , Cholesterol , Reoviridae , Virus Internalization , Cholesterol/metabolism , Capsid/metabolism , Cell Membrane/virology , Cell Membrane/metabolism , Reoviridae/physiology , Humans , Animals , Capsid Proteins/metabolism , Capsid Proteins/chemistry
18.
Virus Res ; 346: 199413, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38848818

ABSTRACT

The conversion of Adenosine (A) to Inosine (I), by Adenosine Deaminases Acting on RNA or ADARs, is an essential post-transcriptional modification that contributes to proteome diversity and regulation in metazoans including humans. In addition to its transcriptome-regulating role, ADARs also play a major part in immune response to viral infection, where an interferon response activates interferon-stimulated genes, such as ADARp150, in turn dynamically regulating host-virus interactions. A previous report has shown that infection from reoviruses, despite strong activation of ADARp150, does not influence the editing of some of the major known editing targets, while likely editing others, suggesting a potentially nuanced editing pattern that may depend on different factors. However, the results were based on a handful of selected editing sites and did not cover the entire transcriptome. Thus, to determine whether and how reovirus infection specifically affects host ADAR editing patterns, we analyzed a publicly available deep-sequenced RNA-seq dataset, from murine fibroblasts infected with wild-type and mutant reovirus strains that allowed us to examine changes in editing patterns on a transcriptome-wide scale. To the best of our knowledge, this is the first transcriptome-wide report on host editing changes after reovirus infection. Our results demonstrate that reovirus infection induces unique nuanced editing changes in the host, including introducing sites uniquely edited in infected samples. Genes with edited sites are overrepresented in pathways related to immune regulation, cellular signaling, metabolism, and growth. Moreover, a shift in editing targets has also been observed, where the same genes are edited in infection and control conditions but at different sites, or where the editing rate is increased for some and decreased for other differential targets, supporting the hypothesis of dynamic and condition-specific editing by ADARs.


Subject(s)
Adenosine Deaminase , Fibroblasts , Inosine , RNA Editing , Transcriptome , Animals , Mice , Fibroblasts/virology , Fibroblasts/metabolism , Inosine/metabolism , Inosine/genetics , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Adenosine/metabolism , Adenosine/genetics , Reoviridae Infections/virology , Reoviridae Infections/genetics , Host-Pathogen Interactions , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Reoviridae/genetics , Reoviridae/physiology
19.
PLoS Pathog ; 20(6): e1012318, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38865374

ABSTRACT

Many plant arboviruses are persistently transmitted by piercing-sucking insect vectors. However, it remains largely unknown how conserved insect Toll immune response exerts antiviral activity and how plant viruses antagonize it to facilitate persistent viral transmission. Here, we discover that southern rice black-streaked dwarf virus (SRBSDV), a devastating planthopper-transmitted rice reovirus, activates the upstream Toll receptors expression but suppresses the downstream MyD88-Dorsal-defensin cascade, resulting in the attenuation of insect Toll immune response. Toll pathway-induced the small antibacterial peptide defensin directly interacts with viral major outer capsid protein P10 and thus binds to viral particles, finally blocking effective viral infection in planthopper vector. Furthermore, viral tubular protein P7-1 directly interacts with and promotes RING E3 ubiquitin ligase-mediated ubiquitinated degradation of Toll pathway adaptor protein MyD88 through the 26 proteasome pathway, finally suppressing antiviral defensin production. This virus-mediated attenuation of Toll antiviral immune response to express antiviral defensin ensures persistent virus infection without causing evident fitness costs for the insects. E3 ubiquitin ligase also is directly involved in the assembly of virus-induced tubules constructed by P7-1 to facilitate viral spread in planthopper vector, thereby acting as a pro-viral factor. Together, we uncover a previously unknown mechanism used by plant arboviruses to suppress Toll immune response through the ubiquitinated degradation of the conserved adaptor protein MyD88, thereby facilitating the coexistence of arboviruses with their vectors in nature.


Subject(s)
Arboviruses , Insect Vectors , Signal Transduction , Toll-Like Receptors , Animals , Arboviruses/immunology , Toll-Like Receptors/metabolism , Insect Vectors/virology , Insect Vectors/immunology , Plant Diseases/virology , Plant Diseases/immunology , Reoviridae/physiology , Reoviridae/immunology , Hemiptera/virology , Hemiptera/immunology , Oryza/virology , Oryza/immunology , Insect Proteins/metabolism , Immunity, Innate
20.
Cell Host Microbe ; 32(6): 945-946, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38870904

ABSTRACT

In this issue of Cell Host & Microbe, Shang et al. identify murine neuropilin 1 as a host factor that binds reovirus particles, directing cell entry and contributing to viral dissemination and neurovirulence. This study highlights the reovirus model system to investigate host receptors and their significance in viral pathogenesis.


Subject(s)
Neurons , Neuropilin-1 , Reoviridae , Virus Internalization , Animals , Mice , Neurons/virology , Neuropilin-1/metabolism , Reoviridae/physiology , Reoviridae/genetics , Reoviridae/pathogenicity , Humans , Host-Pathogen Interactions , Reoviridae Infections/virology , Receptors, Virus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL