SlidePlayer

  • My presentations

Auth with social network:

Download presentation

We think you have liked this presentation. If you wish to download it, please recommend it to your friends in any social system. Share buttons are a little bit lower. Thank you!

Presentation is loading. Please wait.

REPLICATION OF THE VIRUS

Published by Stanley Freeman Modified over 9 years ago

Similar presentations

Presentation on theme: "REPLICATION OF THE VIRUS"— Presentation transcript:

REPLICATION OF THE VIRUS

Ch. 19 Viruses Objective: EK 3.C.3: Viral replication results in genetic variation, and viral infection can introduce genetic variation into the hosts.

presentation on viral replication

Essentials of Virology

presentation on viral replication

LECTURE PRESENTATIONS For CAMPBELL BIOLOGY, NINTH EDITION Jane B. Reece, Lisa A. Urry, Michael L. Cain, Steven A. Wasserman, Peter V. Minorsky, Robert.

presentation on viral replication

Viruses (Ch. 18).

presentation on viral replication

Max Sanam.  Understand stages in animal virus replication  Compare and contrast the multiplication cycle of DNA and RNA-containing animal viruses 

presentation on viral replication

Viruses of Bacteria Chapter 13. General Characteristics of Viruses Non-living entities Not considered organisms Can infect organisms of every domain All.

presentation on viral replication

 Non-living entities  Can infect organisms of every domain  Commonly referred to by organism they infect  Viruses that infect bacteria: Bacteriophage.

presentation on viral replication

Viruses, Viroids, and Prions

presentation on viral replication

Viruses: Bacterial and Animal Types

presentation on viral replication

Viruses, part 2.

presentation on viral replication

Viruses.  What is a virus? Defined by their inability to replicate/multiply without utilizing a host cells reproductive mechanisms. Only contain ONE.

presentation on viral replication

Copyright © 2004 Pearson Education, Inc., publishing as Benjamin Cummings PowerPoint ® Lecture Slide Presentation prepared by Christine L. Case Microbiology.

presentation on viral replication

Lecture 29: Viruses 0.5 m.

presentation on viral replication

Bacteriophages ( a.k.a. Phages) Viruses that target bacteria Virus defining characteristics: parasitic entities Nucleic acid molecules protected by protein.

presentation on viral replication

Associate professor in microbiology

presentation on viral replication

Viruses: a kind of “borrowed life” HIV infected T-cell.

presentation on viral replication

Viruses Part II. It was not until the 1950s that scientists were able to isolate and identify how viruses multiplied in a host It was not until the 1950s.

presentation on viral replication

Scene from the 1918 influenza pandemic.. Scene from the 2003 SARS Scare.

presentation on viral replication

Lecture 9 Viruses, Viroids, Prions

presentation on viral replication

Viruses Viral Structure Growing (Cultivating) Prokaryote Types Eukaryote Types Obligate Intracellular Parasites: Require living host cell in order to replicate.

About project

© 2024 SlidePlayer.com Inc. All rights reserved.

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • View all journals
  • Explore content
  • About the journal
  • Publish with us
  • Sign up for alerts
  • Open access
  • Published: 23 August 2024

Regulation viral RNA transcription and replication by higher-order RNA structures within the nsp1 coding region of MERS coronavirus

  • Yutaka Terada 1 , 2 ,
  • Sodbayasgalan Amarbayasgalan 3 ,
  • Yoshiharu Matsuura 4 , 5 , 6 &
  • Wataru Kamitani 1 , 3  

Scientific Reports volume  14 , Article number:  19594 ( 2024 ) Cite this article

100 Accesses

Metrics details

  • Microbiology

Coronavirus (CoV) possesses numerous functional cis-acting elements in its positive-strand genomic RNA. Although most of these RNA structures participate in viral replication, the functions of RNA structures in the genomic RNA of CoV in viral replication remain unclear. In this study, we investigated the functions of the higher-order RNA stem-loop (SL) structures SL5B, SL5C, and SL5D in the ORF1a coding region of Middle East respiratory syndrome coronavirus (MERS-CoV) in viral replication. Our approach, using reverse genetics of a bacterial artificial chromosome system, revealed that SL5B and SL5C play essential roles in the discontinuous transcription of MERS-CoV. In silico analyses predicted that SL5C interacts with a bulged stem-loop (BSL) in the 3′ untranslated region, suggesting that the RNA structure of SL5C is important for viral RNA transcription. Conversely, SL5D did not affect transcription, but mediated the synthesis of positive-strand genomic RNA. Additionally, the RNA secondary structure of SL5 in the revertant virus of the SL5D mutant was similar to that of the wild-type, indicating that the RNA structure of SL5D can finely tune RNA replication in MERS-CoV. Our data indicate novel regulatory mechanisms of viral RNA transcription and replication by higher-order RNA structures in the MERS-CoV genomic RNA.

Similar content being viewed by others

presentation on viral replication

Structures and functions of coronavirus replication–transcription complexes and their relevance for SARS-CoV-2 drug design

presentation on viral replication

Structure of replicating SARS-CoV-2 polymerase

presentation on viral replication

Structural insights into ribonucleoprotein dissociation by nucleocapsid protein interacting with non-structural protein 3 in SARS-CoV-2

Introduction.

Coronaviruses (CoVs) (order Nidovirales, family Coronaviridae, subfamily Coronavirinae) are enveloped and contain large single-stranded, positive-sense RNA. Most CoVs cause enteric and/or respiratory diseases in both mammals and birds 1 . In humans, most CoVs cause mild illnesses, including the common cold 2 , 3 , 4 , 5 . Three lethal human CoVs have emerged as zoonotic pathogens within this century. One is the severe acute respiratory syndrome CoV (SARS-CoV), which emerged in 2002 in China 6 , 7 , and second is the Middle East respiratory syndrome CoV (MERS-CoV), which emerged in Saudi Arabia in 2012 8 . MERS-CoV remains endemic, with more than 2000 cases and 858 deaths reported. Third, most recently, coronavirus disease 2019 (COVID-19), caused by SARS-CoV-2, became a global pandemic 9 , 10 , 11 .

Numerous RNA viruses utilize functional RNAs in their genomes as cis-acting elements for efficient viral replication 12 , 13 , 14 , 15 . One of the most well-known functional RNA structures is the internal ribosome entry site (IRES) 16 , 17 , which induces efficient translation of viral proteins by directly recruiting the host translational system. As CoVs have the largest RNA genome among all RNA viruses, they possess several types of RNA structures 18 , 19 . The stem-loop (SL) structure within the non-structural protein 15 (nsp15)-coding region of the mouse hepatitis virus (MHV) acts as a packaging signal 20 . Similar to other viruses, higher-order RNA structures within the 5′ or 3′ untranslated region (UTR) of the CoV genome have been proposed to be cis-acting elements 21 , 22 , 23 , 24 , 25 , 26 . We discovered that nsp1 from SARS-CoV and MERS-CoV are RNA-binding proteins that bind and utilize the SL1 region within the 5′ UTR of viral RNA as a gene marker for viral RNA. This recognition plays an important role in evading translational shutoff and RNA degradation induced by nsp1 27 , 28 . RNA structures in CoV genomic RNA have been proposed to act as cis-acting elements for replication based on experiments using defective interfering (DI)-RNA 22 , 24 , 25 , 29 . However, the detailed mechanisms through which cis-acting RNA in the CoV genome regulate viral replication remain unclear.

Recently, we used a reverse genetics approach in a bacterial artificial chromosome (BAC) system and discovered that RNA structures of the SL5 region in MERS-CoV RNA participate in viral replication 28 . The SL5 RNA region, which consists of SL5B, SL5C, and SL5D, encodes the 5′ UTR and the nsp1 coding region. However, the infectious virus could not be recovered from the cDNA clone because of an amino acid substitution at position 13 of nsp1 in the SL5 region, which disrupted the RNA structure and impaired viral replication. In contrast, the cDNA clone that retained the RNA structure of SL5 produced an infectious virus. Our previous results suggest that higher-order RNA structures in SL5 of the MERS-CoV genome play critical roles in viral replication. Although the importance of RNA structures for viral replication has been discussed, the functions of specific RNA structures during the CoV life cycle remain unclear.

In this study, we examined the biological effects of RNA structures in SL5 of the MERS-CoV genome on viral RNA transcription and replication using a reverse genetics approach in a BAC system. Our mutant viruses showed that the RNA structures of both SL5B and SL5C were important for the production of subgenomic mRNAs. In contrast, the RNA structure of SL5D proved to be critical for the replication of positive-stranded genomic RNA. Our results suggest that the higher-order RNA structures of SL5 in the MERS-CoV RNA genome play critical roles in regulating RNA transcription and replication during the viral life cycle.

Effects of higher-order RNA structures, SL5B, SL5C, and SL5D in viral propagation

In a previous report, the RNA sequence consisting of SL5B, SL5C, and SL5D within the nsp1-coding region was shown to likely function as a cis-acting element in viral propagation 28 . To examine the roles of SL5B, SL5C, and SL5D in viral propagation, we constructed cDNA clones carrying mutations in these regions (Fig.  1 A,B). The SL5B-Loop and SL5C-Loop mutants (pBAC-MERS-SL5B-Loop and pBAC-MERS-SL5C-Loop, respectively) were designed to retain their RNA structures within the loop regions. SL5B-R, SL5C-R, and SL5D-R, which contain mutations on the right side of each stem region, were designed to disrupt the RNA structures (pBAC-MERS-SL5B-R, pBAC-MERS-SL5C-R, and pBAC-MERS-SL5D-R, respectively). To retain the secondary RNA structures in each SL, mutations were introduced to the left side of each stem region, yielding SL5B-RL, SL5C-RL, and SL5D-RL (pBAC-MERS-SL5B-RL, pBAC-MERS-SL5C-RL, and pBAC-MERS-SL5D-RL, respectively). Nucleotide substitutions in SL5C-RL and SL5D-RL caused non-synonymous mutations in the nsp1 protein. The secondary RNA structures of the mutants used in this study using Mfold analysis 30 are shown in Fig.  1 C. As expected, SL5B-Loop, SL5B-RL, SL5C-Loop, SL5C-RL, and SL5D-RL retained the same RNA structure as their wild-type (WT) counterparts. In contrast, SL5B-R, SL5C-R, and SL5D-R were predicted to disrupt the RNA structures of SL5.

figure 1

Construction of SL5 structure disrupting mutants. ( A ) RNA structures predicted by Mfold. Numbers indicate the nucleotide positions in the MERS-CoV EMC strain (DDBJ accession No. NC_019843). Circles indicate the mutation sites for each of the SL5 mutants shown in ( B ). ATG: Start codon of ORF1a. ( B ) Nucleotide mutations of SL5B, SL5C, and SL5D mutants used in this study. ( C ) Predicted secondary RNA structures of each mutant.

To investigate the functional significance of SL5 structures in viral propagation, we transfected full-length cDNA clones of either WT or mutant viruses into Huh7 cells, and viral titers in the culture supernatants were determined using a 50% tissue culture infectious dose (TCID 50 ) assay with Vero cells (Fig.  2 A). Viral infectivity in cells transfected with pBAC-MERS-WT reached 3.16E+05 TCID 50 /mL 72 h post-transfection. Although the viral infectivity levels in cells transfected with pBAC-MERS-SL5B-Loop and pBAC-MERS-SL5B-RL were low at 48 h post-transfection, both values increased to approximately 1.0E+04 TCID 50 /mL at 72 h post-transfection (Fig.  2 A). In particular, there was a statistically significant (two-way ANOVA) difference in the infectious virus titer in the 5B-R, 5C-R, and 5D-R samples at 72 h compared to the WT. The predicted RNA secondary structures of SL5B-Loop and SL5B-RL indicated that both mutants retained the RNA secondary structure from nts 1–460, which contains the nsp1-coding region (Fig.  1 C). In contrast, the SL5B-R mutation disrupted the RNA secondary structure (Fig.  1 C). In combination with our finding that no viral progeny was recovered from the pBAC-MERS-SL5B-R transfected cells (Fig.  2 A), data suggest that the RNA structure of SL5B is important for efficient viral replication.

figure 2

Effects of SL5B, SL5C, and SL5D RNA structures on viral propagation. ( A ) Viral growth of SL5 mutants. Huh7 cells were transfected with the MERS-CoV cDNA clones and cultured. Experiments were carried out in triplicate and representative results were shown. Culture supernatants were collected to harvest the virus, and viral titers were determined by TCID 50 assay using Vero cells. Mock indicates the transfected Huh7 cells with backbone BAC plasmid without any viral sequence. **** P < 0.0001. ( B ) Real-time RT-PCR results evaluating viral RNA transcription. Huh7 cells transfected with each cDNA clone were cultured for the indicated times. Total RNA was extracted from each infected cell, and real-time RT-PCR targeting sg N mRNA was performed. The sg N mRNA levels were normalized to the GAPDH mRNA levels. **** P < 0.0001. # represents the limit of detection.  ( C ) Immunoblot analysis for the detection of MERS-CoV N protein. Cell lysates of Huh7 cells transfected with each cDNA clone were subjected to western blot analysis using anti-MERS-CoV N antibody (a-N) and anti-actin antibody (a-Actin). ( D ) Luciferase activities of replicons carrying each of the SL5 mutants. 293T cells were transfected with each cDNA replicon and cultured for 48 h. Nluc activities in cultured cells were measured using confocal microscopy. Firefly luciferase activities from pGL3-control plasmid were also measured as an internal control. Polymerase dead mutant (SAA) was used as a negative control.* P <0.05.

As shown in Fig.  2 A, SL5C mutants presented features similar to those of SL5B. Both SL5C-Loop and SL5C-RL produced viral progeny in the culture supernatants, whereas SL5C-R, which had a disrupted secondary structure, produced no viral progeny (Figs.  1 C and 2 A). As expected, SL5D-RL, which retained its RNA secondary structure, produced viral progeny (Fig.  2 A). Unlike SL5B-R and SL5C-R, SL5D-R released viral progeny into the culture supernatant; however, the viral titer was lower than that of the WT construct (Fig.  2 A). These results suggest that the RNA structures of SL5B, SL5C, and SL5D are important for efficient viral propagation, and that the SL5D structure regulates viral propagation through an unknown mechanism that is different from that of SL5B and SL5C.

Effect of SL5 RNA secondary structure on viral RNA transcription and viral protein expression

To examine the functional significance of SL5 RNA secondary structures in viral RNA transcription, we extracted total RNA from Huh7 cells transfected with cDNA clones and performed real-time RT-PCR to measure the expression levels of subgenomic (sg) mRNA for nucleocapsid (N) protein (sg N mRNA) (Fig.  2 B). As expected, the four mutants that retained their RNA secondary structures, SL5B-Loop, SL5B-RL, SL5C-Loop, and SL5C-RL–efficiently produced sg N mRNA (Fig.  2 B). In contrast, the mutants with disrupted RNA secondary structures, SL5B-R and SL5C-R, did not produce sg N mRNA (Fig.  2 B). A statistically significant difference (two-way ANOVA) was observed between the amount of 5B-R and 5D-R viral RNA at 72 h compared to the WT. Notably, the amount of 5C-R viral RNA was below the detection limit. Although the infectious viral titers in pBAC-MERS-SL5D-R-transfected cells were substantially lower than those in pBAC-MERS-SL5D-RL-transfected cells (Fig.  2 A), the expression levels of sg N mRNA in pBAC-MERS-SL5D-R-transfected cells were similar to those in the SL5D-RL mutant (Fig.  2 B). These results indicate that the disruption of SL5B and SL5C, but not SL5D, reduces sg N mRNA transcription.

Next, we measured the expression levels of the viral N protein in Huh7 cells transfected with cDNA clones using immunoblotting (Fig.  2 C). The amount of N protein increased in pBAC-MERS-WT-transfected cells in a time-dependent manner. N protein expression was detected in cells transfected with SL5B-Loop, SL5B-RL, SL5C-Loop, and SL5C-RL at 72 h post-transfection. However, the levels of the N protein were lower than those in cells expressing the WT virus (Fig.  2 C, left panels). In contrast, no expression of the N protein was observed in cells expressing the RNA-disrupting mutants SL5B-R and SL5C-R (Fig.  2 C, left panels), which was due to a significant reduction in sg N mRNA production (Fig.  2 B). Unlike SL5B-R and SL5C-R mutants, the SL5D-R mutant produced the N protein (Fig.  2 C, right panel); however, the amount of N protein produced was lower than that in cells expressing SL5D-RL (Fig.  2 C, right panels). These results suggest that the RNA structures in SL5 regulate the transcription of sg N mRNA.

To confirm the functional significance of the SL5 RNA structure in viral RNA transcription, we used a MERS-CoV RNA replicon system in which MERS-CoV RNA efficiently replicates but does not produce any infectious particles. The MERS-CoV replicon system was engineered to contain the nano luciferase (Nluc) gene and the TRS-M 100 nt upstream of the Nluc gene to increase Nluc expression (designated pBAC-MERS-Rep). As a negative control, we used a replicon cDNA clone containing dead RNA-dependent RNA polymerase (RdRP; nsp12). To generate a series of MERS-CoV replicon cDNA clones with SL5 mutants, pBAC-MERS-Rep was used as the template. Consistent with the expression levels of sg N mRNA (Fig.  2 B), luciferase activity in 293T cell lysates at 48 h post-transfection with replicon cDNA, excluding SL5B-R and SL5C-R, was significantly increased (Fig.  2 D). There was also a significant difference in the amount of replication of the replicon DNA of 5B-R and 5C-R as compared to that of the WT. These results suggest that the RNA structures of SL5B and SL5C, but not that of SL5D, play critical roles in viral RNA transcription. In contrast, the RNA structure of SL5D may function only after viral RNA transcription.

Effects of SL5B and SL5C RNA structures on viral RNA transcription

Using a reverse genetics system for MERS-CoV, we discovered that mutations in SL5B-R and SL5C-R arrested the viral RNA transcription (Fig.  2 B). This may be due to the low stability of the genomic RNA conferred by the introduction of nucleotide substitutions. To test this possibility, we evaluated the levels of genomic RNA at earlier time points after the transfection of WT, SL5B-R, SL5B-RL, SL5C-R, and SL5C-RL cDNA into Huh7 cells. Based on real-time RT-PCR analyses, the expression levels of genomic RNA at 0, 4, 8, and 12 h post-transfection were similar in all conditions (Fig.  3 A); thus, suggesting that nucleotide substitutions in SL5B and SL5C did not affect viral genomic RNA stability.

figure 3

Functional step of SL5B and SL5C in MERS-CoV lifecycle. ( A ) Genomic RNA stabilities of viruses containing mutant SL5B or SL5C regions with disrupted secondary structures. Huh7 cells were transfected with each cDNA clone and cultured for 0, 4, 8, and 12 h. Total RNA were extracted from the cells and subjected to real-time RT-PCR targeting ORF1a region to measure the levels of genomic RNA. The levels of genomic RNAs were normalized to the GAPDH mRNA levels. ( B ) Schematic of pcDNA3.1-SL5-fluc plasmid. MERS-CoV sequence from nts 265–343 was inserted between the CMV promoter and the firefly luciferase gene (fluc). Gray squares indicate the nucleotide regions of SL5B, SL5C, and SL5D. ( C ) Effects of SL5B and SL5C structures on protein translation. 293T cells were transfected with pcDNA3.1-SL5-fluc or SL5B/SL5C mutants. pcDNA3.1-fluc without the viral sequence was used as a positive control. Renilla luciferase reporter plasmid, pRL-SV40, was used as the internal control. After 24 h of incubation, the transfected cells were collected, and luciferase activities were measured. Firefly luciferase activities were normalized to the Renilla luciferase activities. ( D ) Schematic diagram of the reporter assay for detecting the MERS-CoV protease, nsp5. cDNA clones of WT, SL5B mutant, or SL5C mutant were co-transfected into Huh7 cells with the pGlo-VRLQS biosensor plasmid. Translated proteins from pGlo-VRLQS do not show luciferase activity. Cleavage caused by MERS-CoV nsp5 increases luciferase activity. Transfected Huh7 cells were cultured for 24 and 48 h, and luciferase activities in cells were measured using confocal microscopy. ( E ) Expression of nsp5 in SL5B and SL5C mutants. The results of the MERS-CoV nsp5 biosensor assay described in ( D ). The Nsp5 deletion mutant, pBAC-MERS-nsp5-rpsL, was used as a negative control (Control).

In the CoV lifecycle, after uncapsidation, genomic RNA functions as an mRNA to produce nsps that are critical for subsequent viral RNA transcription and replication 31 . Next, we tested the effects of nucleotide substitutions in SL5B or SL5C on genomic RNA translation using a firefly luciferase reporter plasmid carrying nts 265–348 of MERS-CoV, including SL5B, SL5C, and SL5D, upstream of the firefly luciferase gene (Fig.  3 B). A reporter plasmid without the viral sequence was used as a control (fluc). Luciferase activity in 293T cells at 24 h post-transfection indicated that the levels in all SL5 mutants were comparable to those associated with the WT sequence (SL5) (Fig.  3 C). These results indicated that nucleotide substitutions in SL5B and SL5C did not affect genomic RNA translation.

As shown in Fig.  3 A,C, the impairment of viral RNA replication by SL5B-R and SL5C-R cDNA was not due to decreased RNA stability or loss of genomic RNA translation. Next, we investigated whether the secondary RNA structures of SL5B and SL5C affected viral nsp(s) expression using a luciferase-based biosensor system to detect nsp5 protease activity, as shown in Fig.  3 D 32 . The specific amino acid sequence (VRLQS) recognized and cleaved by MERS-CoV nsp5 was inserted into a firefly luciferase reporter plasmid to produce pGlo-30F-VRLQS 32 . Luciferase activities of Huh7 cells co-transfected with pGlo-30F-VRLQS and MERS-CoV cDNA clones are shown in Fig.  3 E. Similar to the WT, all SL5B and SL5C mutants showed increased luciferase activity, suggesting that nucleotide substitutions in SL5B and SL5C did not impair genomic RNA translation. Thus, the structures of SL5B and SL5C are key factors in viral RNA transcription after the first round of translation and they play a role in nsps production.

Identification of the RNA-RNA interaction region between SL5 and 3′ UTR

In several positive-stranded RNA viruses, RNA-RNA interactions in genomic RNA are important for regulating viral RNA transcription or translation 33 . As shown in (Figs. 1 , 2 , 3 ), the RNA structures of SL5B and SL5C regulate viral RNA transcription. To identify the RNA-RNA interaction sites of SL5 in the genomic RNA of MERS-CoV, we used the LRIscan software 33 ; 30,258 RNA-RNA interactions were predicted to occur within the MERS-CoV genome (Fig.  4 A). Although the p-value above a commonly used threshold (0.277) (Fig.  4 B), the stem region of SL5C (nts 301–305) was predicted to interact with the bulged stem-loop (BSL, nts 29,829–29,833) of the 3′ UTR (Fig.  4 B). These results suggest that SL5C may play an important role in regulating viral RNA transcription through the circularization of genomic RNA via its interaction with the BSL in the 3′ UTR.

figure 4

Prediction of long RNA-RNA interaction with SL5. ( A ) Plot of all predicted LRI values with P < 0.005 observed in four MERS-CoV genomes. The outer circle represents the genome information of MERS-CoV. The inner circle shows all predicted interactions between all genome positions. The plot was created with Circos 50 . ( B ) Predicted RNA-RNA interaction between SL5C and the BSL within the 3′UTR based on the LRIscan analysis. The nucleotide positions in MERS-CoV EMC are indicated (DDBJ accession No. NC_019843).

SL5D regulates positive-strand genomic RNA replication

Although the infectious viral titer was low in pBAC-MERS-SL5D-R-transfected Huh7 cells (Fig.  2 A), the expression levels of sg N mRNA and protein were comparable to those in pBAC-MERS-SL5D-RL-transfected cells (Fig.  2 B,C). Furthermore, RNA transcription and replication occurred efficiently in the SL5D-R replicon DNA-transfected cells (Fig.  2 D). These results led us to hypothesize that viral assembly was impaired in the pBAC-MERS-SL5D-R-transfected cells. We further believe that the identification of infected cells in the SL5D-R sample was difficult because of the low infectious viral titer in the pBAC-MERS-SL5D-R-transfected cells (Fig.  2 A).

To overcome this difficulty, we used confocal microscopy to identify infected cells using reporter viruses carrying the fluorescent protein ZsGreen (data not shown). In addition, we utilized transmembrane serine protease 2 (TMPRSS2)-expressing Vero-TMPRSS2 cells 34 to obtain a reasonable amount of rMERS-ZsGreen virus containing the SL5D-R mutation for transmission electron microscopy (TEM) analysis. The infectious viral titer of the WT and mutant viruses was higher in Vero-TMPRSS2 cells than that in Vero cells (data not shown). Vero cells inoculated with WT or SL5D-R reporter viruses (MOI = 0.1) were cultured for 24 h and fixed to identify infected cells by confocal microscopy. The infected cells were subsequently analyzed using TEM. As shown in Fig.  5 B, the SL5D-R virus particles were approximately 100 nm in size, similar to those observed in infected WT cells. These results suggest that viral particles were produced in SL5D-R-infected cells.

figure 5

Functional step of SL5D in MERS-CoV lifecycle. ( A , B ) Correlative light and electron microscopy (CLEM) analysis of rMERS-ZsGreen or rMERS-ZsGreen-SL5D-R mutants. Vero cells were inoculated with rMERS-ZsGreen or rMERS-ZsGreen-SL5D-R (MOI = 0.1) and cultured for 24 h. ( A ) Infected cells were identified by green fluorescence and (B) subjected to transmission electron microscopy (TEM) analysis. Black arrows indicate the viral particles inside the infected cells. White arrows indicate the viral particles on the cell surface. Scale bar = 1 µm. N nucleolus. Bottom: Zoomed image of red square areas from top TEM image. ( C ) Viral release of rMERS-ZsGreen or rMERS-ZsGreen-SL5D-R mutants from Vero cells. Vero cells were inoculated with rMERS-ZsGreen or rMERS-ZsGreen-SL5D-R (MOI = 0.01) and cultured. At the indicated time, culture supernatants were collected. Cells were collected in fresh medium, stored frozen, and then thawed before use. Viral titers of both supernatants (Sup) and cell (Cell) samples were measured to determine the levels of released virus relative to that inside the cell. * P < 0.05  ( D ) Examination of the levels of strand-specific genomic RNA in wildtype or the SL5D-R mutant virus. Huh7 cells were transfected with pBAC-MERS or pBAC-MERS-SL5D-R and cultured for 72 h, and total RNAs were extracted. RNA samples were reverse-transcribed using positive- or negative-strand specific primers and then treated with RNaseH. The strand-specific cDNAs were subjected to real-time PCR targeting the ORF1a region. * P < 0.05.

Next, we evaluated the effects of the SL5D secondary structure on viral release from infected cells by inoculating Vero cells with either rMERS-ZsGreen or rMERS-ZsGreen-SL5D-R and measuring viral titers in the culture supernatants. The viral release was calculated by dividing the infectious viral titer in the culture supernatants by that in the cells (Fig.  5 C). Statistically, there was a significant difference in the amount of virus released into the supernatant of the cells infected with the WT and SL5D-R virus. Although the viral release from cells infected with the WT virus increased in a time-dependent manner, no increase was observed for the SL5D-R-infected cells (Fig.  5 C). These results suggest that the loss of the secondary structure in SL5D attenuates viral release.

Although viral particles were observed in the SL5D-R infected cells (Fig.  5 B), the amount of virus released from the infected cells was low (Fig.  5 C). Next, we examined the amount of positive-strand viral genomic RNA in SL5D-R-infected cells using real-time RT-PCR. Because positive-stranded genomic RNA is incorporated into infectious viral particles, we hypothesized that the loss of viral release was due to the decreased production of genomic RNA in SL5D-R-infected cells. To test this hypothesis, Huh7 cells transfected with pBAC-MERS-wt or pBAC-MERS-SL5D-R were cultured for 72 h, and total RNA was extracted and subjected to real-time RT-PCR to detect either positive- or negative-strand genomic RNA. The amount of positive-strand genomic RNA was divided by that of the negative-strand genomic RNA. As shown in Fig.  5 D, the amount of positive-stranded genomic RNA relative to negative-stranded genomic RNA in cells transfected with pBAC-MERS-WT was > 1.0, which was a statistically significant increase. In contrast, the value in pBAC-MERS-SL5D-R-transfected cells was 1.0, indicating that the synthesis of positive-strand genomic RNA was impaired in SL5D-R-infected cells. These results suggest that the structure of SL5D plays an important role in the synthesis of positive-stranded genomic RNA.

Emergence of revertant virus from SL5D-R

Although the infectious viral titer in the culture supernatants released from pBAC-MERS-SL5D-R-transfected Huh7 cells was low, we successfully recovered the mutant virus from these cells (Fig.  2 A). To understand the biological significance of the SL5D structure in viral RNA replication, we sought to obtain a revertant virus by serially passaging Vero cells. Although the cytopathic effect (CPE) was still weak in infected Vero cells at passages 1 and 2, it became much stronger in cells after passage 10. The nucleotide sequences of the SL5D mutation sites in the P1, P2, P5, and P10 virus samples were examined. Although the introduced mutation (G335C/C338A) in SL5D-R was retained in the P1 and P2 viruses, a revertant mutation (C338) at position 338 was identified in the P5 virus (Fig.  6 A,B). Additionally, the revertant mutation (C338) was predominant after passage 10 (Fig.  6 B). The C population reached 50% (5/10), as shown in Fig.  6 B. The emergence of the revertant virus indicates that the SL5D structure has beneficial effects on MERS-CoV propagation.

figure 6

Emergence of revertant SL5D-R mutant virus in Vero cells. ( A ) Nucleotide sequences from 330–344 of WT and SL5D-R mutant viruses. Mutated nucleotides are shown in red. ( B ) Nucleotide sequences of rMERS-SL5D-R viruses passaged in Vero cells. Vero cells were inoculated with rMERS-SL5D-R virus and passaged. After ten passages in Vero cells, viral RNAs were extracted from the P1, P2, P5, and P10 virus stocks, and RT-PCR was performed to amplify the SL5D region and read the nucleotide sequences. The amplified PCR products were cloned into a Blunt vector and transformed to E. coli . The cloned vector DNA from ten clones was extracted and the nucleotide sequences were identified. ( C ) Growth kinetics of passaged rMERS-SL5D-R viruses in Vero cells. Vero cells were inoculated with rMERS or passaged rMERS-SL5D-R viruses (MOI = 0.01) and cultured. Viral titers of culture supernatants were determined using the TCID 50 assay. ( D ) Plaque morphology of the passaged rMERS-SL5D-R viruses. Seeded Vero cells were inoculated with 25 TCID 50 of each virus. Infected Vero cells were fixed with phosphate-buffered formaldehyde at 3 d post infection and stained using crystal violet. White arrow indicates small plaques. White arrowheads indicate large plaques. Scale bar = 1 cm. ( E ) RNA structures of positive-strand RNA of P10 virus predicted following Mfold analysis. ( F ) RNA structures of negative-strand RNA predicted following Mfold analysis. Predicted structures were constructed using the reverse-complement sequences of nts 1–460 from the WT, SL5D-R mutant, and SL5D-R P10 viruses. Numbers indicate the corresponding nucleotide position of positive-strand genomic RNA.

Next, we investigated the growth of the passaged SL5D-R virus in Vero cells. Vero cells inoculated with P2, P5, or P10 viruses (MOI = 0.01) were cultured for 24, 48, and 72 h, and infectious viral titers in the supernatants were measured. The results show that the highly passaged viruses (P5 and P10) exhibited increased growth kinetics compared to the low-passage virus P2 (Fig.  6 C). The P2 virus produced the smallest plaques, the P5 virus produced both small and large plaques, and the P10 virus produced large plaques (Fig.  6 D). Thus, plaque size correlated with the ratio of revertant to non-revertant viruses (Fig.  6 B). These results suggest that the RNA structure of SL5D is important for efficient viral replication as it regulates the synthesis of positive-strand genomic RNA.

To clarify the molecular mechanisms underlying efficient viral replication in cells infected with the revertant virus (P10 virus), we predicted the RNA secondary structure of SL5D in the P10 virus using Mfold. As expected, the predicted RNA structures of the positive and negative strands of the P10 virus were analogous to those of the WT virus (Fig.  6 E,F, right panel). Furthermore, the predicted RNA secondary structure of the negative strand of SL5D-R was different from that of the WT and P10 viruses (Fig.  6 F). As shown in Fig.  6 F, one stem-loop structure within the negative-strand RNA corresponding to the SL5D region (nts 328–338) was predicted for all three viruses (WT, SL5D-R, and P10). A short stem-loop with a stem structure consisting of seven base pairs was predicted in the negative strand of the WT virus. In contrast, the stem structure in the short stem-loop of the negative-strand of SL5D-R consisted of four base pairs (Fig.  6 F, middle panel), whereas this structure consisted of six base pairs in the P10 virus (Fig.  6 F, right panel). This suggests that the stem-loop structure of SL5D in the negative-strand is important for regulating positive-strand genomic RNA synthesis.

Several studies had reported the importance of RNA structures at the 5′ UTR, its adjacent ORF1a coding region, and the 3′ UTR of CoV genomes for viral and/or DI-RNA replication 21 , 24 , 25 , 35 , 36 . However, the detailed functions of these RNA structures in the viral life cycle have not yet been fully elucidated. In this study, we revealed the significant roles of the higher-order RNA structures SL5B, SL5C, and SL5D in the MERS-CoV lifecycle using a BAC-based reverse genetics system. Our results show that SL5B and SL5C play essential roles in regulating viral RNA transcription to synthesize sg mRNAs (Fig.  3 ). In contrast, SL5D is important for regulating the synthesis of the positive-strand genomic RNA (Fig.  5 ).

Higher-order RNA structures in viral genomic RNA are important for increasing its stability and evading host RNA degradation machinery 37 , 38 . We hypothesized that disrupting the RNA structure of the SL5 impairs viral replication by decreasing the stability of viral genomic RNA. However, the levels of genomic RNA in cells transfected with mutant viruses lacking the secondary structure (SL5B and SL5C) were similar to those in the WT (Fig.  3 A); thus, suggesting that the higher-order structures in these regions do not directly affect genomic RNA stability. Furthermore, our luciferase reporter assay indicated that the disruption of RNA structures in the SL5B and SL5C mutants did not impair the translational ability of their own mRNAs (Fig.  3 C). The protease activity of nsp5 in cells transfected with SL5B or SL5C mutant cDNA was similar to that in cells transfected with WT cDNA (Fig.  3 E). Thus, the RNA structures of SL5B and SL5C are responsible for regulating viral RNA transcription during the MERS-CoV life cycle.

CoV has been shown to synthesize sg mRNAs via discontinuous transcription, which is a unique mechanism 39 . Given the lack of sg N mRNA, the RNA structures of SL5B and SL5C are essential for sg N mRNA synthesis. In this discontinuous transcription model, the transcription regulatory sequence (TRS) located at the leader sequence (TRS-L) and preceding each viral gene (TRS-B) plays a critical role in mediating long RNA-RNA interactions (leader-body joining) and switching the template 40 , 41 , 42 . Circularization of the CoV genome through the interaction between the 5′ and 3′UTRs is important for forming the leader-body joining and subsequent viral RNA elongation 42 . We predicted a specific interaction between SL5C and BSL at the 3′UTR (Fig.  4 ). Although further studies are required to investigate the interaction between SL5C and BSL, our results suggest that the RNA structure of SL5C is critical for the circularization of viral genomic RNA. Although a binding interaction was not predicted for SL5B, the SL5B disrupting mutant showed a phenotype similar to that of the SL5C mutant. These results indicate that the RNA structure of SL5B neighboring SL5C may affect the interaction between SL5C and BSL.

CoVs use both host proteins and viral proteins to replicate and transcribe viral RNA. Poly A binding protein is involved in the RNA circularization of CoV 43 , and the viral N proteins of the MHV and the transmissible gastroenteritis virus participate in viral RNA transcription through RNA-RNA interactions between TRS-L and TRS-Bs 44 , 45 , 46 . The MHV N protein also interacts with RNA sequences to modulate the packaging of MHV genomic RNA 47 . Although further studies are needed to identify the host factor(s) or viral proteins that interact with the SL5 region of MERS-CoV, our results suggest that these higher-order RNA structures interact with the host and/or viral proteins to play important roles in viral RNA transcription and replication. Our data indicates that the RNA structures of SL5 have drastic effects on MERS-CoV replication; hence, host proteins that interact with SL5 are attractive candidates for developing therapeutic agents against MERS-CoV infection.

Interestingly, SL5D mutants showed a different phenotype than that of SL5B and SL5C mutants (Fig.  2 ), even though SL5D is adjacent to both SL5B and SL5C in the MERS-CoV genomic RNA. Although the SL5D-R mutant synthesized sg mRNA, it did not efficiently produce viral progeny (Fig.  2 A,B). These results indicate that SL5D functions at a different step in the viral lifecycle than SL5B and SL5C. Our results suggested that SL5D is important for the synthesis of positive-strand genomic RNAs (Fig.  5 C). In addition, we recovered a revertant SL5D-R virus. We introduced two nucleotide substitutions (G335C and C338A) into the SL5D region to construct the SL5D-R mutant (Fig.  1 B); however, the P10 virus acquired one revertant substitution at position 338 (Fig.  6 B). As expected, the predicted RNA secondary structure of the positive-strand RNA in SL5 of the P10 virus was similar to that of the WT (Fig.  6 E). Furthermore, the negative-strand RNA of the WT and passaged P10 viruses was predicted to contain longer stem-loop structures within SL5D than those associated with the SL5D-R mutant (Fig.  6 F). Thus, the stem-loop structure of the negative-strand RNA in the region corresponding to SL5D is important for regulating the synthesis of genomic (positive-strand) RNA from negative-strand RNA.

In conclusion, we identified that higher-order RNA structures within the nsp1 coding region play critical roles in the MERS-CoV lifecycle through several steps. Our approach using a BAC-based reverse genetics system revealed that higher-order RNA structures within viral RNA regulate viral RNA transcription and replication. Furthermore, these RNA structures are conserved among betacoronaviruses despite their low nucleotide homology 19 , 21 , 48 , thereby suggesting that these RNA structures are potential targets for developing novel antivirals against betacoronaviruses. Further studies are required to reveal the mechanisms through which the highly structured SL5 RNA regulates CoV RNA transcription and replication through RNA-RNA and/or RNA–protein interactions.

Cells and viruses

Huh7 (human hepatocellular carcinoma, JCRB0403 from JCRB cell bank) and 293T (human embryonic kidney, CRL-3216 from ATCC) cells were maintained in Dulbecco's modified minimum essential medium (DMEM) (Nacalai Tesque, Kyoto, Japan) containing 10% heat-inactivated fetal bovine serum (FBS), 100 U/mL penicillin, and 100 µg/mL streptomycin (Nacalai Tesque). Vero cells (African green monkey kidney) and Vero-TMPRSS2 34 were maintained in DMEM containing 5% FBS, 100 U/mL penicillin, and 100 µg/mL streptomycin (Nacalai Tesque). Vero-TMPRSS2 cells were a kind gift from Dr. Shutoku Matsuyama (National Institute of Infectious Diseases, Tokyo, Japan). Cells were cultured in a humidified, 5% CO 2 atmosphere at 37 °C.

BAC construction

We previously established a reverse genetics system for MERS-CoV using the BAC system: pBAC-MERS-WT 28 . To construct cDNA clones of several SL5 mutants from pBAC-MERS-WT, we used the Red/ET Recombination System Counter-Selection BAC Modification Kit (Gene Bridges, Heidelberg, Germany). The nucleotide mutations in each SL5 mutant are shown in Fig.  1 B. To generate SL5B and SL5C mutants, synonymous mutations that retained the RNA structure were introduced within the loop regions. These constructs were designated as pBAC-MERS-SL5B-Loop and pBAC-MERS-SL5C-Loop, respectively. To generate SL5B-R, SL5C-R, and SL5D-R mutants, synonymous mutations that disrupted RNA structures were introduced into the right side of each stem region, which were designated as pBAC-MERS-SL5B-R, pBAC-MERS-SL5C-R, and pBAC-MERS-SL5D-R, respectively. To retain the RNA secondary structures in each SL, mutations were introduced to the left side of each stem region, yielding SL5B-RL, SL5C-RL, and SL5D-RL, these cDNA clones were designated as pBAC-MERS-SL5B-RL, pBAC-MERS-SL5C-RL, and pBAC-MERS-SL5D-RL, respectively (Fig.  1 A,B). Sequence analyses were performed by Eurofins Scientific (Tokyo, Japan) to confirm substitutions.

We established a MERS-CoV replicon system by replacing the genomic sequence between the S and M genes with that of Nluc. A red/ET recombination system counterselection BAC modification kit was used to construct the MERS replicon. To increase Nluc activity, we inserted nts 27,737–27,861 of the MERS-CoV EMC strain, which contain the TRS of the M gene, upstream of Nluc. The cDNA clone of the replicon with the WT sequence was designated as pBAC-MERS-Rep. To construct a polymerase-dead mutant of the MERS replicon, we introduced substitutions (SDD to SAA) within the active site of the viral RNA-dependent RNA polymerase (RdRp; nsp12). In addition, cDNA clones of SL5 mutants were constructed using the method described above.

To establish the reporter MERS coronavirus carrying the ZsGreen gene, the ORF5 gene of the pBAC-MERS-WT was replaced with the ZsGreen gene using the recombination method described above and designated as pBAC-MERS-ZsGreen. Recombination was performed to construct a pBAC-MERS-ZsGreen-SL5D-R cDNA clone carrying the SL5D-R mutation.

Transfection

Huh7 cells were seeded into 6-well plates (Violamo; Misumi, Schaumberg, IL, USA) at 4.0 × 10 5 cells/well. After incubation at 37 °C overnight, the cells were transfected with 4 µg of cDNA clones using XtremeGene 9 DNA Transfection Reagent (Roche, Basel, Switzerland) according to manufacturer’s instructions. After incubating at 37 °C for the indicated time, the culture supernatants and cell pellets were collected and stored at − 80 °C until further use.

Virus rescue and propagation

To propagate reporter viruses, rMERS-ZsGreen and rMERS-ZsGreen-SL5D-R, pBAC-MERS-ZsGreen, or pBAC-MERS-ZsGreen-SL5D-R were transfected into Huh7 cells as described above. After incubating for 3 days, culture supernatants were collected and stored at − 80 °C. Viruses were stored at P0. The rMERS-ZsGreen P0 virus was propagated twice in Vero cells and stored as the P2 virus. The rMERS-ZsGreen-SL5D-R P0 virus was propagated twice in Vero-TMPRSS2 cells and stored as the P2 virus. Viral titers were determined using the titration assay described below.

The TCID 50 method using Vero cells was used to determine the infection titer of each virus, as described previously 28 .

Realtime RT-PCR

Total RNAs from transfected Huh7 cells were extracted using a PureLink RNA Mini Kit (Thermo Fisher Scientific, Waltham, MA, USA) according to manufacturer’s instructions and stored at − 80 °C until use. First-strand cDNA was synthesized using a ReverTra Ace qPCR RT kit (TOYOBO, Osaka, Japan) according to the manufacturer's instructions. For cDNA synthesis, a primer mix (TOYOBO) was used for sg N mRNA, YT750 (5′-GGGCTTGAGGCTTCTCCAATG-3′) was used for positive-strand genomic RNA, and YT749 (5′-ACACTTTTCTTGTTGCCTGTGG-3′) was used for negative-strand genomic RNA. The reverse-transcribed samples were treated with RNase H at 37 °C for 20 min. The level of each cDNA was determined using a Thunderbird Probe qPCR Mix (TOYOBO), and the reaction was performed using a StepOne Real-Time PCR System (Applied Biosystems, Waltham, MA, USA). To quantify the sg N mRNA, we used the primer pair, WK1350 (5′-TCGTTCTCTTGCAGAACTTTG-3′) and WK1351 (5′-TTGGATTACGTCCTCTACCTC-3′), and the FAM-labeled specific probe, WK1352 (5′-CCTCGTGCTGTTTCCTTTGCCGAT-3′). To quantify the positive- or negative-strand genomic RNA, we used the primer pair, YT727 (5′-CCACTACTCCCATTTCGTCAG-3′) and YT728 (5′-CAGTATGTGTAGTGCGCATATAAGCA-3′), and the FAM-labeled specific probe, YT729 (5′-TTGCAAATTGGCTTGCCCCCACT-3′). For quantification of GAPDH mRNA, we used the primer pair, WK1288 (5′-GAAGGTGAAGGTCGGAGT-3′) and WK1289 (5′-GAAGATGGTGATGGGATTTC-3′), and the FAM-labeled specific probe, WK1290 (5′-CAAGCTTCCCGTTCTCAGCC-3′). Cycling conditions were 95 °C for 1 min, followed by 40 cycles of 95 °C for 15 s and 58 °C for 45 s. The RNA levels of GAPDH were used to normalize viral RNA levels.

Immunoblotting

Transfected Huh7 cells were lysed in lysis buffer (100 mM Tris–HCl, pH 8.0, 150 mM NaCl, and 1% TritonX-100). After centrifugation at 16,000× g , the supernatants were collected and mixed with 2× sample buffer (0.1 M Tris–HCl pH 6.8, 4% sodium dodecyl sulfate [SDS], 20% glycerol, 0.004% bromophenol blue, and 10% 2-mercaptoethanol). Next, samples were boiled. Proteins were separated by 10% SDS-polyacrylamide gel electrophoresis (SDS-PAGE) and transferred onto a polyvinylidene difluoride (PVDF) membrane (Merck Millipore, Billerica, MA, USA). The membranes were blocked with 3% skim milk in PBS containing 0.05% Tween 20 (Nacalai Tesque). Rabbit anti-MERS-N (Sino Biological, Beijing, China) or mouse anti-β-Actin (Sigma-Aldrich, St. Louis, MO, USA) were used as primary antibodies, and goat anti-mouse IgG-horseradish peroxidase (HRP; Sigma-Aldrich) or goat anti-mouse IgG-HRP (Sigma-Aldrich) were used as secondary antibodies, respectively. ChemiLumi One Ultra (Nacalai Tesque) was used for visualization with the LAS-4000 image analyzer system (Fujifilm, Tokyo, Japan).

Replicon assay

293T cells were seeded into 24-well plates (Violamo) at 1.5 × 10 5 cells/well and cultured at 37 °C overnight. Next, cDNA clones of the replicon were transfected into seeded 293T cells using XtremeGene 9 DNA Transfection Reagent (Roche) according to the manufacturer’s instructions. The firefly luciferase reporter plasmid pGL3-control was co-transfected with each cDNA clone as an internal control. After incubation for 48 h, the transfected cells were lysed using passive buffer (Promega, Madison, WI, USA). Luciferase activity was measured using a PowerScan HT (DS Pharma Biomedical, Osaka, Japan). The Nano-Glo Luciferase Assay System (Promega) was used to measure Nluc activity. A luciferase assay system (Promega) was used to measure firefly luciferase activity. Luciferase activity was normalized to that of firefly luciferase. The experiments were performed in triplicates.

Nsp5 reporter assay

To examine the expression levels of nsp5 in transfected Huh7 cells, we used the biosensor expression plasmid, pGlo-30F-VRLQS 32 , which was a kind gift from Dr. Susan Baker (Loyola University, Chicago, IL, USA). pGlo-30F-VRLQS, encoding Photuris pennsylvanica luciferase with the inserted amino acid sequence VRLQS, was recognized and cleaved by the MERS nsp5 protease 3CLpro.

Huh7 cells were seeded into 24 well-plates (Violamo) at 1.0 × 10 5 cells/well and cultured overnight. The seeded cells were transfected with pGlo-30F-VRLQS and cDNA clones using the XtremeGene 9 DNA Transfection Reagent (Roche) according to the manufacturer's instructions. As a control, pBAC-MERS-nsp5-rpsL, in which nsp5 was replaced with an rpsL cassette (Red/ET Recombination System Counter-Selection BAC Modification Kit; Gene Bridges), was used. Transfected cells were cultured for 24 and 48 h and then lysed with passive buffer (Promega). Luciferase activity was measured as previously described.

Correlative light and electron microscopy (CLEM) analysis

Vero cells were seeded into 35-mm glass bottom dishes (P35G-1.5-14-CGRD: MatTek) at 5.0 × 10 4 cells/well and cultured overnight. Cells were inoculated with rMERS-ZsGreen or rMERS-ZsGreen-SL5D-R at an MOI of 0.1. After adsorption at 37 °C for 1 h, the infected cells were washed with DMEM without FBS and DMEM containing 2% FBS was added. After incubation for 24 h at 37 °C, the laser scanning confocal microscope FluoView FV1000 (Olympus, Tokyo, Japan) was used to observe ZsGreen fluorescence to identify infected cells. Cells were fixed in 4% formaldehyde in 0.1 M phosphate buffer for 30 min at room temperature. Fixed cells were washed three times with washing buffer (4% sucrose in 0.2 M phosphate buffer). After washing, 2.5% glutaraldehyde in 0.1 M phosphate buffer was applied for TEM analysis. Cells were post-fixed for 1 h with 1% osmium tetroxide and 0.5% potassium ferrocyanide in 0.1 M cacodylate buffer (pH 7.4), dehydrated in a graded series of ethanol, and embedded in Epon812 (TAAB, Reading, UK). Ultrathin (80 nm) sections were stained with saturated uranyl acetate and lead citrate solutions. Electron micrographs were acquired using a JEM-1011 transmission electron microscope (JEOL, Tokyo, Japan).

Viral titers in supernatants and cells

Vero cells were seeded into 12-well plates (Violamo) at 2.0 × 10 5 cells/well and cultured overnight at 37 °C. Cells were inoculated with rMERS-ZsGreen or rMERS-ZsGreen-SL5D-R at a MOI of 0.01. After adsorption at 37 °C for 1 h, the inoculum was removed and 1 mL/well of DMEM containing 2% FBS was added. The infected cells were cultured at 37 °C for 24, 48, and 72 h. Culture supernatants were collected and stored at − 80 °C until use. DMEM containing 2% FBS (1 mL) was added to the cell supernatants, which were then stored at − 80 °C. Freeze–thaw cycles were performed three times, and the samples were centrifuged at 2500× g for 10 min. Supernatants were collected as cell samples. The infectious viral titers in the supernatants and cell samples were measured using the TCID 50 method described above.

Plasmid construction

To construct a luciferase reporter plasmid carrying the MERS-CoV SL5 nucleotide sequence upstream of the firefly luciferase gene, pcDNA3.1-SL5-fluc, we first constructed the plasmid pcDNA3.1-MERS460nt-fluc carrying the first 460 nts of MERS-CoV. Next, the pBAC-MERS was used as a template to amplifying the 460-nt sequence. The PCR product was cloned into pcDNA3.1-fluc 27 , and the resulting vector was designated as pcDNA3.1-MERS-460nt-fluc. To construct pcD-SL5-fluc, we deleted nucleotides 1–264 and 349–460 of the MERS-CoV from the pcDNA3.1-MERS-460nt-fluc using the PrimeSTAR mutagenesis kit (Takara Bio, Tokyo, Japan), according to the manufacturer’s instructions. To construct the SL5B and SL5C mutants pcDNA3.1-SL5B-R-fluc, pcDNA3.1-SL5B-RL-fluc, pcDNA3.1-SL5C-R-fluc, and pcDNA3.1-SL5C-RL-fluc, we used the PrimeSTAR mutagenesis kit (Takara) and pcDNA3.1-SL5-fluc as the template.

Luciferase assay

293T cells were seeded into 24-well plates (Violamo) at 1.5 × 10 5 cells/well and cultured overnight at 37 °C. Cultured cells were transfected with pcDNA3.1, using TransIT LT1 (Mirus, Madison, WI, USA) according to the manufacturer's instructions. Renilla luciferase reporter plasmid pRL-SV40 was used as an internal control. At 24 h post-transfection, the transfected cells were lysed with passive buffer (Promega) and luciferase activity was measured as described above. The firefly luciferase activity was normalized to the Renilla luciferase activity.

Viral passage in Vero cells

Vero cells were seeded into 10 cm dishes (Violamo) at 2.0 × 10 6 cells/well and cultured overnight at 37 °C. Seeded Vero cells were inoculated with rMERS-SL5D-R P0 virus and allowed to adsorb for 1 h. After adsorption, the inoculated cells were washed twice with DMEM and 10 mL of DMEM containing 2% FBS was added. The cells were cultured for 3 days and cytopathic effects (CPEs) were observed. The supernatant was collected and stored at − 80 °C as P1 virus. The rMERS-SL5D-R P1 virus was further amplified nine times in Vero cells (P10 virus) and viruses from each amplification were stored at − 80 °C. Viral titers were determined using the TCID 50 assay described above.

Growth kinetics of passaged rMERS-SL5D-R viruses in Vero cells

Vero cells were seeded into 6-well plates (Violamo) at 4.0 × 10 5  cells/well and cultured overnight at 37 °C. Cultured cells were inoculated with rMERS-WT or rMERS-SL5D-R (P1, P2, P5, or P10) viruses at an MOI of 0.01. After adsorption, the cells were washed twice with DMEM and 2 mL of DMEM containing 2% FBS was added to each well. The infected cells were cultured for 24, 48, and 72 h, and the supernatants were collected and stored at − 80 °C until use. Viral titers of the culture supernatants were determined using the TCID 50 assay described above.

Plaque assay

Vero cells were seeded into 6-well plates (Violamo) at 6.0 × 10 5 cells/well and cultured overnight at 37 °C. Cultured Vero cells were inoculated at a TCID 50 of 25 and passaged rMERS-SL5D-R (P2, P5, or P10) viruses. After adsorption at 37 °C for 1 h, the cells were washed twice with DMEM and then overlaid with 0.8% agarose (Seaplaque ® GTG Agarose; Lonza, Switzerland) in DMEM containing 2% FBS. Infected cells were cultured at 37 °C for 3 days and then fixed with phosphate-buffered formalin. Fixed cells were stained with crystal violet.

To construct the RNA alignment data for LRIscan 33 , we used Molecular Evolutionary Genetics Analysis version 6.0 (MEGA6 49 ). To construct the MERS-CoV alignment data, we used the full-length sequences of four MERS-CoV strains: MERS-CoV_EMC (Accession No.: NC_019843), MERS-CoV Hu/Oman_50_2015 (Accession No.: KY673148.1), MERS-CoV Al-Hasa_1_2013 (Accession No.: KY673148.1), and MERS-CoV Korea/Seoul/SNU1-035/2015 (Accession No.: KY673148.1). The cluster file containing MERS-CoV alignment data was analyzed using LRIscan 33 and plotted with Circos 50 .

Statistical analyses

A two-way analysis of variance (ANOVA) test was conducted to determine statistical significance using GraphPad Prism ver. 10 (GraphPad Software, San Diego, CA, USA). A p value of < 0.05 was considered statistically significant.

Data availability

The datasets used and/or analysed during the current study available from the corresponding author on reasonable request.

Weiss, S. R. & Navas-Martin, S. Coronavirus pathogenesis and the emerging pathogen severe acute respiratory syndrome coronavirus. Microbiol. Mol. Biol. Rev. 69 , 635–664. https://doi.org/10.1128/MMBR.69.4.635-664.2005 (2005).

Article   CAS   PubMed   PubMed Central   Google Scholar  

Bradburne, A. F., Bynoe, M. L. & Tyrrell, D. A. Effects of a “new” human respiratory virus in volunteers. Br. Med. J. 3 , 767–769. https://doi.org/10.1136/bmj.3.5568.767 (1967).

van der Hoek, L., Pyrc, K. & Berkhout, B. Human coronavirus NL63, a new respiratory virus. FEMS Microbiol. Rev. 30 , 760–773. https://doi.org/10.1111/j.1574-6976.2006.00032.x (2006).

Article   CAS   PubMed   Google Scholar  

van der Hoek, L. et al. Identification of a new human coronavirus. Nat. Med. 10 , 368–373. https://doi.org/10.1038/nm1024 (2004).

Woo, P. C. et al. Characterization and complete genome sequence of a novel coronavirus, coronavirus HKU1, from patients with pneumonia. J. Virol. 79 , 884–895. https://doi.org/10.1128/JVI.79.2.884-895.2005 (2005).

Drosten, C. et al. Identification of a novel coronavirus in patients with severe acute respiratory syndrome. N. Engl. J. Med. 348 , 1967–1976. https://doi.org/10.1056/NEJMoa030747 (2003).

Ksiazek, T. G. et al. A novel coronavirus associated with severe acute respiratory syndrome. N. Engl. J. Med. 348 , 1953–1966. https://doi.org/10.1056/NEJMoa030781 (2003).

Zaki, A. M., van Boheemen, S., Bestebroer, T. M., Osterhaus, A. D. & Fouchier, R. A. Isolation of a novel coronavirus from a man with pneumonia in Saudi Arabia. N. Engl. J. Med. 367 , 1814–1820. https://doi.org/10.1056/NEJMoa1211721 (2012).

Chen, N. et al. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: A descriptive study. Lancet 395 , 507–513. https://doi.org/10.1016/S0140-6736(20)30211-7 (2020).

Huang, C. et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 395 , 497–506. https://doi.org/10.1016/S0140-6736(20)30183-5 (2020).

Wang, D. et al. Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus-infected pneumonia in Wuhan, China. JAMA 323 , 1061–1069. https://doi.org/10.1001/jama.2020.1585 (2020).

Ferhadian, D. et al. Structural and functional motifs in influenza virus RNAs. Front. Microbiol. 9 , 559. https://doi.org/10.3389/fmicb.2018.00559 (2018).

Article   PubMed   PubMed Central   Google Scholar  

Ito, T., Tahara, S. M. & Lai, M. M. The 3′-untranslated region of hepatitis C virus RNA enhances translation from an internal ribosomal entry site. J. Virol. 72 , 8789–8796. https://doi.org/10.1128/JVI.72.11.8789-8796.1998 (1998).

Tsukiyama-Kohara, K., Iizuka, N., Kohara, M. & Nomoto, A. Internal ribosome entry site within hepatitis C virus RNA. J. Virol. 66 , 1476–1483. https://doi.org/10.1128/JVI.66.3.1476-1483.1992 (1992).

Villordo, S. M., Carballeda, J. M., Filomatori, C. V. & Gamarnik, A. V. RNA structure duplications and flavivirus host adaptation. Trends Microbiol. 24 , 270–283. https://doi.org/10.1016/j.tim.2016.01.002 (2016).

Jang, S. K. et al. A segment of the 5′ nontranslated region of encephalomyocarditis virus RNA directs internal entry of ribosomes during in vitro translation. J. Virol. 62 , 2636–2643. https://doi.org/10.1128/JVI.62.8.2636-2643.1988 (1988).

Pelletier, J. & Sonenberg, N. Internal initiation of translation of eukaryotic mRNA directed by a sequence derived from poliovirus RNA. Nature 334 , 320–325. https://doi.org/10.1038/334320a0 (1988).

Article   ADS   CAS   PubMed   Google Scholar  

Sola, I., Almazan, F., Zuniga, S. & Enjuanes, L. Continuous and discontinuous RNA synthesis in coronaviruses. Annu. Rev. Virol. 2 , 265–288. https://doi.org/10.1146/annurev-virology-100114-055218 (2015).

Yang, D. & Leibowitz, J. L. The structure and functions of coronavirus genomic 3′ and 5′ ends. Virus Res. 206 , 120–133. https://doi.org/10.1016/j.virusres.2015.02.025 (2015).

Fosmire, J. A., Hwang, K. & Makino, S. Identification and characterization of a coronavirus packaging signal. J. Virol. 66 , 3522–3530. https://doi.org/10.1128/JVI.66.6.3522-3530.1992 (1992).

Chen, S. C. & Olsthoorn, R. C. Group-specific structural features of the 5′-proximal sequences of coronavirus genomic RNAs. Virology 401 , 29–41. https://doi.org/10.1016/j.virol.2010.02.007 (2010).

Kim, Y. N. & Makino, S. Characterization of a murine coronavirus defective interfering RNA internal cis-acting replication signal. J. Virol. 69 , 4963–4971. https://doi.org/10.1128/JVI.69.8.4963-4971.1995 (1995).

Liu, P. et al. A U-turn motif-containing stem-loop in the coronavirus 5′ untranslated region plays a functional role in replication. RNA 13 , 763–780. https://doi.org/10.1261/rna.261807 (2007).

Raman, S., Bouma, P., Williams, G. D. & Brian, D. A. Stem-loop III in the 5′ untranslated region is a cis-acting element in bovine coronavirus defective interfering RNA replication. J. Virol. 77 , 6720–6730. https://doi.org/10.1128/jvi.77.12.6720-6730.2003 (2003).

Raman, S. & Brian, D. A. Stem-loop IV in the 5′ untranslated region is a cis-acting element in bovine coronavirus defective interfering RNA replication. J. Virol. 79 , 12434–12446. https://doi.org/10.1128/JVI.79.19.12434-12446.2005 (2005).

Yang, D., Liu, P., Wudeck, E. V., Giedroc, D. P. & Leibowitz, J. L. SHAPE analysis of the RNA secondary structure of the Mouse Hepatitis Virus 5′ untranslated region and N-terminal nsp1 coding sequences. Virology 475 , 15–27. https://doi.org/10.1016/j.virol.2014.11.001 (2015).

Tanaka, T., Kamitani, W., DeDiego, M. L., Enjuanes, L. & Matsuura, Y. Severe acute respiratory syndrome coronavirus nsp1 facilitates efficient propagation in cells through a specific translational shutoff of host mRNA. J. Virol. 86 , 11128–11137. https://doi.org/10.1128/JVI.01700-12 (2012).

Terada, Y., Kawachi, K., Matsuura, Y. & Kamitani, W. MERS coronavirus nsp1 participates in an efficient propagation through a specific interaction with viral RNA. Virology 511 , 95–105. https://doi.org/10.1016/j.virol.2017.08.026 (2017).

Kim, Y. N., Jeong, Y. S. & Makino, S. Analysis of cis-acting sequences essential for coronavirus defective interfering RNA replication. Virology 197 , 53–63. https://doi.org/10.1006/viro.1993.1566 (1993).

Zuker, M. Mfold web server for nucleic acid folding and hybridization prediction. Nucleic Acids Res. 31 , 3406–3415. https://doi.org/10.1093/nar/gkg595 (2003).

Snijder, E. J., Decroly, E. & Ziebuhr, J. The nonstructural proteins directing coronavirus RNA synthesis and processing. Adv. Virus Res. 96 , 59–126. https://doi.org/10.1016/bs.aivir.2016.08.008 (2016).

Kilianski, A., Mielech, A. M., Deng, X. & Baker, S. C. Assessing activity and inhibition of Middle East respiratory syndrome coronavirus papain-like and 3C-like proteases using luciferase-based biosensors. J. Virol. 87 , 11955–11962. https://doi.org/10.1128/JVI.02105-13 (2013).

Fricke, M. & Marz, M. Prediction of conserved long-range RNA–RNA interactions in full viral genomes. Bioinformatics 32 , 2928–2935. https://doi.org/10.1093/bioinformatics/btw323 (2016).

Shirogane, Y. et al. Efficient multiplication of human metapneumovirus in Vero cells expressing the transmembrane serine protease TMPRSS2. J. Virol. 82 , 8942–8946. https://doi.org/10.1128/JVI.00676-08 (2008).

Goebel, S. J., Hsue, B., Dombrowski, T. F. & Masters, P. S. Characterization of the RNA components of a putative molecular switch in the 3′ untranslated region of the murine coronavirus genome. J. Virol. 78 , 669–682. https://doi.org/10.1128/jvi.78.2.669-682.2004 (2004).

Goebel, S. J., Taylor, J. & Masters, P. S. The 3′ cis-acting genomic replication element of the severe acute respiratory syndrome coronavirus can function in the murine coronavirus genome. J. Virol. 78 , 7846–7851. https://doi.org/10.1128/JVI.78.14.7846-7851.2004 (2004).

Dickson, A. M. & Wilusz, J. Strategies for viral RNA stability: Live long and prosper. Trends Genet. 27 , 286–293. https://doi.org/10.1016/j.tig.2011.04.003 (2011).

Ford, L. P. & Wilusz, J. 3′-Terminal RNA structures and poly(U) tracts inhibit initiation by a 3′–>5′ exonuclease in vitro. Nucleic Acids Res. 27 , 1159–1167. https://doi.org/10.1093/nar/27.4.1159 (1999).

Sawicki, S. G. & Sawicki, D. L. A new model for coronavirus transcription. Adv. Exp. Med. Biol. 440 , 215–219. https://doi.org/10.1007/978-1-4615-5331-1_26 (1998).

Sola, I., Mateos-Gomez, P. A., Almazan, F., Zuniga, S. & Enjuanes, L. RNA–RNA and RNA–protein interactions in coronavirus replication and transcription. RNA Biol. 8 , 237–248. https://doi.org/10.4161/rna.8.2.14991 (2011).

Sola, I., Moreno, J. L., Zuniga, S., Alonso, S. & Enjuanes, L. Role of nucleotides immediately flanking the transcription-regulating sequence core in coronavirus subgenomic mRNA synthesis. J. Virol. 79 , 2506–2516. https://doi.org/10.1128/JVI.79.4.2506-2516.2005 (2005).

Zuniga, S., Sola, I., Alonso, S. & Enjuanes, L. Sequence motifs involved in the regulation of discontinuous coronavirus subgenomic RNA synthesis. J. Virol. 78 , 980–994. https://doi.org/10.1128/jvi.78.2.980-994.2004 (2004).

Spagnolo, J. F. & Hogue, B. G. Host protein interactions with the 3′ end of bovine coronavirus RNA and the requirement of the poly(A) tail for coronavirus defective genome replication. J. Virol. 74 , 5053–5065. https://doi.org/10.1128/jvi.74.11.5053-5065.2000 (2000).

Nelson, G. W., Stohlman, S. A. & Tahara, S. M. High affinity interaction between nucleocapsid protein and leader/intergenic sequence of mouse hepatitis virus RNA. J. Gen. Virol. 81 , 181–188. https://doi.org/10.1099/0022-1317-81-1-181 (2000).

Zuniga, S. et al. Coronavirus nucleocapsid protein facilitates template switching and is required for efficient transcription. J. Virol. 84 , 2169–2175. https://doi.org/10.1128/JVI.02011-09 (2010).

Zuniga, S. et al. Coronavirus nucleocapsid protein is an RNA chaperone. Virology 357 , 215–227. https://doi.org/10.1016/j.virol.2006.07.046 (2007).

Kuo, L., Koetzner, C. A., Hurst, K. R. & Masters, P. S. Recognition of the murine coronavirus genomic RNA packaging signal depends on the second RNA-binding domain of the nucleocapsid protein. J. Virol. 88 , 4451–4465. https://doi.org/10.1128/JVI.03866-13 (2014).

Guan, B. J., Wu, H. Y. & Brian, D. A. An optimal cis-replication stem-loop IV in the 5′ untranslated region of the mouse coronavirus genome extends 16 nucleotides into open reading frame 1. J. Virol. 85 , 5593–5605. https://doi.org/10.1128/JVI.00263-11 (2011).

Tamura, K., Stecher, G., Peterson, D., Filipski, A. & Kumar, S. MEGA6: Molecular Evolutionary Genetics Analysis version 6.0. Mol. Biol. Evol. 30 , 2725–2729. https://doi.org/10.1093/molbev/mst197 (2013).

Krzywinski, M. et al. Circos: An information aesthetic for comparative genomics. Genome Res. 19 , 1639–1645. https://doi.org/10.1101/gr.092759.109 (2009).

Download references

Acknowledgements

We thank Dr. Luis Enjuanes (CNB-CSIC, Madrid, Spain) for providing the BAC DNA reagent, and Dr. Bart L. Haagmens (Erasmus Medical Center, Rotterdam, Netherlands) for providing the MERS-CoV/EMC2012 strain through Dr. Makoto Takeda (National Institute of Infectious Diseases). We also thank Dr. Susan C. Baker (Loyola University, Chicago, IL, USA) for providing us with the MERS protease reporter system. We thank Ms. Kaede Yukawa and Marie Shitara for secretarial assistance and Ms. Kanako Yoshizawa, Nozomi Shimada, Yuki Shida, Yukari Shiba, and Naho Takashima for their technical assistance. We also thank Dr. Naohisa Goto for advice on the LRIscan analyses and Dr. Hiroko Omori for the electron microscopy analyses.

This study was supported by a Grant for Joint Research Project of the Research Institute for Microbial Diseases, Osaka University. This work was supported in part by grants-in-aid from the Japanese Society for the Promotion of Science (JSPS), KAKENHI grant (JP16K08811and JP15J07066), from the Ministry of Health, Labor and Welfare of Japan, and the Japan Agency for Medical Research and Development (AMED, JP18fk0108058, JP18fm028101j0002, JP21fk0108560h, and JP20fk0108267h). This work was supported in part by the Japan Science and Technology Agency (Moonshot Research & Development) (Grant Number JPMJMS2025). YT was supported by a JSPS Research Fellowship for Young Scientists.

Author information

Authors and affiliations.

Laboratory of Clinical Research on Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Suita, Japan

Yutaka Terada & Wataru Kamitani

Center for Vaccine Research and Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA

Yutaka Terada

Department of Infectious Diseases and Host Defense, Graduate School of Medicine, Gunma University, Maebashi, Japan

Sodbayasgalan Amarbayasgalan & Wataru Kamitani

Center for Infectious Disease Education and Research (CiDER), Suita, Japan

Yoshiharu Matsuura

Research Institute for Microbial Diseases (RIMD), Suita, Japan

Center for Advanced Modalities and DDS (CAMaD), Osaka University, Suita, Japan

You can also search for this author in PubMed   Google Scholar

Contributions

Y.T. and W.K. conceived and designed the experiments. Y. T., and S. A. performed the experiments. Y.T., Y.M., and W.K. analyzed the data and wrote the manuscript.

Corresponding author

Correspondence to Wataru Kamitani .

Ethics declarations

Competing interests.

The authors declare no competing interests.

Additional information

Publisher's note.

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Supplementary information., rights and permissions.

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/ .

Reprints and permissions

About this article

Cite this article.

Terada, Y., Amarbayasgalan, S., Matsuura, Y. et al. Regulation viral RNA transcription and replication by higher-order RNA structures within the nsp1 coding region of MERS coronavirus. Sci Rep 14 , 19594 (2024). https://doi.org/10.1038/s41598-024-70601-5

Download citation

Received : 08 February 2024

Accepted : 19 August 2024

Published : 23 August 2024

DOI : https://doi.org/10.1038/s41598-024-70601-5

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

By submitting a comment you agree to abide by our Terms and Community Guidelines . If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Quick links

  • Explore articles by subject
  • Guide to authors
  • Editorial policies

Sign up for the Nature Briefing: Microbiology newsletter — what matters in microbiology research, free to your inbox weekly.

presentation on viral replication

Information

  • Author Services

Initiatives

You are accessing a machine-readable page. In order to be human-readable, please install an RSS reader.

All articles published by MDPI are made immediately available worldwide under an open access license. No special permission is required to reuse all or part of the article published by MDPI, including figures and tables. For articles published under an open access Creative Common CC BY license, any part of the article may be reused without permission provided that the original article is clearly cited. For more information, please refer to https://www.mdpi.com/openaccess .

Feature papers represent the most advanced research with significant potential for high impact in the field. A Feature Paper should be a substantial original Article that involves several techniques or approaches, provides an outlook for future research directions and describes possible research applications.

Feature papers are submitted upon individual invitation or recommendation by the scientific editors and must receive positive feedback from the reviewers.

Editor’s Choice articles are based on recommendations by the scientific editors of MDPI journals from around the world. Editors select a small number of articles recently published in the journal that they believe will be particularly interesting to readers, or important in the respective research area. The aim is to provide a snapshot of some of the most exciting work published in the various research areas of the journal.

Original Submission Date Received: .

  • Active Journals
  • Find a Journal
  • Proceedings Series
  • For Authors
  • For Reviewers
  • For Editors
  • For Librarians
  • For Publishers
  • For Societies
  • For Conference Organizers
  • Open Access Policy
  • Institutional Open Access Program
  • Special Issues Guidelines
  • Editorial Process
  • Research and Publication Ethics
  • Article Processing Charges
  • Testimonials
  • Preprints.org
  • SciProfiles
  • Encyclopedia

pathogens-logo

Article Menu

presentation on viral replication

  • Subscribe SciFeed
  • Recommended Articles
  • Google Scholar
  • on Google Scholar
  • Table of Contents

Find support for a specific problem in the support section of our website.

Please let us know what you think of our products and services.

Visit our dedicated information section to learn more about MDPI.

JSmol Viewer

Advancement in the antigenic epitopes and vaccine adjuvants of african swine fever virus.

presentation on viral replication

1. Introduction

2. advancement in the development of antigenic epitopes of asfv, 2.1. antigenic epitope-identified proteins in asfv, 2.2. current methods for the identification of antigenic epitopes of the asfv, 2.2.1. methods for the identification of b-cell epitopes, 2.2.2. methods for the identification of t-cell epitopes, 2.3. research progress on b-cell epitopes in asfv proteins, 2.3.1. p72-derived b-cell epitopes, 2.3.2. cd2v-derived b-cell epitopes, 2.3.3. p30-derived b-cell epitopes, 2.3.4. p54-derived b-cell epitopes, 2.3.5. dp96r-derived b-cell epitopes, 2.3.6. e120r-derived b-cell epitopes, 2.3.7. pa104r-derived b-cell epitopes, 2.3.8. e184l-derived b-cell epitopes, 2.4. research progress on asfv-specific t-cell epitopes, 2.4.1. cd2v-derived t-cell epitopes, 2.4.2. p72-derived t-cell epitopes, 2.4.3. f317l-derived t-cell epitopes, 2.4.4. c129r-derived t-cell epitopes, 3. advancement in the development of vaccine adjuvants of asfv, 3.1. the functions and mechanism of adjuvants in asf vaccines, 3.2. types of adjuvants for asf vaccines, 3.2.1. montanide™ isa, 3.2.2. polygen™, 3.2.3. zoetis, 3.2.4. biomize ®, 3.2.5. mf59 ®, 3.2.6. adjuvants from bacterial component, 3.2.7. nano-adjuvants, 4. prospects of antigenic epitopes and adjuvants in the development of novel asfv vaccines and diagnostic reagents, 5. conclusions, author contributions, institutional review board statement, informed consent statement, data availability statement, acknowledgments, conflicts of interest.

  • Dixon, L.K.; Sun, H.; Roberts, H. African swine fever. Antiviral Res. 2019 , 165 , 34–41. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Penrith, M.L. African swine fever. Onderstepoort J. Vet. Res. 2009 , 76 , 91–95. [ Google Scholar ]
  • Vu, H.L.X.; McVey, D.S. Recent progress on gene-deleted live-attenuated African swine fever virus vaccines. NPJ Vaccines 2024 , 9 , 60. [ Google Scholar ] [ CrossRef ]
  • Wu, K.; Liu, J.; Wang, L.; Fan, S.; Li, Z.; Li, Y.; Yi, L.; Ding, H.; Zhao, M.; Chen, J. Current State of Global African Swine Fever Vaccine Development under the Prevalence and Transmission of ASF in China. Vaccines 2020 , 8 , 531. [ Google Scholar ] [ CrossRef ]
  • Sereda, A.D.; Balyshev, V.M.; Kazakova, A.S.; Imatdinov, A.R.; Kolbasov, D.V. Protective Properties of Attenuated Strains of African Swine Fever Virus Belonging to Seroimmunotypes I–VIII. Pathogens 2020 , 9 , 274. [ Google Scholar ] [ CrossRef ]
  • Schafer, A.; Franzoni, G.; Netherton, C.L.; Hartmann, L.; Blome, S.; Blohm, U. Adaptive Cellular Immunity against African Swine Fever Virus Infections. Pathogens 2022 , 11 , 274. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Canter, J.A.; Aponte, T.; Ramirez-Medina, E.; Pruitt, S.; Gladue, D.P.; Borca, M.V.; Zhu, J.J. Serum Neutralizing and Enhancing Effects on African Swine Fever Virus Infectivity in Adherent Pig PBMC. Viruses 2022 , 14 , 1249. [ Google Scholar ] [ CrossRef ]
  • Escribano, J.M.; Galindo, I.; Alonso, C. Antibody-mediated neutralization of African swine fever virus: Myths and facts. Virus Res. 2013 , 173 , 101–109. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Gomez-Puertas, P.; Escribano, J.M. Blocking antibodies inhibit complete African swine fever virus neutralization. Virus Res. 1997 , 49 , 115–122. [ Google Scholar ] [ CrossRef ]
  • Xu, Z.; Hu, Y.; Li, J.; Wang, A.; Meng, X.; Chen, L.; Wei, J.; Tong, W.; Kong, N.; Yu, L.; et al. Screening and identification of the dominant antigens of the African swine fever virus. Front. Vet. Sci. 2023 , 10 , 1175701. [ Google Scholar ] [ CrossRef ]
  • Zhang, G.; Liu, W.; Yang, S.; Song, S.; Ma, Y.; Zhou, G.; Liang, X.; Miao, C.; Li, J.; Liu, Y.; et al. Evaluation of humoral and cellular immune responses induced by a cocktail of recombinant African swine fever virus antigens fused with OprI in domestic pigs. Virol. J. 2023 , 20 , 104. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Song, J.; Wang, M.; Zhou, L.; Tian, P.; Sun, Z.; Sun, J.; Wang, X.; Zhuang, G.; Jiang, D.; Wu, Y.; et al. A candidate nanoparticle vaccine comprised of multiple epitopes of the African swine fever virus elicits a robust immune response. J. Nanobiotechnol. 2023 , 21 , 424. [ Google Scholar ] [ CrossRef ]
  • Jenson, J.S.; Childerstone, A.; Takamatsu, H.; Dixon, L.K.; Parkhouse, R.M. The cellular immune recognition of proteins expressed by an African swine fever virus random genomic library. J. Immunol. Methods 2000 , 242 , 33–42. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Lacasta, A.; Ballester, M.; Monteagudo, P.L.; Rodriguez, J.M.; Salas, M.L.; Accensi, F.; Pina-Pedrero, S.; Bensaid, A.; Argilaguet, J.; Lopez-Soria, S.; et al. Expression library immunization can confer protection against lethal challenge with African swine fever virus. J. Virol. 2014 , 88 , 13322–13332. [ Google Scholar ] [ CrossRef ]
  • Yang, G.; Wang, J.; Sun, P.; Qin, J.; Yang, X.; Chen, D.; Zhang, Y.; Zhong, N.; Wang, Z. SARS-CoV-2 epitope-specific T cells: Immunity response feature, TCR repertoire characteristics and cross-reactivity. Front. Immunol. 2023 , 14 , 1146196. [ Google Scholar ] [ CrossRef ]
  • Huang, C.; Cao, C.; Xu, Z.; Lin, Y.; Wu, J.; Weng, Q.; Liu, Z.; Jin, Y.; Chen, P.; Hua, Q. A blocking ELISA based on virus-like nanoparticles chimerized with an antigenic epitope of ASFV P54 for detecting ASFV antibodies. Sci. Rep. 2023 , 13 , 19928. [ Google Scholar ] [ CrossRef ]
  • Xie, C.; Yao, R.; Xia, X. The advances of adjuvants in mRNA vaccines. NPJ Vaccines 2023 , 8 , 162. [ Google Scholar ] [ CrossRef ]
  • Lavelle, E.C.; Ward, R.W. Mucosal vaccines—Fortifying the frontiers. Nat. Rev. Immunol. 2022 , 22 , 236–250. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Pulendran, B.; S Arunachalam, P.; O’Hagan, D.T. Emerging concepts in the science of vaccine adjuvants. Nat. Rev. Drug Discov. 2021 , 20 , 454–475. [ Google Scholar ] [ CrossRef ]
  • Liu, S.; Ding, P.; Du, Y.; Ren, D.; Chen, Y.; Li, M.; Sun, X.; Wang, S.; Chang, Z.; Li, R.; et al. Development and characterization of monoclonal antibodies against the extracellular domain of African swine fever virus structural protein, CD2v. Front. Microbiol. 2022 , 13 , 1056117. [ Google Scholar ] [ CrossRef ]
  • Wang, A.; Chen, Z.; Zhou, J.; Chen, Y.; Liu, Y.; Liu, H.; Liang, C.; Zhu, X.; Zhang, Y.; Xin, C.; et al. Development and characterization of monoclonal antibodies against p37 protein of African swine fever virus. Int. J. Biol. Macromol. 2024 , 264 , 130689. [ Google Scholar ] [ CrossRef ]
  • Mima, K.A.; Katorkina, E.I.; Katorkin, S.A.; Tsybanov, S.Z.; Malogolovkin, A.S. In silico prediction of B- and T-cell epitopes in the CD2v protein of african swine fever virus (African swine fever virus, Asfivirus, Asfarviridae). Vopr. Virusol. 2020 , 65 , 103–112. [ Google Scholar ] [ CrossRef ]
  • Huang, Y.; Zhai, W.; Wang, Z.; He, Y.; Tao, C.; Chu, Y.; Pang, Z.; Zhu, H.; Jia, H. Analysis of the Immunogenicity of African Swine Fever F317L Protein and Screening of T Cell Epitopes. Animals 2024 , 14 , 1331. [ Google Scholar ] [ CrossRef ]
  • Heimerman, M.E.; Murgia, M.V.; Wu, P.; Lowe, A.D.; Jia, W.; Rowland, R.R. Linear epitopes in African swine fever virus p72 recognized by monoclonal antibodies prepared against baculovirus-expressed antigen. J. Vet. Diagn. Invest. 2018 , 30 , 406–412. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Miao, C.; Yang, S.; Shao, J.; Zhou, G.; Ma, Y.; Wen, S.; Hou, Z.; Peng, D.; Guo, H.; Liu, W.; et al. Identification of p72 epitopes of African swine fever virus and preliminary application. Front. Microbiol. 2023 , 14 , 1126794. [ Google Scholar ] [ CrossRef ]
  • Tesfagaber, W.; Wang, W.; Wang, L.; Zhao, R.; Zhu, Y.; Li, F.; Sun, E.; Liu, R.; Bu, Z.; Meng, G.; et al. A highly efficient blocking ELISA based on p72 monoclonal antibody for the detection of African swine fever virus antibodies and identification of its linear B cell epitope. Int. J. Biol. Macromol. 2024 , 268 , 131695. [ Google Scholar ] [ CrossRef ]
  • Ren, D.; Ding, P.; Liu, S.; Zhang, N.; Chen, Y.; Li, Q.; Fan, L.; Chang, Z.; Zhang, G. Development and characterization of recombinant ASFV CD2v protein nanoparticle-induced monoclonal antibody. Int. J. Biol. Macromol. 2022 , 209 , 533–541. [ Google Scholar ] [ CrossRef ]
  • Jiang, W.; Jiang, D.; Li, L.; Wang, J.; Wang, P.; Shi, X.; Zhao, Q.; Liu, B.; Ji, P.; Zhang, G. Identification of Two Novel Linear B Cell Epitopes on the CD2v Protein of African Swine Fever Virus Using Monoclonal Antibodies. Viruses 2022 , 15 , 131. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Liu, H.; Wang, A.; Yang, W.; Liang, C.; Zhou, J.; Chen, Y.; Liu, Y.; Zhou, Y.; Zhang, G. Expression of extracellular domain of ASFV CD2v protein in mammalian cells and identification of B cell epitopes. Virus Res. 2023 , 323 , 199000. [ Google Scholar ] [ CrossRef ]
  • Jia, R.; Zhang, G.; Bai, Y.; Liu, H.; Chen, Y.; Ding, P.; Zhou, J.; Feng, H.; Li, M.; Tian, Y.; et al. Identification of Linear B Cell Epitopes on CD2V Protein of African Swine Fever Virus by Monoclonal Antibodies. Microbiol. Spectr. 2022 , 10 , e0105221. [ Google Scholar ] [ CrossRef ]
  • Song, J.; Wang, M.; Du, Y.; Wan, B.; Zhang, A.; Zhang, Y.; Zhuang, G.; Ji, P.; Wu, Y.; Zhang, G. Identification of a linear B-cell epitope on the African swine fever virus CD2v protein. Int. J. Biol. Macromol. 2023 , 232 , 123264. [ Google Scholar ] [ CrossRef ]
  • Lu, W.; Bai, Y.; Zhang, S.; Zhao, X.; Jin, J.; Zhu, X.; Wang, R.; Wu, Y.; Zhang, A.; Zhang, G.; et al. An Intracellular Epitope of ASFV CD2v Protein Elicits Humoral and Cellular Immune Responses. Animals 2023 , 13 , 1967. [ Google Scholar ] [ CrossRef ]
  • Neilan, J.G.; Zsak, L.; Lu, Z.; Burrage, T.G.; Kutish, G.F.; Rock, D.L. Neutralizing antibodies to African swine fever virus proteins p30, p54, and p72 are not sufficient for antibody-mediated protection. Virology 2004 , 319 , 337–342. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Murgia, M.V.; Mogler, M.; Certoma, A.; Green, D.; Monaghan, P.; Williams, D.T.; Rowland, R.R.R.; Gaudreault, N.N. Evaluation of an African swine fever (ASF) vaccine strategy incorporating priming with an alphavirus-expressed antigen followed by boosting with attenuated ASF virus. Arch. Virol. 2019 , 164 , 359–370. [ Google Scholar ] [ CrossRef ]
  • Petrovan, V.; Yuan, F.; Li, Y.; Shang, P.; Murgia, M.V.; Misra, S.; Rowland, R.R.R.; Fang, Y. Development and characterization of monoclonal antibodies against p30 protein of African swine fever virus. Virus Res. 2019 , 269 , 197632. [ Google Scholar ] [ CrossRef ]
  • Tian, P.; Sun, Z.; Wang, M.; Song, J.; Sun, J.; Zhou, L.; Jiang, D.; Zhang, A.; Wu, Y.; Zhang, G. Identification of a novel linear B-cell epitope on the p30 protein of African swine fever virus using monoclonal antibodies. Virus Res. 2024 , 341 , 199328. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Rodriguez, F.; Alcaraz, C.; Eiras, A.; Yanez, R.J.; Rodriguez, J.M.; Alonso, C.; Rodriguez, J.F.; Escribano, J.M. Characterization and molecular basis of heterogeneity of the African swine fever virus envelope protein p54. J. Virol. 1994 , 68 , 7244–7252. [ Google Scholar ] [ CrossRef ]
  • Desmet, C.; Coelho-Cruz, B.; Mehn, D.; Colpo, P.; Ruiz-Moreno, A. ASFV epitope mapping by high density peptides microarrays. Virus Res. 2024 , 339 , 199287. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Zheng, N.; Li, C.; Hou, H.; Chen, Y.; Zhang, A.; Han, S.; Wan, B.; Wu, Y.; He, H.; Wang, N.; et al. A Novel Linear B-Cell Epitope on the P54 Protein of African Swine Fever Virus Identified Using Monoclonal Antibodies. Viruses 2023 , 15 , 867. [ Google Scholar ] [ CrossRef ]
  • Zhao, H.; Wang, G.; Dong, H.; Wu, S.; Du, Y.; Wan, B.; Ji, P.; Wu, Y.; Jiang, D.; Zhuang, G.; et al. Identification of a Linear B Cell Epitope on p54 of African Swine Fever Virus Using Nanobodies as a Novel Tool. Microbiol. Spectr. 2023 , 11 , e0336222. [ Google Scholar ] [ CrossRef ]
  • Dodantenna, N.; Cha, J.W.; Chathuranga, K.; Chathuranga, W.A.G.; Weerawardhana, A.; Ranathunga, L.; Kim, Y.; Jheong, W.; Lee, J.S. The African Swine Fever Virus Virulence Determinant DP96R Suppresses Type I IFN Production Targeting IRF3. Int. J. Mol. Sci. 2024 , 25 , 2099. [ Google Scholar ] [ CrossRef ]
  • Qi, X.; Feng, T.; Ma, Z.; Zheng, L.; Liu, H.; Shi, Z.; Shen, C.; Li, P.; Wu, P.; Ru, Y.; et al. Deletion of DP148R, DP71L, and DP96R Attenuates African Swine Fever Virus, and the Mutant Strain Confers Complete Protection against Homologous Challenges in Pigs. J. Virol. 2023 , 97 , e0024723. [ Google Scholar ] [ CrossRef ]
  • O’Donnell, V.; Risatti, G.R.; Holinka, L.G.; Krug, P.W.; Carlson, J.; Velazquez-Salinas, L.; Azzinaro, P.A.; Gladue, D.P.; Borca, M.V. Simultaneous Deletion of the 9GL and UK Genes from the African Swine Fever Virus Georgia 2007 Isolate Offers Increased Safety and Protection against Homologous Challenge. J. Virol. 2017 , 91 . [ Google Scholar ] [ CrossRef ]
  • Li, C.; Si, X.Y.; Wang, X.G.; Yan, Z.W.; Hou, H.Y.; You, L.Q.; Chen, Y.L.; Zhang, A.K.; Wang, N.; Sun, A.J.; et al. Preparation and epitope analysis of monoclonal antibodies against African swine fever virus DP96R protein. BMC Vet. Res. 2024 , 20 , 191. [ Google Scholar ] [ CrossRef ]
  • Liu, H.; Zhu, Z.; Feng, T.; Ma, Z.; Xue, Q.; Wu, P.; Li, P.; Li, S.; Yang, F.; Cao, W.; et al. African Swine Fever Virus E120R Protein Inhibits Interferon Beta Production by Interacting with IRF3 To Block Its Activation. J. Virol. 2021 , 95 , e0082421. [ Google Scholar ] [ CrossRef ]
  • Liu, R.; Sun, Y.; Chai, Y.; Li, S.; Li, S.; Wang, L.; Su, J.; Yu, S.; Yan, J.; Gao, F.; et al. The structural basis of African swine fever virus pA104R binding to DNA and its inhibition by stilbene derivatives. Proc. Natl. Acad. Sci. USA 2020 , 117 , 11000–11009. [ Google Scholar ] [ CrossRef ]
  • Frouco, G.; Freitas, F.B.; Coelho, J.; Leitao, A.; Martins, C.; Ferreira, F. DNA-Binding Properties of African Swine Fever Virus pA104R, a Histone-Like Protein Involved in Viral Replication and Transcription. J. Virol. 2017 , 91 . [ Google Scholar ] [ CrossRef ]
  • Chen, Q.; Liu, L.; Guo, S.; Li, L.; Yu, Y.; Liu, Z.; Tan, C.; Chen, H.; Wang, X. Characterization of the monoclonal antibody and the immunodominant B-cell epitope of African swine fever virus pA104R by using mouse model. Microbiol. Spectr. 2024 , 12 , e0140123. [ Google Scholar ] [ CrossRef ]
  • Zhu, Z.; Li, S.; Ma, C.; Yang, F.; Cao, W.; Liu, H.; Chen, X.; Feng, T.; Shi, Z.; Tian, H.; et al. African Swine Fever Virus E184L Protein Interacts with Innate Immune Adaptor STING to Block IFN Production for Viral Replication and Pathogenesis. J. Immunol. 2023 , 210 , 442–458. [ Google Scholar ] [ CrossRef ]
  • Ramirez-Medina, E.; Vuono, E.; Rai, A.; Pruitt, S.; Espinoza, N.; Velazquez-Salinas, L.; Pina-Pedrero, S.; Zhu, J.; Rodriguez, F.; Borca, M.V.; et al. Deletion of E184L, a Putative DIVA Target from the Pandemic Strain of African Swine Fever Virus, Produces a Reduction in Virulence and Protection against Virulent Challenge. J. Virol. 2022 , 96 , e0141921. [ Google Scholar ] [ CrossRef ]
  • Tesfagaber, W.; Lan, D.; Wang, W.; Zhao, R.; Yin, L.; Yang, M.; Zhu, Y.; Sun, E.; Liu, R.; Lin, W.; et al. Identification of two novel B cell epitopes on E184L protein of African swine fever virus using monoclonal antibodies. Virus Res. 2024 , 346 , 199412. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Urbano, A.C.; Ferreira, N.; Jordao, N.; Boinas, F.; Martins, C.; Ferreira, F. Targeted mutagenesis of the beta-strand DNA binding region of African swine fever virus histone-like protein (pA104R) impairs DNA-binding activity and antibody recognition. Antiviral Res. 2024 , 221 , 105784. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Song, J.; Wang, M.; Zhou, L.; Tian, P.; Sun, J.; Sun, Z.; Guo, C.; Wu, Y.; Zhang, G. A novel conserved B-cell epitope in pB602L of African swine fever virus. Appl. Microbiol. Biotechnol. 2024 , 108 , 78. [ Google Scholar ] [ CrossRef ]
  • Shi, L.F.; Ren, H.; Zhang, B.; Shi, S.Y.; Shao, H.C.; Xing, H.; Zhao, Y.Y.; Lin, Z.Z.; Zhang, Y.; Han, S.; et al. Preparation and epitope mapping of monoclonal antibodies against African swine fever virus p22 protein. Int. J. Biol. Macromol. 2024 , 255 , 128111. [ Google Scholar ] [ CrossRef ]
  • Zhang, S.J.; Niu, B.; Liu, S.M.; Zhu, Y.M.; Zhao, D.M.; Bu, Z.G.; Hua, R.H. Identification of Two Linear Epitopes on MGF_110-13L Protein of African Swine Fever Virus with Monoclonal Antibodies. Animals 2024 , 14 , 1951. [ Google Scholar ] [ CrossRef ]
  • Burmakina, G.; Malogolovkin, A.; Tulman, E.R.; Xu, W.; Delhon, G.; Kolbasov, D.; Rock, D.L. Identification of T-cell epitopes in African swine fever virus CD2v and C-type lectin proteins. J. Gen. Virol. 2019 , 100 , 259–265. [ Google Scholar ] [ CrossRef ]
  • Liu, Q.; Ma, B.; Qian, N.; Zhang, F.; Tan, X.; Lei, J.; Xiang, Y. Structure of the African swine fever virus major capsid protein p72. Cell Res. 2019 , 29 , 953–955. [ Google Scholar ] [ CrossRef ]
  • Sun, W.; Zhang, H.; Fan, W.; He, L.; Chen, T.; Zhou, X.; Qi, Y.; Sun, L.; Hu, R.; Luo, T.; et al. Evaluation of Cellular Immunity with ASFV Infection by Swine Leukocyte Antigen (SLA)-Peptide Tetramers. Viruses 2021 , 13 , 2264. [ Google Scholar ] [ CrossRef ]
  • Yang, J.; Li, S.; Feng, T.; Zhang, X.; Yang, F.; Cao, W.; Chen, H.; Liu, H.; Zhang, K.; Zhu, Z.; et al. African Swine Fever Virus F317L Protein Inhibits NF-kappaB Activation To Evade Host Immune Response and Promote Viral Replication. mSphere 2021 , 6 , e0065821. [ Google Scholar ] [ CrossRef ]
  • Xu, L.; Hao, F.; Jeong, D.G.; Chen, R.; Gan, Y.; Zhang, L.; Yeom, M.; Lim, J.W.; Yu, Y.; Bai, Y.; et al. Mucosal and cellular immune responses elicited by nasal and intramuscular inoculation with ASFV candidate immunogens. Front. Immunol. 2023 , 14 , 1200297. [ Google Scholar ] [ CrossRef ]
  • Dodantenna, N.; Ranathunga, L.; Chathuranga, W.A.G.; Weerawardhana, A.; Cha, J.W.; Subasinghe, A.; Gamage, N.; Haluwana, D.K.; Kim, Y.; Jheong, W.; et al. African Swine Fever Virus EP364R and C129R Target Cyclic GMP-AMP to Inhibit the cGAS-STING Signaling Pathway. J. Virol. 2022 , 96 , e0102222. [ Google Scholar ] [ CrossRef ]
  • Zhai, W.; Huang, Y.; He, Y.; Chu, Y.; Tao, C.; Pang, Z.; Wang, Z.; Zhu, H.; Jia, H. Immunogenicity Analysis and Identification of Potential T-Cell Epitopes in C129R Protein of African Swine Fever Virus. Microorganisms 2024 , 12 , 1056. [ Google Scholar ] [ CrossRef ]
  • Lokhandwala, S.; Petrovan, V.; Popescu, L.; Sangewar, N.; Elijah, C.; Stoian, A.; Olcha, M.; Ennen, L.; Bray, J.; Bishop, R.P.; et al. Adenovirus-vectored African Swine Fever Virus antigen cocktails are immunogenic but not protective against intranasal challenge with Georgia 2007/1 isolate. Vet. Microbiol. 2019 , 235 , 10–20. [ Google Scholar ] [ CrossRef ]
  • Zajac, M.D.; Trujillo, J.D.; Yao, J.; Kumar, R.; Sangewar, N.; Lokhandwala, S.; Sang, H.; Mallen, K.; McCall, J.; Burton, L.; et al. Immunization of pigs with replication-incompetent adenovirus-vectored African swine fever virus multi-antigens induced humoral immune responses but no protection following contact challenge. Front. Vet. Sci. 2023 , 10 , 1208275. [ Google Scholar ] [ CrossRef ]
  • Levast, B.; Awate, S.; Babiuk, L.; Mutwiri, G.; Gerdts, V.; van Drunen Littel-van den Hurk, S. Vaccine Potentiation by Combination Adjuvants. Vaccines 2014 , 2 , 297–322. [ Google Scholar ] [ CrossRef ]
  • Pikalo, J.; Porfiri, L.; Akimkin, V.; Roszyk, H.; Pannhorst, K.; Kangethe, R.T.; Wijewardana, V.; Sehl-Ewert, J.; Beer, M.; Cattoli, G.; et al. Vaccination With a Gamma Irradiation-Inactivated African Swine Fever Virus Is Safe But Does Not Protect against a Challenge. Front. Immunol. 2022 , 13 , 832264. [ Google Scholar ] [ CrossRef ]
  • Andrianarivo, A.G.; Rowe, J.D.; Barr, B.C.; Anderson, M.L.; Packham, A.E.; Sverlow, K.W.; Choromanski, L.; Loui, C.; Grace, A.; Conrad, P.A. A POLYGEN-adjuvanted killed Neospora caninum tachyzoite preparation failed to prevent foetal infection in pregnant cattle following i.v./i.m. experimental tachyzoite challenge. Int. J. Parasitol. 2000 , 30 , 985–990. [ Google Scholar ] [ CrossRef ]
  • Blome, S.; Gabriel, C.; Beer, M. Modern adjuvants do not enhance the efficacy of an inactivated African swine fever virus vaccine preparation. Vaccine 2014 , 32 , 3879–3882. [ Google Scholar ] [ CrossRef ]
  • Hofacre, C.L.; Rosales, A.G.; Costa, M.D.; Cookson, K.; Schaeffer, J.; Jones, M.K. Immunity and Protection Provided by Live Modified Vaccines Against Paratyphoid Salmonella in Poultry-An Applied Perspective. Avian Dis. 2021 , 65 , 295–302. [ Google Scholar ] [ CrossRef ]
  • Mancera Gracia, J.C.; Pearce, D.S.; Masic, A.; Balasch, M. Influenza A Virus in Swine: Epidemiology, Challenges and Vaccination Strategies. Front. Vet. Sci. 2020 , 7 , 647. [ Google Scholar ] [ CrossRef ]
  • Renukaradhya, G.J.; Meng, X.J.; Calvert, J.G.; Roof, M.; Lager, K.M. Live porcine reproductive and respiratory syndrome virus vaccines: Current status and future direction. Vaccine 2015 , 33 , 4069–4080. [ Google Scholar ] [ CrossRef ]
  • Lokhandwala, S.; Waghela, S.D.; Bray, J.; Martin, C.L.; Sangewar, N.; Charendoff, C.; Shetti, R.; Ashley, C.; Chen, C.H.; Berghman, L.R.; et al. Induction of Robust Immune Responses in Swine by Using a Cocktail of Adenovirus-Vectored African Swine Fever Virus Antigens. Clin. Vaccine Immunol. 2016 , 23 , 888–900. [ Google Scholar ] [ CrossRef ]
  • Cadenas-Fernandez, E.; Sanchez-Vizcaino, J.M.; Kosowska, A.; Rivera, B.; Mayoral-Alegre, F.; Rodriguez-Bertos, A.; Yao, J.; Bray, J.; Lokhandwala, S.; Mwangi, W.; et al. Adenovirus-vectored African Swine Fever Virus Antigens Cocktail Is Not Protective against Virulent Arm07 Isolate in Eurasian Wild Boar. Pathogens 2020 , 9 , 171. [ Google Scholar ] [ CrossRef ]
  • O’Hagan, D.T.; Ott, G.S.; Nest, G.V.; Rappuoli, R.; Giudice, G.D. The history of MF59((R)) adjuvant: A phoenix that arose from the ashes. Expert. Rev. Vaccines 2013 , 12 , 13–30. [ Google Scholar ] [ CrossRef ]
  • Cadenas-Fernandez, E.; Sanchez-Vizcaino, J.M.; van den Born, E.; Kosowska, A.; van Kilsdonk, E.; Fernandez-Pacheco, P.; Gallardo, C.; Arias, M.; Barasona, J.A. High Doses of Inactivated African Swine Fever Virus Are Safe, but Do Not Confer Protection against a Virulent Challenge. Vaccines 2021 , 9 , 242. [ Google Scholar ] [ CrossRef ]
  • Leitao, A.; Malur, A.; Cornelis, P.; Martins, C.L. Identification of a 25-aminoacid sequence from the major African swine fever virus structural protein VP72 recognised by porcine cytotoxic T lymphocytes using a lipoprotein based expression system. J. Virol. Methods 1998 , 75 , 113–119. [ Google Scholar ] [ CrossRef ]
  • Leitao, A.; Malur, A.; Cartaxeiro, C.; Vasco, G.; Cruz, B.; Cornelis, P.; Martins, C.L. Bacterial lipoprotein based expression vectors as tools for the characterisation of African swine fever virus (ASFV) antigens. Arch. Virol. 2000 , 145 , 1639–1657. [ Google Scholar ] [ CrossRef ]
  • Liu, W.; Jiang, P.; Song, T.; Yang, K.; Yuan, F.; Gao, T.; Liu, Z.; Li, C.; Guo, R.; Xiao, S.; et al. A Recombinant Chimera Vaccine Composed of LTB and Mycoplasma hyopneumoniae Antigens P97R1, mhp390 and P46 Elicits Cellular Immunologic Response in Mice. Vaccines 2023 , 11 , 1291. [ Google Scholar ] [ CrossRef ]
  • Zhang, H.; Zhao, S.; Zhang, H.; Shen, Y.; Zhang, P.; Shan, H.; Cai, X. Orally administered recombinant Lactobacillus expressing African swine fever virus antigens that induced immunity responses. Front. Microbiol. 2022 , 13 , 1103327. [ Google Scholar ] [ CrossRef ]
  • Liao, Z.; Huang, J.; Lo, P.C.; Lovell, J.F.; Jin, H.; Yang, K. Self-adjuvanting cancer nanovaccines. J. Nanobiotechnol. 2022 , 20 , 345. [ Google Scholar ] [ CrossRef ]
  • Lu, H.; Zhou, X.; Wu, Z.; Zhang, X.; Zhu, L.; Guo, X.; Zhang, Q.; Zhu, S.; Zhu, H.; Sun, H. Comparison of the mucosal adjuvanticities of two Toll-like receptor ligands for recombinant adenovirus-delivered African swine fever virus fusion antigens. Vet. Immunol. Immunopathol. 2021 , 239 , 110307. [ Google Scholar ] [ CrossRef ]
  • Huang, Q.; Niu, T.; Zou, B.; Wang, J.; Xin, J.; Niu, H.; Li, N.; Jiang, Y.; Bao, J.; Zhang, D.; et al. Lactobacillus plantarum Surface-Displayed ASFV (p14.5) Can Stimulate Immune Responses in Mice. Vaccines 2022 , 10 , 355. [ Google Scholar ] [ CrossRef ]
  • Gallardo, C.; Fernandez-Pinero, J.; Arias, M. African swine fever (ASF) diagnosis, an essential tool in the epidemiological investigation. Virus Res. 2019 , 271 , 197676. [ Google Scholar ] [ CrossRef ]
  • Li, L.; Qiao, S.; Li, G.; Tong, W.; Dong, S.; Liu, J.; Guo, Z.; Zheng, H.; Zhao, R.; Tong, G.; et al. The Indirect ELISA and Monoclonal Antibody against African Swine Fever Virus p17 Revealed Efficient Detection and Application Prospects. Viruses 2022 , 15 , 50. [ Google Scholar ] [ CrossRef ]
  • Afayibo, D.J.A.; Zhang, Z.; Sun, H.; Fu, J.; Zhao, Y.; Amuda, T.O.; Wu, M.; Du, J.; Guan, G.; Niu, Q.; et al. Establishment of an ELISA Based on a Recombinant Antigenic Protein Containing Multiple Prominent Epitopes for Detection of African Swine Fever Virus Antibodies. Microorganisms 2024 , 12 , 943. [ Google Scholar ] [ CrossRef ]
  • Li, D.; Zhang, Q.; Liu, Y.; Wang, M.; Zhang, L.; Han, L.; Chu, X.; Ding, G.; Li, Y.; Hou, Y.; et al. Indirect ELISA Using Multi-Antigenic Dominants of p30, p54 and p72 Recombinant Proteins to Detect Antibodies against African Swine Fever Virus in Pigs. Viruses 2022 , 14 , 2660. [ Google Scholar ] [ CrossRef ]
  • Rossjohn, J.; Gras, S.; Miles, J.J.; Turner, S.J.; Godfrey, D.I.; McCluskey, J. T cell antigen receptor recognition of antigen-presenting molecules. Annu. Rev. Immunol. 2015 , 33 , 169–200. [ Google Scholar ] [ CrossRef ]
  • Wang, Z.; Zhang, J.; Li, F.; Zhang, Z.; Chen, W.; Zhang, X.; Sun, E.; Zhu, Y.; Liu, R.; He, X.; et al. The attenuated African swine fever vaccine HLJ/18-7GD provides protection against emerging prevalent genotype II variants in China. Emerg. Microbes Infect. 2024 , 13 , 2300464. [ Google Scholar ] [ CrossRef ]
  • Argilaguet, J.M.; Perez-Martin, E.; Nofrarias, M.; Gallardo, C.; Accensi, F.; Lacasta, A.; Mora, M.; Ballester, M.; Galindo-Cardiel, I.; Lopez-Soria, S.; et al. DNA vaccination partially protects against African swine fever virus lethal challenge in the absence of antibodies. PLoS ONE 2012 , 7 , e40942. [ Google Scholar ] [ CrossRef ]
  • Urbano, A.C.; Ferreira, F. African swine fever control and prevention: An update on vaccine development. Emerg. Microbes Infect. 2022 , 11 , 2021–2033. [ Google Scholar ] [ CrossRef ]
  • Borca, M.V.; Ramirez-Medina, E.; Silva, E.; Rai, A.; Espinoza, N.; Velazquez-Salinas, L.; Gladue, D.P. ASF Vaccine Candidate ASFV-G-∆I177L Does Not Exhibit Residual Virulence in Long-Term Clinical Studies. Pathogens 2023 , 12 , 805. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Shawan, M.; Sharma, A.R.; Halder, S.K.; Arian, T.A.; Shuvo, M.N.; Sarker, S.R.; Hasan, M.A. Advances in Computational and Bioinformatics Tools and Databases for Designing and Developing a Multi-Epitope-Based Peptide Vaccine. Int. J. Pept. Res. Ther. 2023 , 29 , 60. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Tang, M.; Cai, J.H.; Diao, H.Y.; Guo, W.M.; Yang, X.; Xing, S. The progress of peptide vaccine clinical trials in gynecologic oncology. Hum. Vaccin. Immunother. 2022 , 18 , 2062982. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Simbulan, A.M.; Banico, E.C.; Sira, E.; Odchimar, N.M.O.; Orosco, F.L. Immunoinformatics-guided approach for designing a pan-proteome multi-epitope subunit vaccine against African swine fever virus. Sci. Rep. 2024 , 14 , 1354. [ Google Scholar ] [ CrossRef ]
  • Nguyen, T.L.; Samuel Leon Magdaleno, J.; Rajjak Shaikh, A.; Choowongkomon, K.; Li, V.; Lee, Y.; Kim, H. Designing a multi-epitope candidate vaccine by employing immunoinformatics approaches to control African swine fever spread. J. Biomol. Struct. Dyn. 2023 , 41 , 10214–10229. [ Google Scholar ] [ CrossRef ]

Click here to enlarge figure

ASFV
(Protein)
SequenceApplicationReferences
VaccineDiagnosis
p72 TLVDPFGRPI
QRTCSHTNPKFLSQHF
FPENSHNIQTAGKQD
AGKQDITPITDATY
+[ ]
PENSHNIQTA +[ ]
NSHNIQ +[ ]
CD2v SILE ++[ ]
LDSNITNDNNINGVSWNFFNNSF +[ ]
DINGVSWN
GTNTNIY
+[ ]
TCKKNNGTNT
VKYTNESILE
+[ ]
FVKYT
EYNWN
SSNY
++[ ]
WNNSNINNFT + [ ]
EPSPREP ++[ ]
FNKVIRAHNFIQTIYGTPLK +[ ]
p54 IVLIYLFSSRKKKAA +[ ]
TMSAIENLR +[ ]
QQWVEV +[ ]
p37 LGDAIAGRLMQLD
DWKATVSAIELEY
TTGDTLAQVFESFPT
AGRLMQLDTEKNARI
+[ ]
E120REEFEPIPDYDTTST+[ ]
DP96R THDCSLKEK
YWKGIKRGND
+[ ]
pA104RKPTITKOELYSI++[ ]
KFTVVTVKA
KAVKIRALK
+[ ]
E184L IQRQGFL
DPTEFF
++[ ]
pB602L SKENLTPDE ++[ , ]
p22 KVCKVDKDCGSGEHC
VYNNPHHPVLKYGKDHIIALP
+[ ]
MGF_110-13L WDCQDGICKNKITESRFIDS
GDHQQLSIKQ
+[ ]
ASFV
(Protein)
SequenceApplicationReference
VaccineDiagnosis
F317L SRRSLVNPWT +[ ]
CD2v
Intracellular Epitope
EPSPREP +[ ]
p72P351(SRISNIKNNKY)
P334(SDYTL)
P366(SSYGGAK)
+[ ]
C129R LQNPYEAVI
GHVTWAVPY
AKPDAIMLT
ALNQNVLTL
+[ ]
Adjuvant NameThe Components of ASFV VaccineImmune RoutReference
Montanide ISAOPMT + OPET + OCET + MontanideTM ISA206™i.m.[ ]
Ad5-ASFV Mix + Montanide ISA-201™i.m.[ ]
30 kGy Irradiated ASFV + Montanide ISA-201™i.m.[ ]
Inactivated ASFV + MontanideTM ISA201™i.m.[ ]
Polygen™30 kGy Irradiated ASFV + Polygen™i.m.[ ]
BEI-inactivated ASFV + Polygen™i.m.[ ]
ZoetisAd-ASFV cocktail-II + Zoetisi.m.[ ]
Ad5-ASFV 4-way cocktail + Zoetisi.m.[ ]
ENABLAd5-ASFV 4-way cocktail + ENABLi.m.[ ]
BioMize Ad-ASFV cocktail-I + BioMizei.m.[ ]
Ad5-ASFV + BioMizei.m.[ ]
MF59 Inactivated ASFV + MF59 i.m.[ ]
Emulsigen DBEI-inactivated ASFV + Emulsigen Di.m.[ ]
Silica oilInactivated ASFV + Silica oili.m.[ ]
mGNEInactivated ASFV + mGNEi.m.[ ]
LTBMG1363/pMG36e-p30 + p54 + p72-LTB-HisOral[ ]
OprIOPMT + OPET + OCET + OprIi.m.[ ]
FlaBrAd-ASFV CD2v-p30-p54-FlaBi.n.[ ]
Hsp70rAd-ASFV CD2v-p30-p54-Hsp70i.n.[ ]
IL-33NC8-pLP-S-p14.5-IL-33-MusOral[ ]
CTA1-DDNC8-pLP-S-CTA1-p14.5-CTA1-DDOral[ ]
SpyTag-SpyCatcherSpyTag/SpyCatcher + ASFV epitopes + SpyTag-SpyCatcheri.m.[ ]
The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

Wu, Q.; Li, C.; Zhu, B.; Zhu, J.; Yang, K.; Liu, Z.; Liu, W.; Gao, T.; Yuan, F.; Guo, R.; et al. Advancement in the Antigenic Epitopes and Vaccine Adjuvants of African Swine Fever Virus. Pathogens 2024 , 13 , 706. https://doi.org/10.3390/pathogens13080706

Wu Q, Li C, Zhu B, Zhu J, Yang K, Liu Z, Liu W, Gao T, Yuan F, Guo R, et al. Advancement in the Antigenic Epitopes and Vaccine Adjuvants of African Swine Fever Virus. Pathogens . 2024; 13(8):706. https://doi.org/10.3390/pathogens13080706

Wu, Qiong, Chang Li, Bo Zhu, Jiajia Zhu, Keli Yang, Zewen Liu, Wei Liu, Ting Gao, Fangyan Yuan, Rui Guo, and et al. 2024. "Advancement in the Antigenic Epitopes and Vaccine Adjuvants of African Swine Fever Virus" Pathogens 13, no. 8: 706. https://doi.org/10.3390/pathogens13080706

Article Metrics

Article access statistics, further information, mdpi initiatives, follow mdpi.

MDPI

Subscribe to receive issue release notifications and newsletters from MDPI journals

slide1

Virus Replication

Aug 16, 2014

220 likes | 453 Views

Virus Replication. What is a Virus?. Virus : A biological particle composed of nucleic acid and protein Intracellular Parasites : organism that must “live” inside a host. Reproduce Have nucleic acid Adapt to surroundings Have organization. Not made of cells or organelles

Share Presentation

  • release same
  • transcription translation
  • high risk activities
  • nucleic acid
  • low risk activities

wilda

Presentation Transcript

What is a Virus? • Virus: A biological particle composed of nucleic acid and protein • Intracellular Parasites: organism that must “live” inside a host

Reproduce Have nucleic acid Adapt to surroundings Have organization Not made of cells or organelles Can’t reproduce on own Don’t metabolize energy Don’t perform cellular processes Are viruses alive? No Yes

Virus Parts • All Have: • 1) Capsid: coat of protein that surrounds nucleic acid • 2) Nucleic Acid: RNA or DNA • Some Have: • Tail Fibers: Used for attachment (not legs) • Shapes vary

Viral Replication • Bacteriophages (infect bacteria) are often studied • Replication is similar with many animal viruses

The Lytic Cycle 1st Step: Attachment • Virus lands on cell membrane • Virus attaches to a cell receptor • No attachment = No infection • Virus acts as a “key” to the receptor

The Lytic Cycle 2nd Step: Entry • Virus enzyme weakens cell membrane • Genetic material (DNA or RNA) enters host cell endocytosis

The Lytic Cycle 3rd Step: Replication • Virus DNA/RNA uses ribosomes to make virus proteins • Virus proteins created by transcription/ translation

The Lytic Cycle 4th Step: Assembly • New virus proteins are assembled in the cytoplasm

The Lytic Cycle 5th Step: Release • Virus enzyme causes cell membrane to burst • Viruses are released to find new host • Cycle repeats

The Lysogenic Cycle 1st step: Attachment = Same 2nd Step: Entry = Same

Lysogenic Cycle 3rd step: Replication • Virus DNA combines with cell DNA, and waits • Provirus = cell DNA + viral DNA • Cell divides by mitosis • Each new cell will contain the provirus

The Lysogenic Cycle 4th Step: Assembly Same, except many new viruses are being assembled in many cells 5th Step: Release Same, except many cells burst releasing many more viruses

RNA viruses Contains the enzyme Reverse Transcriptase Steps 1) Virus RNA enters host cell 2) Cell’s ribosomes create virus DNA 3) Virus DNA combines with cell DNA (becomes dormant) 4) Cells divide as usual 5) Once active, normal steps of transcription/translation followed Retroviruses

High risk activities: 1) Sex (oral, vaginal, anal) 2) Sharing needles (tattoos, piercings, drugs) Low risk activities: 3) Mother to child in womb 4) Breast milk 5) Blood transfusions How is HIV contracted?

New Exposures to HIV (2006) What is AIDS? Gender of those living with HIV (2003) • Acquired Immunodeficiency Syndrome • AIDS results when: • 1) Less then 200 T cells per 1mm³ blood • 2) Multiple symptoms/infections from HIV appear • Therefore, the HIV virus causes the disease AIDS

Female Transmission of HIV (2006) Male Transmission of HIV (2006) Want more stats? Go to the Centers for Disease Control’s Website http://www.cdc.gov/hiv/topics/surveillance/united_states.htm

AIDS Quilt 3ft x 6ft quilt panel can be created as a memorial tribute

AIDS Quilt Panels from across the world are sewn together

AIDS Quilt Panels travel around the world to help remember and raise awareness

AIDS Quilt Over 44,000 panels exist today

  • More by User

Virus structure, classification and replication

Virus structure, classification and replication

Virus structure, classification and replication. Viruses defined. Obligate intracellular parasites. Under attack!. (From Medical Microbiology, 4 th ed., Murray, Rosenthal, Kobayashi &amp; Pfaller, Mosby Inc., 2002, Fig. 65-1.). Structures compared.

1.54k views • 24 slides

VIRUS REPLICATION

VIRUS REPLICATION

VIRUS REPLICATION. PETER H. RUSSELL, BVSc, PhD, FRCPath, MRCVS Department of Pathology and Infectious Diseases, The Royal Veterinary College, Royal College Street, London NW1 OTU. E-mail Web site. Objectives Students should be able to:. describe in outline how to cultivate viruses

401 views • 19 slides

Replication

Replication

Ondřej Ševeček | GOPAS a.s. | MCM: Directory Services | MVP: Enterprise Security | ondrej@ sevecek.com | www.sevecek.com |. Replication. Active Directory Replication. Intro. Central Database. LDAP – Lightweight Directory Access Protocol database query language similar to SQL

1.54k views • 126 slides

Lec . 8 Virus genome replication Dr. Ahmed K. Ali

Lec . 8 Virus genome replication Dr. Ahmed K. Ali

Lec . 8 Virus genome replication Dr. Ahmed K. Ali. The genome of the infecting virus is replicated so that viral transcription can be amplified and to provide copies of the genome for progeny virions .

454 views • 19 slides

Replication of Small DNA Virus

Replication of Small DNA Virus

Replication of Small DNA Virus. Animal Virus DNA Genomes. Larger size range (5 – 200 kbp) than RNA viruses Similar to host genome, may use cell machinery for DNA replication &amp; transcription Problems of dependence on host cell for DNA replication: Cell must be in S phase for DNA synthesis

781 views • 29 slides

Replication

Updating replicated data. F. Alice. Bob. . . F'. F''. . . Bob. Alice. Update and consistency are primary issues.. shared. Separate replicas. . . Passive replication. At most one replica can be the primary server Each client maintains a variable L(leader) that specifies the replica towhich it wi

559 views • 43 slides

RNA VIRUS REPLICATION STRATEGIES

RNA VIRUS REPLICATION STRATEGIES

RNA VIRUS REPLICATION STRATEGIES. Objectives. Classes of RNA genomes Baltimore Classification Different strategies used by RNA viruses RNA replication and mRNA synthesis Initiation, elongation and termination of replication Examples . RNA -&gt; RNA. RNA-dependent RNA polymerase . RNA -&gt; DNA.

889 views • 24 slides

RNA VIRUS REPLICATION/ TRNSLATION STRATEGIES

RNA VIRUS REPLICATION/ TRNSLATION STRATEGIES

RNA VIRUS REPLICATION/ TRNSLATION STRATEGIES. Sadia Anjum Virology lecture 5. RNA -&gt; RNA. RNA-dependent RNA polymerase . RNA -&gt; DNA. RNA-dependent DNA polymerase - reverse transcriptase . Host cell DNA -&gt; RNA. DNA-dependent RNA polymerase . RNA VIRUS STRATEGIES. AAA.

762 views • 53 slides

Replication

Replication . Topics. Why Replication? System Model Consistency Models One approach to consistency management and dealing with failures. Readings. Van Steen and Tanenbaum: 6.1, 6.2 and 6.3, 6.4 Coulouris: 11,14. Fault-tolerance / High availability

396 views • 25 slides

Virus Genome Replication

Virus Genome Replication

LECTURE 12:. Virus Genome Replication. Waqas Nasir Chaudhry. Viro100: Virology 3 Credit hours NUST Centre of Virology &amp; Immunology. Transcription of virus genomes. There are four main categories of virus genome: dsDNA , ssDNA , dsRNA and ssRNA

461 views • 8 slides

DNA Virus Replication Chapter 9

DNA Virus Replication Chapter 9

DNA Virus Replication Chapter 9. Figure 9.1 Mapping ori for SV40. Figure 9.2 Recognition and unwinding. Figure 9.5 Leading and Lagging strands. Figure 9.7 SV40. Fox 9.4 Spooling of DNA in SV40 replication. Figure 9.10 Replication of Adenoviruses. Figure 9.11 HSV genome.

510 views • 25 slides

Replication of Large DNA Virus

Replication of Large DNA Virus

Replication of Large DNA Virus. Herpesvirus, Poxvirus. Family Herpesviridae. “creeping” spread of rash &amp; vesicle lesions Widely found in nature – plants, fungi, animals, humans Highly infectious Infections – acute, persistent, transform Eight Human herpesvirus (HHV 1-8)

803 views • 41 slides

Replication

Replication. دانشگاه علوم پزشكي وخدمات بهداشتي درماني تهران. Dr. Parvin Pasalar Tehran University of Medical Sciences. Objectives: To know and explain What are needed for Replication Template properties Building Blocks Enzymes Protein Factors Different Stages of Replication

665 views • 36 slides

Replication of Double-Stranded RNA Virus; Subviral Pathogens

Replication of Double-Stranded RNA Virus; Subviral Pathogens

Replication of Double-Stranded RNA Virus; Subviral Pathogens. Family Reoviridae. “ r espiratory, e nteric, o rphan” dsRNA Double icosadehral capsid, 60 nm Outer capsid + short spikes Inner nucleocapsid core Infects plants, insects, animals. Genus: Orthoreovirus.

800 views • 39 slides

Inhibition of rabies virus replication by microRNA

Inhibition of rabies virus replication by microRNA

Inhibition of rabies virus replication by microRNA. N.ISRASENA , N. RATANASETYUTH , P. SUPAVONWONG, P. VIROJANAPIROM, T.HEMACHUDHA  Department of Pharmacology, Chulalongkorn University Hospital. Queen Saovabha Memorial Institute, Thai Red Cross Society, Bangkok, Thailand .

530 views • 30 slides

Human Viral Disease; Virus Replication Cycle

Human Viral Disease; Virus Replication Cycle

Human Viral Disease; Virus Replication Cycle. Human-Virus Interaction. Virus extinction Clear virus, immunity Large number of deaths Small Population Favors persistent virus infection Virus infection with an immunological naïve person i.e. herpes simplex virus, parent to newborn

1.01k views • 52 slides

Replication of Negative-Sense RNA Virus (Monopartite)

Replication of Negative-Sense RNA Virus (Monopartite)

Replication of Negative-Sense RNA Virus (Monopartite). (-)RNA Virus Replication. Family Rhabdoviridae. “rod” (-)RNA viruses Coiled nucleocapsid into cylindrical structure Envelope, large spikes, 70x180 nm, “bullet-shape”. Genus: Vesiculovirus. “vesicles” – fluid filled lesions

1.28k views • 31 slides

Replication of Reverse-Transcribing Hepatitis B Virus

Replication of Reverse-Transcribing Hepatitis B Virus

Replication of Reverse-Transcribing Hepatitis B Virus. Family Hepadnaviridae. Hepatitis – inflammation of liver (injury, chemical/drug, infectious agent) DNA virus – tropism for hepatocytes Unusual genome architecture and mode of replication ( DNA pol;reverse transcription )

607 views • 37 slides

VIRUS VIRUS VIRUS

VIRUS VIRUS VIRUS

VIRUS VIRUS VIRUS. VIRUS VIRUS VIRUS. VIRUS VIRUS VIRUS. VIRUS VIRUS VIRUS. VIRUS VIRUS VIRUS. VIRUS VIRUS VIRUS. VIRUS VIRUS VIRUS. VIRUS VIRUS VIRUS. VIRUS VIRUS VIRUS. VIRUS VIRUS VIRUS. VIRUS VIRUS VIRUS. VIRUS VIRUS VIRUS. VIRUS VIRUS VIRUS. VIRUS VIRUS

271 views • 8 slides

Replication-Competent Virus Testing

Replication-Competent Virus Testing

Recombinant viruses are especially useful for gene transfer and protein expression in vivo and in vitro. During viral vector production, particles may be generated which are replication competent through recombination with host sequences or by contamination.https://www.creative-biogene.com/Services/Custom-Viral-Service/Replication-Competent-Virus-Testing.html

20 views • 1 slides

VIRUS REPLICATION STRATEGIES Dr. Sobia Manzoor MV, Lecture 06

VIRUS REPLICATION STRATEGIES Dr. Sobia Manzoor MV, Lecture 06

VIRUS REPLICATION STRATEGIES Dr. Sobia Manzoor MV, Lecture 06. Introduction. Replication of genetic information is the single most distinctive characteristic of living organisms, which is accomplished with great economy and simplicity among viruses.

244 views • 21 slides

Replication

Replication. CSE 593 - Transaction Processing Philip A. Bernstein. Outline. 1. Introduction 2. Singe-Master Primary-Copy Replication 3. Multi-Master Replication 4. Other Approaches. 1. Introduction.

296 views • 27 slides

IMAGES

  1. Viral replication infographic for education Vector Image

    presentation on viral replication

  2. Replication Of Virus

    presentation on viral replication

  3. PPT

    presentation on viral replication

  4. Reproduction in Viruses

    presentation on viral replication

  5. PPT

    presentation on viral replication

  6. PPT

    presentation on viral replication

COMMENTS

  1. PDF Viral Replication: Basic Concepts

    Steps in Viral Replication: Assembly and Release. (Sixth and Seventh Steps) Process involves bringing together newly formed genomic nucleic acid and structural proteins to form the nucleocapsid of the virus. Nonenveloped viruses exhibit full maturation in the cytoplasm or nucleus with disintegration of cell.

  2. PDF PowerPoint Presentation

    the progress of understanding virus replication. • Every virus family employs a unique strategy for replication. • One important concept to unify and simply the replication process was proposed by David Baltimore at 1978, to assign viruses to one of six classes based on their genome structure and the pathways they use to produce their mRNAs.

  3. Virus replication

    4. Replication: After the viral genome has been uncoated, transcription or translation of the viral genome is initiated. It is this stage of viral replication that differs greatly between DNA and RNA viruses and viruses with opposite nucleic acid polarity. This process culminates in the de novo synthesis of viral proteins and genome.

  4. Viral replication

    Viral replication is the formation of biological viruses during the infection process in the target host cells. Viruses must first get into the cell before viral replication can occur. Through the generation of abundant copies of its genome and packaging these copies, the virus continues infecting new hosts. Replication between viruses is greatly varied and depends on the type of genes ...

  5. REPLICATION OF THE VIRUS

    13 The viral replication cycle can be separated into several phases. Early phase of infection, the virus must recognize an appropriate target cell; attache to the cell; penetrate the plasma membrane and be taken up by the cell; then release (uncoat) its genome into the cytoplasm, and if necessary, deliver the genome to the host nucleus Late phase begins with the start of genome replication and ...

  6. Coronavirus Biology

    In this presentation, you will learn general principles of viral biology, with specific exemplars from the SARS-CoV-2 novel coronavirus. By the end of the presentation you will be able to answer the following questions: ... The general method of viral replication results in the destruction of the infected cell and the release of new viruses in ...

  7. PDF Virus Replication

    Virus Replication John Goulding, Imperial College London, UK As viruses are obligate intracellular pathogens they cannot replicate without the machinery and metabolism of a host cell . Although the replicative life cycle of viruses differs greatly between species and category of virus, there are six basic stages that are essential for viral ...

  8. Viral and host heterogeneity and their effects on the viral ...

    The textbook depiction of the viral replication cycle 1,2 contains four relatively uniform core steps: entry, replication, assembly and egress (Fig. 1a).Innovative studies have uncovered novel and ...

  9. The Viral Life Cycle

    This CLEAR & SIMPLE tutorial describes the steps of the viral life cycle, including 1) Attachment 2) Entry 3) Replication 4) Biosynthesis 5) Assembly and 6...

  10. PPT

    Presentation Transcript. Viral Replication Scott M. Hammer, M.D. Viral Replication: Basic Concepts • Viruses are obligate intracellular parasites • Viruses carry their genome (RNA or DNA) and sometimes functional proteins required for early steps in replication cycle • Viruses depend on host cell machinery to complete replication cycle ...

  11. Viruses

    Viruses are transferred as particles, known as virions. Once the virion enters a host cell, it disassembles and the viral genome begins to interfere with cel...

  12. Ch. 17 Viruses 2018.ppt

    Dengue Virus Life Cycle. -Painful, debilitating -caused by any one of four closely related viruses. -related to the viruses that cause West Nile and yellow fever. Dengue Fever. (pronounced DENgee) Retrovirus. RNA virus that uses reverse transcriptase (RNA → DNA) Newly made viral DNA inserted into chromosome of host.

  13. Coronavirus biology and replication: implications for SARS-CoV-2

    Virus-host interactions in this context are multifaceted and include strategies to hide viral pathogen-associated molecular patterns, such as replication intermediates (dsRNA), that may be ...

  14. Structures and functions of coronavirus replication-transcription

    Viral RNA synthesis occurs within double-membrane vesicles that are part of virus-induced membranous replication organelles (Box 1). The RTCs produce new gRNAs and a set of subgenomic mRNAs (sg ...

  15. PPT

    VIRUS REPLICATION PETER H. RUSSELL, BVSc, PhD, FRCPath, MRCVS Department of Pathology and Infectious Diseases, The Royal Veterinary College, Royal College Street, London NW1 OTU. E-mailWeb site. ObjectivesStudents should be able to: • describe in outline how to cultivate viruses • list the stages of virus replication in a host cell with reference to how these differ between RNA and DNA ...

  16. PPT

    Presentation Transcript. VIRAL REPLICATION Viruses replicate in one of two ways: • Lytic • Lysogenic. Lytic Cycle • For each step use the description and picture to determine a verb that would describe what is going on in each step. Lytic Cycle • 1.Proteins on the surface of the virus bind with proteins on the surface of the host cell ...

  17. PPT

    Lytic cycle - Step 2 • Step 2 - Entry • Once attached to the exterior of the host cell, enzymes begin to break it down and the core (DNA/RNA) is injected into the host cell. Lytic cycle • Step 3 - Replication and Transcription • Using the cell's enzymes and organelles, the virus begins to produce multiple copies of the viral ...

  18. The Hemagglutinin of Influenza A Virus Induces Ferroptosis to

    To clarify the correlation between IAV-induced ferroptosis and the viral replication, we demonstrated that Fer-1 treatment significantly reduced viral titer levels ... In this study, statistical analysis and presentation graphics were carried out by the GraphPad Prism 8.0 software. Results were shown as mean ± SEM from at least three ...

  19. Regulation viral RNA transcription and replication by higher ...

    Coronavirus (CoV) possesses numerous functional cis-acting elements in its positive-strand genomic RNA. Although most of these RNA structures participate in viral replication, the functions of RNA ...

  20. Advancement in the Antigenic Epitopes and Vaccine Adjuvants of ...

    African swine fever virus (ASFV), a highly virulent double-stranded DNA virus, poses a significant threat to global pig farming, with mortality rates in domestic pigs reaching up to 100%. Originating in Kenya in 1921, ASFV has since proliferated to Western Europe, Latin America, Eastern Europe, and most recently China in 2018, resulting in substantial global agricultural losses. Antigenic ...

  21. PPT

    The Lysogenic Cycle 1st step: Attachment = Same 2nd Step: Entry = Same. Lysogenic Cycle 3rd step: Replication • Virus DNA combines with cell DNA, and waits • Provirus = cell DNA + viral DNA • Cell divides by mitosis • Each new cell will contain the provirus. The Lysogenic Cycle 4th Step: Assembly Same, except many new viruses are being ...