frog virus 3
Recently Published Documents


TOTAL DOCUMENTS

224
(FIVE YEARS 28)

H-INDEX

31
(FIVE YEARS 3)

Viruses ◽  
2021 ◽  
Vol 13 (11) ◽  
pp. 2299
Author(s):  
Muhammad Riadul Haque Hossainey ◽  
Amulya Yaparla ◽  
Kelsey A. Hauser ◽  
Tyler E. Moore ◽  
Leon Grayfer

Infections by Frog Virus 3 (FV3) and other ranavirus genus members are significantly contributing to global amphibian decline. The Xenopus laevis frog is an ideal research platform upon which to study the roles of distinct frog leukocyte populations during FV3 infections. Frog macrophages (MΦs) are integrally involved during FV3 infection, as they facilitate viral dissemination and persistence but also participate in immune defense against this pathogen. In turn, MΦ differentiation and functionality depend on the colony-stimulating factor-1 receptor (CSF-1R), which is ligated by CSF-1 and iterleukin-34 (IL-34) cytokines. Our past work indicated that X. laevis CSF-1 and IL-34 give rise to morphologically and functionally distinct frog MΦ subsets, and that these CSF-1- and IL-34-MΦs respectively confer susceptibility and antiviral resistance to FV3. Because FV3 targets the frog kidneys and establishes chronic infections therein, presently we examined the roles of the frog CSF-1- and IL-34-MΦs in seeding and maintaining these chronic kidney infections. Our findings indicate that the frog CSF-1-MΦs result in more prominent kidney FV3 infections, which develop into greater reservoirs of lingering FV3 marked by infiltrating leukocytes, fibrosis, and overall immunosuppressive states. Moreover, the antiviral effects of IL-34-MΦs are short-lived and are lost as FV3 infections progress.


2021 ◽  
Vol 10 (46) ◽  
Author(s):  
Cody R. K. Conrad ◽  
Kuttichantran Subramaniam ◽  
V. Gregory Chinchar ◽  
Thomas B. Waltzek

Two ranavirus isolates were recovered during a wildlife disease investigation in California in 1996. Preliminary testing at the time of sample collection indicated that the two isolates were identical. Phylogenetic analysis of the full genomes of these two isolates revealed that they are a single strain of frog virus 3.


2021 ◽  
Author(s):  
Oliver Lung ◽  
Ayooluwa J. Bolaji ◽  
Michelle Nebroski ◽  
Mat Fisher ◽  
Cody Buchanan ◽  
...  

Abstract Ranaviruses are emerging pathogens that threaten the biodiversity of wild and captive cold-blooded vertebrates. Reports of ranavirus-induced mortality events are increasing and ranavirus disease is reportable to the World Organization for Animal Health. Previous studies have suggested interclass transmission of ranaviruses and Frog virus 3 (FV3)-like viruses are of particular interest. This study presents the whole-genome assembly of a 106 kb FV3-like genome obtained from the liver tissue of a reptile (wild Chelydra serpentina, common snapping turtle) that died of ranavirus disease in Canada. The FV3-like ON turtle/2018 strain shares the highest genome-wide nucleotide identity (99.71%) with the wild-type FV3 virus detected in the USA from a Northern leopard frog and an FV3-like strain identified from a wood frog in 2017 in Alberta, Canada. The novel genome contains all 26 Iridoviridae core genes, 11 FV3-like genes, and 9 unique truncations, three of which are core Iridoviridae ORFs. Additionally, the two most closely related FV3-like strains from amphibians, were compared to a non-FV3-like amphibian infecting and a fish infecting ranavirus species that showed similar codon usage patterns. G/C-ending codons were the preferred codons for all five strains. Investigation of putative recombination events identified four potential recombination events in the FV3-like ON turtle/2018 genome consistent with this FV3-like reptile infecting strain originating from an amphibian infecting FV3-like ranavirus. Altogether, this study provides insights into the genome structure and the differences in the novel FV3-like genome compared to other ranavirus genomes.


2021 ◽  
Author(s):  
Lauren A. Todd ◽  
Barbara A. Katzenback

Frog virus 3 (FV3, genus Ranavirus) causes devastating disease in amphibian populations and is capable of subverting host immune responses. Evidence suggests that virus-encoded microRNAs (v-miRNAs) are implicated in host immunoevasion tactics. Thus, we sought to discover FV3-encoded v-miRNAs and to uncover their putative roles in immunoevasion. Small RNA libraries were generated from FV3-infected Xela DS2, a Xenopus laevis dorsal skin epithelial-like cell line, at 24- and 72-hours post-infection (hpi). We discovered 43 FV3 v-miRNAs and identified that 15 are upregulated at 24 hpi, while 18 are upregulated at 72 hpi. Target prediction analyses revealed that FV3 v-miRNAs target host genes involved in key antiviral signaling pathways, while gene ontology analyses suggest that FV3 v-miRNAs may broadly impact host cell function. This is the first study to experimentally detect mature v-miRNAs produced by FV3. Our findings highlight the possibility that ranaviral v-miRNAs facilitate immunoevasion of frog antiviral responses.


2021 ◽  
Vol 12 ◽  
Author(s):  
Kelsey A. Hauser ◽  
Julia C. Singer ◽  
Muhammad Riadul H. Hossainey ◽  
Tyler E. Moore ◽  
Emily S. Wendel ◽  
...  

The global amphibian declines are compounded by ranavirus infections such as Frog Virus 3 (FV3), and amphibian tadpoles more frequently succumb to these pathogens than adult animals. Amphibian gastrointestinal tracts represent a major route of ranavirus entry, and viral pathogenesis often leads to hemorrhaging and necrosis within this tissue. Alas, the differences between tadpole and adult amphibian immune responses to intestinal ranavirus infections remain poorly defined. As interferon (IFN) cytokine responses represent a cornerstone of vertebrate antiviral immunity, it is pertinent that the tadpoles and adults of the anuran Xenopus laevis frog mount disparate IFN responses to FV3 infections. Presently, we compared the tadpole and adult X. laevis responses to intestinal FV3 infections. Our results indicate that FV3-challenged tadpoles mount more robust intestinal type I and III IFN responses than adult frogs. These tadpole antiviral responses appear to be mediated by myeloid cells, which are recruited into tadpole intestines in response to FV3 infections. Conversely, myeloid cells bearing similar cytology already reside within the intestines of healthy (uninfected) adult frogs, possibly accounting for some of the anti-FV3 resistance of these animals. Further insight into the differences between tadpole and adult frog responses to ranaviral infections is critical to understanding the facets of susceptibility and resistance to these pathogens.


Viruses ◽  
2021 ◽  
Vol 13 (7) ◽  
pp. 1325
Author(s):  
Yun Tian ◽  
Francisco De Jesús Andino ◽  
Collins N. Khwatenge ◽  
Jiuyi Li ◽  
Jacques Robert ◽  
...  

Ranaviruses (Iridoviridae), including Frog Virus 3 (FV3), are large dsDNA viruses that cause devastating infections globally in amphibians, fish, and reptiles, and contribute to catastrophic amphibian declines. FV3’s large genome (~105 kb) contains at least 98 putative open reading frames (ORFs) as annotated in its reference genome. Previous studies have classified these coding genes into temporal classes as immediate early, delayed early, and late viral transcripts based on their sequential expression during FV3 infection. To establish a high-throughput characterization of ranaviral gene expression at the genome scale, we performed a whole transcriptomic analysis (RNA-Seq) using total RNA samples containing both viral and cellular transcripts from FV3-infected Xenopus laevis adult tissues using two FV3 strains, a wild type (FV3-WT) and an ORF64R-deleted recombinant (FV3-∆64R). In samples from the infected intestine, liver, spleen, lung, and especially kidney, an FV3-targeted transcriptomic analysis mapped reads spanning the full-genome coverage at ~10× depth on both positive and negative strands. By contrast, reads were only mapped to partial genomic regions in samples from the infected thymus, skin, and muscle. Extensive analyses validated the expression of almost all of the 98 annotated ORFs and profiled their differential expression in a tissue-, virus-, and temporal class-dependent manner. Further studies identified several putative ORFs that encode hypothetical proteins containing viral mimicking conserved domains found in host interferon (IFN) regulatory factors (IRFs) and IFN receptors. This study provides the first comprehensive genome-wide viral transcriptome profiling during infection and across multiple amphibian host tissues that will serve as an instrumental reference. Our findings imply that Ranaviruses like FV3 have acquired previously unknown molecular mimics, interfering with host IFN signaling during evolution.


Author(s):  
Maxwell P. Bui-Marinos ◽  
Lauren A. Todd ◽  
Marie-Claire D. Wasson ◽  
Brandon E.E. Morningstar ◽  
Barbara A. Katzenback

2021 ◽  
Vol 12 ◽  
Author(s):  
Yun Tian ◽  
Collins N. Khwatenge ◽  
Jiuyi Li ◽  
Francisco De Jesus Andino ◽  
Jacques Robert ◽  
...  

BackgroundFrog Virus 3 (FV3) is a large dsDNA virus belonging to Ranaviruses of family Iridoviridae. Ranaviruses infect cold-blood vertebrates including amphibians, fish and reptiles, and contribute to catastrophic amphibian declines. FV3 has a genome at ~105 kb that contains nearly 100 coding genes and 50 intergenic regions as annotated in its reference genome. Previous studies have mainly focused on coding genes and rarely addressed potential non-coding regulatory role of intergenic regions.ResultsUsing a whole transcriptomic analysis of total RNA samples containing both the viral and cellular transcripts from FV3-infected frog tissues, we detected virus-specific reads mapping in non-coding intergenic regions, in addition to reads from coding genes. Further analyses identified multiple cis-regulatory elements (CREs) in intergenic regions neighboring highly transcribed coding genes. These CREs include not only a virus TATA-Box present in FV3 core promoters as in eukaryotic genes, but also viral mimics of CREs interacting with several transcription factors including CEBPs, CREBs, IRFs, NF-κB, and STATs, which are critical for regulation of cellular immunity and cytokine responses. Our study suggests that intergenic regions immediately upstream of highly expressed FV3 genes have evolved to bind IRFs, NF-κB, and STATs more efficiently. Moreover, we found an enrichment of putative microRNA (miRNA) sequences in more than five intergenic regions of the FV3 genome. Our sequence analysis indicates that a fraction of these viral miRNAs is targeting the 3’-UTR regions of Xenopus genes involved in interferon (IFN)-dependent responses, including particularly those encoding IFN receptor subunits and IFN-regulatory factors (IRFs).ConclusionsUsing the FV3 model, this study provides a first genome-wide analysis of non-coding regulatory mechanisms adopted by ranaviruses to epigenetically regulate both viral and host gene expressions, which have co-evolved to interact especially with the host IFN response.


2021 ◽  
Author(s):  
Maxwell P. Bui-Marinos ◽  
Lauren A. Todd ◽  
Marie-Claire D. Wasson ◽  
Brandon E. E. Morningstar ◽  
Barbara A. Katzenback

Frog virus 3 (FV3) causes mortality in a range of amphibian species. Despite the importance of the skin epithelium as a first line of defence against FV3, the interaction between amphibian skin epithelial cells and FV3 remains largely uncharacterized. Here, we used newly established Xenopus laevis skin epithelial-like cell lines, Xela DS2 and Xela VS2, to study the susceptibility and permissiveness of frog skin epithelial cells to FV3, and the innate immune antiviral and proinflammatory gene regulatory responses of these cells to FV3. Both cell lines are susceptible and permissive to FV3, yet do not exhibit appreciable transcript levels of scavenger receptors recently demonstrated to be used by FV3 for cellular entry. Xela DS2 and Xela VS2 upregulate antiviral and proinflammatory cytokine transcripts in response to poly(I:C) but not to FV3 or UV-inactivated FV3. Poly(I:C) pretreatment limited FV3 replication and FV3-induced cytopathic effects in both cell lines. Thus, Xela DS2 and Xela VS2 can support FV3 propagation, represent in vitro systems to investigate antiviral responses of frog skin epithelial cells, and are novel tools for screening compounds that initiate effective antiviral programs to limit FV3 replication.


Author(s):  
Claudia Maris Ferreira ◽  
Kuttichantran Subramaniam ◽  
Ricardo Luiz Moro de Sousa ◽  
Loiane S. Tavares ◽  
Thaís C. Corrêa ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document