Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
1.
J Virol ; 96(11): e0063422, 2022 06 08.
Article in English | MEDLINE | ID: mdl-35575553

ABSTRACT

The global amphibian declines are compounded by infections with members of the Ranavirus genus such as Frog Virus 3 (FV3). Premetamorphic anuran amphibians are believed to be significantly more susceptible to FV3 while this pathogen targets the kidneys of both pre- and postmetamorphic animals. Paradoxically, FV3-challenged Xenopus laevis tadpoles exhibit lower kidney viral loads than adult frogs. Presently, we demonstrate that X. laevis tadpoles are intrinsically more resistant to FV3 kidney infections than cohort-matched metamorphic and postmetamorphic froglets and that this resistance appears to be epigenetically conferred by endogenous retroviruses (ERVs). Using a X. laevis kidney-derived cell line, we show that enhancing ERV gene expression activates cellular double-stranded RNA-sensing pathways, resulting in elevated mRNA levels of antiviral interferon (IFN) cytokines and thus greater anti-FV3 protection. Finally, our results indicate that large esterase-positive myeloid-lineage cells, rather than renal cells, are responsible for the elevated ERV/IFN axis seen in the tadpole kidneys. This conclusion is supported by our observation that CRISPR-Cas9 ablation of colony-stimulating factor-3 results in abolished homing of these myeloid cells to tadpole kidneys, concurrent with significantly abolished tadpole kidney expression of both ERVs and IFNs. We believe that the manuscript marks an important step forward in understanding the mechanisms controlling amphibian antiviral defenses and thus susceptibility and resistance to pathogens like FV3. IMPORTANCE Global amphibian biodiversity is being challenged by pathogens like the Frog Virus 3 (FV3) ranavirus, underlining the need to gain a greater understanding of amphibian antiviral defenses. While it was previously believed that anuran (frog/toad) amphibian tadpoles are more susceptible to FV3, we demonstrated that tadpoles are in fact more resistant to this virus than metamorphic and postmetamorphic froglets. We showed that this resistance is conferred by large myeloid cells within the tadpole kidneys (central FV3 target), which possess an elevated expression of endogenous retroviruses (ERVs). In turn, these ERVs activate cellular double-stranded RNA-sensing pathways, resulting in a greater expression of antiviral interferon cytokines, thereby offering the observed anti-FV3 protection.


Subject(s)
DNA Virus Infections , Endogenous Retroviruses , Ranavirus , Xenopus laevis , Animals , Cell Line , DNA Virus Infections/immunology , DNA Virus Infections/veterinary , Disease Resistance , Endogenous Retroviruses/immunology , Interferons/immunology , Kidney/virology , Larva/immunology , Larva/virology , RNA, Double-Stranded , Ranavirus/pathogenicity , Xenopus laevis/virology
2.
Front Immunol ; 12: 737403, 2021.
Article in English | MEDLINE | ID: mdl-34489981

ABSTRACT

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.


Subject(s)
Amphibian Proteins/metabolism , DNA Virus Infections/virology , Interferons/metabolism , Intestines/virology , Myeloid Cells/virology , Ranavirus/pathogenicity , Xenopus laevis/virology , Age Factors , Animals , DNA Virus Infections/immunology , DNA Virus Infections/metabolism , Disease Susceptibility , Female , Host-Pathogen Interactions , Intestines/embryology , Intestines/immunology , Larva/immunology , Larva/metabolism , Larva/virology , Male , Myeloid Cells/immunology , Myeloid Cells/metabolism , Ranavirus/immunology , Viral Load , Xenopus laevis/embryology , Xenopus laevis/immunology , Xenopus laevis/metabolism
3.
Sci Rep ; 9(1): 4453, 2019 03 14.
Article in English | MEDLINE | ID: mdl-30872735

ABSTRACT

Ranavirus is the second most common infectious cause of amphibian mortality. These viruses affect caudates, an order in which information regarding Ranavirus pathogenesis is scarce. In the Netherlands, two strains (CMTV-NL I and III) were suspected to possess distinct pathogenicity based on field data. To investigate susceptibility and disease progression in urodeles and determine differences in pathogenicity between strains, 45 adult smooth newts (Lissotriton vulgaris) were challenged via bath exposure with these ranaviruses and their detection in organs and feces followed over time by PCR, immunohistochemistry and in situ hybridization. Ranavirus was first detected at 3 days post infection (p.i.) in the oral cavity and upper respiratory mucosa. At 6 days p.i, virus was found in connective tissues and vasculature of the gastrointestinal tract. Finally, from 9 days p.i onwards there was widespread Ranavirus disease in various organs including skin, kidneys and gonads. Higher pathogenicity of the CMTV-NL I strain was confirmed by higher correlation coefficient of experimental group and mortality of challenged animals. Ranavirus-exposed smooth newts shed virus in feces intermittently and infection was seen in the absence of lesions or clinical signs, indicating that this species can harbor subclinical infections and potentially serve as disease reservoirs.


Subject(s)
DNA Virus Infections/veterinary , Host-Pathogen Interactions/physiology , Ranavirus/pathogenicity , Salamandridae/virology , Animals , Feces/virology , Mouth/virology , Netherlands , Polymerase Chain Reaction , Ranavirus/physiology , Virus Replication , Virus Shedding
4.
Viruses ; 11(1)2019 01 03.
Article in English | MEDLINE | ID: mdl-30609806

ABSTRACT

Ranaviruses are the second deadliest pathogens for amphibian populations throughout the world. Despite their wide distribution in America, these viruses have never been reported in Mexico, the country with the fifth highest amphibian diversity in the world. This paper is the first to address an outbreak of ranavirus in captive American bullfrogs (Lithobates catesbeianus) from Sinaloa, Mexico. The farm experienced high mortality in an undetermined number of juveniles and sub-adult bullfrogs. Affected animals displayed clinical signs and gross lesions such as lethargy, edema, skin ulcers, and hemorrhages consistent with ranavirus infection. The main microscopic lesions included mild renal tubular necrosis and moderate congestion in several organs. Immunohistochemical analyses revealed scant infected hepatocytes and renal tubular epithelial cells. Phylogenetic analysis of five partial ranavirus genes showed that the causative agent clustered within the Frog virus 3 clade. Risk assessment with the Pandora⁺ protocol demonstrated a high risk for the pathogen to affect amphibians from neighboring regions (overall Pandora risk score: 0.619). Given the risk of American bullfrogs escaping and spreading the disease to wild amphibians, efforts should focus on implementing effective containment strategies and surveillance programs for ranavirus at facilities undertaking intensive farming of amphibians.


Subject(s)
DNA Virus Infections/epidemiology , Disease Outbreaks/veterinary , Rana catesbeiana/virology , Ranavirus/pathogenicity , Animals , Animals, Wild/virology , Aquaculture , DNA Virus Infections/mortality , Edema/epidemiology , Edema/virology , Mexico/epidemiology , Phylogeny , Ranavirus/genetics , Ranavirus/isolation & purification , Risk Assessment , Skin/pathology , Skin/virology , Viral Proteins/genetics
5.
J Wildl Dis ; 54(4): 716-725, 2018 10.
Article in English | MEDLINE | ID: mdl-29878878

ABSTRACT

The pathogenicity of frog virus 3 (FV3)-like ranavirus varies in adult chelonian species at different environmental temperatures, but differences in pathogenicity at different temperatures has yet to be determined in juveniles. Our objective was to determine the susceptibility to FV3-like ranavirus in four species of juvenile chelonians: red-eared sliders (RES; Trachemys scripta elegans), Mississippi map turtles ( Graptemys pseudogeographica kohnii), false map turtles (FMT; Graptemys pseudogeographica), and eastern river cooters ( Pseudemys concinna concinna) at two environmental temperatures. Two simultaneous trials ( n=8 treatment and n=4 controls of each species) were conducted in separate temperature-controlled rooms with animals maintained at 22 C or 27 C. All of the inoculated animals of each species at each temperature died, but no mortality was observed in control animals. Median survival times varied between 8 d and 11 d, based on species and temperature, with RES in the 27 C trial surviving the shortest time and the FMT in the 22 C trial surviving the longest. Combining all species, turtles in the 27 C trial survived for fewer days than those housed at 22 C, despite all turtles in both trials having similar viral copies detected in postmortem tissues. Lesions in inoculated turtles resembled those noted in natural and experimental FV3-like ranavirus infections and included vasculitis, thrombosis, hemorrhage in multiple organs, renal tubular necrosis, and hepatic necrosis. Myositis was not present in any juvenile, infected turtles in this study. This study confirmed that juvenile chelonians have a high susceptibility to ranaviral disease.


Subject(s)
DNA Virus Infections/veterinary , Ranavirus/pathogenicity , Temperature , Turtles , Animals , DNA Virus Infections/mortality , DNA Virus Infections/transmission , DNA Virus Infections/virology
6.
Bull Math Biol ; 80(7): 1937-1961, 2018 07.
Article in English | MEDLINE | ID: mdl-29785520

ABSTRACT

For epidemic models, it is shown that fatal infectious diseases cannot drive the host population into extinction if the incidence function is upper density-dependent. This finding holds even if a latency period is included and the time from infection to disease-induced death has an arbitrary length distribution. However, if the incidence function is also lower density-dependent, very infectious diseases can lead to a drastic decline of the host population. Further, the final population size after an epidemic outbreak can possibly be substantially affected by the infection-age distribution of the initial infectives if the life expectations of infected individuals are an unbounded function of infection age (time since infection). This is the case for lognormal distributions, which fit data from infection experiments involving tiger salamander larvae and ranavirus better than gamma distributions and Weibull distributions.


Subject(s)
Communicable Diseases/epidemiology , Communicable Diseases/mortality , Epidemics/statistics & numerical data , Models, Biological , Ambystoma/virology , Animals , DNA Virus Infections/veterinary , Disease Outbreaks/statistics & numerical data , Host Microbial Interactions , Host-Pathogen Interactions , Humans , Incidence , Likelihood Functions , Mathematical Concepts , Population Density , Ranavirus/pathogenicity , Survival Analysis , Time Factors
7.
Dev Comp Immunol ; 85: 108-114, 2018 08.
Article in English | MEDLINE | ID: mdl-29626489

ABSTRACT

Andrias davidianus is a large and economically important amphibian in China. Ranavirus infection causes serious losses in A. davidianus farming industry. MicroRNA mediated host-pathogen interactions are important in antiviral defense. In this study, five small-RNA libraries from ranavirus infected and non-infected A. davidianus spleens were sequenced using high throughput sequencing. The miRNA expression pattern, potential functions, and target genes were investigated. In total, 1356 known and 431 novel miRNAs were discovered. GO and KEGG analysis revealed that certain miRNA target genes are associated with apoptotic, signal pathway, and immune response categories. Analysis identified 82 downregulated and 9 upregulated differentially expressed miRNAs, whose putative target genes are involved in pattern-recognition receptor signaling pathways and immune response. These findings suggested miRNAs play key roles in A. davidianus's response to ranavirus and could provide a reference for further miRNA functional identification, leading to novel approaches to improve A. davidianus ranavirus resistance.


Subject(s)
MicroRNAs/genetics , Ranavirus/pathogenicity , Urodela/genetics , Urodela/virology , Animals , Apoptosis/genetics , China , Down-Regulation/genetics , High-Throughput Nucleotide Sequencing/methods , Host-Pathogen Interactions/genetics , Immunity/genetics , Membrane Proteins/genetics , Serine Endopeptidases/genetics , Up-Regulation/genetics
8.
Emerg Microbes Infect ; 7(1): 56, 2018 Apr 04.
Article in English | MEDLINE | ID: mdl-29615625

ABSTRACT

Ranaviruses are pathogenic viruses for poikilothermic vertebrates worldwide. The identification of a common midwife toad virus (CMTV) associated with massive die-offs in water frogs (Pelophylax spp.) in the Netherlands has increased awareness for emerging viruses in amphibians in the country. Complete genome sequencing of 13 ranavirus isolates collected from ten different sites in the period 2011-2016 revealed three CMTV groups present in distinct geographical areas in the Netherlands. Phylogenetic analysis showed that emerging viruses from the northern part of the Netherlands belonged to CMTV-NL group I. Group II and III viruses were derived from the animals located in the center-east and south of the country, and shared a more recent common ancestor to CMTV-amphibian associated ranaviruses reported in China, Italy, Denmark, and Switzerland. Field monitoring revealed differences in water frog host abundance at sites where distinct ranavirus groups occur; with ranavirus-associated deaths, host counts decreasing progressively, and few juveniles found in the north where CMTV-NL group I occurs but not in the south with CMTV-NL group III. Investigation of tandem repeats of coding genes gave no conclusive information about phylo-geographical clustering, while genetic analysis of the genomes revealed truncations in 17 genes across CMTV-NL groups II and III compared to group I. Further studies are needed to elucidate the contribution of these genes as well as environmental variables to explain the observed differences in host abundance.


Subject(s)
DNA Virus Infections/veterinary , Ranavirus/genetics , Ranidae/virology , Animals , DNA Virus Infections/virology , Genotype , Netherlands , Phylogeny , Ranavirus/classification , Ranavirus/isolation & purification , Ranavirus/pathogenicity , Virulence
9.
Mol Ecol ; 27(6): 1413-1427, 2018 03.
Article in English | MEDLINE | ID: mdl-29420865

ABSTRACT

Ranaviruses are responsible for a lethal, emerging infectious disease in amphibians and threaten their populations throughout the world. Despite this, little is known about how amphibian populations respond to ranaviral infection. In the United Kingdom, ranaviruses impact the common frog (Rana temporaria). Extensive public engagement in the study of ranaviruses in the UK has led to the formation of a unique system of field sites containing frog populations of known ranaviral disease history. Within this unique natural field system, we used RNA sequencing (RNA-Seq) to compare the gene expression profiles of R. temporaria populations with a history of ranaviral disease and those without. We have applied a RNA read-filtering protocol that incorporates Bloom filters, previously used in clinical settings, to limit the potential for contamination that comes with the use of RNA-Seq in nonlaboratory systems. We have identified a suite of 407 transcripts that are differentially expressed between populations of different ranaviral disease history. This suite contains genes with functions related to immunity, development, protein transport and olfactory reception among others. A large proportion of potential noncoding RNA transcripts present in our differentially expressed set provide first evidence of a possible role for long noncoding RNA (lncRNA) in amphibian response to viruses. Our read-filtering approach also removed significantly more bacterial reads from libraries generated from positive disease history populations. Subsequent analysis revealed these bacterial read sets to represent distinct communities of bacterial species, which is suggestive of an interaction between ranavirus and the host microbiome in the wild.


Subject(s)
Animals, Wild/genetics , DNA Virus Infections/genetics , Rana temporaria/virology , Ranavirus/pathogenicity , Animals , Animals, Wild/microbiology , DNA Virus Infections/virology , Microbiota/genetics , Rana temporaria/genetics , Sequence Analysis, RNA , United Kingdom
10.
Microb Pathog ; 112: 269-273, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28987623

ABSTRACT

Ranavirus has become a noticeable threat to both farmed and natural populations of fish and amphibians. Herein, we reported that 3 strains of novel viruses, designated as ScRIV-GM-20150902, CmRIV-XT-20150917 and ScRIV-ZS-20151201, were isolated from diseased Chinese perch and snakehead fish in China. Efficient propagation of these isolates were determined in Chinese perch brain (CPB) cell line by the means of cytopathic effect observation, PCR amplification and electron microscopy observation. And their viral titers in CPB cells reached 108.13 TCID50 ml-1, 107.71 TCID50 ml-1 and 107.94 TCID50 ml-1, respectively. While the challenge experiment results showed that 3 isolates resulted in 100% mortality of Chinese perch after virus infection. Electron microscopy analysis showed that two kinds of viral inclusion bodies (intracytoplasmic and intranuclear inclusion body) were observed in infected CPB cells. Sequence alignment and phylogenetic analysis of major capsid protein gene sequences of isolates revealed that these isolates belonged to the species Santee-Cooper Ranavirus.


Subject(s)
DNA Virus Infections/veterinary , Fish Diseases/virology , Fishes/virology , Perches/virology , Ranavirus/classification , Ranavirus/isolation & purification , Ranavirus/pathogenicity , Animals , Ascites/pathology , Ascites/virology , Base Sequence , Brain/pathology , Brain/virology , Capsid Proteins/genetics , Cell Line , China , DNA Virus Infections/pathology , DNA Virus Infections/virology , DNA, Viral , Inclusion Bodies, Viral , Mesentery/pathology , Mesentery/virology , Microscopy, Electron, Transmission , Phylogeny , Ranavirus/genetics , Sequence Alignment , Virulence
11.
Virology ; 511: 290-299, 2017 11.
Article in English | MEDLINE | ID: mdl-28919326

ABSTRACT

Ambystoma tigrinum virus (ATV) (family Iridoviridae, genus Ranavirus) was isolated from diseased tiger salamanders (Ambystoma tigrinum stebbinsi) from the San Rafael Valley in southern Arizona, USA in 1996. Genomic sequencing of ATV, as well as other members of the genus, identified an open reading frame that has homology to the eukaryotic translation initiation factor, eIF2α (ATV eIF2α homologue, vIF2αH). Therefore, we asked if the ATV vIF2αH could also inhibit PKR. To test this hypothesis, the ATV vIF2αH was cloned into vaccinia virus (VACV) in place of the well-characterized VACV PKR inhibitor, E3L. Recombinant VACV expressing ATV vIF2αH partially rescued deletion of the VACV E3L gene. Rescue coincided with rapid degradation of PKR in infected cells. These data suggest that the salamander virus, ATV, contains a novel gene that may counteract host defenses, and this gene product may be involved in the presentation of disease caused by this environmentally important pathogen.


Subject(s)
Protein Kinase Inhibitors/metabolism , Ranavirus/pathogenicity , Recombinant Proteins/metabolism , Viral Proteins/metabolism , Virulence Factors/metabolism , eIF-2 Kinase/antagonists & inhibitors , Ambystoma/virology , Animals , Arizona , Gene Expression , Genetic Vectors , Host-Pathogen Interactions , Immune Evasion , Ranavirus/genetics , Recombinant Proteins/genetics , Vaccinia virus/genetics , Viral Proteins/genetics , Virulence Factors/genetics
12.
Virology ; 511: 272-279, 2017 11.
Article in English | MEDLINE | ID: mdl-28860047

ABSTRACT

Ranaviruses are pathogens of ectothermic vertebrates, including amphibians. We reviewed patterns of host range and virulence of ranaviruses in the context of virus genotype and postulate that patterns reflect significant variation in the historical and current host range of three groups of Ranavirus: FV3-like, CMTV-like and ATV-like ranaviruses. Our synthesis supports previous hypotheses about host range and jumps: FV3s are amphibian specialists, while ATVs are predominantly fish specialists that switched once to caudate amphibians. The most recent common ancestor of CMTV-like ranaviruses and FV3-like forms appears to have infected amphibians but CMTV-like ranaviruses may circulate in both amphibian and fish communities independently. While these hypotheses are speculative, we hope that ongoing efforts to describe ranavirus genetics, increased surveillance of host species and targeted experimental assays of susceptibility to infection and/or disease will facilitate better tests of the importance of hypothetical evolutionary drivers of ranavirus virulence and host range.


Subject(s)
Amphibians/virology , Fishes/virology , Host Specificity , Ranavirus/isolation & purification , Ranavirus/physiology , Animals , Biological Evolution , Ranavirus/pathogenicity , Virulence
13.
Virology ; 511: 309-319, 2017 11.
Article in English | MEDLINE | ID: mdl-28625407

ABSTRACT

We first review fundamental insights into anti-ranavirus immunity learned with the Xenopus laevis/ranavirus FV3 model that are generally applicable to ectothermic vertebrates. We then further investigate FV3 genes involved in immune evasion. Focusing on FV3 knockout (KO) mutants defective for a putative viral caspase activation and recruitment domain-containing (CARD)-like protein (Δ64R-FV3), a ß-hydroxysteroid dehydrogenase homolog (Δ52L-FV3), and an immediate-early18kDa protein (FV3-Δ18K), we assessed the involvement of these viral genes in replication, dissemination and interaction with peritoneal macrophages in tadpole and adult frogs. Our results substantiate the role of 64R and 52L as critical immune evasion genes, promoting persistence and dissemination in the host by counteracting type III IFN in tadpoles and type I IFN in adult frogs. Comparably, the substantial accumulation of genome copy numbers and exacerbation of type I and III IFN gene expression responses but deficient release of infectious virus suggests that 18K is a viral regulatory gene.


Subject(s)
Host-Pathogen Interactions , Immune Evasion , Ranavirus/pathogenicity , Xenopus laevis/immunology , Xenopus laevis/virology , Animals , Gene Knockout Techniques , Interferons/antagonists & inhibitors , Macrophages/virology , Viral Proteins/genetics , Viral Proteins/metabolism , Virulence Factors/genetics , Virulence Factors/metabolism , Virus Replication
14.
J Anim Ecol ; 86(4): 921-931, 2017 07.
Article in English | MEDLINE | ID: mdl-28317105

ABSTRACT

Coinfections are increasingly recognized as important drivers of disease dynamics. Consequently, greater emphasis has been placed on integrating principles from community ecology with disease ecology to understand within-host interactions among parasites. Using larval amphibians and two amphibian parasites (ranaviruses and the trematode Echinoparyphium sp.), we examined the influence of coinfection on disease outcomes. Our first objective was to examine how priority effects (the timing and sequence of parasite exposure) influence infection and disease outcomes in the laboratory. We found that interactions between the parasites were asymmetric; prior infection with Echinoparyphium reduced ranaviral loads by 9% but there was no reciprocal effect of prior ranavirus infection on Echinoparyphium load. Additionally, survival rates of hosts (larval gray treefrogs; Hyla versicolor) infected with Echinoparyphium 10 days prior to virus exposure were 25% greater compared to hosts only exposed to virus. Our second objective was to determine whether these patterns were generalizable to multiple amphibian species under more natural conditions. We conducted a semi-natural mesocosm experiment consisting of four larval amphibian hosts [gray treefrogs, American toads (Anaxyrus americanus), leopard frogs (Lithobates pipiens) and spring peepers (Pseudacris crucifer)] to examine how prior Echinoparyphium infection influenced ranavirus transmission within the community, using ranavirus-infected larval wood frogs (Lithobates sylvaticus) as source of ranavirus. Consistent with the laboratory experiment, we found that prior Echinoparyphium infection reduced ranaviral loads by 19 to 28% in three of the four species. Collectively, these results suggest that macroparasite infection can reduce microparasite replication rates across multiple amphibian species, possibly through cross-reactive immunity. Although the immunological mechanisms driving this outcome are in need of further study, trematode infections appear to benefit hosts that are exposed to ranaviruses. Additionally, these results suggest that consideration of priority effects and timing of exposure are vital for understanding parasite interactions within hosts and disease outcomes.


Subject(s)
Anura , Coinfection , Ranavirus/pathogenicity , Trematoda/virology , Animals , Anura/microbiology , Anura/virology , Bufonidae
15.
Sci Rep ; 7: 43260, 2017 02 27.
Article in English | MEDLINE | ID: mdl-28240267

ABSTRACT

Emerging diseases have been increasingly associated with population declines, with co-infections exhibiting many types of interactions. The chytrid fungus (Batrachochytrium dendrobatidis) and ranaviruses have extraordinarily broad host ranges, however co-infection dynamics have been largely overlooked. We investigated the pattern of co-occurrence of these two pathogens in an amphibian assemblage in Serra da Estrela (Portugal). The detection of chytridiomycosis in Portugal was linked to population declines of midwife-toads (Alytes obstetricans). The asynchronous and subsequent emergence of a second pathogen - ranavirus - caused episodes of lethal ranavirosis. Chytrid effects were limited to high altitudes and a single host, while ranavirus was highly pathogenic across multiple hosts, life-stages and altitudinal range. This new strain (Portuguese newt and toad ranavirus - member of the CMTV clade) caused annual mass die-offs, similar in host range and rapidity of declines to other locations in Iberia affected by CMTV-like ranaviruses. However, ranavirus was not always associated with disease, mortality and declines, contrasting with previous reports on Iberian CMTV-like ranavirosis. We found little evidence that pre-existing chytrid emergence was associated with ranavirus and the emergence of ranavirosis. Despite the lack of cumulative or amplified effects, ranavirus drove declines of host assemblages and changed host community composition and structure, posing a grave threat to all amphibian populations.


Subject(s)
Chytridiomycota/pathogenicity , DNA Virus Infections/veterinary , Life Cycle Stages , Mycoses/veterinary , Ranavirus/pathogenicity , Altitude , Animals , Anura/growth & development , Anura/microbiology , Anura/virology , Chytridiomycota/physiology , Coinfection , DNA Virus Infections/epidemiology , DNA Virus Infections/mortality , DNA Virus Infections/virology , Mycoses/epidemiology , Mycoses/microbiology , Mycoses/mortality , Portugal/epidemiology , Prevalence , Ranavirus/physiology , Salamandridae/growth & development , Salamandridae/microbiology , Salamandridae/virology , Survival Analysis
16.
Vet Pathol ; 54(3): 531-548, 2017 05.
Article in English | MEDLINE | ID: mdl-28060677

ABSTRACT

Wood frogs ( Rana sylvatica) are highly susceptible to infection with Frog virus 3 (FV3, Ranavirus, Iridoviridae), a cause of mass mortality in wild populations. To elucidate the pathogenesis of FV3 infection in wood frogs, 40 wild-caught adults were acclimated to captivity, inoculated orally with a fatal dose of 104.43 pfu/frog, and euthanized at 0.25, 0.5, 1, 2, 4, 9, and 14 days postinfection (dpi). Mild lesions occurred sporadically in the skin (petechiae) and bone marrow (necrosis) during the first 2 dpi. Severe lesions occurred 1 to 2 weeks postinfection and consisted of necrosis of medullary and extramedullary hematopoietic tissue, lymphoid tissue in spleen and throughout the body, and epithelium of skin, mucosae, and renal tubules. Viral DNA was first detected (polymerase chain reaction) in liver at 4 dpi; by dpi 9 and 14, all viscera tested (liver, kidney, and spleen), skin, and feces were positive. Immunohistochemistry (IHC) first detected viral antigen in small areas devoid of histologic lesions in the oral mucosa, lung, and colon at 4 dpi; by 9 and 14 dpi, IHC labeling of viral antigen associated with necrosis was found in multiple tissues. Based on IHC staining intensity and lesion severity, the skin, oral, and gastrointestinal epithelium and renal tubular epithelium were important sites of viral replication and shedding, suggesting that direct contact (skin) and fecal-oral contamination are effective routes of transmission and that skin tissue, oral, and cloacal swabs may be appropriate antemortem diagnostic samples in late stages of disease (>1 week postinfection) but poor samples to detect infection in clinically healthy frogs.


Subject(s)
DNA Virus Infections/veterinary , Ranavirus , Ranidae/virology , Animals , Animals, Wild/virology , DNA Virus Infections/pathology , DNA Virus Infections/virology , Male , Ranavirus/pathogenicity , Ranidae/genetics , Reverse Transcriptase Polymerase Chain Reaction/veterinary
17.
Chemosphere ; 170: 169-175, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27988452

ABSTRACT

Water pollutants associated with agriculture may contribute to the increased prevalence of infectious diseases caused by ranaviruses. We have established the amphibian Xenopus laevis and the ranavirus Frog Virus 3 (FV3) as a reliable experimental platform for evaluating the effects of common waterborne pollutants, such as the insecticide carbaryl. Following 3 weeks of exposure to 10 ppb carbaryl, X. laevis tadpoles exhibited a marked increase in mortality and accelerated development. Exposure at lower concentrations (0.1 and 1.0 ppb) was not toxic, but it impaired tadpole innate antiviral immune responses, as evidenced by significantly decreased TNF-α, IL-1ß, IFN-I, and IFN-III gene expression. The defect in IFN-I and IL-1ß gene expression levels persisted after metamorphosis in froglets, whereas only IFN-I gene expression in response to FV3 was attenuated when carbaryl exposure was performed at the adult stage. These findings suggest that the agriculture-associated carbaryl exposure at low but ecologically-relevant concentrations has the potential to induce long term alterations in host-pathogen interactions and antiviral immunity.


Subject(s)
Carbaryl/toxicity , DNA Virus Infections/immunology , Immunity, Innate/drug effects , Larva/drug effects , Ranavirus/pathogenicity , Water Pollutants, Chemical/toxicity , Animals , DNA Virus Infections/genetics , DNA Virus Infections/virology , Gene Expression/drug effects , Gene Expression Profiling , Immunity, Innate/immunology , Larva/growth & development , Larva/immunology , Metamorphosis, Biological/drug effects , Metamorphosis, Biological/immunology , Time Factors , Tumor Necrosis Factor-alpha/genetics , Xenopus laevis
18.
Ecohealth ; 13(4): 775-783, 2016 12.
Article in English | MEDLINE | ID: mdl-27682604

ABSTRACT

Amphibians face an extinction crisis with no precedence. Two emerging infectious diseases, ranaviral disease caused by viruses within the genus Ranavirus and chytridiomycosis due to Batrachochytrium dendrobatidis (Bd), have been linked with amphibian mass mortalities and population declines in many regions of the globe. The African clawed frog (Xenopus laevis) has been indicated as a vector for the spread of these pathogens. Since the 1970s, this species has been invasive in central Chile. We collected X. laevis and dead native amphibians in Chile between 2011 and 2013. We conducted post-mortem examinations and molecular tests for Ranavirus and Bd. Eight of 187 individuals (4.3 %) tested positive for Ranavirus: seven X. laevis and a giant Chilean frog (Calyptocephallela gayi). All positive cases were from the original area of X. laevis invasion. Bd was found to be more prevalent (14.4 %) and widespread than Ranavirus, and all X. laevis Bd-positive animals presented low to moderate levels of infection. Sequencing of a partial Ranavirus gene revealed 100 % sequence identity with Frog Virus 3. This is the first report of Ranavirus in Chile, and these preliminary results are consistent with a role for X. laevis as an infection reservoir for both Ranavirus and Bd.


Subject(s)
Xenopus laevis/virology , Amphibians , Animals , Anura , Chile , Chytridiomycota/pathogenicity , Ranavirus/pathogenicity , Xenopus laevis/microbiology
19.
Viruses ; 8(7)2016 07 06.
Article in English | MEDLINE | ID: mdl-27399758

ABSTRACT

Ranaviruses (Iridoviridae) are large DNA viruses that are causing emerging infectious diseases at an alarming rate in both wild and captive cold blood vertebrate species all over the world. Although the general biology of these viruses that presents some similarities with poxvirus is characterized, many aspects of their replication cycles, host cell interactions and evolution still remain largely unclear, especially in vivo. Over several years, strategies to generate site-specific ranavirus recombinant, either expressing fluorescent reporter genes or deficient for particular viral genes, have been developed. We review here these strategies, the main ranavirus recombinants characterized and their usefulness for in vitro and in vivo studies.


Subject(s)
DNA Virus Infections/veterinary , Host-Pathogen Interactions , Molecular Biology/methods , Ranavirus/pathogenicity , Virology/methods , Animals , DNA Virus Infections/pathology , DNA Virus Infections/virology , Ranavirus/genetics , Reverse Genetics , Vertebrates
20.
PLoS One ; 11(6): e0157473, 2016.
Article in English | MEDLINE | ID: mdl-27315226

ABSTRACT

In the four years following the first detection of ranavirus (genus Ranavirus, family Iridoviridae) infection in Dutch wildlife in 2010, amphibian mortality events were investigated nationwide to detect, characterize and map ranaviruses in amphibians over time, and to establish the affected host species and the clinico-pathological presentation of the disease in these hosts. The ultimate goal was to obtain more insight into ranavirus disease emergence and ecological risk. In total 155 dead amphibians from 52 sites were submitted between 2011 and 2014, and examined using histopathology, immunohistochemistry, virus isolation and molecular genetic characterization. Ranavirus-associated amphibian mortality events occurred at 18 sites (35%), initially only in proximity of the 2010 index site. Specimens belonging to approximately half of the native amphibian species were infected, including the threatened Pelobates fuscus (spadefoot toad). Clustered massive outbreaks involving dead adult specimens and ranavirus genomic identity indicated that one common midwife toad virus (CMTV)-like ranavirus strain is emerging in provinces in the north of the Netherlands. Modelling based on the spatiotemporal pattern of spread showed a high probability that this emerging virus will continue to be detected at new sites (the discrete reproductive power of this outbreak is 0.35). Phylogenetically distinct CMTV-like ranaviruses were found in the south of the Netherlands more recently. In addition to showing that CMTV-like ranaviruses threaten wild amphibian populations not only in Spain but also in the Netherlands, the current spread and risk of establishment reiterate that understanding the underlying causes of CMTV-like ranavirus emergence requires international attention.


Subject(s)
Amphibians/virology , DNA Virus Infections/genetics , Phylogeny , Ranavirus/pathogenicity , Animals , Animals, Wild , DNA Virus Infections/epidemiology , Epidemics , Necrosis/virology , Netherlands , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...