Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS Pathog ; 20(3): e1012060, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38442126

RESUMO

The recent discovery of Hepatitis D (HDV)-like viruses across a wide range of taxa led to the establishment of the Kolmioviridae family. Recent studies suggest that kolmiovirids can be satellites of viruses other than Hepatitis B virus (HBV), challenging the strict HBV/HDV-association dogma. Studying whether kolmiovirids are able to replicate in any animal cell they enter is essential to assess their zoonotic potential. Here, we compared replication of three kolmiovirids: HDV, rodent (RDeV) and snake (SDeV) deltavirus in vitro and in vivo. We show that SDeV has the narrowest and RDeV the broadest host cell range. High resolution imaging of cells persistently replicating these viruses revealed nuclear viral hubs with a peculiar RNA-protein organization. Finally, in vivo hydrodynamic delivery of viral replicons showed that both HDV and RDeV, but not SDeV, efficiently replicate in mouse liver, forming massive nuclear viral hubs. Our comparative analysis lays the foundation for the discovery of specific host factors controlling Kolmioviridae host-shifting.


Assuntos
Hepatite D , Vírus Delta da Hepatite , Camundongos , Animais , Humanos , Roedores , Vírus da Hepatite B/genética , Serpentes , Replicação Viral , RNA Viral/genética
2.
Vet Pathol ; 61(2): 256-268, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37681312

RESUMO

The European hedgehog (Erinaceus europaeus) is a common wildlife species in European countries. Populations are declining due to anthropogenic factors and natural diseases. Verminous pneumonia has been observed as a frequent infectious disease in hedgehogs submitted for diagnostic postmortem examination. This prompted the present in-depth investigation on the lungs of 27 necropsied hedgehogs with confirmed lungworm infections, with or without antiparasitic treatment prior to death. The histological and/or parasitic (fecal samples) examination identified Capillaria aerophila infection in most animals (82%). The parasites were found free in the airway lumen and/or within the airway epithelium, from the larynx to bronchioles. Embedded worms and eggs were associated with epithelial hyperplasia or metaplasia, and long-term inflammation. More than half of the animals (59%) carried Crenosoma striatum, and 41% had a coinfection. C striatum adults were predominantly found free in the lumen of bronchi and bronchioles, and larvae were occasionally seen in granulomas in the pulmonary interstitium, the liver, and the intestine. Independent of the parasite species, a lymphoplasmacytic peribronchitis and, less frequently, interstitial infiltration of eosinophils, neutrophils, and macrophages as well as pneumocyte type II hyperplasia was seen. Interestingly, the extent of pneumonia was not correlated with age, respiratory clinical signs, antiparasitic treatment, or single or coinfection. Verminous pneumonia appeared to be the cause of death in over 25% of the animals, indicating that these parasites not only coexist with hedgehogs but can also be a primary pathogen in this species.


Assuntos
Coinfecção , Pneumonia , Animais , Ouriços/parasitologia , Coinfecção/veterinária , Hiperplasia/veterinária , Pneumonia/parasitologia , Pneumonia/veterinária , Antiparasitários
3.
Viruses ; 15(12)2023 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-38140554

RESUMO

Reptarenaviruses cause Boid Inclusion Body Disease (BIBD), a fatal disease of boid snakes with an economic and ecological impact, as it affects both captive and wild constrictor snakes. The clinical picture of BIBD is highly variable but often only limited. Intracytoplasmic inclusion bodies (IB), which develop in most cell types including blood cells, are the pathognomonic hallmark of BIBD; their detection represents the diagnostic gold standard of the disease. However, IBs are not consistently present in clinically healthy reptarenavirus carriers, which can, if undetected, lead to and maintain the spread of the disease within and between snake populations. Sensitive viral detection tools are required for screening and control purposes; however, the genetic diversity of reptarenaviruses hampers the reverse transcription (RT) PCR-based diagnostics. Here, we describe a multiplex RT-PCR approach for the molecular diagnosis of reptarenavirus infection in blood samples. The method allows the detection of a wide range of reptarenaviruses with the detection limit reaching 40 copies per microliter of blood. Using 245 blood samples with a reference RT-PCR result, we show that the technique performs as well as the segment-specific RT-PCRs in our earlier studies. It can identify virus carriers and serve to limit reptarenavirus spreading in captive snake collections.


Assuntos
Infecções por Arenaviridae , Arenaviridae , Boidae , Animais , Arenaviridae/genética , Transcrição Reversa , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Boidae/genética
4.
Microbiol Spectr ; 11(3): e0506522, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-37212675

RESUMO

Reptarenaviruses cause boid inclusion body disease (BIBD), a fatal disease particularly impacting captive boa constrictor collections. The development of cytoplasmic inclusion bodies (IBs) comprising reptarenavirus nucleoprotein (NP) in many cell types of affected snakes is characteristic of BIBD. However, snakes can harbor reptarenaviruses without showing IBs, hence representing carriers and a potential source of transmission. The RNA genome of reptarenaviruses comprises a small (S) and a large (L) segment, and the snakes with BIBD commonly carry a swarm of reptarenavirus segments. To design sensitive and reliable tools for the diagnosis of reptarenavirus infection in snake colonies, we used metatranscriptomics to determine the reptarenavirus segments present in a large boa constrictor breeding colony. The analysis identified one reptarenavirus S segment and three L segments in the colony. The sequence data served to design real-time reverse transcription-PCR (RT-PCR) targeting the found S segment. This allowed us to identify all infected animals and to quantify the S segment RNA levels, which we found to correlate with the presence of IBs. We further found a positive correlation between the number of L segments and the S segment RNA level, which could suggest that L segment excess also contributes to the IB formation. Information on cohousing of the snakes showed a clear association of reptarenavirus infection with cohousing in general and cohousing with infected animals. Information on breeding and offspring confirmed that vertical transmission occurred. Furthermore, our data suggest that some animals might be able to clear the infection or at least exhibit transient or intermittent viremia. IMPORTANCE Boid inclusion body disease (BIBD) is caused by reptarenavirus infection, and while reptarenavirus nucleoprotein is the main component of the inclusion bodies (IBs) characteristic of BIBD, not all reptarenavirus-infected snakes demonstrate IBs in their cells. Identification of infected individuals is critical for controlling the spread of the disease; however, the genetic divergence of reptarenaviruses complicates reverse transcription-PCR (RT-PCR)-based diagnostics. Here, we tested a next-generation-sequencing-based approach to establish a tailored "colony-specific" set of diagnostic tools for the detection of reptarenavirus small (S) and large (L) genome segments. With this approach, we could demonstrate that an S-segment-specific RT-PCR is highly effective in identifying the infected individuals. We further found the S segment RNA level to positively correlate with the presence of IBs and the number of L segments, which could direct future studies to identify the BIBD pathogenetic mechanisms.


Assuntos
Arenaviridae , Boidae , Corpos de Inclusão , Animais , Arenaviridae/genética , Boidae/genética , Nucleoproteínas/genética , RNA Viral/genética
5.
Anim Genet ; 54(3): 412-415, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36786090

RESUMO

Primary ciliary dyskinesia (PCD) represents a group of diseases characterized by impaired movement of cilia and subsequent health problems in diverse organ systems, notably the respiratory tract. Almost 50 candidate genes for PCD are known in humans. In this study, we investigated an Australian Shepherd dog with a history of recurrent respiratory infections and nasal discharge. A transmission electron microscopy investigation led to the diagnosis of PCD with central pair defect, in which the normal 9:2 arrangement of respiratory cilia was altered and reduced to a 9:0 arrangement. Whole genome sequencing data from the affected dog was obtained and searched for variants in PCD candidate genes that were not present in 918 control genomes from different breeds. This revealed a homozygous single base pair exchange at a splice site of STK36, XM_038585732.1:c.2868-1G>A. The mutant allele was absent from 281 additionally genotyped Australian Shepherd dogs. RT-PCR confirmed aberrant splicing in the affected dog with the skipping of exon 20 and the insertion of a cryptic exon, which is predicted to lead to a premature stop codon and truncation of 36% of the STK36 wild-type open reading frame, XP_038441660.1:(p.Met957Profs*11). STK36 variants were previously reported to cause PCD in humans and mice. The knowledge from other species together with the absence of the mutant allele in more than 1000 control dogs suggests STK36:c.2868-1G>A as the most likely candidate variant for PCD in the investigated case.


Assuntos
Transtornos da Motilidade Ciliar , Doenças do Cão , Animais , Cães , Transtornos da Motilidade Ciliar/genética , Transtornos da Motilidade Ciliar/veterinária , Genótipo , Homozigoto , Proteínas Serina-Treonina Quinases/genética
6.
J Comp Pathol ; 200: 12-17, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36610082

RESUMO

We present a case that illustrates the complex contexts in which forensic veterinary pathology examinations may be carried out. A wild muskrat (Ondatra zibethicus) had died after a putative bite attack from a domestic dog. Bite attacks by privately owned dogs on wild animals in Switzerland violate the Swiss civil (and/or hunting) laws, and dog owners are generally punished with a monetary fine; hence, this case appeared to be straightforward. However, the results of the subsequent post-mortem examination of the muskrat not only confirmed the presence of injuries related to the bite attack, but also detected alveolar echinococcosis (ie, infestation with Echinococcus multilocularis). Therefore, as an intermediate host of the parasite, the muskrat could have contributed to further spread of a severe helminthic zoonosis had it not been killed by the dog. It was probably an easy prey for the dog as it probably had been weakened by the disease. Furthermore, muskrats are considered as pests and invasive species, and are non-indigenous huntable game in Switzerland and other European countries in which programmes for the prevention of their further spread and endangerment of native wildlife are established. The role of the forensic veterinary pathologist in such a complex scenario is to adopt an unbiased approach and establish the facts, which in this case was to determine the cause of death and suspected perpetrator, identify any concomitant and/or underlying diseases and consider potential animal welfare issues.


Assuntos
Doenças do Cão , Equinococose , Doenças dos Roedores , Animais , Cães , Humanos , Arvicolinae/parasitologia , Prova Pericial , Patologistas , Equinococose/veterinária , Equinococose/parasitologia , Animais Selvagens , Doenças do Cão/parasitologia
7.
Microbiol Spectr ; 10(5): e0170522, 2022 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-36094085

RESUMO

Reptarenaviruses cause boid inclusion body disease (BIBD), a potentially fatal disease, occurring in captive constrictor snakes boas and pythons worldwide. Classical BIBD, characterized by the formation of pathognomonic cytoplasmic inclusion bodies (IBs), occurs mainly in boas, whereas in pythons, for example, reptarenavirus infection most often manifests as central nervous system signs with limited IB formation. The natural hosts of reptarenaviruses are unknown, although free-ranging/wild constrictor snakes are among the suspects. Here, we report BIBD with reptarenavirus infection in indigenous captive and wild boid snakes in Costa Rica using histology, immunohistology, transmission electron microscopy, and next-generation sequencing (NGS). The snakes studied represented diagnostic postmortem cases of captive and wild-caught snakes since 1989. The results from NGS on archival paraffin blocks confirm that reptarenaviruses were already present in wild boa constrictors in Costa Rica in the 1980s. Continuous sequences that were de novo assembled from the low-quality RNA obtained from paraffin-embedded tissue allowed the identification of a distinct pair of reptarenavirus S and L segments in all studied animals; in most cases, reference assembly could recover almost complete segments. Sampling of three prospective cases in 2018 allowed an examination of fresh blood or tissues and resulted in the identification of additional reptarenavirus segments and hartmanivirus coinfection. Our results show that BIBD is not only a disease of captive snakes but also occurs in indigenous wild constrictor snakes in Costa Rica, suggesting boa constrictors to play a role in natural reptarenavirus circulation. IMPORTANCE The literature describes cases of boid inclusion body disease (BIBD) in captive snakes since the 1970s, and in the 2010s, others and ourselves identified reptarenaviruses as the causative agent. BIBD affects captive snakes globally, but the origin and the natural host of reptarenaviruses remain unknown. In this report, we show BIBD and reptarenavirus infections in two native Costa Rican constrictor snake species, and by studying archival samples, we show that both the viruses and the disease have been present in free-ranging/wild snakes in Costa Rica at least since the 1980s. The diagnosis of BIBD in wild boa constrictors suggests that this species plays a role in the circulation of reptarenaviruses. Additional sample collection and analysis would help to clarify this role further and the possibility of, e.g., vector transmission from an arthropod host.


Assuntos
Infecções por Arenaviridae , Arenaviridae , Boidae , Doenças Transmissíveis , Animais , Boidae/genética , Infecções por Arenaviridae/veterinária , Parafina , Arenaviridae/genética , Corpos de Inclusão , RNA
8.
Microbiol Spectr ; 10(4): e0158522, 2022 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-35862992

RESUMO

Mammarenaviruses establish a persistent infection in their rodent and bat hosts, and the evidence suggests that reptarenaviruses and hartmaniviruses found in captive snakes act similarly. In snakes, reptarenaviruses cause boid inclusion body disease (BIBD), which is often associated with secondary infections. Snakes with BIBD usually carry more than a single pair of reptarenavirus S and L segments and occasionally demonstrate hartmanivirus coinfection. Here, we reported the generation of cell lines persistently infected with a single or two reptarenavirus(es) and a cell line with persistent reptarenavirus-hartmanivirus coinfection. By RT-PCR we demonstrated that the amount of viral RNA within the persistently infected cells remains at levels similar to those observed following initial infection. Using antibodies against the glycoproteins (GPs) and nucleoprotein (NP) of reptarenaviruses, we studied the levels of viral protein in cells passaged 10 times after the original inoculation and observed that the expression of GPs declines dramatically during persistent infection, unlike the expression of NP. Immunofluorescence (IF) staining served to demonstrate differences in the distribution of NP within the persistently infected compared to freshly infected cells. IF staining of cells inoculated with the viruses secreted from the persistently infected cell lines produced similar NP staining compared to cells infected with a traditionally passaged virus, suggesting that the altered NP expression pattern of persistently infected cells does not relate to changes in the virus. The cell cultures described herein can serve as tools for studying the coinfection and superinfection interplay between reptarenaviruses and studying the BIBD pathogenesis mechanisms. IMPORTANCE Mammarenaviruses cause a persistent infection in their natural rodent and bat hosts. Reptarenaviruses cause boid inclusion body disease (BIBD) in constrictor snakes, but it is unclear whether snakes are the natural host of these viruses. In this study, we showed that reptarenaviruses established a persistent infection in cultured Boa constrictor cells and that the persistently infected cells continued to produce infectious virus. Our results showed that persistent infection results from subsequent passaging of cells inoculated with a single reptarenavirus, two reptarenaviruses, or even when inoculating the cells with reptarenavirus and hartmanivirus (another arenavirus genus). The results further suggested that coinfection would not result in overt competition between the different reptarenaviruses, thus helping to explain the frequent reptarenavirus coinfections in snakes with BIBD. The established cell culture models of persistent infection could help to elucidate the role of coinfection and superinfection and potential immunosuppression as the pathogenic mechanisms behind BIBD.


Assuntos
Arenaviridae , Boidae , Quirópteros , Coinfecção , Superinfecção , Animais , Arenaviridae/genética , Linhagem Celular
9.
Anim Genet ; 53(3): 416-421, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35233794

RESUMO

Inherited forms of cataract are a heterogeneous group of eye disorders known in livestock species. Clinicopathological analysis of a single case of impaired vision in a newborn Original Braunvieh calf revealed nuclear cataract. Whole-genome sequencing of the parent-offspring trio revealed a de novo mutation of ADAMTSL4 in this case. The heterozygous p.Arg776His missense variant affects a conserved residue of the ADAMTSL4 gene that encodes a secreted glycoprotein expressed in the lens throughout embryonic development. In humans, ADAMTSL4 genetic variants cause recessively inherited forms of subluxation of the lens. Given that ADAMTSL4 is a functional candidate gene for inherited disorders of the lens, we suggest that heterozygosity for the identified missense variant may have caused the congenital cataract in the affected calf. Cattle populations should be monitored for unexplained cataract cases, with subsequent DNA sequencing a hypothesized pathogenic effect of heterozygous ADAMTSL4 variants could be confirmed.


Assuntos
Catarata , Doenças dos Bovinos , Animais , Catarata/genética , Catarata/veterinária , Bovinos/genética , Doenças dos Bovinos/genética , Mutação de Sentido Incorreto , Linhagem , Sequenciamento Completo do Genoma
10.
Mol Ther Methods Clin Dev ; 24: 268-279, 2022 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-35211639

RESUMO

Hepatic gene therapy by delivering non-integrating therapeutic vectors in newborns remains challenging due to the risk of dilution and loss of efficacy in the growing liver. Previously we reported on hepatocyte transfection in piglets by intraportal injection of naked DNA vectors. Here, we established delivery of naked DNA vectors to target periportal hepatocytes in weaned pigs by hydrodynamic retrograde intrabiliary injection (HRII). The surgical procedure involved laparotomy and transient isolation of the liver. For vector delivery, a catheter was placed within the common bile duct by enterotomy. Under optimal conditions, no histological abnormalities were observed in liver tissue upon pressurized injections. The transfection of hepatocytes in all tested liver samples was observed with vectors expressing luciferase from a liver-specific promoter. However, vector copy number and luciferase expression were low compared to hydrodynamic intraportal injection. A 10-fold higher number of vector genomes and luciferase expression was observed in pigs using a non-integrating naked DNA vector with the potential for replication. In summary, the HRII application was less efficient (i.e., lower luciferase activity and vector copy numbers) than the intraportal delivery method but was significantly less distressful for the piglets and has the potential for injection (or re-injection) of vector DNA by endoscopic retrograde cholangiopancreatography.

11.
Viruses ; 14(1)2022 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-35062311

RESUMO

Human hepatitis D virus (HDV) depends on hepatitis B virus co-infection and its glycoproteins for infectious particle formation. HDV was the sole known deltavirus for decades and believed to be a human-only pathogen. However, since 2018, several groups reported finding HDV-like agents from various hosts but without co-infecting hepadnaviruses. In vitro systems enabling helper virus-independent replication are key for studying the newly discovered deltaviruses. Others and we have successfully used constructs containing multimers of the deltavirus genome for the replication of various deltaviruses via transfection in cell culture. Here, we report the establishment of deltavirus infectious clones with 1.2× genome inserts bearing two copies of the genomic and antigenomic ribozymes. We used Swiss snake colony virus 1 as the model to compare the ability of the previously reported "2× genome" and the "1.2× genome" infectious clones to initiate replication in cell culture. Using immunofluorescence, qRT-PCR, immuno- and northern blotting, we found the 2× and 1.2× genome clones to similarly initiate deltavirus replication in vitro and both induced a persistent infection of snake cells. The 1.2× genome constructs enable easier introduction of modifications required for studying deltavirus replication and cellular interactions.


Assuntos
Boidae/virologia , Células Clonais , Coinfecção/genética , Vírus Delta da Hepatite/genética , Replicação Viral , Animais , Boidae/genética , Genoma Viral , Vírus Auxiliares/genética , Hepadnaviridae/genética , Hepatite B/genética , Vírus da Hepatite B/genética , Hepatite D/virologia , RNA Catalítico , RNA Viral/genética , Transfecção
12.
Vet Pathol ; 59(2): 269-283, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34955067

RESUMO

The sequence of pathological events in feline hypertrophic cardiomyopathy (fHCM) is still largely unknown, although we know that fHCM is characterized by interstitial remodeling in a macrophage-driven pro-inflammatory environment and that myocardial ischemia might contribute to its progression. This study aimed to gain further insights into the structural changes associated with interstitial remodeling in fHCM with special focus on the myocardial microvasculature and the phenotype of the interstitial cells. Twenty-eight hearts (16 hearts with fHCM and 12 without cardiac disease) were evaluated in the current study, with immunohistochemistry, RNA-in situ hybridization, and transmission electron microscopy. Morphometrical evaluations revealed a statistically significant lower microvascular density in fHCM. This was associated with structural alterations in capillaries that go along with a widening of the interstitium due to the accumulation of edema fluid, collagen fibers, and mononuclear cells that also proliferated locally. The interstitial cells were mainly of fibroblastic or vascular phenotype, with a substantial contribution of predominantly resident macrophages. A large proportion expressed CD34 mRNA, which suggests a progenitor cell potential. Our results indicate that microvascular alterations are key events in the pathogenesis of fHCM and that myocardial interstitial cell populations with CD34+ phenotype play a role in the pathogenesis of the disease.


Assuntos
Cardiomiopatias , Cardiomiopatia Hipertrófica , Doenças do Gato , Animais , Cardiomiopatias/patologia , Cardiomiopatias/veterinária , Cardiomiopatia Hipertrófica/patologia , Cardiomiopatia Hipertrófica/veterinária , Doenças do Gato/patologia , Gatos , Densidade Microvascular , Miocárdio/patologia , Miócitos Cardíacos/patologia
13.
Sci Rep ; 11(1): 21048, 2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34702948

RESUMO

Viruses need cells for their replication and, therefore, ways to hijack cellular functions. Mitochondria play fundamental roles within the cell in metabolism, immunity and regulation of homeostasis due to which some viruses aim to alter mitochondrial functions. Herein we show that the nucleoprotein (NP) of arenaviruses enters the mitochondria of infected cells, affecting the mitochondrial morphology. Reptarenaviruses cause boid inclusion body disease (BIBD) that is characterized, especially in boas, by the formation of cytoplasmic inclusion bodies (IBs) comprising reptarenavirus NP within the infected cells. We initiated this study after observing electron-dense material reminiscent of IBs within the mitochondria of reptarenavirus infected boid cell cultures in an ultrastructural study. We employed immuno-electron microscopy to confirm that the mitochondrial inclusions indeed contain reptarenavirus NP. Mutations to a putative N-terminal mitochondrial targeting signal (MTS), identified via software predictions in both mamm- and reptarenavirus NPs, did not affect the mitochondrial localization of NP, suggesting that it occurs independently of MTS. In support of MTS-independent translocation, we did not detect cleavage of the putative MTSs of arenavirus NPs in reptilian or mammalian cells. Furthermore, in vitro translated NPs could not enter isolated mitochondria, suggesting that the translocation requires cellular factors or conditions. Our findings suggest that MTS-independent mitochondrial translocation of NP is a shared feature among arenaviruses. We speculate that by targeting the mitochondria arenaviruses aim to alter mitochondrial metabolism and homeostasis or affect the cellular defense.


Assuntos
Arenaviridae/metabolismo , Boidae/virologia , Corpos de Inclusão Viral/metabolismo , Mitocôndrias/metabolismo , Mitocôndrias/virologia , Nucleoproteínas/metabolismo , Animais , Arenaviridae/classificação , Arenaviridae/genética , Chlorocebus aethiops , Corpos de Inclusão Viral/genética , Mitocôndrias/genética , Nucleoproteínas/genética , Células Vero
14.
Toxicol Pathol ; 49(6): 1174-1192, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34060347

RESUMO

Antisense oligonucleotides (ASOs) are chemically modified nucleic acids with therapeutic potential, some of which have been approved for marketing. We performed a study in rats to investigate mechanisms of toxicity after administration of 3 tool locked nucleic acid (LNA)-containing ASOs with differing established safety profiles. Four male rats per group were dosed once, 3, or 6 times subcutaneously, with 7 days between dosing, and sacrificed 3 days after the last dose. These ASOs were either unconjugated (naked) or conjugated with N-acetylgalactosamine for hepatocyte-targeted delivery. The main readouts were in-life monitoring, clinical and anatomic pathology, exposure assessment and metabolite identification in liver and kidney by liquid chromatography coupled to tandem mass spectrometry, ASO detection in liver and kidney by immunohistochemistry, in situ hybridization, immune electron microscopy, and matrix-assisted laser desorption/ionization mass spectrometry imaging. The highly toxic compounds showed the greatest amount of metabolites and a low degree of tissue accumulation. This study reveals different patterns of cell death associated with toxicity in liver (apoptosis and necrosis) and kidney (necrosis only) and provides new ultrastructural insights on the tissue accumulation of ASOs. We observed that the immunostimulatory properties of ASOs can be either primary from sequence-dependent properties or secondary to cell necrosis.


Assuntos
Oligonucleotídeos Antissenso , Oligonucleotídeos , Acetilgalactosamina , Animais , Masculino , Oligonucleotídeos Antissenso/toxicidade , Ratos , Distribuição Tecidual
15.
Vet Anim Sci ; 10: 100150, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33089008

RESUMO

This article presents radiologic examinations of a deeply sedated Boa constrictor with boid inclusion body disease (BIBD) as an adjunction to the subsequent necropsy. This method is known as virtopsy. The Boa constrictor in the present case was gravid. Computed tomography (CT) allowed for the detailed depiction of a fetal skeleton at the rear end of the adult snake. Furthermore, tiny gas formation was detected inside the cranium of the fetus, which was deemed a radiologic sign for decomposition. Magnetic resonance imaging (MRI) delineated the soft tissue at high resolution. This article illustrates the use of CT and MRI for the examination of a gravid Boa constrictor before necropsy and demonstrates the detection of "normal" postmortem findings leading to the confirmation of fetal death in situ.

16.
Microsc Microanal ; 26(4): 731-740, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32627730

RESUMO

Concurrent three-dimensional imaging of the renal vascular and tubular systems on the whole-kidney scale with capillary level resolution is labor-intensive and technically difficult. Approaches based on vascular corrosion casting and X-ray micro computed tomography (µCT), for example, suffer from vascular filling artifacts and necessitate imaging with an additional modality to acquire tubules. In this work, we report on a new sample preparation, image acquisition, and quantification protocol for simultaneous vascular and tubular µCT imaging of whole, uncorroded mouse kidneys. The protocol consists of vascular perfusion with the water-soluble, aldehyde-fixable, polymeric X-ray contrast agent XlinCA, followed by laboratory-source µCT imaging and structural analysis using the freely available Fiji/ImageJ software. We achieved consistent filling of the entire capillary bed and staining of the tubules in the cortex and outer medulla. After imaging at isotropic voxel sizes of 3.3 and 4.4 µm, we segmented vascular and tubular systems and quantified luminal volumes, surface areas, diffusion distances, and vessel path lengths. This protocol permits the analysis of vascular and tubular parameters with higher reliability than vascular corrosion casting, less labor than serial sectioning and leaves tissue intact for subsequent histological examination with light and electron microscopy.


Assuntos
Túbulos Renais/irrigação sanguínea , Túbulos Renais/diagnóstico por imagem , Modelos Anatômicos , Microtomografia por Raio-X/métodos , Animais , Meios de Contraste/farmacologia , Feminino , Interpretação de Imagem Assistida por Computador/métodos , Imageamento Tridimensional/métodos , Rim/irrigação sanguínea , Rim/diagnóstico por imagem , Testes de Função Renal , Camundongos , Camundongos Endogâmicos C57BL
17.
J Virol ; 94(18)2020 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-32641481

RESUMO

In recent years, nidoviruses have emerged as important respiratory pathogens of reptiles, affecting captive python populations. In pythons, nidovirus (recently reclassified as serpentovirus) infection induces an inflammation of the upper respiratory and alimentary tract which can develop into a severe, often fatal proliferative pneumonia. We observed pyogranulomatous and fibrinonecrotic lesions in organ systems other than the respiratory tract during full postmortem examinations on 30 serpentovirus reverse transcription-PCR (RT-PCR)-positive pythons of varying species originating from Switzerland and Spain. The observations prompted us to study whether this not yet reported wider distribution of lesions is associated with previously unknown serpentoviruses or changes in the serpentovirus genome. RT-PCR and inoculation of Morelia viridis cell cultures served to recruit the cases and obtain virus isolates. Immunohistochemistry and immunofluorescence staining against serpentovirus nucleoprotein demonstrated that the virus infects not only a broad spectrum of epithelia (respiratory and alimentary epithelium, hepatocytes, renal tubules, pancreatic ducts, etc.), but also intravascular monocytes, intralesional macrophages, and endothelial cells. With next-generation sequencing we obtained a full-length genome for a novel serpentovirus species circulating in Switzerland. Analysis of viral genomes recovered from pythons showing serpentovirus infection-associated respiratory or systemic disease did not reveal sequence association to phenotypes; however, functional studies with different strains are needed to confirm this observation. The results indicate that serpentoviruses have a broad cell and tissue tropism, further suggesting that the course of infection could vary and involve lesions in a broad spectrum of tissues and organ systems as a consequence of monocyte-mediated viral systemic spread.IMPORTANCE During the last years, python nidoviruses (now reclassified as serpentoviruses) have become a primary cause of fatal disease in pythons. Serpentoviruses represent a threat to captive snake collections, as they spread rapidly and can be associated with high morbidity and mortality. Our study indicates that, different from previous evidence, the viruses do not only affect the respiratory tract, but can spread in the entire body with blood monocytes, have a broad spectrum of target cells, and can induce a variety of lesions. Nidovirales is an order of animal and human viruses that comprises important zoonotic pathogens such as Middle East respiratory syndrome coronavirus (MERS-CoV), severe acute respiratory syndrome coronavirus (SARS-CoV), and SARS-CoV-2. Serpentoviruses belong to the same order as the above-mentioned human viruses and show similar characteristics (rapid spread, respiratory and gastrointestinal tropism, etc.). The present study confirms the relevance of natural animal diseases to better understand the complexity of viruses of the order Nidovirales.


Assuntos
Infecções por Nidovirales/virologia , Nidovirales/fisiologia , Infecções Respiratórias/virologia , Doenças dos Animais/diagnóstico , Doenças dos Animais/virologia , Animais , Biópsia , Boidae/virologia , Suscetibilidade a Doenças , Humanos , Imuno-Histoquímica , Nidovirales/isolamento & purificação , Infecções por Nidovirales/diagnóstico , Especificidade de Órgãos , Fenótipo , Filogenia , Recombinação Genética , Infecções Respiratórias/diagnóstico , Tropismo Viral , Eliminação de Partículas Virais
18.
Am J Physiol Lung Cell Mol Physiol ; 319(3): L562-L575, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32579393

RESUMO

The poorly understood tolerance toward high tidal volume (VT) ventilation observed in critically ill children and age-equivalent animal models may be explained by surfactant homeostasis. The aim of our prospective animal study was to test whether high VT with adequate positive end-expiratory pressure (PEEP) is associated with surfactant de novo synthesis and secretion, leading to improved lung function, and whether extreme mechanical ventilation affects intracellular lamellar body formation and exocytosis. Rats (14 days old) were allocated to five groups: nonventilated controls, PEEP 5 cmH2O with VT of 8, 16, and 24 mL/kg, and PEEP 1 cmH2O with VT 24 mL/kg. Following 6 h of ventilation, lung function, surfactant proteins and phospholipids, and lamellar bodies were assessed by forced oscillation technique, quantitative real-time polymerase chain reaction, mass spectrometry, immunohistochemistry, and transmission electron microscopy. High VT (24 mL/kg) with PEEP of 5 cmH2O improved respiratory system mechanics and was not associated with lung injury, elevated surfactant protein expression, or surfactant phospholipid content. Extreme ventilation with VT 24 mL/kg and PEEP 1 cmH2O produced a mild inflammatory response and correlated with higher surfactant phospholipid concentrations in bronchoalveolar lavage fluid without affecting lamellar body count and morphology. Elevated phospholipid concentrations in the potentially most injurious strategy (VT 24 mL/kg, PEEP 1 cmH2O) need further evaluation and might reflect accumulation of biophysically inactive small aggregates. In conclusion, our data confirm the resilience of infant rats toward high VT-induced lung injury and challenge the relevance of surfactant synthesis, storage, and secretion as protective factors.


Assuntos
Lesão Pulmonar/metabolismo , Lesão Pulmonar/fisiopatologia , Surfactantes Pulmonares/metabolismo , Volume de Ventilação Pulmonar/fisiologia , Animais , Líquido da Lavagem Broncoalveolar/citologia , Pulmão/metabolismo , Pulmão/fisiopatologia , Ratos , Mecânica Respiratória/fisiologia , Tensoativos/metabolismo
19.
Viruses ; 12(4)2020 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-32252443

RESUMO

Reptarenaviruses cause Boid Inclusion Body Disease (BIBD), and co-infections by several reptarenaviruses are common in affected snakes. Reptarenaviruses have only been found in captive snakes, and their reservoir hosts remain unknown. In affected animals, reptarenaviruses appear to replicate in most cell types, but their complete host range, as well as tissue and cell tropism are unknown. As with other enveloped viruses, the glycoproteins (GPs) present on the virion's surface mediate reptarenavirus cell entry, and therefore, the GPs play a critical role in the virus cell and tissue tropism. Herein, we employed single cycle replication, GP deficient, recombinant vesicular stomatitis virus (VSV) expressing the enhanced green fluorescent protein (scrVSV∆G-eGFP) pseudotyped with different reptarenavirus GPs to study the virus cell tropism. We found that scrVSV∆G-eGFPs pseudotyped with reptarenavirus GPs readily entered mammalian cell lines, and some mammalian cell lines exhibited higher, compared to snake cell lines, susceptibility to reptarenavirus GP-mediated infection. Mammarenavirus GPs used as controls also mediated efficient entry into several snake cell lines. Our results confirm an important role of the virus surface GP in reptarenavirus cell tropism and that mamma-and reptarenaviruses exhibit high cross-species transmission potential.


Assuntos
Arenaviridae/fisiologia , Vesiculovirus/fisiologia , Proteínas do Envelope Viral , Tropismo Viral , Células A549 , Animais , Arenaviridae/genética , Linhagem Celular , Chlorocebus aethiops , Proteínas de Fluorescência Verde/genética , Células HEK293 , Humanos , Serpentes , Células Vero , Vesiculovirus/genética , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo
20.
J Virol ; 94(11)2020 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-32238580

RESUMO

Boid inclusion body disease (BIBD) is a transmissible viral disease of captive snakes that causes severe losses in snake collections worldwide. It is caused by reptarenavirus infection, which can persist over several years without overt signs but is generally associated with the eventual death of the affected snakes. Thus far, reports have confirmed the existence of reptarenaviruses in captive snakes in North America, Europe, Asia, and Australia, but there is no evidence that it also occurs in wild snakes. BIBD affects boa species within the subfamily Boinae and pythons in the family Pythonidae, the habitats of which do not naturally overlap. Here, we studied Brazilian captive snakes with BIBD using a metatranscriptomic approach, and we report the identification of novel reptarenaviruses, hartmaniviruses, and a new species in the family Chuviridae The reptarenavirus L segments identified are divergent enough to represent six novel species, while we found only a single novel reptarenavirus S segment. Until now, hartmaniviruses had been identified only in European captive boas with BIBD, and the present results increase the number of known hartmaniviruses from four to six. The newly identified chuvirus showed 38.4%, 40.9%, and 48.1% amino acid identity to the nucleoprotein, glycoprotein, and RNA-dependent RNA polymerase, respectively, of its closest relative, Guangdong red-banded snake chuvirus-like virus. Although we cannot rule out the possibility that the found viruses originated from imported snakes, the results suggest that the viruses could circulate in indigenous snake populations.IMPORTANCE Boid inclusion body disease (BIBD), caused by reptarenavirus infection, affects captive snake populations worldwide, but the reservoir hosts of reptarenaviruses remain unknown. Here, we report the identification of novel reptarenaviruses, hartmaniviruses, and a chuvirus in captive Brazilian boas with BIBD. Three of the four snakes studied showed coinfection with all three viruses, and one of the snakes harbored three novel reptarenavirus L segments and one novel S segment. The samples originated from collections with Brazilian indigenous snakes only, which could indicate that these viruses circulate in wild snakes. The findings could further indicate that boid snakes are the natural reservoir of reptarena- and hartmaniviruses commonly found in captive snakes. The snakes infected with the novel chuvirus all suffered from BIBD; it is therefore not possible to comment on its potential pathogenicity and contribution to the observed changes in the present case material.


Assuntos
Arenaviridae , Boidae/virologia , Proteínas Virais , Animais , Arenaviridae/classificação , Arenaviridae/genética , Arenaviridae/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...