Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
Viruses ; 16(5)2024 05 01.
Article in English | MEDLINE | ID: mdl-38793603

ABSTRACT

Bovine viral diarrhea virus (BVDV) infections cause USD 1.5-2 billion in losses annually. Maternal BVDV after 150 days of gestation causes transient fetal infection (TI) in which the fetal immune response clears the virus. The impact of fetal TI BVDV infections on postnatal growth and white blood cell (WBC) methylome as an index of epigenetic modifications was examined by inoculating pregnant heifers with noncytopathic type 2 BVDV or media (sham-inoculated controls) on Day 175 of gestation to generate TI (n = 11) and control heifer calves (n = 12). Fetal infection in TI calves was confirmed by virus-neutralizing antibody titers at birth and control calves were seronegative. Both control and TI calves were negative for BVDV RNA in WBCs by RT-PCR. The mean weight of the TI calves was less than that of the controls (p < 0.05). DNA methyl seq analysis of WBC DNA demonstrated 2349 differentially methylated cytosines (p ≤ 0.05) including 1277 hypomethylated cytosines, 1072 hypermethylated cytosines, 84 differentially methylated regions based on CpGs in promoters, and 89 DMRs in islands of TI WBC DNA compared to controls. Fetal BVDV infection during late gestation resulted in epigenomic modifications predicted to affect fetal development and immune pathways, suggesting potential consequences for postnatal growth and health of TI cattle.


Subject(s)
Bovine Virus Diarrhea-Mucosal Disease , DNA Methylation , Diarrhea Viruses, Bovine Viral , Epigenesis, Genetic , Leukocytes , Animals , Cattle , Bovine Virus Diarrhea-Mucosal Disease/virology , Bovine Virus Diarrhea-Mucosal Disease/genetics , Female , Pregnancy , Leukocytes/virology , Diarrhea Viruses, Bovine Viral/genetics , Antibodies, Viral/blood , Fetal Diseases/virology , Fetal Diseases/veterinary , Fetal Diseases/genetics , Diarrhea Virus 2, Bovine Viral/genetics , Fetus/virology
2.
Curr Microbiol ; 79(12): 356, 2022 Oct 16.
Article in English | MEDLINE | ID: mdl-36245036

ABSTRACT

Bovine viral diarrhea virus (BVDV) is an important animal pathogen and has a negative economic impact on cattle industries worldwide. In this study, the BVDV strain named BJ175170 was detected, isolated, and identified from cattle in Beijing, China, during herd screening by BVDV antigen-ELISA and indirect immunofluorescence assay (IFA). To investigate its genomic features, the characteristic 5'UTR region of the isolates were sequenced and BLAST analyzed. BVDV BJ175170 belongs to the BVDV-1c subtype, which differs from the Beijing prevalent BVDV strains. The BVDV particles were further observed by transmission electron microscopy (TEM). To evaluate the virulence of the BVDV BJ175170, the BVDV seronegative rabbits were intraperitoneally inoculated with the virus suspension. Blood samples were analyzed for changes in leukocyte number and antibody titer, and tissue samples were taken for histopathology analysis. These data confirmed again that rabbits could act as the reservoir of BVDV, which poses a small but non-zero risk of re-infection for BVDV-free cattle herds. To our knowledge, this is the first report of pathological changes in rabbits after exposure to BVDV-1c subtype, which could act as experimental reference. Meanwhile, the results of this study indicate that rabbits could act as a potential model for studying the mechanism of BVDV in vivo.


Subject(s)
Bovine Virus Diarrhea-Mucosal Disease , Diarrhea Virus 1, Bovine Viral , Diarrhea Viruses, Bovine Viral , 5' Untranslated Regions , Animals , Bovine Virus Diarrhea-Mucosal Disease/genetics , Bovine Virus Diarrhea-Mucosal Disease/prevention & control , Cattle , Diarrhea , Diarrhea Virus 1, Bovine Viral/genetics , Diarrhea Viruses, Bovine Viral/genetics , Phylogeny , Rabbits , Virulence
3.
J Virol ; 96(17): e0111322, 2022 09 14.
Article in English | MEDLINE | ID: mdl-35993735

ABSTRACT

Bovine viral diarrhea virus (BVDV) is the causative agent of the bovine viral diarrhea-mucosal disease, which is a leading cause of economic losses in the cattle industry worldwide. To date, many underlying mechanisms involved in BVDV-host interactions remain unclear, especially the functions of long noncoding RNAs (lncRNAs). In our previous study, the lncRNA expression profiles of BVDV-infected Madin-Darby bovine kidney (MDBK) cells were obtained by RNA-seq, and a significantly downregulated lncRNA IALNCR targeting MAPK8/JNK1 (a key regulatory factor of apoptosis) was identified through the lncRNA-mRNA coexpression network analysis. In this study, the function of IALNCR in regulating apoptosis to affect BVDV replication was further explored. Our results showed that BVDV infection-induced downregulation of the lncRNA IALNCR in the host cells could suppress the expression of MAPK8/JNK1 at both the mRNA and protein levels, thereby indirectly promoting the activation of caspase-3, leading to cell-autonomous apoptosis to antagonize BVDV replication. This was further confirmed by the small interfering RNA (siRNA)-mediated knockdown of the lncRNA IALNCR. However, the overexpression of the lncRNA IALNCR inhibited apoptosis and promoted BVDV replication. In conclusion, our findings demonstrated that the lncRNA IALNCR plays an important role in regulating host antiviral innate immunity against BVDV infection. IMPORTANCE Bovine viral diarrhea-mucosal disease caused by BVDV is an important viral disease in cattle, causing severe economic losses to the cattle industry worldwide. The molecular mechanisms of BVDV-host interactions are complex. To date, most studies focused only on how BVDV escapes host innate immunity. By contrast, how the host cell regulates anti-BVDV innate immune responses is rarely reported. In this study, a significantly downregulated lncRNA, with a potential function of inhibiting apoptosis (inhibiting apoptosis long noncoding RNA, IALNCR), was obtained from the lncRNA expression profiles of BVDV-infected cells and was experimentally evaluated for its function in regulating apoptosis and affecting BVDV replication. We demonstrated that downregulation of BVDV infection-induced lncRNA IALNCR displayed antiviral function by positively regulating the MAPK8/JNK1 pathway to promote cell apoptosis. Our data provided evidence that host lncRNAs regulate the innate immune response to BVDV infection.


Subject(s)
Apoptosis , Bovine Virus Diarrhea-Mucosal Disease , Diarrhea Viruses, Bovine Viral , Down-Regulation , Mitogen-Activated Protein Kinase 8 , RNA, Long Noncoding , Virus Replication , Animals , Bovine Virus Diarrhea-Mucosal Disease/genetics , Bovine Virus Diarrhea-Mucosal Disease/immunology , Bovine Virus Diarrhea-Mucosal Disease/virology , Cattle , Cell Line , Diarrhea Viruses, Bovine Viral/growth & development , Diarrhea Viruses, Bovine Viral/immunology , Immunity, Innate , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 8/metabolism , RNA, Long Noncoding/genetics , RNA, Messenger/genetics
4.
Virol J ; 19(1): 134, 2022 08 19.
Article in English | MEDLINE | ID: mdl-35986298

ABSTRACT

BACKGROUND: Bovine viral diarrhea virus 1 (BVDV-1) of the pestivirus genus is an economically crippling virus in the cattle industry; this positive RNA virus causes mucosal disease resulting in reproductive losses and other disease syndromes. The pathogenesis mechanism of the disease caused by BVDV infection is not well understood; for a better understanding of in vivo host BVDV-1 interactions, we conducted a transcriptomic study of infected cells at different times post-infection. METHODS: We compared the permissiveness and cellular response of a BVDV-1 cytopathogenic strain on Madin-Darby Bovine Kidney cells (MDBK) and bovine lung primary cells, a model closer to in vivo infection. Then a RNAseq analysis was realized on the infected bovine lung primary cells, at 10 hpi and 30 hpi (hours post-infection), to identify transcriptomic signatures. RESULTS: RNAseq analysis on BVDV-1 infected bovine primary cells showed 2,759 and 5,376 differentially expressed genes at respectively 10 hpi and 30 hpi with an absolute Fold Change ≥ 2. Among the different pathways deregulated, data analysis revealed a deregulation of Wnt signaling pathway, a conserved process that play a critical role in embryogenesis, cellular proliferation, and differentiation as well as in viral responses against viruses such as Influenza or Hepatitis C. We demonstrated here that the deregulation of the Wnt/ßcatenin signaling pathway plays a role in viral replication of BVDV cp strain. Interestingly, we showed that the inhibition of this Wnt pathway using two inhibitors, FZM1 and iCRT14, induced a delay in onset of the establishment of a cytopathic effect of primary cells. CONCLUSIONS: Thereby, this study highlighted a role of the Wnt signaling pathway in the BVDV-1 viral replication in bovine cells, suggesting an interesting option to explore as a new therapeutic target.


Subject(s)
Bovine Virus Diarrhea-Mucosal Disease , Diarrhea Viruses, Bovine Viral , Animals , Bovine Virus Diarrhea-Mucosal Disease/genetics , Cattle , Cell Line , Cytopathogenic Effect, Viral , Diarrhea Viruses, Bovine Viral/genetics , Virus Replication/genetics , Wnt Signaling Pathway
5.
Emerg Microbes Infect ; 11(1): 60-72, 2022 Dec.
Article in English | MEDLINE | ID: mdl-34839792

ABSTRACT

The genus Pestivirus within the family Flaviviridae comprises highly relevant animal pathogens such as bovine viral diarrhoea virus 1 and 2 (BVDV-1 and -2) classified into the two species Pestivirus A and Pestivirus B, respectively. First described in 2004, HoBi-like pestiviruses (HoBiPeV) represent emerging bovine pathogens that belong to a separate species (Pestivirus H), but share many similarities with BVDV-1 and -2. Additionally, two giraffe pestivirus (GPeV) strains both originating from Kenya represent another distinct species (Pestivirus G), whose members replicate very efficiently in bovine cells. In this study, we investigated the role of bovine complement regulatory protein 46 (CD46bov), the receptor of BVDV-1 and -2, in the entry of HoBiPeV and GPeV. For this purpose, bovine CD46-knockout and CD46-rescue cell lines were generated by CRISPR/Cas9 technology and subsequent trans-complementation, respectively. Our results provide strong evidence that the impact of CD46bov differs between viruses belonging to Pestivirus H and viruses representing Pestivirus G: CD46bov revealed to be a major cellular entry factor for HoBiPeV strain HaVi-20. In contrast, GPeV strain PG-2 presented as largely independent of CD46bov, suggesting a different entry mechanism involving other molecular determinants which remain to be identified. In addition, we demonstrated that, similar to BVDV-1 and -2, virus isolates of both Pestivirus H and Pestivirus G are able to adapt to cell culture conditions by using heparan sulfate to enter the host cell. In conclusion, our findings show that different bovine pestiviruses use diverse mechanisms of host cell entry.


Subject(s)
Bovine Virus Diarrhea-Mucosal Disease/metabolism , Diarrhea Viruses, Bovine Viral/physiology , Membrane Cofactor Protein/metabolism , Receptors, Virus/metabolism , Animals , Bovine Virus Diarrhea-Mucosal Disease/genetics , Bovine Virus Diarrhea-Mucosal Disease/virology , Cattle , Cell Line , Diarrhea Viruses, Bovine Viral/classification , Diarrhea Viruses, Bovine Viral/genetics , Membrane Cofactor Protein/genetics , Receptors, Virus/genetics , Virus Internalization
6.
Virology ; 567: 34-46, 2022 02.
Article in English | MEDLINE | ID: mdl-34953294

ABSTRACT

The bovine viral diarrhea virus 1 (BVDV-1), belonging to the Pestivirus genus, is characterized by the presence of two biotypes, cytopathogenic (cp) or non-cytopathogenic (ncp). For a better understanding of the host pathogen interactions, we set out to identify transcriptomic signatures of bovine lung primary cells (BPCs) infected with a cp or a ncp strain. For this, we used both a targeted approach by reverse transcription droplet digital PCR and whole genome approach using RNAseq. Data analysis showed 3571 differentially expressed transcripts over time (Fold Change >2) and revealed that the most deregulated pathways for cp strain are signaling pathways involved in responses to viral infection such as inflammatory response or apoptosis pathways. Interestingly, our data analysis revealed a deregulation of Wnt signaling pathway, a pathway described in embryogenesis, that was specifically seen with the BVDV-1 cp but not the ncp suggesting a role of this pathway in viral replication.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Bovine Virus Diarrhea-Mucosal Disease/genetics , Cytopathogenic Effect, Viral/genetics , Diarrhea Virus 1, Bovine Viral/genetics , Transcriptome , Wnt Signaling Pathway/genetics , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/metabolism , Bovine Virus Diarrhea-Mucosal Disease/metabolism , Bovine Virus Diarrhea-Mucosal Disease/pathology , Bovine Virus Diarrhea-Mucosal Disease/virology , Cattle , Diarrhea Virus 1, Bovine Viral/metabolism , Diarrhea Virus 1, Bovine Viral/pathogenicity , Epithelial Cells/metabolism , Epithelial Cells/virology , Gene Expression Profiling , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Interleukins/genetics , Interleukins/metabolism , Lung/metabolism , Lung/virology , Membrane Potential, Mitochondrial , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/virology , NF-kappa B/genetics , NF-kappa B/metabolism , Primary Cell Culture , Respiratory Mucosa/metabolism , Respiratory Mucosa/virology , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism , Virus Replication
7.
RNA Biol ; 18(1): 1-15, 2021 01.
Article in English | MEDLINE | ID: mdl-32615049

ABSTRACT

Circular RNAs (circRNAs) are ubiquitously expressed, covalently closed rings, produced by pre-mRNA splicing in a reversed order during post-transcriptional processing. Circularity endows 3'-5'-linked circRNAs with stability and resistance to exonucleolytic degradation which raises the question whether circRNAs may be relevant as potential therapeutic targets or agents. High stability in biological systems is the most remarkable property and a major criterion for why circRNAs could be exploited for a range of RNA-centred medical applications. Even though various biological roles and regulatory functions of circRNAs have been reported, their in-depth study is challenging because of their circular structure and sequence-overlap with linear mRNA counterparts. Moreover, little is known about their role in viral infections and in antiviral immune responses. We believe that an in-depth and detailed understanding of circRNA mediated viral protein regulations will increase our knowledge of the biology of these novel molecules. In this review, we aimed to provide a comprehensive basis and overview on the biogenesis, significance and regulatory roles of circRNAs in the context of antiviral immune responses and viral infections including hepatitis C virus infection, hepatitis B virus infection, hepatitis delta virus infection, influenza A virus infection, Epstein-Barr virus infection, kaposi's sarcoma herpesvirus infection, human cytomegalovirus infection, herpes simplex virus infection, human immunodeficiency virus infection, porcine epidemic diarrhoea virus infection, ORF virus infection, avian leukosis virus infection, simian vacuolating virus 40 infection, transmissible gastroenteritis coronavirus infection, and bovine viral diarrhoea virus infection. We have also discussed the critical regulatory role of circRNAs in provoking antiviral immunity, providing evidence for implications as therapeutic agents and as diagnostic markers.


Subject(s)
Host-Pathogen Interactions/physiology , Precision Medicine/methods , RNA, Circular/immunology , Virus Diseases/genetics , Virus Diseases/immunology , Animals , Biomarkers/analysis , Bovine Virus Diarrhea-Mucosal Disease/genetics , Cattle , Coronavirus Infections/genetics , Coronavirus Infections/veterinary , HIV Infections/genetics , Hepatitis C/genetics , Herpesviridae Infections/genetics , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Influenza, Human/genetics , RNA Viruses/genetics , RNA, Circular/physiology , Swine , Swine Diseases/virology
8.
Mol Immunol ; 128: 33-40, 2020 12.
Article in English | MEDLINE | ID: mdl-33053462

ABSTRACT

The bovine viral diarrhea virus (BVDV-1) is a pathogen with the capacity to modulate the interferon type I system. To further investigate the effects of BVDV-1 on the production of the immune response, the Madin-Darby bovine kidney cell line was infected with the cytopathic CH001 field isolate of BVDV-1, and the IFNbeta expression profiles were analyzed. The results showed that cpBVDV-1 was able to induce the production of IFNbeta in a way similar to polyinosinic-polycytidylic acid, but with less intensity. Interestingly, all cpBVDV-1 activities were blocked by pharmacological inhibitors of the IRF-1, IRF-7, and NF-κB signaling pathway, and the level of IFNbeta decreased at the level of transcript and protein. These results, together with in silico analyses showing the presence of several regulatory consensus target motifs, suggest that cpBVDV-1 regulates IFNbeta expression in bovines through the activation of several key transcription factors. Collectively, the results suggest that during cpBVDV-1 infection, cross talk is evident between various signaling pathways involved in transcriptional activation of IFNbeta in cattle.


Subject(s)
Bovine Virus Diarrhea-Mucosal Disease/genetics , Diarrhea Virus 1, Bovine Viral/immunology , Gene Expression Regulation/genetics , Gene Expression/genetics , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-7/genetics , NF-kappa B/genetics , Animals , Bovine Virus Diarrhea-Mucosal Disease/virology , Cattle , Cell Line , Epithelial Cells/immunology , Epithelial Cells/virology , Gene Expression/immunology , Gene Expression Regulation/immunology , Interferon Regulatory Factor-1/immunology , Interferon Regulatory Factor-7/immunology , NF-kappa B/immunology , Signal Transduction/genetics , Signal Transduction/immunology
9.
Vopr Virusol ; 65(2): 95-102, 2020.
Article in Russian | MEDLINE | ID: mdl-32515565

ABSTRACT

INTRODUCTION: Pestiviruses are the cause of reproductive problems, diseases of the gastrointestinal and respiratory tracts of animals. Three species are important for cattle: Pestivirus A, B, and H. Fast and reliable methods of differentiation of these pathogens are currently needed. Aims and objectives of the study: the development of multiplex real time PCR for the simultaneous detection and differentiation of three viruses. MATERIAL AND METHODS: The nucleotide sequences of the conserved regions of the 5´-UTR genes of pes tivirusesA, B, and H served as a target. RESULTS: The reaction showed high specificity, sensitivity, reproducibility and was able to detect virus RNA at a concentration of not less than 0.6-1.2 lg TCID50/cm3. Cross-reactions with other pestiviruses wer e not observed. Real time PCR confirmed the results obtained previously in RT-PCR with gel electrophoresis detection. In a parallel study of 1823 biological samples, the results of the two reactions were completely consistent. Pestivirus spp. was detectedin 76 samples, Pestivirus A was present in 73 samples, Pestivirus B - in 3 samples, and Pestivirus H was not detected. DISCUSSION: A two-step real time PCR was developed for the simultaneous detection and differentiation of three pestiviruses. Modified pan primers of S. Vilcek et al. were used for the first reaction, and primers and probes of our own design were used for virus typing, which resulted in high reaction efficiency. CONCLUSION: On the big dairy farms for livestock maintenance, there are favorable conditions for the circulation of pathogenic viruses. In this situation, rapid diagnostic methods are needed to quickly identify of several viruses. Real-time triplex analysis can be recommended as the rapid method for mass epidemiological studies, as well as for screening fetal calf serum used for virus cultivation in medicine and veterinary practice.


Subject(s)
Bovine Virus Diarrhea-Mucosal Disease/genetics , Diarrhea Viruses, Bovine Viral/genetics , Real-Time Polymerase Chain Reaction , Animals , Bovine Virus Diarrhea-Mucosal Disease/diagnosis , Bovine Virus Diarrhea-Mucosal Disease/virology , Cattle , Diarrhea Viruses, Bovine Viral/isolation & purification
10.
J Vet Med Sci ; 82(5): 607-614, 2020 May 20.
Article in English | MEDLINE | ID: mdl-32173670

ABSTRACT

The objective of this study was to provide a screening scheme of persistently infected (PI) cattle on dairy herds by combining reverse-transcription polymerase chain reaction (RT-PCR) to detect bovine viral diarrhea virus (BVDV) in milk tanker samples and commercial enzyme-linked immunosorbent assay to detect BVDV antibodies in bulk tank milk. We conducted a pilot survey and regional survey targeting all dairy farms in Ibaraki Prefecture by using milk tanker and bulk tank milk samples to screen PI cattle. Farms with positive samples underwent a follow-up test to identify PI cattle. In the pilot study, all virus-positive samples in bulk tank milk were included in the positive milk tanker samples. The RT-PCR assay successfully detected BVDV at dilutions of 1:1,600 by using two PI cows' milk. In the regional survey, 5 of 79 milk tanker samples were virus-positive. The virus was detected in three PI lactating cows and one PI calf on three farms. Antibody screening using bulk tank milk samples revealed 15 of 363 samples were positive, and 12 of 348 farms were BVDV antibody-positive. Follow-up tests on one farm identified three PI calves. Thus, eight PI cattle on five farms were identified in this study. In conclusion, combining BVDV detection using milk tanker samples and antibody detection using bulk tank milk is a feasible and economical method to efficiently screen PI cattle and confirm the PI-free status among dairy herds.


Subject(s)
Antibodies, Viral/analysis , Bovine Virus Diarrhea-Mucosal Disease/epidemiology , Diarrhea Viruses, Bovine Viral/isolation & purification , Milk/virology , RNA, Viral/analysis , Animals , Bovine Virus Diarrhea-Mucosal Disease/blood , Bovine Virus Diarrhea-Mucosal Disease/genetics , Cattle , Dairying/methods , Diarrhea Viruses, Bovine Viral/immunology , Enzyme-Linked Immunosorbent Assay/veterinary , Female , Japan/epidemiology , Pilot Projects , Reverse Transcriptase Polymerase Chain Reaction/veterinary
11.
Viruses ; 12(1)2019 12 18.
Article in English | MEDLINE | ID: mdl-31861316

ABSTRACT

In cattle, conceptus-derived interferon tau (IFNT) is the pregnancy recognition (PR) signal. Our previous studies showed that non-cytopathic bovine viral diarrhoea virus (ncpBVDV) infection inhibited IFNT-induced interferon stimulated gene (ISG) expression, potentially causing early embryonic death. This study investigated the effect of bovine viral diarrhoea virus (BVDV) infection on upstream regulatory pathways of ISG production using an established PR model. Uterine endometrial cells from 10 apparently healthy and BVDV free cows were cultured and treated with 0 or 100 ng/mL IFNT for 24 h in the presence or absence of ncpBVDV infection. Microarray and pathway analysis were used to determine the IFNT-induced upstream regulators. Expression of the genes associated with the identified pathways were quantified with qPCR. IFNT challenge activated the signalling pathways associated with IFN receptors, JAK1/TYK2, IRFs and STATs and ncpBVDV infection inhibited the activation of IFNT on this pathway. Inhibition of this upstream signalling pathway may thus reduce ISG production to disrupt maternal PR. In addition, the reduction of uterine immunity by ncpBVDV infection may predispose the animals to uterine infection, which in turn impairs their reproductive performance. This provides a mechanism of how BVDV infection leads to early pregnancy failure in cows.


Subject(s)
Bovine Virus Diarrhea-Mucosal Disease/genetics , Bovine Virus Diarrhea-Mucosal Disease/virology , Diarrhea Viruses, Bovine Viral/physiology , Pregnancy Complications, Infectious/veterinary , Animals , Bovine Virus Diarrhea-Mucosal Disease/diagnosis , Bovine Virus Diarrhea-Mucosal Disease/metabolism , Cattle , Cell Line , Cell Survival , Cells, Cultured , Female , Gene Expression Regulation , Gene Regulatory Networks , Host-Pathogen Interactions/genetics , Models, Biological , Pregnancy , Signal Transduction
12.
Virology ; 538: 97-110, 2019 12.
Article in English | MEDLINE | ID: mdl-31590058

ABSTRACT

Our previous study reported that persistently infected (PI) cattle of bovine viral diarrhea virus (BVDV) have co-infected with BVDV/END- and /END+ that promote and inhibit host's type-I interferon (IFN) production, respectively. However, the relationship between co-infection of immunologically distinct BVDVs and persistent infection as well as the biological significance of END- viruses remains unknown. Experiments using cultured cells revealed that END+ virus, which is unable to propagate in situations where the host's immune response is induced by IFN-α addition, is able to propagate under those conditions when co-infecting with END- virus. These results indicate that BVDV/END- can coexist with BVDV/END+ and that co-infection with END- viruses supports the propagation of END+ viruses. Our in vitro experiments strongly suggest that co-infection with END- virus is involved in the maintenance of persistent infection of BVDV.


Subject(s)
Bovine Virus Diarrhea-Mucosal Disease/immunology , Bovine Virus Diarrhea-Mucosal Disease/virology , Diarrhea Viruses, Bovine Viral/physiology , Animals , Bovine Virus Diarrhea-Mucosal Disease/genetics , Cattle , Classical Swine Fever/genetics , Classical Swine Fever/immunology , Classical Swine Fever/virology , Classical Swine Fever Virus/genetics , Classical Swine Fever Virus/physiology , Diarrhea Viruses, Bovine Viral/genetics , Diarrhea Viruses, Bovine Viral/immunology , Guinea Pigs , Immunity, Innate , Interferon-alpha/genetics , Interferon-alpha/immunology , Swine
13.
J Anim Sci ; 97(2): 559-568, 2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30412254

ABSTRACT

Genetic selection is an inexpensive and complementary strategy to traditional methods of improving animal health and welfare. Nonetheless, endeavors to incorporate animal health and welfare traits in international breeding programs have been hampered by the availability of informative phenotypes. The recent eradication program for bovine viral diarrhea (BVD) in the Republic of Ireland has provided an opportunity to quantify the potential benefits that genetic selection could offer BVD eradication programs elsewhere, as well as inform possible eradication programs for other diseases in the Republic of Ireland. Using a dataset of 188,085 Irish calves, the estimated direct and maternal heritability estimates for the birth of persistently infected calves following likely in utero exposure to BVD virus ranged from not different from zero (linear model) to 0.29 (SE = 0.075; threshold model) and from essentially zero (linear model) to 0.04 (SE = 0.033; threshold model), respectively. The corresponding genetic SD for the direct and maternal effect of the binary trait (0, 1) ranged from 0.005 (linear model) to 0.56 (threshold model) units and ranged from 0.00008 (linear model) to 0.20 (threshold model) units, respectively. The coefficient of direct genetic variation based on the linear model was 2.56% indicating considerable genetic variation could be exploited. Based on results from the linear model in the present study, there is the potential to reduce the incidence of persistent infection in cattle by on average 0.11 percentage units per year which is cumulative and permanent. Therefore, genetic selection can contribute to reducing the incidence of persistent infection in cattle. Moreover, where populations are free from persistent infection, inclusion of the estimated genetic merit for BVD in national breeding indexes could contribute to a preservation of a BVD-free status. Results from the present study can be used to inform breeding programs of the potential genetic gains achievable. Moreover, the approaches used in the present study can be applied to other diseases when data become available.


Subject(s)
Bovine Virus Diarrhea-Mucosal Disease/transmission , Diarrhea Viruses, Bovine Viral/physiology , Genetic Variation , Infectious Disease Transmission, Vertical/veterinary , Animals , Bovine Virus Diarrhea-Mucosal Disease/genetics , Bovine Virus Diarrhea-Mucosal Disease/prevention & control , Bovine Virus Diarrhea-Mucosal Disease/virology , Breeding , Cattle , Disease Eradication , Female , Ireland/epidemiology , Linear Models , Male , Phenotype , Selection, Genetic
14.
J Virol ; 93(3)2019 02 01.
Article in English | MEDLINE | ID: mdl-30404802

ABSTRACT

After initiation of an infective cycle, spread of virus infection can occur in two fundamentally different ways: (i) viral particles can be released into the external environment and diffuse through the extracellular space until they interact with a new host cell, and (ii) virions can remain associated with infected cells, promoting the direct passage between infected and uninfected cells that is referred to as direct cell-to-cell transmission. Although evidence of cell-associated transmission has accumulated for many different viruses, the ability of members of the genus Pestivirus to use this mode of transmission has not been reported. In the present study, we used a novel recombinant virus expressing the envelope glycoprotein E2 fused to mCherry fluorescent protein to monitor the spreading of bovine viral diarrhea virus (BVDV) (the type member of the pestiviruses) infection. To demonstrate direct cell-to-cell transmission of BVDV, we developed a cell coculture system that allowed us to prove direct transmission from infected to uninfected cells in the presence of neutralizing antibodies. This mode of transmission requires cell-cell contacts and clathrin-mediated receptor-dependent endocytosis. Notably, it overcomes antibody blocking of the BVDV receptor CD46, indicating that cell-to-cell transmission of the virus involves the engagement of coreceptors on the target cell.IMPORTANCE BVDV causes one of the most economically important viral infections for the cattle industry. The virus is able to cross the placenta and infect the fetus, leading to the birth of persistently infected animals, which are reservoirs for the spread of BVDV. The occurrence of persistent infection has hampered the efficacy of vaccination because it requires eliciting levels of protection close to sterilizing immunity to prevent fetal infections. While vaccination prevents disease, BVDV can be detected if animals with neutralizing antibodies are challenged with the virus. Virus cell-to-cell transmission allows the virus to overcome barriers to free virus dissemination, such as antibodies or epithelial barriers. Here we show that BVDV exploits cell-cell contacts to propagate infection in a process that is resistant to antibody neutralization. Our results provide new insights into the mechanisms underlying the pathogenesis of BVDV infection and can aid in the design of effective control strategies.


Subject(s)
Bovine Virus Diarrhea-Mucosal Disease/virology , Cell Communication , Diarrhea Virus 1, Bovine Viral/pathogenicity , Host-Pathogen Interactions , Viral Envelope Proteins/metabolism , Virus Replication , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Bovine Virus Diarrhea-Mucosal Disease/genetics , Bovine Virus Diarrhea-Mucosal Disease/metabolism , Cattle , Cells, Cultured , Viral Envelope Proteins/genetics , Virus Internalization
15.
PLoS One ; 13(12): e0207604, 2018.
Article in English | MEDLINE | ID: mdl-30517140

ABSTRACT

The first records of smallpox and rabies date back thousands of years and foot-and-mouth disease in cattle was described in the 16th century. These diseases stood out by their distinct signs, dramatic way of transmission from rabid dogs to humans, and sudden appearance in cattle herds. By contrast, infectious diseases that show variable signs and affect few individuals were identified only much later. Bovine viral diarrhea (BVD), endemic in cattle worldwide, was first described in 1946, together with the eponymous RNA virus as its cause. There is general agreement that BVD was not newly emerging at that time, but its history remains unknown. A search for associations between the nucleotide sequences of over 7,000 BVD viral strains obtained during a national campaign to eradicate BVD and features common to the hosts of these strains enabled us to trace back in time the presence of BVD in the Swiss cattle population. We found that animals of the two major traditional cattle breeds, Fleckvieh and Swiss Brown, were infected with strains of only four different subgenotypes of BVDV-1. The history of these cattle breeds and the events that determined the current distribution of the two populations are well documented. Specifically, Fleckvieh originates from the Bernese and Swiss Brown from the central Alps. The spread to their current geographic distribution was determined by historic events during a major expansion of the Swiss Confederation during the 15th and 16th centuries. The association of the two cattle populations with different BVD viral subgenotypes may have been preserved by a lack of cattle imports, trade barriers within the country, and unique virus-host interactions. The congruent traces of history in the distribution of the two cattle breeds and distinct viral subgenotypes suggests that BVD may have been endemic in Switzerland for at least 600 years.


Subject(s)
Bovine Virus Diarrhea-Mucosal Disease/genetics , Bovine Virus Diarrhea-Mucosal Disease/history , Diarrhea Viruses, Bovine Viral/genetics , Animals , Base Sequence/genetics , Cattle , Diarrhea/veterinary , Diarrhea/virology , Diarrhea Viruses, Bovine Viral/pathogenicity , Genetic Variation/genetics , History, 15th Century , History, 16th Century , History, Medieval , Phylogeography , RNA Viruses/genetics , Switzerland
16.
Virus Genes ; 54(6): 768-778, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30218293

ABSTRACT

Bovine viral diarrhea virus 1, reclassified as Pestivirus A, causes an economically important cattle disease that is distributed worldwide. Pestivirus A may cause persistent infection in that calves excrete the virus throughout their lives, spreading the infection in the herd. Many persistently infected (PI) calves die in the first 2 years of life from mucosal disease (MD) or secondary infections, probably as a consequence of virus-induced immune depression. Here, high-throughput sequencing (HTS) was applied for evaluation of the total virome in sera of (i) PI calves displaying clinically apparent MD (n = 8); (ii) PI calves with no signs of MD (n = 8); and (iii) control, Pestivirus A-free calves (n = 8). All the groups were collected at the same time and from the same herd. Serum samples from calves in each of the groups were pooled, submitted to viral RNA/DNA enrichment, and sequenced by HTS. Viral genomes of Pestivirus A, Ungulate erythroparvovirus 1, bosavirus (BosV), and hypothetical circular Rep-encoding single-stranded DNA (CRESS-DNA) viruses were identified. Specific real-time PCR assays were developed to determine the frequency of occurrence of such viruses in each of the groups. The absolute number of distinct viral genomes detected in both PI calf groups was higher than in the control group, as revealed by higher number of reads, contigs, and genomes, representing a wider range of taxons. Genomes representing members of the family Parvoviridae, such as U. erythroparvovirus 1 and BosV, were most frequently detected in all the three groups of calves. Only in MD-affected PI calves, we found two previously unreported Hypothetical single-stranded DNA genomes clustered along with CRESS-DNA viruses. These findings reveal that parvoviruses were the most frequently detected viral genomes in cattle serum; its frequency of detection bears no statistical correlation with the status of calves in relation to Pestivirus A infection, since clinically normal or MD-affected/non-affected PI calves were infected with similar U. erythroparvovirus 1 genome loads. Moreover, MD-affected PI calves were shown to support viremia of CRESS-DNA viral genomes; however, the meaning of such correlation remains to be established.


Subject(s)
Bovine Virus Diarrhea-Mucosal Disease/genetics , Diarrhea Virus 1, Bovine Viral/genetics , High-Throughput Nucleotide Sequencing , Pestivirus/genetics , Animals , Antibodies, Viral/blood , Bovine Virus Diarrhea-Mucosal Disease/blood , Bovine Virus Diarrhea-Mucosal Disease/physiopathology , Bovine Virus Diarrhea-Mucosal Disease/virology , Cattle , DNA, Viral/genetics , Diarrhea Virus 1, Bovine Viral/isolation & purification , Diarrhea Virus 1, Bovine Viral/pathogenicity , Genome, Viral/genetics , Pestivirus/classification , Pestivirus/isolation & purification , Pestivirus/pathogenicity , RNA, Viral/genetics
17.
Virology ; 507: 123-134, 2017 07.
Article in English | MEDLINE | ID: mdl-28432927

ABSTRACT

Bovine viral diarrhea virus (BVDV) infection induces immunosuppression and in conjunction with bovine respiratory syncytial virus (BRSV) contributes to the bovine respiratory disease complex. Bovine turbinate cells were single or co-infected with type 2 BVDV wild-type (BVDV2-wt), its dysfunctional Npro mutant (BVDV2-E), and/or BRSV. BVDV2-E significantly up-regulated PKR, IRF-7, TBK-1, IRF-3, and IFN-ß mRNAs based on real-time Q-RT-PCR. BRSV-infected cells expressed significantly up-regulated PKR, IRF-3, IRF-7, and IFN-ß mRNAs, whereas BVDV2-wt, but not BVDV2-E, abolished this up-regulation in co-infection. No significant differences were observed in MAVS, NF-κB, and PIN-1 mRNAs. A dual-luciferase reporter assay showed that BVDV2-wt significantly increased NF-κB activity compared to BVDV2-E, while BVDV2-E significantly increased IFN-ß activity compared to BVDV2-wt. The BRSV titer and RNA levels significantly increased in cells co-infected with BRSV/BVDV2-wt compared to cells co-infected with BRSV/BVDV2-E or infected with BRSV alone. This data supports the synergistic action of BVDV2-wt and BRSV inhibition of IFN-1.


Subject(s)
Bovine Virus Diarrhea-Mucosal Disease/metabolism , Diarrhea Virus 2, Bovine Viral/physiology , Interferon-beta/metabolism , Respiratory Syncytial Virus Infections/veterinary , Signal Transduction , Animals , Bovine Virus Diarrhea-Mucosal Disease/genetics , Bovine Virus Diarrhea-Mucosal Disease/virology , Cattle , Coinfection/genetics , Coinfection/immunology , Coinfection/virology , Diarrhea Virus 2, Bovine Viral/genetics , Interferon-beta/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Respiratory Syncytial Virus Infections/genetics , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Bovine/genetics , Respiratory Syncytial Virus, Bovine/physiology , Virus Replication
18.
PLoS One ; 11(7): e0159491, 2016.
Article in English | MEDLINE | ID: mdl-27420479

ABSTRACT

Bovine viral diarrhea virus (BVDV) is a member of the Flaviviridae family. BVDV isolates are classified into two biotypes based on the development of cytopathic (cp) or non-cytopathic (ncp) effects in epithelial cell culture. BVDV isolates are further separated into species, BVDV1 and 2, based on genetic differences. Symptoms of BVDV infection range from subclinical to severe, depending on strain virulence, and may involve multiple organ systems and induction of a generalized immunosuppression. During BVDV-induced immune suppression, macrophages, critical to innate immunity, may have altered pathogen recognition receptor (PRR) signaling, including signaling through toll-like receptors (TLRs). Comparison of BVDV 2 strains with different biotypes and virulence levels is valuable to determining if there are differences in host macrophage cellular responses between viral phenotypes. The current study demonstrates that cytopathic (cp), noncytopathic (ncp), high (hv) or low virulence (lv) BVDV2 infection of bovine monocyte-derived macrophages (MDMΦ) result in differential expression of pro-inflammatory cytokines compared to uninfected MDMΦ. A hallmark of cp BVDV2 infection is IL-6 production. In response to TLR2 or 4 ligation, as might be observed during secondary bacterial infection, cytokine secretion was markedly decreased in BVDV2-infected MDMΦ, compared to non-infected MDMΦ. Macrophages were hyporesponsive to viral TLR3 or TLR8 ligation. However, TLR7 stimulation of BVDV2-infected MDMΦ induced cytokine secretion, unlike results observed for other TLRs. Together, these data suggest that BVDV2 infection modulated mRNA responses and induced a suppression of proinflammatory cytokine protein responses to TLR ligation in MDMΦ with the exception of TLR7 ligation. It is likely that there are distinct differences in TLR pathways modulated following BVDV2 infection, which have implications for macrophage responses to secondary infections.


Subject(s)
Bovine Virus Diarrhea-Mucosal Disease/immunology , Diarrhea Virus 2, Bovine Viral/immunology , Macrophages/immunology , Toll-Like Receptor 7/immunology , Animals , Bovine Virus Diarrhea-Mucosal Disease/genetics , Bovine Virus Diarrhea-Mucosal Disease/pathology , Cattle , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , Cytopathogenic Effect, Viral , Diarrhea Virus 2, Bovine Viral/pathogenicity , Diarrhea Virus 2, Bovine Viral/physiology , Gene Expression Regulation , Interleukin-6/genetics , Interleukin-6/immunology , Macrophages/pathology , Macrophages/virology , RNA, Messenger/genetics , Toll-Like Receptors/immunology
19.
Mol Immunol ; 71: 192-202, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26919728

ABSTRACT

The bovine viral diarrhea virus (BVDV) is responsible for significant economic losses in the dairy and cattle industry; however, little is known about the protective and pathological responses of hosts to infection. The present study determined the principal molecular markers implicated in viral infection through meta-transcriptomic analysis using MDBK cells infected for two hours with a field isolate of BVDV-1. While several immune regulator genes were induced, genes involved in cell signaling, metabolic processes, development, and integrity were down-regulated, suggesting an isolation of infected cells from cell-to-cell interactions and responses to external signals. Analysis through RT-qPCR confirmed the expression of more than one hundred markers. Interestingly, there was a significant up-regulation of two negative NF-κB regulators, IER3 and TNFAIP3, indicating a possible blocking of this signaling pathway mediated by BVDV-1 infection. Additionally, several genes involved in the metabolism of reactive oxygen species were down-regulated, suggesting increased oxidative stress. Notably, a number of genes involved in cellular growth and development were also regulated during infection, including MTHFD1L, TGIF1, and Brachyury. Moreover, there was an increased expression of the genes ß-catenin, caprin-2, GSK3ß, and MMP-7, all of which are crucial to the Wnt signaling pathway that is implicated in the embryonic development of a variety of organisms. This meta-transcriptomic analysis provides the first data towards understanding the infection mechanisms of cytopathic BVDV-1 and the putative molecular relationship between viral and host components.


Subject(s)
Bovine Virus Diarrhea-Mucosal Disease/genetics , Bovine Virus Diarrhea-Mucosal Disease/immunology , Transcriptome , Animals , Cattle , Cell Line , Diarrhea Virus 1, Bovine Viral , Fluorescent Antibody Technique , Gene Expression Profiling , Reverse Transcriptase Polymerase Chain Reaction
20.
Gene ; 581(2): 117-29, 2016 May 01.
Article in English | MEDLINE | ID: mdl-26809100

ABSTRACT

Cytokine production for immunological process is tightly regulated at the transcriptional and posttranscriptional levels. The NF-κB signaling pathway maintains immune homeostasis in the cell through the participation of molecules such as A20 (TNFAIP3), which is a key regulatory factor in the immune response, hematopoietic differentiation, and immunomodulation. Although A20 has been identified in mammals, and despite recent efforts to identify A20 members in other higher vertebrates, relatively little is known about the composition of this regulator in other classes of vertebrates, particularly for bovines. In this study, the genetic context of bovine A20 was explored and compared against homologous genes in the human, mouse, chicken, dog, and zebrafish chromosomes. Through in silico analysis, several regions of interest were found conserved between even phylogenetically distant species. Additionally, a protein-deduced sequence of bovine A20 evidenced many conserved domains in humans and mice. Furthermore, all potential amino acid residues implicated in the active site of A20 were conserved. Finally, bovine A20 mRNA expression as mediated by the bovine viral diarrhea virus and poly (I:C) was evaluated. These analyses evidenced a strong fold increase in A20 expression following virus exposure, a phenomenon blocked by a pharmacological NF-κB inhibitor (BAY 117085). Interestingly, A20 mRNA had a half-life of only 32min, likely due to adenylate- and uridylate-rich elements in the 3'-untranslated region. Collectively, these data identify bovine A20 as a regulator of immune marker expression. Finally, this is the first report to find the bovine viral diarrhea virus modulating bovine A20 activation through the NF-κB pathway.


Subject(s)
Bovine Virus Diarrhea-Mucosal Disease/genetics , Bovine Virus Diarrhea-Mucosal Disease/immunology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , NF-kappa B/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Animals , Bovine Virus Diarrhea-Mucosal Disease/virology , Catalytic Domain , Cattle , Cell Line , DNA-Binding Proteins/chemistry , Gene Expression Regulation/drug effects , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Mice , Nitriles/pharmacology , Nuclear Proteins/chemistry , RNA, Messenger/metabolism , Sequence Homology, Amino Acid , Signal Transduction/drug effects , Sulfones/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL