Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 136
Filter
1.
Viruses ; 16(3)2024 03 05.
Article in English | MEDLINE | ID: mdl-38543767

ABSTRACT

Bovine parainfluenza virus type 3 (BPIV-3) is one of the major pathogens of the bovine respiratory disease complex (BRDC). BPIV-3 surveillance in China has been quite limited. In this study, we used PCR to test 302 cattle in China, and found that the positive rate was 4.64% and the herd-level positive rate was 13.16%. Six BPIV-3C strains were isolated and confirmed by electron microscopy, and their titers were determined. Three were sequenced by next-generation sequencing (NGS). Phylogenetic analyses showed that all isolates were most closely related to strain NX49 from Ningxia; the genetic diversity of genotype C strains was lower than strains of genotypes A and B; the HN, P, and N genes were more suitable for genotyping and evolutionary analyses of BPIV-3. Protein variation analyses showed that all isolates had mutations at amino acid sites in the proteins HN, M, F, and L. Genetic recombination analyses provided evidence for homologous recombination of BPIV-3 of bovine origin. The virulence experiment indicated that strain Hubei-03 had the highest pathogenicity and could be used as a vaccine candidate. These findings apply an important basis for the precise control of BPIV-3 in China.


Subject(s)
Parainfluenza Virus 3, Bovine , Parainfluenza Virus 3, Human , Animals , Cattle , Virulence , Phylogeny , Prevalence , Parainfluenza Virus 3, Bovine/genetics , China/epidemiology
2.
Vet Microbiol ; 290: 109972, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38183839

ABSTRACT

Bovine Parainfluenza virus Type 3 (BPIV3) is one of the most important pathogens in cattle, capable of causing severe respiratory symptoms. Numerous studies have shown that autophagy plays a diverse role in the infection process of various pathogens. The influence of autophagy machinery on BPIV3 infection has not yet been confirmed. In the present study, we initially demonstrated that the expression of LC3 was significantly increased and exhibited a notable increase in double or single-membrane vesicles under a transmission electron microscope during BPIV3 infection. These observations unequivocally establish the induction of steady-state autophagy in vitro consequent to BPIV3 infection. Furthermore, quantification of autophagic flux substantiates the induction of an incomplete autophagic process during BPIV3 infection. Additionally, through targeted interventions, we demonstrate the regulatory impact of pharmacological agents influencing autophagy and RNA interference targeting an autophagy-associated protein on viral replication. Intriguingly, our data revealed that BPIV3 infection enhanced the phosphorylation of rapamycin kinase (mTOR). This result demonstrated that mTOR does not operate as a counteractive regulator of BPIV3-induced autophagy. Instead, we discern an augmentation in the expression of Beclin1, a key autophagy initiator, which complexes with Vps34, constituting a Class III phosphatidylinositol 3-kinase. This phenomenon serves as a hallmark in the inaugural phase of autophagy initiation during BPIV3 infection. Collectively, these discernments underscore that BPIV3 infection actively stimulates autophagy, thereby enhancing viral replication through the activation of Beclin1, independently of the mTOR signaling pathway. This nuanced comprehension significantly contributes to unraveling the intricate molecular mechanisms governing BPIV3-induced autophagy.


Subject(s)
Cattle Diseases , Paramyxoviridae Infections , Animals , Cattle , Beclin-1/genetics , Parainfluenza Virus 3, Bovine/physiology , TOR Serine-Threonine Kinases/metabolism , Virus Replication/genetics , Autophagy , Paramyxoviridae Infections/veterinary
3.
Microb Pathog ; 185: 106444, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37951410

ABSTRACT

Bovine parainfluenza virus type 3 (BPIV3) is a viral respiratory pathogen of cattle that causes substantial economic losses. A replicating-defective recombinant human adenovirus type 5 (HAd5), carrying a fusion protein of BPIV3 genotype C (HAd5-F), was constructed and evaluated for its immunogenicity and protective efficacy in mice. After intramuscular injection with the HAd5-F, the IgG titers against F proteins increased to 1:102,400, and virus-neutralizing titers increased to 1:256, significantly higher than those in the group injected with inactivated BPIV3C in mice (p<0.05). The splenic CD4+/CD8+T lymphocytes and IFN-γ+/IL-4+ cytokine percentages were more significant in the HAd5-F group than those in the control group. A BPIV3C challenge in a mouse model was used to assess protective efficacy of the HAd5-F. The viral loads in the lungs and tracheas of mice immunized with the HAd5-F were significantly lower than those in the control group (p<0.0001). There were no significant histopathological alterations in the lungs of mice vaccinated with the HAd5-F. These findings suggested that the HAd5-F elicited excellent immunity against BPIV3C infection.


Subject(s)
Adenoviridae , Parainfluenza Virus 3, Human , Animals , Cattle , Humans , Mice , Adenoviridae/genetics , Antibodies, Viral , Parainfluenza Virus 3, Bovine/genetics , Recombinant Proteins/genetics , Genotype
4.
Braz J Microbiol ; 54(4): 3237-3244, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37700145

ABSTRACT

Bovine viral diarrhea virus (BVDV), bovine alphaherpesvirus 1 (BoAHV1), bovine respiratory syncytial virus (BRSV), and bovine parainfluenza virus 3 (BPIV-3) are involved in bovine respiratory disease. These viruses can infect the respiratory system and cause considerable economic losses to beef and dairy cattle herds. This study aimed to determine the serological profiles of steers for BVDV, BoAHV1, BRSV, and BPIV-3 upon their arrival at Brazilian feedlot facilities. A total of 1,282 serum samples from unvaccinated steers were obtained on the first day of feeding. Samples were collected from 31 beef cattle herds reared in an extensive rearing system in six Brazilian states. Antibodies against BVDV, BoAHV1, BRSV, and BPIV-3 were detected using a virus neutralization test. The steers were distributed in agreement with their age and the Brazilian state of origin. The highest seropositivity was for BoAHV1 and BPIV-3 at 92.1% (1,154/1,253) and 86.6% (1,100/1,270), respectively. The seropositivity of BRSV was 77.1% (959/1,244). BVDV presented a lower rate, at slightly more than 50% (51.8%; 656/1,266). Age was a risk factor for the presence of antibodies against BVDV, BoAHV1, and BPIV-3 but not BRSV. A positive correlation was identified between BoAHV1 and BPIV-3 (P = 0.85) and between BRSV and BPIV-3 (P = 0.47). The high rate of seropositive steers for these four respiratory viruses on the first day of confinement identified in this serological survey provides important epidemiological information on respiratory infections, as the seropositivity of the four main bovine respiratory viruses in Brazilian beef cattle herds in an extensive rearing system.


Subject(s)
Cattle Diseases , Diarrhea Viruses, Bovine Viral , Herpesvirus 1, Bovine , Viruses , Animals , Cattle , Brazil/epidemiology , Cattle Diseases/microbiology , Parainfluenza Virus 3, Bovine , Antibodies, Viral
5.
Infect Genet Evol ; 113: 105483, 2023 09.
Article in English | MEDLINE | ID: mdl-37482235

ABSTRACT

Bovine Parainfluenza Type 3 virus (BPIV-3) is an enveloped, non-segmented single-stranded, negative-sense RNA virus belonging to the Paramyxoviridae family (genus Respirovirus) with a well-known role in Bovine Respiratory Disease (BRD) onset. Being isolated for the first time in 1959, BPIV-3 currently circulates worldwide in cattle herds and is routinely tested in suspected BRD cases. Different commercial vaccines are available to prevent infection and/or to reduce the clinical signs associated with BPIV-3 infection, which are essential to prevent secondary infections. Despite years of molecular surveillance, a very limited number of complete genome sequences were made publicly available, preventing thus the understanding of the genetic diversity of the circulating strains in the field. In addition, no data about the genetic identity between field and vaccine strains is currently available. In this study, we sequenced the full-genome and genetically characterized BPIV-3 strains isolated from animals displaying respiratory illness in France and Sweden, as well as the vaccine strains contained in three different commercialized vaccines. Our results show that the sequences from France and Sweden belong to genotype C. However, a third sequence from Sweden from 2017 clustered within genotype A. The sequencing of vaccine strains revealed that two of the vaccine strains clustered within genotype C, whereas the third vaccine strain belonged to genotype A. Altogether, our findings suggest that both genotypes A and C circulate in Europe and that BPIV-3 field and vaccine strains are genetically divergent. Our sequencing results could be useful to better understand the genetic differences between the circulating field and vaccine BPIV-3 strains. This is crucial for a correct interpretation of diagnostic findings and for the assessment of BPIV-3 prevalence in cattle population.


Subject(s)
Cattle Diseases , Paramyxoviridae Infections , Viral Vaccines , Cattle , Animals , Respirovirus/genetics , Parainfluenza Virus 3, Bovine/genetics , Viral Vaccines/genetics , Europe , Cattle Diseases/epidemiology , Cattle Diseases/prevention & control
6.
Microbiol Immunol ; 67(4): 204-209, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36609846

ABSTRACT

Bovine parainfluenza virus type 3 (BPIV3) is a promising vaccine vector against various respiratory virus infections, including the human PIV3, respiratory syncytial virus, and severe acute respiratory syndrome-coronavirus 2 infections. In this study, we combined the Magnet system and reverse genetic approach to generate photocontrollable BPIV3. An optically controllable Magnet gene was inserted into the H2 region of the BPIV3 large protein gene, which encodes an RNA-dependent RNA polymerase. The generated photocontrollable BPIV3 grew in specific regions of the cell sheet only when illuminated with blue light, suggesting that spatiotemporal control can aid in safe clinical applications of BPIV3.


Subject(s)
COVID-19 , Respiratory Syncytial Virus, Human , Animals , Cattle , Humans , Parainfluenza Virus 3, Human/genetics , Cell Line , Virus Replication , Parainfluenza Virus 3, Bovine/genetics
7.
Arch Razi Inst ; 78(6): 1779-1786, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38828184

ABSTRACT

Aluminum-containing adjuvants are extensively used in inactive human and animal vaccines owing to their favorable immunostimulatory and safe properties. Nonetheless, there is controversy over the effects of different aluminum salts as an adjuvant for the bovine parainfluenza virus type 3 (BPIV3) vaccine. In order to find a suitable adjuvant, we studied the effects of two adjuvants (i.e., aluminum hydroxide [Al(OH)3] and aluminum potassium sulfate [AlPO4]) on the production of neutralizing antibodies (NAbs) for an experimental BPIV3 vaccine. The animals under study (Guinea pigs) were randomly assigned to five groups of experimental vaccines containing Al(OH)3 (AH), AlPO4 (AP), Al(OH)3-AlPO4 mixture (MIX), commercial vaccine (COM), and control (NS). The treatment groups were immunized with two doses of vaccine 21 days apart (on days 0 and 21), and the control group received normal saline under the same conditions. The animals were monitored for 42 days, and blood samples were then taken. The results indicated that all vaccines were able to induce the production of NAbs at levels higher than the minimum protective titer (0.6). An increase in titer was observed throughout the monitoring period. Moreover, an increase in both the level and mean titer of NAbs obtained from the vaccine containing Al(OH)3 adjuvant was significantly higher than in the other studied groups (P≤0.005). The comparison of NAbs titer in other groups did not display a significant difference. Considering the speed of rising and the optimal titer of NAbs production in the experimental vaccine, the Al(OH)3 adjuvant is a suitable candidate for preparing a vaccine against BPIV3 for immunization.


Subject(s)
Adjuvants, Immunologic , Aluminum Hydroxide , Antibodies, Neutralizing , Parainfluenza Virus 3, Bovine , Animals , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/administration & dosage , Aluminum Hydroxide/pharmacology , Aluminum Hydroxide/administration & dosage , Antibodies, Neutralizing/blood , Guinea Pigs , Parainfluenza Virus 3, Bovine/immunology , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/pharmacology , Antibodies, Viral/blood , Random Allocation , Aluminum Compounds/pharmacology , Aluminum Compounds/administration & dosage , Female
8.
Viruses ; 14(10)2022 10 09.
Article in English | MEDLINE | ID: mdl-36298776

ABSTRACT

Bovine parainfluenza virus 3 (BPIV3) is one of several viruses that contribute to bovine respiratory disease complex (BRDC). During this study, isolation of BPIV3 was attempted from 20 PCR-positive swabs by Madin-Darby Bovine Kidney (MDBK) cells. Nine samples showed obvious cytopathic lesions identified as BPIV3 by reverse-transcription polymerase chain reaction amplification and sequencing. The genomes of isolates XJ21032-1 and XJ20055-3 were sequenced using Illumina sequencing technology and determined to have lengths of 15,512 bp and 15,479 bp, respectively. Phylogenetic analysis revealed that isolate XJ21032-1 was genotype B, and isolate XJ20055-3 was genotype C. In addition, the two isolates had multiple amino acid changes in nucleocapsid protein, fusion protein, and hemagglutinin/neuraminidase, major antigenic proteins. This allows the further recognition of the presence of BPIV3 type B in Chinese cattle herds. We hope this will help trace the origin of BPIV3, improve the understanding of differences between genotypes, and provide data support for vaccine development.


Subject(s)
Parainfluenza Virus 3, Bovine , Paramyxoviridae Infections , Cattle , Animals , Parainfluenza Virus 3, Bovine/genetics , Phylogeny , Hemagglutinins , Neuraminidase/genetics , Genotype , Nucleocapsid Proteins/genetics , Amino Acids/genetics
9.
Virol J ; 19(1): 116, 2022 07 13.
Article in English | MEDLINE | ID: mdl-35831876

ABSTRACT

BACKGROUND: Bovine parainfluenza virus type 3 (BPIV3) infection often causes respiratory tissue damage and immunosuppression and further results in bovine respiratory disease complex (BRDC), one of the major diseases in dairy cattle, caused huge economical losses every year. However, the pathogenetic and immunoregulatory mechanisms involved in the process of BPIV3 infection remain unknown. However, the pathogenetic and immunoregulatory mechanisms involved in the process of BPIV3 infection remain unknown. Proteomics is a powerful tool for high-throughput identification of proteins, which has been widely used to understand how viruses interact with host cells. METHODS: In the present study, we report a proteomic analysis to investigate the whole cellular protein alterations of MDBK cells infected with BPIV3. To investigate the infection process of BPIV3 and the immune response mechanism of MDBK cells, isobaric tags for relative and absolute quantitation analysis (iTRAQ) and Q-Exactive mass spectrometry-based proteomics were performed. The differentially expressed proteins (DEPs) involved in the BPIV3 invasion process in MDBK cells were identified, annotated, and quantitated. RESULTS: A total of 116 proteins, which included 74 upregulated proteins and 42 downregulated proteins, were identified as DEPs between the BPIV3-infected and the mock-infected groups. These DEPs included corresponding proteins related to inflammatory response, immune response, and lipid metabolism. These results might provide some insights for understanding the pathogenesis of BPIV3. Fluorescent quantitative PCR and western blotting analysis showed results consistent with those of iTRAQ identification. Interestingly, the upregulated protein MKK3 was associated with the p38 MAPK signaling pathway. CONCLUSIONS: The results of proteomics analysis indicated BPIV3 infection could activate the p38 MAPK pathway to promote virus replication.


Subject(s)
Parainfluenza Virus 3, Human , Proteomics , Animals , Cattle , Parainfluenza Virus 3, Bovine/physiology , Virus Replication/physiology , p38 Mitogen-Activated Protein Kinases
10.
Vet Microbiol ; 271: 109488, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35691094

ABSTRACT

Bovine parainfluenza virus 3 (BPIV3) is an important respiratory pathogen of both young and adult cattle. The pathways of cell entry are highly related to viral transmission and pathogenicity. In previous studies, we demonstrated that macropinocytosis and clathrin-dependent endocytosis play critical roles in the entry of BPIV3 into MDBK cells. Macropinocytosis is special endocytic process which need to activate signaling pathways that remodle the actin cytoskeleton. Parainfluenza viruses (PIVs) initiate infection by binding to sialic acid receptors on cell surfaces. Nevertheless, sialic acids are not able to transmit signals across the plasma membrane, indicating the necessity for additional signaling receptors. Here, we have demonstrated that specific inhibitors and siRNAs targeting EGFR inhibit the entry of BPIV3 into MDBK cells. BPIV3 productive infection in MDBK cells led to activation of EGFR. Inactivation of EGFR suppressed BPIV3-induced rearrangement of the F-actin cytoskeleton. In addition, PI3K-Akt and ERK1/2 were activated in an EGFR-dependent manner during BPIV3 infection. Specific inhibitors targeting these canonical downstream effectors of EGFR could significantly reduce viral entry efficacy. Moreover, we also demonstrated that the important regulators of macropinocytosis, Rac1 and Pak1, are downstream mediators of EGFR during BPIV3 internalization. These results indicated that EGFR is a host-entry cofactor used by BPIV3 to enter MDBK cells.


Subject(s)
Parainfluenza Virus 3, Bovine , Parainfluenza Virus 3, Human , Animals , Cattle , ErbB Receptors/genetics , ErbB Receptors/metabolism , Parainfluenza Virus 3, Bovine/physiology , Phosphatidylinositol 3-Kinases , Virus Internalization
11.
Vet Microbiol ; 268: 109415, 2022 May.
Article in English | MEDLINE | ID: mdl-35395543

ABSTRACT

Bovine parainfluenza virus type 3 (BPIV3) is one of the most important viral respiratory pathogens of cattle. No specific therapies are available for BPIV3 infection; vaccination is one of the most effective ways to prevent BPIV3 infection. We therefore prepared the self-assembled BPIV3 nanoparticles by genetically fusing the ectodomain of BPIV3 haemagglutinin-neuraminidase (HN) (HNex) to the NH2 terminus of ferritin (HNex-RFNp) using a baculovirus expression system. It was found that HNex-RFNp-induced bone marrow-derived dendritic cell (BMDC) maturation through the upregulated expression of surface molecules (MHC II, CD80, CD86, and CD40), increased the secretion of inflammatory cytokines (IL-6, IL-12, TNF-α, and IFN-γ), and reduced antigen phagocytosis and T cell activation capacity. HNex-RFNp positively regulated IκBα and NF-κB (p65) phosphorylation and facilitated NF-κB (p65) translocation into the nuclei of mature BMDCs. Incubating RFNp-treated BMDCs with TLR4 and NF-κB (p65) inhibitors, suppressed surface molecule expression as well as pro-inflammatory cytokine production and IκBα and NF-κB (p65) activities. The BPIV3 HNex protein induced BMDC maturation to some extent but was significantly weaker than HNex-RFNp. We found that HNex-RFNp induced a higher titre of specific antibodie, haemagglutinin inhibition (HI) antibody, and virus neutralisation (VN) antibody, and a comprehensive cellular immune response. We examined protection against BPIV3 challenge in a mouse model. Pathological changes were not observed in the lungs of HNex-RFNp-vaccinated mice. Levels of BPIV3 RNA and virus titres in the lungs and trachea were significantly lower in the HNex-RFNp, than HNex, inactivated BPIV3, and PBS groups. In summary, HNex-RFNp elicited better immunogenicity than HNex or inactivated BPIV3 and could be developed as an effective vaccine to protect against BPIV3 infection.


Subject(s)
Dendritic Cells , NF-kappa B , Nanoparticles , Parainfluenza Virus 3, Bovine , Viral Vaccines , Virus Diseases , Animals , Cattle , Dendritic Cells/immunology , Hemagglutinins/metabolism , Immunogenicity, Vaccine , Lymphocyte Activation , Mice , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B/metabolism , Viral Vaccines/immunology , Virus Diseases/prevention & control , Virus Diseases/veterinary
12.
J Dairy Sci ; 105(1): 560-571, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34763911

ABSTRACT

The role of colostrum management in providing adequate immunological protection to neonatal calves has been widely investigated, and thresholds for colostrum quality, as well as optimum volume and timing for colostrum feeding have been established. However, limited information is available on the effect of colostrum source (single dam or pooled) on passive immunity, as well as subsequent antibody survival in the calf. This study aimed to assess the effect of feeding single-dam colostrum (own and other dam) or pooled colostrum on transfer of passive immunity, and also investigate the rate of depletion of disease-specific antibodies among dairy calves. In total, 320 cows and 119 dairy heifer calves were enrolled in the study. Calves were blood-sampled immediately after birth and received either own-dam, other-dam, or pooled colostrum. Calves were blood-sampled at 24 h to assess serum IgG concentrations and at monthly intervals thereafter to document disease-specific antibody survival. Mean colostrum IgG concentration was higher for other-dam treatment group, whereas own-dam and pooled treatments were similar. For all treatment groups, the mean IgG concentration was >80 mg/mL, exceeding the quality threshold of 50 mg/mL. Mean calf serum IgG concentration was lower for calves fed pooled colostrum compared with those that received colostrum from a single cow. There was a negative association with 24-h serum IgG and calf birth bodyweight; calves <30 kg at birth had the highest 24-h serum IgG concentration. Survival of antibodies to bovine viral diarrhea, Salmonella infection, leptospirosis, bovine parainfluenza 3 virus, bovine respiratory syncytical virus, rotavirus, and coronavirus was not associated with colostrum source; however, antibodies to infectious bovine rhinotracheitis had a greater period of survival among calves fed own-dam colostrum. We found that feeding single-dam colostrum can thus improve calf immunity through increased serum IgG levels and antibody survival rates. Furthermore, we hypothesize that immune exclusion may occur with pooled colostrum; therefore, providing pooled colostrum may still be a good practice as long as it can be ensured that enough antibodies are absorbed into the blood stream to deal with pathogens calves may encounter because different dams may have antibodies against different strains of viruses and bacteria, yielding cross protection.


Subject(s)
Colostrum , Parainfluenza Virus 3, Bovine , Animals , Animals, Newborn , Cattle , Female , Immunity, Maternally-Acquired , Immunoglobulins , Parturition , Pregnancy
13.
Proc Natl Acad Sci U S A ; 118(50)2021 12 14.
Article in English | MEDLINE | ID: mdl-34876520

ABSTRACT

Single-dose vaccines with the ability to restrict SARS-CoV-2 replication in the respiratory tract are needed for all age groups, aiding efforts toward control of COVID-19. We developed a live intranasal vector vaccine for infants and children against COVID-19 based on replication-competent chimeric bovine/human parainfluenza virus type 3 (B/HPIV3) that express the native (S) or prefusion-stabilized (S-2P) SARS-CoV-2 S spike protein, the major protective and neutralization antigen of SARS-CoV-2. B/HPIV3/S and B/HPIV3/S-2P replicated as efficiently as B/HPIV3 in vitro and stably expressed SARS-CoV-2 S. Prefusion stabilization increased S expression by B/HPIV3 in vitro. In hamsters, a single intranasal dose of B/HPIV3/S-2P induced significantly higher titers compared to B/HPIV3/S of serum SARS-CoV-2-neutralizing antibodies (12-fold higher), serum IgA and IgG to SARS-CoV-2 S protein (5-fold and 13-fold), and IgG to the receptor binding domain (10-fold). Antibodies exhibited broad neutralizing activity against SARS-CoV-2 of lineages A, B.1.1.7, and B.1.351. Four weeks after immunization, hamsters were challenged intranasally with 104.5 50% tissue-culture infectious-dose (TCID50) of SARS-CoV-2. In B/HPIV3 empty vector-immunized hamsters, SARS-CoV-2 replicated to mean titers of 106.6 TCID50/g in lungs and 107 TCID50/g in nasal tissues and induced moderate weight loss. In B/HPIV3/S-immunized hamsters, SARS-CoV-2 challenge virus was reduced 20-fold in nasal tissues and undetectable in lungs. In B/HPIV3/S-2P-immunized hamsters, infectious challenge virus was undetectable in nasal tissues and lungs; B/HPIV3/S and B/HPIV3/S-2P completely protected against weight loss after SARS-CoV-2 challenge. B/HPIV3/S-2P is a promising vaccine candidate to protect infants and young children against HPIV3 and SARS-CoV-2.


Subject(s)
COVID-19 Vaccines/administration & dosage , COVID-19/prevention & control , SARS-CoV-2/immunology , Administration, Intranasal , Animals , Antibodies, Viral/blood , COVID-19 Vaccines/genetics , COVID-19 Vaccines/immunology , Cricetinae , Genetic Vectors , Immunization , Parainfluenza Virus 3, Bovine/genetics , Parainfluenza Virus 3, Human/genetics , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
14.
Vet Microbiol ; 261: 109185, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34364015

ABSTRACT

Bovine parainfluenza-3 virus (BPIV-3) is one of the main viruses associated with bovine respiratory disease complex (BRDC) worldwide. BPIV-3 infect the bovine respiratory tract causing from subclinical infections to severe pneumonia with significant economic losses in the cattle industry. BPIV-3 is a RNA virus with high genetic variability, nevertheless, the contribution of recombination events to its variability has not been assessed so far. In this study the 25 complete genome sequences (CGS) reported so far and 215 partial sequences of different viral genes of BPIV-3 were analyzed to determine their genotypes and subgenotypes, distribution, and the existence of potential recombination events. Based on the analysis of the HN, M, N, and P genes one hypothetical subgenotype was found (subgenotype A4). Four recombination events between sequences of swine and cattle were detected by RDP4 analysis in conjunction with phylogenetic incongruences in the L gene. In addition, 9 sequences reported from Argentina were found to be miss-classified. These results reveal that homologous recombination events have a relevant role in the evolution of BPIV-3 and highlight the importance of implement advanced molecular characterization to better understand the variability and evolution of BPIV-3 as a component of BRDC.


Subject(s)
Genetic Variation/genetics , Homologous Recombination/genetics , Parainfluenza Virus 3, Bovine/genetics , Viral Proteins/genetics , Animals , Cattle , Cattle Diseases/virology , Genotype , Parainfluenza Virus 3, Bovine/classification , Phylogeny , Respirovirus Infections/virology , Sheep , Sheep Diseases/virology
15.
Viruses ; 13(6)2021 05 31.
Article in English | MEDLINE | ID: mdl-34072688

ABSTRACT

Bovine parainfluenza virus 3 (BPIV3) is a crucial causative agent of respiratory disease in young and adult cattle. No specific therapies are available for BPIV3 infection. Understanding the internalization pathway of the virus will provide a new strategy for the development of antiviral therapy. Here, the mechanism of BPIV3 entry into HeLa cells was analyzed using RNA silencing and pharmacological inhibitors. Treatment of HeLa cells with hypertonic medium prevented BPIV3 internalization. These results indicated that BPIV3 entered HeLa cells via receptor-mediated endocytosis. Moreover, removing cell membrane cholesterol through MßCD treatment hampered viral penetration but not viral replication. In addition, BPIV3 infection was inhibited by pretreatment with dynasore or chlorpromazine (CPZ) or knockdown of dynamin II or clathrin heavy chain. However, virus entry was unaffected by nystatin, EIPA, wortmannin, or cytochalasin D treatment or caveolin-1 knockdown. These data demonstrated that the entry of BPIV3 into HeLa cells was dependent on clathrin-mediated endocytosis but not on caveolae-mediated endocytosis or the macropinocytosis pathway. Many viruses are transported to endosomes, which provide an acidic environment and release their genome upon separation from primary endocytic vesicles. However, we found that BPIV3 infection required endosomal cathepsins, but not a low pH. In summary, we show, for the first time, that BPIV3 enters HeLa cells through the clathrin-mediated endocytosis pathway, presenting novel insights into the invasion mechanism of Paramyxoviridae.


Subject(s)
Cholesterol/metabolism , Clathrin/metabolism , Dynamins/metabolism , Endocytosis , Parainfluenza Virus 3, Bovine/physiology , Virus Internalization , Animals , Cattle , HeLa Cells , Humans
16.
Virology ; 561: 17-27, 2021 09.
Article in English | MEDLINE | ID: mdl-34130198

ABSTRACT

Paramyxovirus matrix (M) proteins are key drivers of virus particle assembly and budding at the plasma membrane. To identify regions important for the M protein function, we generated a series of deletion mutants of the bovine parainfluenza virus type 3 (BPIV3) M protein. We found that M proteins lacking 10 amino acids in the amino-terminal end (ΔN10) or 4 amino acids in the carboxyl-terminal end (ΔC4) did not support M-deficient BPIV3 virion release and M protein-induced virus-like particle (VLP) release. Both ΔN10 and ΔC4 retained M protein-M protein and M protein-nucleocapsid (N) protein interactions. However, neither was transported to the plasma membrane. Our results indicate that both amino- and carboxyl-terminal ends of the BPIV3 M protein are essential for M protein transport to the plasma membrane, where it facilitates virion and VLP release.


Subject(s)
Parainfluenza Virus 3, Bovine/physiology , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/metabolism , Virion/physiology , Virus Release , Animals , Cell Membrane/metabolism , Chlorocebus aethiops , Mutant Proteins/metabolism , Nucleocapsid Proteins/metabolism , Parainfluenza Virus 3, Bovine/chemistry , Protein Transport , Sequence Deletion , Vero Cells , Viral Matrix Proteins/genetics
17.
Vet Microbiol ; 259: 109148, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34147763

ABSTRACT

Bovine parainfluenza virus 3 (BPIV3) is an important respiratory pathogen of both young and adult cattle. No specific therapies are available for BPIV3. Understanding the viral internalization pathway of BPIV3 will provide new strategies for the development of antiviral treatments. Here, the entry mechanism of BPIV3 into MDBK cells was analyzed using chemical inhibitors and RNA silencing. Our data demonstrated that treatment with an inhibitor targeting the clathrin-mediated pathway or clathrin heavy chain (CHC) knockdown suppressed the entry of BPIV3 into MDBK cells. In contrast, sequestration of cellular cholesterol by nystatin or silencing of caveolin-1 had no effect on viral entry. Moreover, inhibition of critical modulators of macropinocytosis significantly reduced BPIV3 uptake. In addition, fluid-phase uptake was significantly increased in cells infected with BPIV3, which is indicative of virus-induced facilitation of macropinocytosis. These results suggest that BPIV3 enters MDBK cells via macropinocytosis and clathrin- but not caveolar-dependent endocytosis. Furthermore, inhibition of endosomal acidification and activation of cathepsin blocked BPIV3 entry, demonstrating that BPIV3 entered MDBK cells in a acid-dependent manner and required cathepsin L. Finally, we demonstrated that macropinocytosis but not clathrin-mediated endocytosis is dependent on actin dynamics during BPIV3 infection.


Subject(s)
Acids/metabolism , Clathrin Heavy Chains/genetics , Clathrin/metabolism , Endocytosis , Parainfluenza Virus 3, Bovine/physiology , Pinocytosis , Virus Internalization , Animals , Cathepsin L/metabolism , Cattle , Cell Line , Clathrin Heavy Chains/metabolism
18.
Vet Med Sci ; 7(5): 1625-1632, 2021 09.
Article in English | MEDLINE | ID: mdl-34031994

ABSTRACT

Bovine parainfluenza virus-3 (BPIV-3), also known as bovine respirovirus 3, causes serious respiratory infection in ungulates, often involving other pathogens, such as viruses, bacteria and mycoplasmas. In this study, we evaluated antibody titers against virus genotypes A (BPIV-3a) and C (BPIV-3c). We conducted a serological survey and comparison analysis of archived serum samples from small and large ruminants reared in four Turkish provinces. A total of 1,307 samples, consisting of sheep (n = 444), cattle (n = 402), water buffalo (n = 261) and goat (n = 200) sera, were randomly selected from stock samples collected between 2015 and 2019 and screened by standard virus neutralisation assay. We found that 49.9% (653/1307) of all samples were positive for neutralising antibody titers. Goats had the highest titer, with total seropositivity of 63% (126/200), followed in descending order by cattle, sheep and water buffalo at 56.2% (226/402), 32.2% (143/444) and 26% (68/261) total seropositivity, respectively. BPIV-3c had the highest neutralising antibody rate at 34.3% (448/1307), whereas BPIV-3a had a 24.3% (317/1307) seropositivity rate. Neutralising antibody titers for positive samples ranged between 1/4 and 1/512 per the SN50 test. Seropositivity rates ranged from a low of 8.9% to a high of 18.3%. Our study was the first to compare antibody seroprevalence for two BPIV-3 genotypes in small and large domestic ruminants, which were shown to be more commonly exposed to BPIV-3c than BPIV-3a. This finding could have significant implications as current vaccines mainly use the BPIV-3a genotype. Further research can determine if current vaccines protect against different BPIV-3 virus genotypes.


Subject(s)
Goats , Parainfluenza Virus 3, Bovine , Animals , Buffaloes , Cattle , Genotype , Parainfluenza Virus 3, Bovine/genetics , Seroepidemiologic Studies , Sheep
19.
Can J Vet Res ; 85(2): 101-105, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33883816

ABSTRACT

Abruptly weaned crossbred steer calves (N = 271) were used in a randomized, blinded 2-arm clinical trial to assess the impact of a long-acting non-steroidal anti-inflammatory drug on bovine herpesvirus type 1, bovine respiratory syncytial virus, parainfluenza virus type 3, and coronavirus titers and health outcomes when administered concurrently with a modified live respiratory vaccine upon arrival at a feedlot. Treatment groups included a control (saline; n = 135) and an experimental group (injectable meloxicam; n = 136). Viral antibody titers and body weight were measured on arrival, day 7, and day 21, along with a final weight on day 45. Body weight and antibody titers for all viruses increased over time (P < 0.001); however, there were no differences by treatment group or a significant group × time interaction when evaluated using repeated measures analysis of variance. Interestingly, the use of meloxicam was associated with increased treatment risk (P < 0.05). In conclusion, the administration of meloxicam may adversely affect health; however, a decreased vaccine response is likely not a contributing factor.


Des bouvillons croisés sevrés rapidement (N = 271) ont été utilisés dans un essai clinique randomisé en aveugle à deux bras pour évaluer l'impact d'un anti-inflammatoire non stéroïdien à action prolongée sur les titres du virus de la rhinotrachéite infectieuse bovine, du virus respiratoire syncytial bovin, du virus parainfluenza 3 et du coronavirus, et les résultats pour la santé lorsqu'administré en même temps qu'un vaccin vivant modifié respiratoire à l'arrivée dans un parc d'engraissement. Les groupes de traitement comprenaient un témoin (solution saline; n = 135) et un groupe expérimental (méloxicam injectable; n = 136). Les titres d'anticorps viraux et le poids corporel ont été mesurés à l'arrivée, au jour 7 et au jour 21, ainsi qu'un poids final au jour 45. Le poids corporel et les titres d'anticorps pour tous les virus ont augmenté avec le temps (P < 0,001); cependant, il n'y avait aucune différence selon le groupe de traitement ou une interaction groupe × temps significative lors de l'évaluation à l'aide de mesures répétées d'analyse de la variance. Fait intéressant, l'utilisation du méloxicam était associée à un risque de traitement accru (P < 0,05). En conclusion, l'administration de méloxicam peut nuire à la santé; cependant, une réponse vaccinale réduite n'est probablement pas un facteur contributif.(Traduit par Docteur Serge Messier).


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Meloxicam/administration & dosage , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antibodies, Viral/blood , Cattle , Coronavirus, Bovine/immunology , Herpesvirus 1, Bovine/immunology , Male , Meloxicam/pharmacology , Meloxicam/therapeutic use , Parainfluenza Virus 3, Bovine/immunology , Respiratory Syncytial Virus, Bovine/immunology , Weaning
20.
Vet Q ; 41(1): 97-106, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33375918

ABSTRACT

BACKGROUND: The knowledge on bovine vaccines against respiratory viruses on bronchoalveolar fluid cells is scarce. OBJECTIVE: To compare the effects of a commercial intranasal (IN) and intramuscular (IM) vaccine against bovine respiratory disease (BRD) complex viruses on bronchoalveolar fluid cells of healthy heifers. METHODS: 21 healthy heifers were assigned to three treatment groups: control (CO, N = 7), intranasally vaccinated (IN) (n = 7), and intramuscularly vaccinated (IM) (n = 7). The IN group received 1 mL of the commercial vaccine in each nostril once containing attenuated BoHV-1, bPIV-3, and BRSV. The IM group was vaccinated with two doses of 2 mL with an interval of 21 days of the commercial vaccine containing attenuated BoHV-1, bPIV-3, and BRSV plus inactivated BVDV. At day 0 (D0), before the first vaccine dose, and at D3, D7, and D21, after the last vaccine dose, airway bronchoscopy was performed to observe local irritation and collect bronchoalveolar lavage fluid (BALF). The bronchoalveolar count, cytological evaluation, bronchoalveolar cell oxidative metabolism, and total bronchoalveolar IgA and IgG were measured. RESULTS: The IN vaccine increased neutrophil cellularity at D7 and D21 and total IgA at D3 in BALF. Total IgA in BALF also increased at D3 and oxidative metabolism of bronchoalveolar cells at D21 lowered compared to the CO group. Following IM vaccination there was no alteration of immunoglobulins or cell oxidative metabolism in BALF. Both vaccines reduced the number of alveolar macrophages. CONCLUSION: Both vaccines induced bronchoalveolar inflammation during the establishment of the vaccine immunity, which was more expressive in the IN protocol.


Subject(s)
Bronchoalveolar Lavage Fluid/cytology , Cattle Diseases/prevention & control , Vaccination/veterinary , Administration, Intranasal/adverse effects , Administration, Intranasal/veterinary , Animals , Bovine Virus Diarrhea-Mucosal Disease/prevention & control , Bronchoalveolar Lavage Fluid/chemistry , Cattle , Cattle Diseases/virology , Diarrhea Viruses, Bovine Viral , Female , Herpesviridae Infections/prevention & control , Herpesvirus 1, Bovine , Immunoglobulin A , Immunoglobulin G , Injections, Intramuscular/adverse effects , Injections, Intramuscular/veterinary , Parainfluenza Virus 3, Bovine , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/veterinary , Respiratory Syncytial Virus, Bovine , Respirovirus Infections/prevention & control , Respirovirus Infections/veterinary , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/adverse effects , Viral Vaccines/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...