Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.513
Filter
1.
Can Vet J ; 65(8): 791-801, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39091482

ABSTRACT

Objective: This study compared clinical and immunological responses to coinfection challenge of beef calves mucosally primed and differentially boosted with commercial combination vaccines containing antigens against bovine coronavirus (BCoV), bovine parainfluenza virus Type 3 (BPIV3), and bovine respiratory syncytial virus (BRSV). Animals: Nineteen commercial beef heifers. Procedure: At birth, calves were mucosally (IN) primed with modified-live virus (MLV) vaccines, differentially boosted by injection of either combination MLV (IN-MLV) or inactivated virus (IN-KV) vaccines at a mean age of 44 d, and then challenged by coinfection with BCoV, BPIV3, and BRSV at weaning. Results: Both groups were similarly protected from clinical disease and had anamnestic neutralizing antibody responses to all 3 viruses. The IN-KV group shed more BCoV, and less BPIV3 and BRSV, than the IN-MLV group. Conclusion: These data indicated similar clinical and immunological protection between IN-MLV and IN-KV; however, shed of virus varied. Clinical relevance: Whereas boosting with KV or MLV appeared to have similar efficacy, viral shed differences may affect disease control.


Efficacité comparative des vaccins vivants modifiés et inactivés pour stimuler les réponses au virus respiratoire syncytial bovin, au virus parainfluenza bovin de type 3 et au coronavirus bovin après amorçage via la muqueuse de veaux de boucherie nouveau-nés. Objectif: Cette étude a comparé les réponses cliniques et immunologiques à une co-infection de veaux de boucherie amorcés par voie muqueuse et différentiellement stimulés avec des vaccins combinés commerciaux contenant des antigènes contre le coronavirus bovin (BCoV), le virus parainfluenza bovin de type 3 (BPIV3) et le virus respiratoire syncytial bovin (BRSV). Animaux: Dix-neuf génisses de boucherie commerciales. Procédure: À la naissance, les veaux ont été vaccinés au niveau des muqueuses (IN) avec des vaccins à virus vivants modifiés (MLV), stimulés de manière différentielle par l'injection de vaccins combinés MLV (IN-MLV) ou de virus inactivés (IN-KV) à un âge moyen de 44 jours. puis provoqué par une co-infection avec BCoV, BPIV3 et BRSV au sevrage. Résultats: Les deux groupes étaient protégés de la même manière contre la maladie clinique et présentaient des réponses anamnestiques en anticorps neutralisants contre les 3 virus. Le groupe IN-KV a excrété plus de BCoV et moins de BPIV3 et de BRSV que le groupe IN-MLV. Conclusion: Ces données indiquent une protection clinique et immunologique similaire entre IN-MLV et IN-KV; cependant, l'excrétion du virus variait. Pertinence clinique: Alors que le rappel avec KV ou MLV semble avoir une efficacité similaire, les différences d'excrétion virale peuvent affecter la limitation de la maladie.(Traduit par Dr Serge Messier).


Subject(s)
Animals, Newborn , Cattle Diseases , Coronavirus, Bovine , Parainfluenza Virus 3, Bovine , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Bovine , Vaccines, Inactivated , Viral Vaccines , Animals , Cattle , Coronavirus, Bovine/immunology , Parainfluenza Virus 3, Bovine/immunology , Respiratory Syncytial Virus, Bovine/immunology , Cattle Diseases/prevention & control , Cattle Diseases/virology , Cattle Diseases/immunology , Female , Vaccines, Inactivated/immunology , Vaccines, Inactivated/administration & dosage , Respiratory Syncytial Virus Infections/veterinary , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/immunology , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Animals, Newborn/immunology , Coronavirus Infections/veterinary , Coronavirus Infections/prevention & control , Coronavirus Infections/immunology , Coronavirus Infections/virology , Antibodies, Viral/blood , Vaccines, Attenuated/immunology , Vaccines, Attenuated/administration & dosage , Respirovirus Infections/veterinary , Respirovirus Infections/prevention & control , Respirovirus Infections/immunology , Immunization, Secondary/veterinary
2.
BMC Vet Res ; 20(1): 342, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095820

ABSTRACT

BACKGROUND: Porcine deltacoronavirus (PDCoV) is a swine enteropathogenic coronavirus that affects young pigs, causing vomiting, acute diarrhea, dehydration, and even death. There is growing evidence that PDCoV can undergo cross-species as well as zoonotic transmissions. Due to the frequent outbreaks of this deadly virus, early detection is essential for effective prevention and control. Therefore, developing a more convenient and reliable method for PDCoV detection is the need of the hour. RESULTS: This study utilized a high-affinity monoclonal antibody as the capture antibody and a horseradish peroxidase labeled polyclonal antibody as the detection antibody to develop an enzyme-linked immunosorbent assay (DAS-ELSA) for PDCoV detection.Both antibodies target the PDCoV nucleocapsid (N) protein. The findings of this study revealed that DAS-ELISA was highly specific to PDCoV and did not cross-react with other viruses to cause swine diarrhea. The limit of detection of the virus titer using this method was 103 TCID50/mL of PDCoV particles. The results of a parallel analysis of 239 known pig samples revealed a coincidence rate of 97.07% (κ = 0.922) using DAS-ELISA and reverse transcriptase PCR (RT-PCR). The DAS-ELISA was used to measure the one-step growth curve of PDCoV in LLC-PK cells and the tissue distribution of PDCoV in infected piglets. The study found that the DAS-ELISA was comparable in accuracy to the TCID50 method while measuring the one-step growth curve. Furthermore, the tissue distribution measured by DAS-ELISA was also consistent with the qRT-PCR method. CONCLUSION: The developed DAS-ELISA method can be conveniently used for the early clinical detection of PDCoV infection in pigs, and it may also serve as an alternative method for laboratory testing of PDCoV.


Subject(s)
Deltacoronavirus , Enzyme-Linked Immunosorbent Assay , Swine Diseases , Animals , Enzyme-Linked Immunosorbent Assay/veterinary , Enzyme-Linked Immunosorbent Assay/methods , Swine , Swine Diseases/virology , Swine Diseases/diagnosis , Swine Diseases/immunology , Deltacoronavirus/isolation & purification , Coronavirus Infections/veterinary , Coronavirus Infections/diagnosis , Coronavirus Infections/virology , Coronavirus Infections/immunology , Antibodies, Monoclonal/immunology , Sensitivity and Specificity , Antigens, Viral/analysis , Antigens, Viral/immunology , Antibodies, Viral/blood
3.
Front Immunol ; 15: 1442160, 2024.
Article in English | MEDLINE | ID: mdl-39100673

ABSTRACT

The COVID-19 pandemic has uncovered the high genetic variability of the SARS-CoV-2 virus and its ability to evade the immune responses that were induced by earlier viral variants. Only a few monoclonal antibodies that have been reported to date are capable of neutralizing a broad spectrum of SARS-CoV-2 variants. Here, we report the isolation of a new broadly neutralizing human monoclonal antibody, iC1. The antibody was identified through sorting the SARS-CoV-1 RBD-stained individual B cells that were isolated from the blood of a vaccinated donor following a breakthrough infection. In vitro, iC1 potently neutralizes pseudoviruses expressing a wide range of SARS-CoV-2 Spike variants, including those of the XBB sublineage. In an hACE2-transgenic mouse model, iC1 provided effective protection against the Wuhan strain of the virus as well as the BA.5 and XBB.1.5 variants. Therefore, iC1 can be considered as a potential component of the broadly neutralizing antibody cocktails resisting the SARS-CoV-2 mutation escape.


Subject(s)
Angiotensin-Converting Enzyme 2 , Antibodies, Monoclonal , Antibodies, Neutralizing , Antibodies, Viral , COVID-19 , Mice, Transgenic , SARS-CoV-2 , Animals , SARS-CoV-2/immunology , Humans , COVID-19/immunology , COVID-19/prevention & control , COVID-19/virology , Angiotensin-Converting Enzyme 2/immunology , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism , Mice , Antibodies, Viral/immunology , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Pandemics/prevention & control , Betacoronavirus/immunology , Betacoronavirus/genetics , Broadly Neutralizing Antibodies/immunology , Disease Models, Animal , Pneumonia, Viral/immunology , Pneumonia, Viral/virology , Pneumonia, Viral/prevention & control , Coronavirus Infections/immunology , Coronavirus Infections/virology , Coronavirus Infections/prevention & control
4.
Front Cell Infect Microbiol ; 14: 1422560, 2024.
Article in English | MEDLINE | ID: mdl-39104852

ABSTRACT

Porcine epidemic diarrhea virus (PEDV) is associated with severe enteritis, which contributes to high mortality in piglets. The aim of this study was to describe molecular mechanisms associated with proinflammatory cytokine(s) production during PEDV infection. We showed that infection of porcine intestine epithelial cell clone J2 (IPEC-J2) with PEDV induces a gradual increase in interleukin 8 (IL-8) production at different time points, as well as infection of Vero E6 with PEDV. The secretion of IL-8 in these two cell lines infected with PEDV is related to the activation of NF-κB. Furthermore, the cells expressing PEDV M or E protein can induce the upregulation of IL-8. These findings suggest that the IL-8 production can be the initiator of inflammatory response by the host cells upon PEDV infection.


Subject(s)
Interleukin-8 , NF-kappa B , Porcine epidemic diarrhea virus , Signal Transduction , Animals , NF-kappa B/metabolism , Swine , Interleukin-8/metabolism , Chlorocebus aethiops , Vero Cells , Cell Line , Swine Diseases/virology , Swine Diseases/metabolism , Epithelial Cells/metabolism , Epithelial Cells/virology , Coronavirus Infections/metabolism , Coronavirus Infections/virology , Coronavirus Infections/immunology
5.
Nat Microbiol ; 9(8): 2038-2050, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39075235

ABSTRACT

Circulating bat coronaviruses represent a pandemic threat. However, our understanding of bat coronavirus pathogenesis and transmission potential is limited by the lack of phenotypically characterized strains. We created molecular clones for the two closest known relatives of SARS-CoV-2, BANAL-52 and BANAL-236. We demonstrated that BANAL-CoVs and SARS-CoV-2 have similar replication kinetics in human bronchial epithelial cells. However, BANAL-CoVs have impaired replication in human nasal epithelial cells and in the upper airway of mice. We also observed reduced pathogenesis in mice and diminished transmission in hamsters. Further, we observed that diverse bat coronaviruses evade interferon and downregulate major histocompatibility complex class I. Collectively, our study demonstrates that despite high genetic similarity across bat coronaviruses, prediction of pandemic potential of a virus necessitates functional characterization. Finally, the restriction of bat coronavirus replication in the upper airway highlights that transmission potential and innate immune restriction can be uncoupled in this high-risk family of emerging viruses.


Subject(s)
COVID-19 , Chiroptera , Immunity, Innate , SARS-CoV-2 , Virus Replication , Animals , Humans , SARS-CoV-2/immunology , SARS-CoV-2/genetics , SARS-CoV-2/physiology , Chiroptera/virology , Chiroptera/immunology , COVID-19/transmission , COVID-19/virology , COVID-19/immunology , Mice , Cricetinae , Immune Evasion , Epithelial Cells/virology , Epithelial Cells/immunology , Coronavirus Infections/transmission , Coronavirus Infections/immunology , Coronavirus Infections/virology , Coronavirus/immunology , Coronavirus/genetics , Coronavirus/classification , Coronavirus/physiology , Coronavirus/pathogenicity , Cell Line , Female
6.
Sheng Wu Gong Cheng Xue Bao ; 40(7): 2150-2161, 2024 Jul 25.
Article in Chinese | MEDLINE | ID: mdl-39044581

ABSTRACT

This study aims to develop an effective bivalent subunit vaccine that is promising to prevent both porcine deltacoronavirus (PDCoV) and porcine epidemic diarrhea virus (PEDV). The receptor-binding domains (RBDs) of PDCoV and PEDV were fused and cloned into the eukaryotic expression vector pCDNA3.1(+). The fusion protein PDCoV-RBD-PEDV-RBD (pdRBD-peRBD) was expressed by the ExpiCHOTM expression system and purified. Mice were immunized with the fusion protein at three different doses (10, 20, and 30 µg). The humoral immune response and cellular immune response induced by the fusion protein were evaluated by ELISA and flow cytometry. The neutralization titers of the serum of immunized mice against PDCoV and PEDV were determined by the microneutralization test. The results showed that high levels of IgG antibodies were induced in the three different dose groups after booster immunization, and there was no significant difference in the antibody level between different dose groups, indicating that the immunization dose of 10 µg could achieve the fine immune effect. The results of flow cytometry showed that the immunization groups demonstrated increased proportion of CD3+CD4+ T cells and decreased proportion of CD3+CD8+ T cells, which was consistent with the expectation about the humoral immune response induced by the subunit vaccine. At the same time, the levels of interleukin (IL)-2, IL-4, and interferon (IFN)-γ in the serum were determined. The results showed that the fusion protein induced both humoral immune effect and cellular immune response. The results of the neutralization test showed that the antibody induced by 10 µg fusion protein neutralized both PDCoV and PEDV in vitro, with the titers of 1:179.25 and 1:141.21, respectively. The above results suggested that the pdRBD-peRBD could induce a high level of humoral immune response at a dose of 10 µg, and the induced antibody could neutralize both PDCoV and PEDV. Therefore, the fusion protein pdRBD-peRBD is expected to be an effective subunit vaccine that can simultaneously prevent PDCoV and PEDV.


Subject(s)
Antibodies, Viral , Coronavirus Infections , Porcine epidemic diarrhea virus , Recombinant Fusion Proteins , Viral Vaccines , Animals , Porcine epidemic diarrhea virus/immunology , Porcine epidemic diarrhea virus/genetics , Mice , Swine , Viral Vaccines/immunology , Viral Vaccines/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/genetics , Coronavirus Infections/prevention & control , Coronavirus Infections/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Deltacoronavirus/immunology , Deltacoronavirus/genetics , Swine Diseases/prevention & control , Swine Diseases/immunology , Vaccines, Subunit/immunology , Vaccines, Subunit/genetics , Mice, Inbred BALB C , Female , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Protein Domains , Immunogenicity, Vaccine , Immunity, Humoral
7.
Int J Mol Sci ; 25(14)2024 Jul 12.
Article in English | MEDLINE | ID: mdl-39062886

ABSTRACT

Porcine deltacoronavirus (PDCoV) is an emerging enteric pathogen that causes substantial economic losses in the swine industry worldwide. The PDCoV NS6 protein is an accessory protein that plays a pivotal role in the viral life cycle and immune evasion. However, the functions of NS6 and its role in PDCoV pathogenesis remain largely unknown. In this study, we prepared a monoclonal antibody (mAb) 5-A11 that specifically recognizes the PDCoV NS6 protein. The mAb 5-A11 exhibited high specificity for PDCoV, with no cross-reactivity with several major porcine pathogenic viruses. Furthermore, the epitope recognized by mAb 5-A11 was precisely mapped to residues 70EYGSIYGKDFI80 of the NS6 protein using Western blot analysis. Notably, this epitope is highly conserved among different PDCoV isolates. Substantial variations were observed when comparing this epitope with the corresponding regions in the NS6 proteins of other δ coronaviruses, suggesting potential differences in the structure, function, and antigenicity of their NS6 proteins. Our findings provide valuable tools and insights for further elucidating the functions of the NS6 protein and its role in PDCoV pathogenesis, as well as for developing diagnostic and therapeutic strategies against PDCoV infection.


Subject(s)
Antibodies, Monoclonal , Deltacoronavirus , Epitopes , Viral Nonstructural Proteins , Animals , Antibodies, Monoclonal/immunology , Swine , Deltacoronavirus/immunology , Viral Nonstructural Proteins/immunology , Viral Nonstructural Proteins/metabolism , Epitopes/immunology , Epitope Mapping , Coronavirus Infections/immunology , Coronavirus Infections/virology , Antibodies, Viral/immunology , Swine Diseases/virology , Swine Diseases/immunology , Mice , Amino Acid Sequence , Mice, Inbred BALB C
8.
Immun Inflamm Dis ; 12(7): e1342, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39023424

ABSTRACT

BACKGROUND: Epidemiological data on seasonal coronaviruses (sCoVs) may provide insight on transmission patterns and demographic factors that favor coronaviruses (CoVs) with greater disease severity. This study describes the incidence of CoVs in several high-risk groups in Ottawa, Canada, from October 2020 to March 2022. METHODS: Serological assays quantified IgG and IgM antibodies to SARS-CoV-2, HCoV-OC43, HCoV-NL63, HCoV-HKU1, and HCoV-229E. Incident infections were compared between four population groups: individuals exposed to children, transit users, immunocompromised, and controls. Associations between antibody prevalence indicative of natural infection and demographic variables were assessed using regression analyses. RESULTS: Transit users and those exposed to children were at no greater risk of infection compared to the control group. Fewer infections were detected in the immunocompromised group (p = .03). SARS-CoV-2 seroprevalence was greater in individuals with low income and within ethnic minorities. CONCLUSIONS: Our findings suggest that nonpharmaceutical interventions intended to reduce SAR-CoV-2 transmission protected populations at high risk of exposure. The re-emergence of sCoVs and other common respiratory viruses alongside SARS-CoV-2 may alter infection patterns and increase the risk in vulnerable populations.


Subject(s)
COVID-19 , SARS-CoV-2 , Seasons , Humans , COVID-19/epidemiology , COVID-19/transmission , COVID-19/immunology , Incidence , Male , Female , SARS-CoV-2/immunology , Child , Adult , Middle Aged , Adolescent , Antibodies, Viral/blood , Child, Preschool , Seroepidemiologic Studies , Young Adult , Aged , Risk Factors , Canada/epidemiology , Infant , Pandemics , Immunoglobulin G/blood , Coronavirus Infections/epidemiology , Coronavirus Infections/transmission , Coronavirus Infections/immunology , Immunoglobulin M/blood
9.
BMC Vet Res ; 20(1): 336, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39080763

ABSTRACT

BACKGROUND: Porcine epidemic diarrhea virus (PEDV) is a highly contagious coronavirus that causes severe diarrhea and death in neonatal piglets, which has brought huge economic losses to the pork industry worldwide since its first discovery in the early 1970s in Europe. Passive immunization with neutralizing antibodies against PEDV is an effective prevention measure. To date, there are no effective therapeutic drugs to treat the PEDV infection. RESULTS: We conducted a screening of specific nanobodies against the S1 protein from a phage display library obtained from immunized alpacas. Through competitive binding to antigenic epitopes, we selected instead of chose nanobodies with high affinity and constructed a multivalent tandem. These nanobodies were shown to inhibit PEDV infectivity by the neutralization assay. The antiviral capacity of nanobody was found to display a dose-dependent pattern, as demonstrated by IFA, TCID50, and qRT-PCR analyses. Notably, biparatopic nanobody SF-B exhibited superior antiviral activity. Nanobodies exhibited low cytotoxicity and high stability even under harsh temperature and pH conditions, demonstrating their potential practical applicability to animals. CONCLUSIONS: Nanobodies exhibit remarkable biological properties and antiviral effects, rendering them a promising candidate for the development of anti-PEDV drugs.


Subject(s)
Antibodies, Neutralizing , Coronavirus Infections , Porcine epidemic diarrhea virus , Single-Domain Antibodies , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Camelids, New World/immunology , Chlorocebus aethiops , Coronavirus Infections/immunology , Coronavirus Infections/prevention & control , Coronavirus Infections/veterinary , Porcine epidemic diarrhea virus/immunology , Single-Domain Antibodies/immunology , Single-Domain Antibodies/pharmacology , Spike Glycoprotein, Coronavirus/immunology , Swine , Swine Diseases/virology , Swine Diseases/immunology , Swine Diseases/prevention & control , Vero Cells
10.
EMBO Rep ; 25(8): 3547-3573, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39009832

ABSTRACT

The COVID-19 pandemic reminded us of the urgent need for new antivirals to control emerging infectious diseases and potential future pandemics. Immunotherapy has revolutionized oncology and could complement the use of antivirals, but its application to infectious diseases remains largely unexplored. Nucleoside analogs are a class of agents widely used as antiviral and anti-neoplastic drugs. Their antiviral activity is generally based on interference with viral nucleic acid replication or transcription. Based on our previous work and computer modeling, we hypothesize that antiviral adenosine analogs, like remdesivir, have previously unrecognized immunomodulatory properties which contribute to their therapeutic activity. In the case of remdesivir, we here show that these properties are due to its metabolite, GS-441524, acting as an Adenosine A2A Receptor antagonist. Our findings support a new rationale for the design of next-generation antiviral agents with dual - immunomodulatory and intrinsic - antiviral properties. These compounds could represent game-changing therapies to control emerging viral diseases and future pandemics.


Subject(s)
Adenosine Monophosphate , Adenosine , Alanine , Antiviral Agents , COVID-19 , SARS-CoV-2 , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , SARS-CoV-2/drug effects , SARS-CoV-2/immunology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine/chemistry , Humans , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Alanine/analogs & derivatives , Alanine/pharmacology , Alanine/chemistry , COVID-19/immunology , COVID-19/virology , Animals , Immunomodulating Agents/pharmacology , Immunomodulating Agents/chemistry , Adenosine A2 Receptor Antagonists/pharmacology , Adenosine A2 Receptor Antagonists/chemistry , Adenosine A2 Receptor Antagonists/therapeutic use , Pandemics , COVID-19 Drug Treatment , Chlorocebus aethiops , Virus Replication/drug effects , Vero Cells , Betacoronavirus/drug effects , Betacoronavirus/immunology , Receptor, Adenosine A2A/metabolism , Coronavirus Infections/drug therapy , Coronavirus Infections/immunology , Coronavirus Infections/virology
11.
Zool Res ; 45(4): 747-766, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-38894519

ABSTRACT

The distribution of the immune system throughout the body complicates in vitro assessments of coronavirus disease 2019 (COVID-19) immunobiology, often resulting in a lack of reproducibility when extrapolated to the whole organism. Consequently, developing animal models is imperative for a comprehensive understanding of the pathology and immunology of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. This review summarizes current progress related to COVID-19 animal models, including non-human primates (NHPs), mice, and hamsters, with a focus on their roles in exploring the mechanisms of immunopathology, immune protection, and long-term effects of SARS-CoV-2 infection, as well as their application in immunoprevention and immunotherapy of SARS-CoV-2 infection. Differences among these animal models and their specific applications are also highlighted, as no single model can fully encapsulate all aspects of COVID-19. To effectively address the challenges posed by COVID-19, it is essential to select appropriate animal models that can accurately replicate both fatal and non-fatal infections with varying courses and severities. Optimizing animal model libraries and associated research tools is key to resolving the global COVID-19 pandemic, serving as a robust resource for future emerging infectious diseases.


Subject(s)
COVID-19 , Disease Models, Animal , Pandemics , SARS-CoV-2 , Animals , COVID-19/immunology , COVID-19/therapy , SARS-CoV-2/immunology , Humans , Mice , Pneumonia, Viral/immunology , Pneumonia, Viral/virology , Pneumonia, Viral/therapy , Coronavirus Infections/immunology , Coronavirus Infections/virology , Coronavirus Infections/therapy , Betacoronavirus/immunology , Cricetinae
12.
BMC Vet Res ; 20(1): 239, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38831363

ABSTRACT

The porcine epidemic diarrhea virus (PEDV) infection inflicted substantial economic losses upon the global pig-breeding industry. This pathogen can infect all pigs and poses a particularly high fatality risk for suckling piglets. The S1 subunit of spike protein is a crucial target protein for inducing the particularly neutralizing antibodies that can intercept the virus-host interaction and neutralize virus infectivity. In the present study, the HEK293F eukaryotic expression system was successfully utilized to express and produce recombinant S1 protein. Through quantitative analysis, five monoclonal antibodies (mAbs) specifically targeting the recombinant S1 protein of PEDV were developed and subsequently evaluated using enzyme-linked immunosorbent assay (ELISA), indirect immunofluorescence assay (IFA), and flow cytometry assay (FCA). The results indicate that all five mAbs belong to the IgG1 isotype, and their half-maximal effective concentration (EC50) values measured at 84.77, 7.42, 0.89, 14.64, and 7.86 pM. All these five mAbs can be utilized in ELISA, FCA, and IFA for the detection of PEDV infection. MAb 5-F9 exhibits the highest sensitivity to detect as low as 0.3125 ng/mL of recombinant PEDV-S1 protein in ELISA, while only 0.096 ng/mL of mAb 5-F9 is required to detect PEDV in FCA. The results from antigen epitope analysis indicated that mAb 8-G2 is the sole antibody capable of recognizing linear epitopes. In conclusion, this study has yielded a highly immunogenic S1 protein and five high-affinity mAbs specifically targeting the S1 protein. These findings have significant implications for early detection of PEDV infection and provide a solid foundation for further investigation into studying virus-host interactions.


Subject(s)
Antibodies, Monoclonal , Coronavirus Infections , Enzyme-Linked Immunosorbent Assay , Porcine epidemic diarrhea virus , Spike Glycoprotein, Coronavirus , Porcine epidemic diarrhea virus/immunology , Antibodies, Monoclonal/immunology , Animals , Spike Glycoprotein, Coronavirus/immunology , Swine , Coronavirus Infections/veterinary , Coronavirus Infections/immunology , Coronavirus Infections/virology , Enzyme-Linked Immunosorbent Assay/veterinary , Antibodies, Viral/immunology , Swine Diseases/virology , Swine Diseases/immunology , HEK293 Cells , Humans , Recombinant Proteins/immunology , Mice, Inbred BALB C , Mice , Fluorescent Antibody Technique, Indirect/veterinary
13.
Vet Res ; 55(1): 74, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38863015

ABSTRACT

Bovine coronavirus (BCoV) is a pneumoenteric virus that can infect the digestive and respiratory tracts of cattle, resulting in economic losses. Despite its significance, information regarding BCoV pathogenesis is limited. Hence, we investigated clinical signs, patterns of viral shedding, changes in antibody abundance, and cytokine/chemokine production in calves inoculated with BCoV via intranasal and oral. Six clinically healthy Korean native calves (< 30 days old), initially negative for BCoV, were divided into intranasal and oral groups and monitored for 15 days post-infection (dpi). BCoV-infected calves exhibited clinical signs such as nasal discharge and diarrhea, starting at 3 dpi and recovering by 12 dpi, with nasal discharge being the most common symptoms. Viral RNA was detected in nasal and fecal samples from all infected calves. Nasal shedding occurred before fecal shedding regardless of the inoculation route; however, fecal shedding persisted longer. Although the number of partitions was very few, viral RNA was identified in the blood of two calves in the oral group at 7 dpi and 9 dpi using digital RT-PCR analysis. The effectiveness of maternal antibodies in preventing viral replication and shedding appeared limited. Our results showed interleukin (IL)-8 as the most common and highly induced chemokine. During BCoV infection, the levels of IL-8, monocyte chemoattractant protein-1, and macrophage inflammatory protein-1ß were significantly affected, suggesting that these emerge as potential and reliable biomarkers for predicting BCoV infection. This study underscores the importance of BCoV as a major pathogen causing diarrhea and respiratory disease.


Subject(s)
Cattle Diseases , Coronavirus Infections , Coronavirus, Bovine , Virus Shedding , Animals , Cattle , Cattle Diseases/virology , Cattle Diseases/immunology , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Coronavirus Infections/immunology , Republic of Korea , Feces/virology , RNA, Viral/analysis , Antibodies, Viral/blood , Cytokines/metabolism , Cytokines/genetics , Male
14.
J Neuroinflammation ; 21(1): 157, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38879499

ABSTRACT

BACKGROUND: Cystatin F is a secreted lysosomal cysteine protease inhibitor that has been implicated in affecting the severity of demyelination and enhancing remyelination in pre-clinical models of immune-mediated demyelination. How cystatin F impacts neurologic disease severity following viral infection of the central nervous system (CNS) has not been well characterized and was the focus of this study. We used cystatin F null-mutant mice (Cst7-/-) with a well-established model of murine coronavirus-induced neurologic disease to evaluate the contributions of cystatin F in host defense, demyelination and remyelination. METHODS: Wildtype controls and Cst7-/- mice were intracranially (i.c.) infected with a sublethal dose of the neurotropic JHM strain of mouse hepatitis virus (JHMV), with disease progression and survival monitored daily. Viral plaque assays and qPCR were used to assess viral levels in CNS. Immune cell infiltration into the CNS and immune cell activation were determined by flow cytometry and 10X genomics chromium 3' single cell RNA sequencing (scRNA-seq). Spinal cord demyelination was determined by luxol fast blue (LFB) and Hematoxylin/Eosin (H&E) staining and axonal damage assessed by immunohistochemical staining for SMI-32. Remyelination was evaluated by electron microscopy (EM) and calculation of g-ratios. RESULTS: JHMV-infected Cst7-/- mice were able to control viral replication within the CNS, indicating that cystatin F is not essential for an effective Th1 anti-viral immune response. Infiltration of T cells into the spinal cords of JHMV-infected Cst7-/- mice was increased compared to infected controls, and this correlated with increased axonal damage and demyelination associated with impaired remyelination. Single-cell RNA-seq of CD45 + cells enriched from spinal cords of infected Cst7-/- and control mice revealed enhanced expression of transcripts encoding T cell chemoattractants, Cxcl9 and Cxcl10, combined with elevated expression of interferon-g (Ifng) and perforin (Prf1) transcripts in CD8 + T cells from Cst7-/- mice compared to controls. CONCLUSIONS: Cystatin F is not required for immune-mediated control of JHMV replication within the CNS. However, JHMV-infected Cst7-/- mice exhibited more severe clinical disease associated with increased demyelination and impaired remyelination. The increase in disease severity was associated with elevated expression of T cell chemoattractant chemokines, concurrent with increased neuroinflammation. These findings support the idea that cystatin F influences expression of proinflammatory gene expression impacting neuroinflammation, T cell activation and/or glia cell responses ultimately impacting neuroinflammation and neurologic disease.


Subject(s)
Coronavirus Infections , Cystatins , Demyelinating Diseases , Mice, Knockout , Murine hepatitis virus , Animals , Mice , Demyelinating Diseases/pathology , Demyelinating Diseases/metabolism , Demyelinating Diseases/virology , Demyelinating Diseases/immunology , Murine hepatitis virus/pathogenicity , Cystatins/genetics , Cystatins/metabolism , Coronavirus Infections/immunology , Coronavirus Infections/pathology , Mice, Inbred C57BL , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/metabolism
15.
Vet Immunol Immunopathol ; 273: 110791, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38824909

ABSTRACT

Infectious bronchitis virus (IBV) strains of the Delmarva (DMV)/1639 genotype have been causing false layer syndrome (FLS) in the Eastern Canadian layer operations since the end of 2015. FLS is characterized by the development of cystic oviducts in layer pullets infected at an early age. Currently, there are no homologous vaccines for the control of this IBV genotype. Our previous research showed that a heterologous vaccination regimen incorporating Massachusetts (Mass) and Connecticut (Conn) IBV types protects layers against DMV/1639 genotype IBV. The aim of this study was to investigate the role of maternal antibodies conferred by breeders received the same vaccination regimen in the protection against the development of DMV/1639-induced FLS in pullets. Maternal antibody-positive (MA+) and maternal antibody-negative (MA-) female progeny chicks were challenged at 1 day of age and kept under observation for 16 weeks. Oviductal cystic formations were observed in 3 of 14 birds (21.4 %) in the MA- pullets, while the lesions were notably absent in the MA+ pullets. Milder histopathological lesions were observed in the examined tissues of the MA+ pullets. However, the maternal derived immunity failed to demonstrate protection against the damage to the tracheal ciliary activity, viral shedding, and viral tissue distribution. Overall, this study underscores the limitations of maternal derived immunity in preventing certain aspects of viral pathogenesis, emphasizing the need for comprehensive strategies to address different aspects of IBV infection.


Subject(s)
Antibodies, Viral , Chickens , Coronavirus Infections , Infectious bronchitis virus , Poultry Diseases , Viral Vaccines , Animals , Infectious bronchitis virus/immunology , Poultry Diseases/prevention & control , Poultry Diseases/immunology , Poultry Diseases/virology , Chickens/immunology , Chickens/virology , Female , Antibodies, Viral/blood , Antibodies, Viral/immunology , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Coronavirus Infections/prevention & control , Coronavirus Infections/veterinary , Coronavirus Infections/immunology , Coronavirus Infections/virology , Immunity, Maternally-Acquired , Trachea/immunology , Trachea/virology , Oviducts/immunology , Oviducts/pathology , Oviducts/virology
16.
Nat Commun ; 15(1): 5330, 2024 Jun 22.
Article in English | MEDLINE | ID: mdl-38909062

ABSTRACT

Porcine deltacoronavirus (PDCoV) is an emerging enteric pathogen that has recently been detected in humans. Despite this zoonotic concern, the antigenic structure of PDCoV remains unknown. The virus relies on its spike (S) protein for cell entry, making it a prime target for neutralizing antibodies. Here, we generate and characterize a set of neutralizing antibodies targeting the S protein, shedding light on PDCoV S interdomain crosstalk and its vulnerable sites. Among the four identified antibodies, one targets the S1A domain, causing local and long-range conformational changes, resulting in partial exposure of the S1B domain. The other antibodies bind the S1B domain, disrupting binding to aminopeptidase N (APN), the entry receptor for PDCoV. Notably, the epitopes of these S1B-targeting antibodies are concealed in the prefusion S trimer conformation, highlighting the necessity for conformational changes for effective antibody binding. The binding footprint of one S1B binder entirely overlaps with APN-interacting residues and thus targets a highly conserved epitope. These findings provide structural insights into the humoral immune response against the PDCoV S protein, potentially guiding vaccine and therapeutic development for this zoonotic pathogen.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Deltacoronavirus , Epitopes , Spike Glycoprotein, Coronavirus , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/chemistry , Animals , Antibodies, Neutralizing/immunology , Swine , Antibodies, Viral/immunology , Epitopes/immunology , Humans , Deltacoronavirus/immunology , Deltacoronavirus/metabolism , CD13 Antigens/metabolism , CD13 Antigens/immunology , Coronavirus Infections/immunology , Coronavirus Infections/virology , Protein Domains , Protein Binding , Swine Diseases/virology , Swine Diseases/immunology , HEK293 Cells
17.
Viruses ; 16(6)2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38932231

ABSTRACT

Porcine hemagglutinating encephalomyelitis virus (PHEV) replicates in the upper respiratory tract and tonsils of pigs. Using an air-liquid interface porcine respiratory epithelial cells (ALI-PRECs) culture system, we demonstrated that PHEV disrupts respiratory epithelia homeostasis by impairing ciliary function and inducing antiviral, pro-inflammatory cytokine, and chemokine responses. This study explores the mechanisms driving early innate immune responses during PHEV infection through host transcriptome analysis. Total RNA was collected from ALI-PRECs at 24, 36, and 48 h post inoculation (hpi). RNA-seq analysis was performed using an Illumina Hiseq 600 to generate 100 bp paired-end reads. Differential gene expression was analyzed using DeSeq2. PHEV replicated actively in ALI-PRECs, causing cytopathic changes and progressive mucociliary disruption. Transcriptome analysis revealed downregulation of cilia-associated genes such as CILK1, DNAH11, LRRC-23, -49, and -51, and acidic sialomucin CD164L2. PHEV also activated antiviral signaling pathways, significantly increasing the expression of interferon-stimulated genes (RSAD2, MX1, IFIT, and ISG15) and chemokine genes (CCL5 and CXCL10), highlighting inflammatory regulation. This study contributes to elucidating the molecular mechanisms of the innate immune response to PHEV infection of the airway epithelium, emphasizing the critical roles of the mucociliary, interferon, and chemokine responses.


Subject(s)
Betacoronavirus 1 , Epithelial Cells , Gene Expression Profiling , Interferons , Animals , Swine , Epithelial Cells/virology , Epithelial Cells/immunology , Interferons/genetics , Interferons/metabolism , Interferons/immunology , Betacoronavirus 1/immunology , Betacoronavirus 1/genetics , Immunity, Innate , Virus Replication , Coronavirus Infections/immunology , Coronavirus Infections/virology , Coronavirus Infections/veterinary , Cytokines/metabolism , Cytokines/genetics , Cytokines/immunology , Transcriptome , Respiratory Mucosa/virology , Respiratory Mucosa/immunology , Swine Diseases/virology , Swine Diseases/immunology , Swine Diseases/genetics , Cells, Cultured , Deltacoronavirus
18.
Open Vet J ; 14(5): 1224-1242, 2024 May.
Article in English | MEDLINE | ID: mdl-38938443

ABSTRACT

Background: Porcine epidemic diarrhea (PED), caused by the porcine epidemic diarrhea virus (PEDV), is associated with high mortality and morbidity rates, especially in neonatal pigs. This has resulted in significant economic losses for the pig industry. PEDV genotype II-based vaccines were found to confer better immunity against both heterologous and homologous challenges; specifically, spike (S) proteins, which are known to play a significant role during infection, are ideal for vaccine development. Aim: This study aims to design a multi-epitope subunit vaccine targeting the S protein of the PEDV GIIa strain using an immunoinformatics approach. Methods: Various bioinformatics tools were used to predict HTL, CTL, and B-cell epitopes. The epitopes were connected using appropriate linkers and conjugated with the CTB adjuvant and M-ligand. The final multiepitope vaccine construct (fMEVc) was then docked to toll-like receptor 4 (TLR4). The stability of the fMEVc-TLR4 complex was then simulated using GROMACS. C-immsim was then used to predict the in vitro immune response of the fMEVc. Results: Six epitopes were predicted to induce antibody production, ten epitopes were predicted to induce CTL responses, and four epitopes were predicted to induce HTL responses. The assembled epitopes conjugated with the CTB adjuvant and M-ligand, fMEVc, is antigenic, non-allergenic, stable, and soluble. The construct showed a favorable binding affinity for TLR4, and the protein complex was shown to be stable through molecular dynamics simulations. A robust immune response was induced after immunization, as demonstrated through immune stimulation. Conclusion: In conclusion, the multi-epitope subunit vaccine construct for PEDV designed in this study exhibits promising antigenicity, stability, and immunogenicity, eliciting robust immune responses and suggesting its potential as a candidate for further vaccine development.


Subject(s)
Computational Biology , Coronavirus Infections , Porcine epidemic diarrhea virus , Spike Glycoprotein, Coronavirus , Swine Diseases , Vaccines, Subunit , Viral Vaccines , Animals , Porcine epidemic diarrhea virus/immunology , Vaccines, Subunit/immunology , Swine , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Coronavirus Infections/prevention & control , Coronavirus Infections/veterinary , Coronavirus Infections/immunology , Coronavirus Infections/virology , Viral Vaccines/immunology , Swine Diseases/prevention & control , Swine Diseases/immunology , Swine Diseases/virology , Genotype , Epitopes/immunology , Epitopes, T-Lymphocyte/immunology , Epitopes, B-Lymphocyte/immunology , Molecular Docking Simulation , Immunoinformatics
19.
Vet Immunol Immunopathol ; 274: 110802, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38924873

ABSTRACT

Controlling pathogenic infections while reducing antibiotic usage is an important challenge during poultry production. In addition to vaccination strategies, several solutions to enhance the immune response against pathogens are evaluated. In this study, we aim to determine the effects of the glycerides of lauric acid (GLA) supplementation in chickens' diets on humoral and cellular immune response pathogenic infections, using an in vivo model of infectious bronchitis virus (IBV). One-day-old Ross 308 broilers were vaccinated with live attenuated IBV and fed diets supplemented with or without GLA at 3 kg/ton. The levels of early (day 7) specific anti-IBV in sera were significantly increased in broilers fed GLA, compared to the control groups (P<0.05), showing a stronger primary humoral response. The secretion levels of main cytokines remained similar in spleens of all the experimental groups. However, the splenocytes from broilers fed GLA showed higher activation and effector abilities when measured by IFN-γ ELISpot in presence of N-261-280 IBV peptide or Concanavalin A (Con A), a pan T lymphocytes mitogen. In response to N-261-280 peptide, GLA group showed a 2-fold increase of spot numbers (P < 0.05) and 3-fold increase of spot surfaces (P < 0.01) compared to the control groups. Similarly, Con A stimulation showed a 2-fold increases in spot surfaces and numbers in the GLA supplemented group compared to the control group (P < 0.01). In summary, GLA supplementation in chicken feed enhances the primary humoral immune response and strengthen the T lymphocytes mediated cellular immune response. These findings demonstrate how GLA can improve chicken resilience against pathogenic challenges by enhancing their immune responses.


Subject(s)
Chickens , Coronavirus Infections , Dietary Supplements , Immunity, Cellular , Immunity, Humoral , Infectious bronchitis virus , Lauric Acids , Poultry Diseases , Animals , Chickens/immunology , Infectious bronchitis virus/immunology , Immunity, Humoral/drug effects , Immunity, Cellular/drug effects , Poultry Diseases/immunology , Poultry Diseases/prevention & control , Poultry Diseases/virology , Coronavirus Infections/veterinary , Coronavirus Infections/immunology , Coronavirus Infections/prevention & control , Lauric Acids/pharmacology , Lauric Acids/administration & dosage , Glycerides/pharmacology , Animal Feed/analysis , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Antibodies, Viral/blood , Diet/veterinary , Cytokines/blood
20.
Front Immunol ; 15: 1381026, 2024.
Article in English | MEDLINE | ID: mdl-38919620

ABSTRACT

Introduction: Porcine deltacoronavirus (PDCoV) is a zoonotic pathogen with a global distribution, capable of infecting both pigs and humans. To mitigate the risk of cross-species transmission and potential outbreaks, it is crucial to characterize novel antiviral genes, particularly those from human hosts. Methods: This research used HIEC-6 to investigate PDCoV infection. HIEC-6 cells were infected with PDCoV. Samples were collected 48 h postinfection for proteomic analysis. Results: We discovered differential expression of MRPS6 gene at 48 h postinfection with PDCoV in HIEC-6 cells. The gene expression initially increased but then decreased. To further explore the role of MRPS6 in PDCoV infection, we conducted experiments involving the overexpression and knockdown of this gene in HIEC-6 and Caco2 cells, respectively. Our findings revealed that overexpression of MRPS6 significantly inhibited PDCoV infection in HIEC-6 cells, while knockdown of MRPS6 in Caco2 cells led to a significant increase of virus titer. Furthermore, we investigated the correlation between PDCoV infection and the expression of MRPS6. Subsequent investigations demonstrated that MRPS6 exerted an augmentative effect on the production of IFN-ß through interferon pathway activation, consequently impeding the progression of PDCoV infection in cellular systems. In conclusion, this study utilized proteomic analysis to investigate the differential protein expression in PDCoV-infected HIEC-6 cells, providing evidence for the first time that the MRPS6 gene plays a restrictive role in PDCoV virus infection. Discussion: Our findings initially provide the validation of MRPS6 as an upstream component of IFN-ß pathway, in the promotion of IRF3, IRF7, STAT1, STAT2 and IFN-ß production of HIEC-6 via dual-activation from interferon pathway.


Subject(s)
Deltacoronavirus , Humans , Animals , Swine , Deltacoronavirus/physiology , Deltacoronavirus/genetics , Caco-2 Cells , Coronavirus Infections/virology , Coronavirus Infections/immunology , Cell Line , Host-Pathogen Interactions/immunology , Proteomics/methods , Signal Transduction , Swine Diseases/virology , Swine Diseases/immunology
SELECTION OF CITATIONS
SEARCH DETAIL