Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.954
Filter
1.
Proc Natl Acad Sci U S A ; 121(21): e2402540121, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38758698

ABSTRACT

All respiratory viruses establish primary infections in the nasal epithelium, where efficient innate immune induction may prevent dissemination to the lower airway and thus minimize pathogenesis. Human coronaviruses (HCoVs) cause a range of pathologies, but the host and viral determinants of disease during common cold versus lethal HCoV infections are poorly understood. We model the initial site of infection using primary nasal epithelial cells cultured at an air-liquid interface (ALI). HCoV-229E, HCoV-NL63, and human rhinovirus-16 are common cold-associated viruses that exhibit unique features in this model: early induction of antiviral interferon (IFN) signaling, IFN-mediated viral clearance, and preferential replication at nasal airway temperature (33 °C) which confers muted host IFN responses. In contrast, lethal SARS-CoV-2 and MERS-CoV encode antagonist proteins that prevent IFN-mediated clearance in nasal cultures. Our study identifies features shared among common cold-associated viruses, highlighting nasal innate immune responses as predictive of infection outcomes and nasally directed IFNs as potential therapeutics.


Subject(s)
Common Cold , Immunity, Innate , Interferons , Nasal Mucosa , SARS-CoV-2 , Signal Transduction , Humans , Nasal Mucosa/virology , Nasal Mucosa/immunology , Nasal Mucosa/metabolism , Interferons/metabolism , Interferons/immunology , Common Cold/immunology , Common Cold/virology , Signal Transduction/immunology , SARS-CoV-2/immunology , Virus Replication , Rhinovirus/immunology , Coronavirus 229E, Human/immunology , Coronavirus Infections/immunology , Coronavirus Infections/virology , Epithelial Cells/virology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Middle East Respiratory Syndrome Coronavirus/immunology , Coronavirus NL63, Human/immunology
2.
Front Cell Infect Microbiol ; 14: 1378804, 2024.
Article in English | MEDLINE | ID: mdl-38736749

ABSTRACT

Introduction: Seasonal human coronavirus NL63 (HCoV-NL63) is a frequently encountered virus linked to mild upper respiratory infections. However, its potential to cause more severe or widespread disease remains an area of concern. This study aimed to investigate a rare localized epidemic of HCoV-NL63-induced respiratory infections among pediatric patients in Guilin, China, and to understand the viral subtype distribution and genetic characteristics. Methods: In this study, 83 pediatric patients hospitalized with acute respiratory infections and positive for HCoV-NL63 were enrolled. Molecular analysis was conducted to identify the viral subgenotypes and to assess genetic variations in the receptor-binding domain of the spiking protein. Results: Among the 83 HCoV-NL63-positive children, three subgenotypes were identified: C4, C3, and B. Notably, 21 cases exhibited a previously unreported subtype, C4. Analysis of the C4 subtype revealed a unique amino acid mutation (I507L) in the receptor-binding domain of the spiking protein, which was also observed in the previously reported C3 genotype. This mutation may suggest potential increases in viral transmissibility and pathogenicity. Discussion: The findings of this study highlight the rapid mutation dynamics of HCoV-NL63 and its potential for increased virulence and epidemic transmission. The presence of a unique mutation in the C4 subtype, shared with the C3 genotype, raises concerns about the virus's evolving nature and its potential public health implications. This research contributes valuable insights into the understanding of HCoV-NL63's epidemiology and pathogenesis, which is crucial for effective disease prevention and control strategies. Future studies are needed to further investigate the biological significance of the observed mutation and its potential impact on the virus's transmissibility and pathogenicity.


Subject(s)
Coronavirus Infections , Coronavirus NL63, Human , Epidemics , Genotype , Phylogeny , Respiratory Tract Infections , Humans , Coronavirus NL63, Human/genetics , China/epidemiology , Coronavirus Infections/epidemiology , Coronavirus Infections/virology , Coronavirus Infections/transmission , Child , Female , Male , Child, Preschool , Respiratory Tract Infections/virology , Respiratory Tract Infections/epidemiology , Infant , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/metabolism , Seasons , Mutation , Adolescent
3.
BMC Vet Res ; 20(1): 187, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38730463

ABSTRACT

BACKGROUND: Porcine epidemic diarrhea virus (PEDV), a type of coronavirus, is one of the main pathogens that can infect pigs of all ages. It causes diarrhea and acute death of newborn piglets, resulting in massive economic losses to the worldwide swine industry. While vaccination remains the primary approach in combating PEDV, it often fails to address all the challenges posed by the infection, particularly in light of the emergence of evolving mutant strains. Therefore, there is a critical need to identify potent antiviral drugs that can effectively safeguard pigs against PEDV infection. RESULTS: In this study, the antiviral efficacy of SP2509, a specific antagonist of Lysine-specific demethylase 1(LSD1), was evaluated in vitro. The RT-qPCR, Western blot, TCID50, and IFA showed that at a concentration of 1µmol/L, SP2509 significantly inhibited PEDV infection. Additionally, viral life cycle assays showed that SP2509 operates by impeding PEDV internalization and replication rather than attachment and release. Regarding mechanism, in Huh-7 cells, knockdowns LSD1 can suppress PEDV replication. This indicated that the inhibition effect of SP2509 on PEDV largely depends on the activity of its target protein, LSD1. CONCLUSION: Our results in vitro show that SP2509 can inhibit PEDV infection during the internalization and replication stage and revealed a role of LSD1 as a restriction factor for PEDV. These imply that LSD1 might be a target for interfering with the viral infection, and SP2509 could be developed as an effective anti-PEDV agent.


Subject(s)
Antiviral Agents , Histone Demethylases , Porcine epidemic diarrhea virus , Virus Replication , Porcine epidemic diarrhea virus/drug effects , Animals , Antiviral Agents/pharmacology , Virus Replication/drug effects , Histone Demethylases/antagonists & inhibitors , Swine , Chlorocebus aethiops , Swine Diseases/virology , Swine Diseases/drug therapy , Coronavirus Infections/veterinary , Coronavirus Infections/drug therapy , Coronavirus Infections/virology , Vero Cells
4.
Biol Pharm Bull ; 47(5): 930-940, 2024.
Article in English | MEDLINE | ID: mdl-38692871

ABSTRACT

The coronavirus disease 2019 (COVID-19) is caused by the etiological agent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). COVID-19, with the recurrent epidemics of new variants of SARS-CoV-2, remains a global public health problem, and new antivirals are still required. Some cholesterol derivatives, such as 25-hydroxycholesterol, are known to have antiviral activity against a wide range of enveloped and non-enveloped viruses, including SARS-CoV-2. At the entry step of SARS-CoV-2 infection, the viral envelope fuses with the host membrane dependent of viral spike (S) glycoproteins. From the screening of cholesterol derivatives, we found a new compound 26,27-dinorcholest-5-en-24-yne-3ß,20-diol (Nat-20(S)-yne) that inhibited the SARS-CoV-2 S protein-dependent membrane fusion in a syncytium formation assay. Nat-20(S)-yne exhibited the inhibitory activities of SARS-CoV-2 pseudovirus entry and intact SARS-CoV-2 infection in a dose-dependent manner. Among the variants of SARS-CoV-2, inhibition of infection by Nat-20(S)-yne was stronger in delta and Wuhan strains, which predominantly invade into cells via fusion at the plasma membrane, than in omicron strains. The interaction between receptor-binding domain of S proteins and host receptor ACE2 was not affected by Nat-20(S)-yne. Unlike 25-hydroxycholesterol, which regulates various steps of cholesterol metabolism, Nat-20(S)-yne inhibited only de novo cholesterol biosynthesis. As a result, plasma membrane cholesterol content was substantially decreased in Nat-20(S)-yne-treated cells, leading to inhibition of SARS-CoV-2 infection. Nat-20(S)-yne having a new mechanism of action may be a potential therapeutic candidate for COVID-19.


Subject(s)
Antiviral Agents , COVID-19 , Cholesterol , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , SARS-CoV-2/drug effects , Antiviral Agents/pharmacology , Humans , COVID-19/virology , Cholesterol/metabolism , Vero Cells , Chlorocebus aethiops , Spike Glycoprotein, Coronavirus/metabolism , Animals , Virus Internalization/drug effects , Betacoronavirus/drug effects , Pandemics , COVID-19 Drug Treatment , Coronavirus Infections/drug therapy , Coronavirus Infections/virology , Angiotensin-Converting Enzyme 2/metabolism , Pneumonia, Viral/drug therapy , Pneumonia, Viral/virology
5.
J Nanobiotechnology ; 22(1): 239, 2024 May 12.
Article in English | MEDLINE | ID: mdl-38735951

ABSTRACT

Widespread distribution of porcine epidemic diarrhea virus (PEDV) has led to catastrophic losses to the global pig farming industry. As a result, there is an urgent need for rapid, sensitive and accurate tests for PEDV to enable timely and effective interventions. In the present study, we develop and validate a floating gate carbon nanotubes field-effect transistor (FG CNT-FET)-based portable immunosensor for rapid identification of PEDV in a sensitive and accurate manner. To improve the affinity, a unique PEDV spike protein-specific monoclonal antibody is prepared by purification, and subsequently modified on FG CNT-FET sensor to recognize PEDV. The developed FET biosensor enables highly sensitive detection (LoD: 8.1 fg/mL and 100.14 TCID50/mL for recombinant spike proteins and PEDV, respectively), as well as satisfactory specificity. Notably, an integrated portable platform consisting of a pluggable FG CNT-FET chip and a portable device can discriminate PEDV positive from negative samples and even identify PEDV and porcine deltacoronavirus within 1 min with 100% accuracy. The portable sensing platform offers the capability to quickly, sensitively and accurately identify PEDV, which further points to a possibility of point of care (POC) applications of large-scale surveillance in pig breeding facilities.


Subject(s)
Biosensing Techniques , Nanotubes, Carbon , Porcine epidemic diarrhea virus , Porcine epidemic diarrhea virus/isolation & purification , Animals , Swine , Biosensing Techniques/methods , Biosensing Techniques/instrumentation , Nanotubes, Carbon/chemistry , Limit of Detection , Immunoassay/methods , Immunoassay/instrumentation , Antibodies, Monoclonal/immunology , Transistors, Electronic , Swine Diseases/diagnosis , Swine Diseases/virology , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/analysis , Coronavirus Infections/diagnosis , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Antibodies, Viral/immunology , Equipment Design
6.
J Med Microbiol ; 73(5)2024 May.
Article in English | MEDLINE | ID: mdl-38771617

ABSTRACT

Infectious bronchitis virus (IBV) is a highly contagious avian Gammacoronavirus that affects mainly chickens (Gallus gallus) but can circulate in other avian species. IBV constitutes a significant threat to the poultry industry, causing reduced egg yield, growth and mortality levels that can vary in impact. The virus can be transmitted horizontally by inhalation or direct/indirect contact with infected birds or contaminated fomites, vehicles, farm personnel and litter (Figure 1). The error-prone viral polymerase and recombination mechanisms mean diverse viral population results, with multiple genotypes, serotypes, pathotypes and protectotypes. This significantly complicates control and mitigation strategies based on vigilance in biosecurity and the deployment of vaccination.


Subject(s)
Chickens , Coronavirus Infections , Infectious bronchitis virus , Poultry Diseases , Infectious bronchitis virus/genetics , Infectious bronchitis virus/classification , Infectious bronchitis virus/physiology , Animals , Chickens/virology , Poultry Diseases/virology , Coronavirus Infections/virology , Coronavirus Infections/veterinary
7.
Sci Rep ; 14(1): 11639, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773161

ABSTRACT

COVID-19 is a kind of coronavirus that appeared in China in the Province of Wuhan in December 2019. The most significant influence of this virus is its very highly contagious characteristic which may lead to death. The standard diagnosis of COVID-19 is based on swabs from the throat and nose, their sensitivity is not high enough and so they are prone to errors. Early diagnosis of COVID-19 disease is important to provide the chance of quick isolation of the suspected cases and to decrease the opportunity of infection in healthy people. In this research, a framework for chest X-ray image classification tasks based on deep learning is proposed to help in early diagnosis of COVID-19. The proposed framework contains two phases which are the pre-processing phase and classification phase which uses pre-trained convolution neural network models based on transfer learning. In the pre-processing phase, different image enhancements have been applied to full and segmented X-ray images to improve the classification performance of the CNN models. Two CNN pre-trained models have been used for classification which are VGG19 and EfficientNetB0. From experimental results, the best model achieved a sensitivity of 0.96, specificity of 0.94, precision of 0.9412, F1 score of 0.9505 and accuracy of 0.95 using enhanced full X-ray images for binary classification of chest X-ray images into COVID-19 or normal with VGG19. The proposed framework is promising and achieved a classification accuracy of 0.935 for 4-class classification.


Subject(s)
COVID-19 , Deep Learning , Neural Networks, Computer , SARS-CoV-2 , COVID-19/diagnostic imaging , COVID-19/virology , COVID-19/diagnosis , Humans , SARS-CoV-2/isolation & purification , Radiography, Thoracic/methods , Pandemics , Pneumonia, Viral/diagnostic imaging , Pneumonia, Viral/virology , Pneumonia, Viral/diagnosis , Coronavirus Infections/diagnostic imaging , Coronavirus Infections/diagnosis , Coronavirus Infections/virology , Betacoronavirus/isolation & purification , Sensitivity and Specificity , Tomography, X-Ray Computed/methods
8.
BMC Vet Res ; 20(1): 209, 2024 May 18.
Article in English | MEDLINE | ID: mdl-38760785

ABSTRACT

BACKGROUND: Bovine coronavirus (BCoV) is implicated in severe diarrhea in calves and contributes to the bovine respiratory disease complex; it shares a close relationship with human coronavirus. Similar to other coronaviruses, remarkable variability was found in the genome and biology of the BCoV. In 2022, samples of feces were collected from a cattle farm. A virus was isolated from 7-day-old newborn calves. In this study, we present the genetic characteristics of a new BCoV isolate. The complete genomic, spike protein, and nucleocapsid protein gene sequences of the BCoV strain, along with those of other coronaviruses, were obtained from the GenBank database. Genetic analysis was conducted using MEGA7.0 and the Neighbor-Joining (NJ) method. The reference strains' related genes were retrieved from GenBank for comparison and analysis using DNAMAN. RESULTS: The phylogenetic tree and whole genome consistency analysis showed that it belonged to the GIIb subgroup, which is epidemic in Asia and America, and was quite similar to the Chinese strains in the same cluster. Significantly, the S gene was highly consistent with QH1 (MH810151.1) isolated from yak. This suggests that the strain may have originated from interspecies transmission involving mutations of wild strains. The N gene was conserved and showed high sequence identity with the epidemic strains in China and the USA. CONCLUSIONS: Genetic characterization suggests that the isolated strain could be a new mutant from a wild-type lineage, which is in the same cluster as most Chinese epidemic strains but on a new branch.


Subject(s)
Cattle Diseases , Coronavirus Infections , Coronavirus, Bovine , Genome, Viral , Phylogeny , Animals , Cattle , Coronavirus, Bovine/genetics , Coronavirus, Bovine/isolation & purification , China/epidemiology , Cattle Diseases/virology , Cattle Diseases/epidemiology , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Coronavirus Infections/epidemiology , Feces/virology , Spike Glycoprotein, Coronavirus/genetics , Animals, Newborn
9.
Vet Microbiol ; 293: 110101, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38718529

ABSTRACT

Cross-species transmission of coronaviruses has been continuously posing a major challenge to public health. Pigs, as the major animal reservoirs for many zoonotic viruses, frequently mediate viral transmission to humans. This study comprehensively mapped the relationship between human and porcine coronaviruses through in-depth bioinformatics analysis. We found that human coronavirus OC43 and porcine coronavirus PHEV share a close phylogenetic relationship, evidenced by high genomic homology, similar codon usage patterns and comparable tertiary structure in spike proteins. Inoculation of infectious OC43 viruses in organoids derived from porcine small and large intestine demonstrated that porcine intestinal organoids (pIOs) are highly susceptible to human coronavirus OC43 infection and support infectious virus production. Using transmission electron microscopy, we visualized OC43 viral particles in both intracellular and extracellular compartments, and observed abnormalities of multiple organelles in infected organoid cells. Robust OC43 infections in pIOs result in a significant reduction of organoids viability and widespread cell death. This study bears essential implications for better understanding the evolutionary origin of human coronavirus OC43, and provides a proof-of-concept for using pIOs as a model to investigate cross-species transmission of human coronavirus.


Subject(s)
Computational Biology , Coronavirus Infections , Coronavirus OC43, Human , Intestines , Organoids , Phylogeny , Animals , Organoids/virology , Swine , Humans , Coronavirus Infections/virology , Coronavirus Infections/transmission , Coronavirus Infections/veterinary , Coronavirus OC43, Human/physiology , Coronavirus OC43, Human/genetics , Intestines/virology , Swine Diseases/virology , Swine Diseases/transmission , Genome, Viral
10.
Vet Microbiol ; 293: 110100, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38718527

ABSTRACT

Recent epidemiological studies have discovered that a lot of cases of porcine epidemic diarrhea virus (PEDV) infection are frequently accompanied by porcine kobuvirus (PKV) infection, suggesting a potential relationship between the two viruses in the development of diarrhea. To investigate the impact of PKV on PEDV pathogenicity and the number of intestinal lymphocytes, piglets were infected with PKV or PEDV or co-infected with both viruses. Our findings demonstrate that co-infected piglets exhibit more severe symptoms, acute gastroenteritis, and higher PEDV replication compared to those infected with PEDV alone. Notably, PKV alone does not cause significant intestinal damage but enhances PEDV's pathogenicity and alters the number of intestinal lymphocytes. These results underscore the complexity of viral interactions in swine diseases and highlight the need for comprehensive diagnostic and treatment strategies addressing co-infections.


Subject(s)
Coinfection , Coronavirus Infections , Intestines , Kobuvirus , Lymphocytes , Porcine epidemic diarrhea virus , Swine Diseases , Animals , Porcine epidemic diarrhea virus/pathogenicity , Porcine epidemic diarrhea virus/physiology , Swine , Swine Diseases/virology , Coinfection/virology , Coinfection/veterinary , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Lymphocytes/virology , Kobuvirus/pathogenicity , Kobuvirus/genetics , Intestines/virology , Diarrhea/virology , Diarrhea/veterinary , Virus Replication , Gastroenteritis/virology , Gastroenteritis/veterinary , Picornaviridae Infections/veterinary , Picornaviridae Infections/virology
11.
N Z Vet J ; 72(4): 191-200, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38650102

ABSTRACT

AIMS: To isolate canine respiratory coronavirus (CRCoV) and canine pneumovirus (CnPnV) in cell culture and to compare partial genomic sequences of CRCoV and CnPnV from New Zealand with those from other countries. METHODS: Oropharyngeal swab samples from dogs affected by canine infectious respiratory disease syndrome that were positive for CnPnV (n = 15) or CRCoV (n = 1) by virus-specific reverse transcriptase quantitative PCR (RT-qPCR) in a previous study comprised the starting material. Virus isolation was performed in HRT-18 cells for CRCoV and RAW 264.7 and Vero cells for CnPnV. The entire sequence of CnPnV G protein (1,266 nucleotides) and most (8,063/9,707 nucleotides) of the 3' region of CRCoV that codes for 10 structural and accessory proteins were amplified and sequenced. The sequences were analysed and compared with other sequences available in GenBank using standard molecular tools including phylogenetic analysis. RESULTS: Virus isolation was unsuccessful for both CRCoV and CnPnV. Pneumovirus G protein was amplified from 3/15 (20%) samples that were positive for CnPnV RNA by RT-qPCR. Two of these (NZ-048 and NZ-049) were 100% identical to each other, and 90.9% identical to the third one (NZ-007). Based on phylogenetic analysis of the G protein gene, CnPnV NZ-048 and NZ-049 clustered with sequences from the USA, Thailand and Italy in group A, and CnPnV NZ-007 clustered with sequences from the USA in group B. The characteristics of the predicted genes (length, position) and their putative protein products (size, predicted structure, presence of N- and O-glycosylation sites) of the New Zealand CRCoV sequence were consistent with those reported previously, except for the region located between open reading frame (ORF)3 (coding for S protein) and ORF6 (coding for E protein). The New Zealand virus was predicted to encode 5.9 kDa, 27 kDa and 12.7 kDa proteins, which differed from the putative coding capacity of this region reported for CRCoV from other countries. CONCLUSIONS: This report represents the first characterisation of partial genomic sequences of CRCoV and CnPnV from New Zealand. Our results suggest that the population of CnPnV circulating in New Zealand is not homogeneous, and that the viruses from two clades described overseas are also present here. Limited conclusions can be made based on only one CRCoV sequence, but the putative differences in the coding capacity of New Zealand CRCoV support the previously reported variability of this region. The reasons for such variability and its biological implications need to be further elucidated.


Subject(s)
Coronavirus, Canine , Dog Diseases , Genome, Viral , Phylogeny , Pneumovirus , Animals , Dogs , New Zealand/epidemiology , Coronavirus, Canine/genetics , Coronavirus, Canine/classification , Coronavirus, Canine/isolation & purification , Dog Diseases/virology , Dog Diseases/epidemiology , Pneumovirus/genetics , Pneumovirus/classification , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Coronavirus Infections/epidemiology , Vero Cells , Chlorocebus aethiops
12.
PLoS Pathog ; 20(4): e1012156, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38598560

ABSTRACT

SARS-CoV-2 has been shown to cause wide-ranging ocular abnormalities and vision impairment in COVID-19 patients. However, there is limited understanding of SARS-CoV-2 in ocular transmission, tropism, and associated pathologies. The presence of viral RNA in corneal/conjunctival tissue and tears, along with the evidence of viral entry receptors on the ocular surface, has led to speculation that the eye may serve as a potential route of SARS-CoV-2 transmission. Here, we investigated the interaction of SARS-CoV-2 with cells lining the blood-retinal barrier (BRB) and the role of the eye in its transmission and tropism. The results from our study suggest that SARS-CoV-2 ocular exposure does not cause lung infection and moribund illness in K18-hACE2 mice despite the extended presence of viral remnants in various ocular tissues. In contrast, intranasal exposure not only resulted in SARS-CoV-2 spike (S) protein presence in different ocular tissues but also induces a hyperinflammatory immune response in the retina. Additionally, the long-term exposure to viral S-protein caused microaneurysm, retinal pigmented epithelium (RPE) mottling, retinal atrophy, and vein occlusion in mouse eyes. Notably, cells lining the BRB, the outer barrier, RPE, and the inner barrier, retinal vascular endothelium, were highly permissive to SARS-CoV-2 replication. Unexpectedly, primary human corneal epithelial cells were comparatively resistant to SARS-CoV-2 infection. The cells lining the BRB showed induced expression of viral entry receptors and increased susceptibility towards SARS-CoV-2-induced cell death. Furthermore, hyperglycemic conditions enhanced the viral entry receptor expression, infectivity, and susceptibility of SARS-CoV-2-induced cell death in the BRB cells, confirming the reported heightened pathological manifestations in comorbid populations. Collectively, our study provides the first evidence of SARS-CoV-2 ocular tropism via cells lining the BRB and that the virus can infect the retina via systemic permeation and induce retinal inflammation.


Subject(s)
Blood-Retinal Barrier , COVID-19 , Retina , SARS-CoV-2 , SARS-CoV-2/immunology , SARS-CoV-2/physiology , Animals , Blood-Retinal Barrier/virology , COVID-19/immunology , COVID-19/virology , Mice , Humans , Retina/virology , Retina/immunology , Retina/metabolism , Angiotensin-Converting Enzyme 2/metabolism , Virus Internalization , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/immunology , Inflammation/immunology , Inflammation/virology , Betacoronavirus/physiology , Viral Tropism , Coronavirus Infections/immunology , Coronavirus Infections/virology , Coronavirus Infections/pathology
13.
Microb Pathog ; 190: 106638, 2024 May.
Article in English | MEDLINE | ID: mdl-38574829

ABSTRACT

Autophagy plays an important role in the lifecycle of viruses. However, there is currently a lack of systematic research on the relationship between Infectious Bronchitis Virus (IBV) and autophagy. This study aims to investigate the impact of IBV on autophagy and the role of autophagy in viral replication. We observed that IBV infection increased the expression of microtubule-associated protein 1 light chain 3, a marker of autophagy, decreased the expression of sequestosome 1, and led to elevated intracellular LC3 puncta levels. These findings suggest that IBV infection activates the autophagic process in cells. To investigate the impact of autophagy on the replication of IBV, we utilized rapamycin as an autophagy activator and 3-methyladenine as an autophagy inhibitor. Our results indicate that IBV promotes viral replication by inducing autophagy. Further investigation revealed that IBV induces autophagosome formation by inhibiting the mTOR-ULK1 pathway and activating the activity of vacuolar protein sorting 34 (VPS34), autophagy-related gene 14, and the Beclin-1 complex. VPS34 plays a crucial role in this process, as inhibiting VPS34 protein activity enhances cell proliferation after IBV infection. Additionally, inhibiting VPS34 significantly improves the survival rate of IBV-infected chicks, suppresses IBV replication in the kidney, and alleviates tracheal, lung, and kidney damage caused by IBV infection. In summary, IBV infection can induce autophagy by modulating the mTOR/ULK1 signaling pathway and activating the VPS34 complex, while autophagy serves to promote virus replication.


Subject(s)
Autophagy , Chickens , Class III Phosphatidylinositol 3-Kinases , Infectious bronchitis virus , Virus Replication , Infectious bronchitis virus/physiology , Animals , Class III Phosphatidylinositol 3-Kinases/metabolism , Chickens/virology , Coronavirus Infections/virology , Coronavirus Infections/metabolism , Sirolimus/pharmacology , Beclin-1/metabolism , Beclin-1/genetics , TOR Serine-Threonine Kinases/metabolism , Signal Transduction , Cell Line , Poultry Diseases/virology , Autophagosomes/metabolism , Autophagosomes/virology , Chlorocebus aethiops , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics
14.
J Med Virol ; 96(5): e29628, 2024 May.
Article in English | MEDLINE | ID: mdl-38682568

ABSTRACT

This study evaluated the potential for antibody-dependent enhancement (ADE) in serum samples from patients exposed to Middle East respiratory syndrome coronavirus (MERS-CoV). Furthermore, we evaluated the effect of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination on ADE in individuals with a MERS infection history. We performed ADE assay in sera from MERS recovered and SARS-CoV-2-vaccinated individuals using BHK cells expressing FcgRIIa, SARS-CoV-2, and MERS-CoV pseudoviruses (PVs). Further, we analyzed the association of ADE to serum IgG levels and neutralization. Out of 16 MERS patients, nine demonstrated ADE against SARS-CoV-2 PV, however, none of the samples demonstrated ADE against MERS-CoV PV. Furthermore, out of the seven patients exposed to SARS-CoV-2 vaccination after MERS-CoV infection, only one patient (acutely infected with MERS-CoV) showed ADE for SARS-CoV-2 PV. Further analysis indicated that IgG1, IgG2, and IgG3 against SARS-CoV-2 S1 and RBD subunits, IgG1 and IgG2 against the MERS-CoV S1 subunit, and serum neutralizing activity were low in ADE-positive samples. In summary, samples from MERS-CoV-infected patients exhibited ADE against SARS-CoV-2 and was significantly associated with low levels of neutralizing antibodies. Subsequent exposure to SARS-CoV-2 vaccination resulted in diminished ADE activity while the PV neutralization assay demonstrated a broadly reactive antibody response in some patient samples.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Antibody-Dependent Enhancement , COVID-19 , Immunoglobulin G , Middle East Respiratory Syndrome Coronavirus , SARS-CoV-2 , Humans , Middle East Respiratory Syndrome Coronavirus/immunology , Antibodies, Viral/blood , SARS-CoV-2/immunology , Immunoglobulin G/blood , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , COVID-19/immunology , Coronavirus Infections/immunology , Coronavirus Infections/virology , Middle Aged , Male , Female , Neutralization Tests , Adult , COVID-19 Vaccines/immunology , Antigens, Viral/immunology , Animals , Aged , Spike Glycoprotein, Coronavirus/immunology , Vaccination
15.
Vet Immunol Immunopathol ; 271: 110753, 2024 May.
Article in English | MEDLINE | ID: mdl-38608406

ABSTRACT

Porcine epidemic diarrhea virus (PEDV) causes immensely large economic losses worldwide in the swine industry. PEDV attacks the intestine, disrupts intestinal epithelium morphology and barrier integrity, and results in profound diarrhea and high mortality. A commercially available isotonic protein solution (IPS) (Tonisity Px) has anecdotally been reported to be effective in supportive treatment of piglets with active PEDV infections. This study evaluated the effects of supplementing (or not) the drinking water of 14 day old PEDV-infected piglets with the IPS on the content of E-cadherin, fibronectin, interferon-alpha (IFN-α), and matrix metalloproteinase 9 (MMP-9) in duodenal tissue. The content of PEDV DNA in feces was also measured. Though both groups had similar PEDV shedding at day 1, IPS piglets had significantly lower PEDV shedding at day 5, 14 and 21. The IPS group also had a shorter duration of PEDV virus shedding. Levels of E-cadherin and fibronectin, both of which are structural proteins in the intestine, remained unchanged from baseline in the IPS group, whereas the same molecules decreased significantly in the control group. IFN-α, an antiviral cytokine, and MMP-9, an enzyme that aids in tissue remodeling, were increased at days 5 and 14 post infection, and then decreased at day 21 post-infection in the IPS group compared to control. Overall, the IPS used in this study enhanced epithelial intercellular adhesion (E-cadherin) and extracellular matrix structure (fibronectin), resulted in significantand favorable changes in MMP-9 activity, and favorably modulated IFN-α production. This is the first report of this panel of biomarkers, especially MMP-9 and IFN-α, in the face of in vivo PEDV infection. This is also the first report to investigate a commercially available swine product that does not need to be administered in solid feed, and that is already registered for use throughout Asia, Europe, South America, and North America. Overall, the results of this study serve to clarify the behavior of 4 key biomarkers in the presence of in vivo PEDV infection. The results also indicate that IPS (Tonisity Px) supplementation is a viable intervention to modulate the porcine intestinal immune response with favorable effects on the intestine.


Subject(s)
Coronavirus Infections , Porcine epidemic diarrhea virus , Swine Diseases , Virus Shedding , Animals , Swine , Porcine epidemic diarrhea virus/physiology , Porcine epidemic diarrhea virus/immunology , Coronavirus Infections/veterinary , Coronavirus Infections/immunology , Coronavirus Infections/virology , Swine Diseases/virology , Swine Diseases/immunology , Fibronectins/metabolism , Matrix Metalloproteinase 9/metabolism , Cadherins/metabolism , Intestines/immunology , Intestines/virology , Interferon-alpha/immunology , Cell Adhesion , Intestinal Mucosa/immunology
16.
J Gen Virol ; 105(4)2024 Apr.
Article in English | MEDLINE | ID: mdl-38656455

ABSTRACT

Porcine epidemic diarrhea (PED) is a serious disease in piglets that leads to high mortality. An effective measure that provides higher IgA levels in the intestine and milk is required to decrease losses. Porcine epidemic diarrhea virus (PEDV) was dissolved in calcium alginate (Alg) and combined with chitosan (CS) via electrostatic interactions between cationic chitosan and anionic alginate to create a porous gel (Alg-CS+PEDV). The gel was used to immunize mice orally or in combination with subcutaneous injections of inactivated PEDV vaccine. At 12 and 24 days after immunization, levels of IgA and IgG in Alg-CS+PEDV were higher than with normal PEDV oral administration. At 24 days after immunization, the concentration of IFN-γ in Alg-CS+PEDV was higher than with normal PEDV oral administration. Furthermore, oral administration combining subcutaneous immunization induced higher levels of IgG and IgA than oral administration alone. Our study provides a new method for the preparation and administration of oral vaccines to achieve enhanced mucosal immunity against PEDV.


Subject(s)
Alginates , Antibodies, Viral , Chitosan , Immunity, Mucosal , Immunoglobulin A , Immunoglobulin G , Porcine epidemic diarrhea virus , Viral Vaccines , Animals , Administration, Oral , Porcine epidemic diarrhea virus/immunology , Alginates/administration & dosage , Chitosan/administration & dosage , Mice , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Antibodies, Viral/immunology , Immunoglobulin A/immunology , Immunoglobulin G/blood , Swine , Coronavirus Infections/immunology , Coronavirus Infections/prevention & control , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology , Swine Diseases/immunology , Swine Diseases/prevention & control , Swine Diseases/virology , Female , Gels/administration & dosage , Mice, Inbred BALB C , Interferon-gamma/immunology , Glucuronic Acid/administration & dosage , Hexuronic Acids/administration & dosage
17.
Vet Microbiol ; 293: 110096, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38636174

ABSTRACT

IgA plays a vital role in defending against the infectious pathogens. However, the specific regulatory pathways involved in IgA secretion in the context of PEDV infection have remained elusive. Therefore, in this study, we explore the molecular mechanisms underlying IgA secretion in response to infection, with a particular focus on PEDV, a devastating enteric virus affecting global swine production. Our investigation begins by examining changes in IgA concentrations in both serum and small intestinal contents following PEDV infection in 2- and 4-week-old pigs. Remarkably, a significant increase in IgA levels in these older pigs post-infection were observed. To delve deeper into the regulatory mechanisms governing IgA secretion in response to PEDV infection, isolated porcine intestinal B cells were co-cultured with monocytes derived DCs (Mo-DCs) in vitro. In the intestinal DC-B cell co-cultures, IgA secretion was found to increase significantly after PEDV infection, as well as upregulating the expression of AID, GLTα and PSTα reflecting isotype switching to IgA. In addition, the expression of TLR9 was upregulated in these cultures, as determined by RT-qPCR and western blotting. Moreover, our findings extend to in vivo observations, where we detected higher levels of TLR9 expression in the ileum of pig post PEDV infection. Collectively, our results highlight the ability of PEDV to stimulate the generation of IgA, particularly in elder pigs, and identify TLR9 as a critical mediator of IgA production within the porcine intestinal microenvironment during PEDV infection.


Subject(s)
Coronavirus Infections , Immunoglobulin A , Intestine, Small , Porcine epidemic diarrhea virus , Swine Diseases , Toll-Like Receptor 9 , Animals , Swine , Porcine epidemic diarrhea virus/immunology , Swine Diseases/immunology , Swine Diseases/virology , Intestine, Small/immunology , Immunoglobulin A/immunology , Toll-Like Receptor 9/metabolism , Toll-Like Receptor 9/genetics , Coronavirus Infections/veterinary , Coronavirus Infections/immunology , Coronavirus Infections/virology , B-Lymphocytes/immunology , Coculture Techniques , Dendritic Cells/immunology
18.
Vet Microbiol ; 293: 110087, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38663176

ABSTRACT

Porcine epidemic diarrhea virus (PEDV) is a devastating pathogen of acute- gastrointestinal infectious diseases, which can cause vomiting, diarrhea, dehydration and high morbidity and mortality among neonatal piglets. Humoral immunity plays a vital role in the host anti-PEDV infection process, but the mechanism of PEDV-induced B-cell immune response remains unknown. In this study, the effects of PEDV infection on CD21+ B cell activation were systematically analyzed through animal experiments. Enzyme-linked immunosorbent assays (ELISA) revealed that low levels of serum-specific IgA, IgM, or IgG were detected in piglets after PEDV infection, respectively. Serum interleukin (IL)-6 levels increased significantly at 4 d after infection, and the levels of IL-4, B-cell activating factor (BAFF), interferon (IFN)-γ, transforming growth factor (TGF)-ß and IL-10 decreased at 7 d after infection. Fluorescence-activated cell sorting (FACS) showed that expression levels of CD21, MHC Ⅱ, CD40, and CD38 on B cell surfaces were significantly higher. In contrast, the proportions of CD21+IgM+ B cells were decreased in peripheral blood mononuclear cells (PBMCs) from the infected piglets. No differences were found in the percentage of CD21+CD80+ and CD21+CD27+ B cells in PBMCs from the infected piglets. In addition, the number of CD21+B cells in PBMCs stimulated with PEDV in vitro was significantly lower. No significant change in the mRNA expression of BCR molecules was found while the expression levels of paired immunoglobulin-like receptor B (PIR-B), B cell adaptor molecule of 32 kDa (Bam32) and BAFF were decreased. In conclusion, our research demonstrates that virulent strains of PEDV profoundly impact B cell activation, leading to alterations in phenotypic expression and BCR signaling molecules. Furthermore, this dysregulation results in compromised specific antibody secretion and perturbed cytokine production, highlighting the intricate immunological dysfunctions induced by PEDV infection.


Subject(s)
B-Lymphocytes , Coronavirus Infections , Lymphocyte Activation , Porcine epidemic diarrhea virus , Receptors, Complement 3d , Swine Diseases , Animals , Porcine epidemic diarrhea virus/immunology , Swine , B-Lymphocytes/immunology , Coronavirus Infections/veterinary , Coronavirus Infections/immunology , Coronavirus Infections/virology , Receptors, Complement 3d/immunology , Receptors, Complement 3d/metabolism , Swine Diseases/virology , Swine Diseases/immunology , Cytokines/immunology , Cytokines/genetics , Cytokines/metabolism , Antibodies, Viral/blood , Immunoglobulin A/blood , Immunoglobulin A/immunology , Immunoglobulin M/blood , Immunoglobulin M/immunology
19.
Vet Microbiol ; 293: 110098, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38677126

ABSTRACT

The infection of canine coronavirus (CCoV) causes a highly contagious disease in dogs with acute gastroenteritis. The efficient serological diagnostics is critical for controlling the disease caused by CCoV. Nucleocapsid (N) protein of CCoV is an important target for developing serological approaches. However, little is known about the antigenic sites in the N protein of CCoV. In this study, we generated a monoclonal antibody (mAb) against the N protein of CCoV, designated as 13E8, through the fusion of the sp2/0 cells with the spleen cells from a mouse immunized with the purified recombinant GST-N protein. Epitope mapping revealed that mAb 13E8 recognized a novel linear B cell epitope in N protein at 294-314aa (named as EP-13E8) by using a serial of truncated N protein through Western blot and ELISA. Sequence analysis showed that the sequence of EP-13E8 was highly conserved (100 %) among different CCoV strains analyzed, but exhibited a low similarity (31.8-63.6 %) with the responding sequence in other coronaviruses of the same genus such as FCoV, PEDV and HCoV except for TGEV (95.5 % identity). Structural assay suggested that the epitope of EP-13E8 were located in the close proximity on the surface of the N protein. Overall, the mAb 13E8 against N protein generated and its epitope EP-13E8 identified here paid the way for further developing epitope-based serological diagnostics for CCoV.


Subject(s)
Antibodies, Monoclonal , Coronavirus, Canine , Epitope Mapping , Epitopes, B-Lymphocyte , Nucleocapsid Proteins , Animals , Antibodies, Monoclonal/immunology , Epitopes, B-Lymphocyte/immunology , Dogs , Mice , Nucleocapsid Proteins/immunology , Coronavirus, Canine/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Mice, Inbred BALB C , Coronavirus Nucleocapsid Proteins/immunology , Dog Diseases/virology , Dog Diseases/immunology , Coronavirus Infections/veterinary , Coronavirus Infections/immunology , Coronavirus Infections/virology , Coronavirus Infections/diagnosis , Amino Acid Sequence
20.
Avian Dis ; 68(1): 10-17, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38687102

ABSTRACT

The relationship between passive immunity and the development of false layer syndrome (FLS) and its associated lesions was investigated in this study by comparing the long-term reproductive effects of an infectious bronchitis virus (IBV) DMV/1639 wild-type strain and the GA08 vaccine in birds with and without maternal antibodies. There was a clear protective effect provided by maternal antibodies against both the early vaccination and challenge. It was also observed that vaccination at an early age, in the absence of maternal antibodies, can induce reproductive issues, such as reduced egg production and FLS-associated lesions (e.g., cystic oviduct and egg yolk coelomitis). This might indicate that maternal antibodies and the timing of IBV infection are more important in the generation of FLS than the IBV strain type.


Mitigación del síndrome de la falsa ponedora mediante anticuerpos maternos contra el virus de la bronquitis infecciosa. En este estudio se investigó la relación entre la inmunidad pasiva y el desarrollo del síndrome de la falsa ponedora (FLS) y sus lesiones asociadas comparando los efectos reproductivos a largo plazo de una cepa de tipo silvestre DMV/1639 del virus de la bronquitis infecciosa (IBV) y la cepa vacunal GA08, en aves con y sin anticuerpos maternos. Hubo un claro efecto protector proporcionado por los anticuerpos maternos tanto contra la vacunación temprana como contra el desafío. También se observó que la vacunación a una edad temprana, en ausencia de anticuerpos maternos, puede inducir problemas reproductivos, como una reducción de la producción de huevo y lesiones asociadas al síndrome de la falsa ponedora (p. ej., oviducto quístico y celomitis de yema de huevo). Esto podría indicar que los anticuerpos maternos y el momento de la infección por el virus de la bronquitis infecciosa son más importantes en la generación del síndrome de la falsa ponedora que el tipo de cepa del virus de la bronquitis infecciosa.


Subject(s)
Antibodies, Viral , Chickens , Coronavirus Infections , Infectious bronchitis virus , Poultry Diseases , Infectious bronchitis virus/immunology , Animals , Poultry Diseases/virology , Poultry Diseases/immunology , Female , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Coronavirus Infections/immunology , Immunity, Maternally-Acquired , Viral Vaccines/immunology , Viral Vaccines/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...