Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.026
Filter
1.
PLoS One ; 19(7): e0306532, 2024.
Article in English | MEDLINE | ID: mdl-38968319

ABSTRACT

This study evaluated the use of endemic enteric coronaviruses polymerase chain reaction (PCR)-negative testing results as an alternative approach to detect the emergence of animal health threats with similar clinical diseases presentation. This retrospective study, conducted in the United States, used PCR-negative testing results from porcine samples tested at six veterinary diagnostic laboratories. As a proof of concept, the database was first searched for transmissible gastroenteritis virus (TGEV) negative submissions between January 1st, 2010, through April 29th, 2013, when the first porcine epidemic diarrhea virus (PEDV) case was diagnosed. Secondly, TGEV- and PEDV-negative submissions were used to detect the porcine delta coronavirus (PDCoV) emergence in 2014. Lastly, encountered best detection algorithms were implemented to prospectively monitor the 2023 enteric coronavirus-negative submissions. Time series (weekly TGEV-negative counts) and Seasonal Autoregressive-Integrated Moving-Average (SARIMA) were used to control for outliers, trends, and seasonality. The SARIMA's fitted and residuals were then subjected to anomaly detection algorithms (EARS, EWMA, CUSUM, Farrington) to identify alarms, defined as weeks of higher TGEV-negativity than what was predicted by models preceding the PEDV emergence. The best-performing detection algorithms had the lowest false alarms (number of alarms detected during the baseline) and highest time to detect (number of weeks between the first alarm and PEDV emergence). The best-performing detection algorithms were CUSUM, EWMA, and Farrington flexible using SARIMA fitted values, having a lower false alarm rate and identified alarms 4 to 17 weeks before PEDV and PDCoV emergences. No alarms were identified in the 2023 enteric negative testing results. The negative-based monitoring system functioned in the case of PEDV propagating epidemic and in the presence of a concurrent propagating epidemic with the PDCoV emergence. It demonstrated its applicability as an additional tool for diagnostic data monitoring of emergent pathogens having similar clinical disease as the monitored endemic pathogens.


Subject(s)
Coronavirus Infections , Porcine epidemic diarrhea virus , Swine Diseases , Transmissible gastroenteritis virus , Animals , Swine , Transmissible gastroenteritis virus/genetics , Transmissible gastroenteritis virus/isolation & purification , Porcine epidemic diarrhea virus/isolation & purification , Porcine epidemic diarrhea virus/genetics , Coronavirus Infections/diagnosis , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Coronavirus Infections/epidemiology , Swine Diseases/virology , Swine Diseases/diagnosis , Retrospective Studies , Gastroenteritis, Transmissible, of Swine/diagnosis , Gastroenteritis, Transmissible, of Swine/virology , Gastroenteritis, Transmissible, of Swine/epidemiology , Polymerase Chain Reaction/methods , Deltacoronavirus/genetics , Deltacoronavirus/isolation & purification , United States/epidemiology
2.
Arch Virol ; 169(8): 158, 2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38970647

ABSTRACT

The highly pathogenic genotype 2b (HP-G2b) of porcine epidemic diarrhea virus (PEDV), which caused a pandemic in 2013-2014, evolved in South Korea and became endemic, affecting the domestic pig industry. This study describes the genotypic traits of novel HP-G2b PEDV strains identified on affected farms experiencing low disease severity with < 10% neonatal mortality. Nucleotide sequencing revealed common deletion patterns, termed S-DEL2, resulting in a two-amino-acid deletion at positions 60 and 61, 61 and 62, or 63 and 64 in the N-terminal domain of the spike (S) protein of all isolates. The S barcode profiles of S-DEL2 variants differed from each other and shared 96.0-99.4% and 98.5-99.6% nt sequence identity with other South Korean HP-G2b PEDV strains in the S gene and in the complete genome sequence, respectively. Genetic and phylogenetic analysis showed that the S-DEL2 strains belonged to diverse domestic clades: CK, CK.1, CK.2, or NC. The emergence of novel S-DEL2 strains suggests that continuous evolution of PEDV occurs under endemic circumstances, resulting in genetic diversity and distinct clinical presentations. This study advances our knowledge regarding the genetic and pathogenic heterogeneity of PEDV and emphasizes the importance of active monitoring and surveillance to identify novel variants and determine their genotypic and phenotypic characteristics.


Subject(s)
Coronavirus Infections , Genotype , Phylogeny , Porcine epidemic diarrhea virus , Spike Glycoprotein, Coronavirus , Swine Diseases , Porcine epidemic diarrhea virus/genetics , Porcine epidemic diarrhea virus/classification , Porcine epidemic diarrhea virus/isolation & purification , Animals , Republic of Korea/epidemiology , Swine , Swine Diseases/virology , Swine Diseases/epidemiology , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Coronavirus Infections/epidemiology , Spike Glycoprotein, Coronavirus/genetics , Genetic Variation , Genome, Viral/genetics , Sequence Deletion
3.
Virol J ; 21(1): 157, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38992629

ABSTRACT

Newborn piglets' health is seriously threatened by the porcine epidemic diarrhea virus (PEDV), which also has a significant effect on the pig industry. The gut microbiota produces butyrate, an abundant metabolite that modulates intestinal function through many methods to improve immunological and intestinal barrier function. The objective of this investigation was to ascertain how elevated butyrate concentrations impacted the host transcriptional profile of PEDV CV777 strain infection. Our findings showed that higher concentrations of butyrate have a stronger inhibitory effect on PEDV CV777 strain infection. According to RNA-seq data, higher concentrations of butyrate induced more significant transcriptional changes in IPEC-J2 cells, and signaling pathways such as PI3K-AKT may play a role in the inhibition of PEDV CV777 strain by high concentrations of butyrate. Ultimately, we offer a theoretical and experimental framework for future research and development of novel approaches to harness butyrate's antiviral infection properties.


Subject(s)
Butyrates , Epithelial Cells , Porcine epidemic diarrhea virus , Animals , Porcine epidemic diarrhea virus/drug effects , Porcine epidemic diarrhea virus/physiology , Swine , Butyrates/pharmacology , Butyrates/metabolism , Epithelial Cells/virology , Epithelial Cells/drug effects , Cell Line , Swine Diseases/virology , Coronavirus Infections/virology , Coronavirus Infections/drug therapy , Coronavirus Infections/veterinary , Antiviral Agents/pharmacology , Signal Transduction/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/virology , Intestinal Mucosa/drug effects , Virus Replication/drug effects , Intestines/virology
4.
Virol J ; 21(1): 155, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38982509

ABSTRACT

BACKGROUND: Canine enteric coronavirus (CECoV) is a prevalent infectious disease among dogs worldwide, yet its epidemiology in mainland China remains poorly understood. This systematic review and meta-analysis aimed to assess the prevalence of CECoV in mainland China and identify factors influencing its prevalence. METHODS: A comprehensive literature search was conducted across multiple databases for studies regarding CECoV epidemiology of China. PubMed, CNKI, Wanfang, and CQVIP were searched to obtain the studies. Eligible studies were selected based on predefined criteria, and data were extracted and synthesized. The quality the studies was assessed using the JBI assessment tool. Heterogeneity was checked using I2 test statistics followed by subgroup and sensitivity analysis. Subgroup analyses were performed to explore variations in CECoV prevalence by factors such as year, region, season, health status, social housing type, gender, age, and breed. Publication bias was assessed using a funnel plot and eggers test that was followed by trim and fill analysis. RESULTS: A total of 27 studies involving 21,034 samples were included in the meta-analysis. The overall pooled prevalence of CECoV in mainland China was estimated to be 0.30 (95% CI 0.24-0.37), indicating persistent circulation of the virus. Subgroup analyses revealed higher prevalence rates in younger dogs, multi-dog households, apparently healthy dogs, and certain regions such as southwest China. Seasonal variations were observed, with lower prevalence rates in summer. However, no significant differences in prevalence were found by gender. CONCLUSIONS: This study provides valuable insights into the epidemiology of CECoV in mainland China, highlighting the persistent circulation of the virus and identifying factors associated with higher prevalence rates. Continuous monitoring and surveillance efforts, along with research into accurate detection methods and preventive measures, are essential for the effective control of CECoV and mitigation of its potential impact on animal and human health.


Subject(s)
Coronavirus Infections , Coronavirus, Canine , Dog Diseases , Animals , Dogs , China/epidemiology , Prevalence , Dog Diseases/epidemiology , Dog Diseases/virology , Coronavirus Infections/epidemiology , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Coronavirus, Canine/genetics , Coronavirus, Canine/isolation & purification
5.
Vopr Virusol ; 69(3): 255-265, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38996374

ABSTRACT

INTRODUCTION: Bats are natural reservoirs of coronaviruses (Coronaviridae), which have caused three outbreaks of human disease SARS, MERS and COVID-19 or SARS-2 over the past decade. The purpose of the work is to study the diversity of coronaviruses among bats inhabiting the foothills and mountainous areas of the Republics of Dagestan, Altai and the Kemerovo region. MATERIALS AND METHODS: Samples of bat oral swabs and feces were tested for the presence of coronavirus RNA by reverse transcription-polymerase chain reaction (RT-PCR). RESULTS: It has been shown that the greater horseshoe bats (Rhinolophus ferrumequinum), inhabiting the Republic of Dagestan, are carriers of two different coronaviruses. One of the two coronaviruses is a member of the Sarbecovius subgenus of the Betacoronavirus genus, which includes the causative agents of SARS and COVID-19. The second coronavirus is assigned to the Decacovirus subgenus of the Alphacoronavirus genus and is most similar to viruses identified among Rhinolophus spp. from European and Middle Eastern countries. In the Altai Republic and Kemerovo region, coronaviruses belonging to the genus Alphacoronavirus, subgenus Pedacovirus, were found in the smooth-nosed bats: Ikonnikov`s bat (Myotis ikonnikovi) and the eastern bat (Myotis petax). The virus from the Altai Republic from M. ikonnikovi is close to viruses from Japan and Korea, as well as viruses from Myotis spp. from European countries. The virus from the Kemerovo region from M. petax groups with coronaviruses from Myotis spp. from Asian countries and is significantly different from coronaviruses previously discovered in the same natural host.


Subject(s)
Chiroptera , Animals , Chiroptera/virology , Siberia/epidemiology , Phylogeny , Disease Reservoirs/virology , Coronavirus/genetics , Coronavirus/isolation & purification , Coronavirus/classification , Humans , Feces/virology , SARS-CoV-2/genetics , SARS-CoV-2/isolation & purification , COVID-19/virology , COVID-19/epidemiology , COVID-19/veterinary , Coronavirus Infections/virology , Coronavirus Infections/veterinary , Coronavirus Infections/epidemiology
6.
Vet Microbiol ; 295: 110163, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38959807

ABSTRACT

Avian influenza virus (AIV) infection and vaccination against live attenuated infectious bronchitis virus (aIBV) are frequent in poultry worldwide. Here, we evaluated the clinical effect of H9N2 subtype AIV and QX genotype aIBV co-infection in specific-pathogen-free (SPF) white leghorn chickens and explored the potential mechanisms underlying the observed effects using by 4D-FastDIA-based proteomics. The results showed that co-infection of H9N2 AIV and QX aIBV increased mortality and suppressed the growth of SPF chickens. In particular, severe lesions in the kidneys and slight respiratory signs similar to the symptoms of virulent QX IBV infection were observed in some co-infected chickens, with no such clinical signs observed in single-infected chickens. The replication of H9N2 AIV was significantly enhanced in both the trachea and kidneys, whereas there was only a slight effect on the replication of the QX aIBV. Proteomics analysis showed that the IL-17 signaling pathway was one of the unique pathways enriched in co-infected chickens compared to single infected-chickens. A series of metabolism and immune response-related pathways linked with co-infection were also significantly enriched. Moreover, co-infection of the two pathogens resulted in the enrichment of the negative regulation of telomerase activity. Collectively, our study supports the synergistic effect of the two pathogens, and pointed out that aIBV vaccines might increased IBV-associated lesions due to pathogenic co-infections. Exacerbation of the pathogenicity and mortality in H9N2 AIV and QX aIBV co-infected chickens possibly occurred because of an increase in H9N2 AIV replication, the regulation of telomerase activity, and the disturbance of cell metabolism and the immune system.


Subject(s)
Chickens , Coinfection , Coronavirus Infections , Infectious bronchitis virus , Influenza A Virus, H9N2 Subtype , Influenza in Birds , Poultry Diseases , Animals , Chickens/virology , Influenza A Virus, H9N2 Subtype/pathogenicity , Influenza A Virus, H9N2 Subtype/genetics , Infectious bronchitis virus/pathogenicity , Infectious bronchitis virus/genetics , Coinfection/virology , Coinfection/veterinary , Influenza in Birds/virology , Poultry Diseases/virology , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Specific Pathogen-Free Organisms , Virus Replication , Vaccines, Attenuated/immunology , Genotype , Virulence , Proteomics , Kidney/virology , Kidney/pathology
7.
PLoS Pathog ; 20(7): e1012039, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38950065

ABSTRACT

The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) not only caused the COVID-19 pandemic but also had a major impact on farmed mink production in several European countries. In Denmark, the entire population of farmed mink (over 15 million animals) was culled in late 2020. During the period of June to November 2020, mink on 290 farms (out of about 1100 in the country) were shown to be infected with SARS-CoV-2. Genome sequencing identified changes in the virus within the mink and it is estimated that about 4000 people in Denmark became infected with these mink virus variants. However, the routes of transmission of the virus to, and from, the mink have been unclear. Phylogenetic analysis revealed the generation of multiple clusters of the virus within the mink. Detailed analysis of changes in the virus during replication in mink and, in parallel, in the human population in Denmark, during the same time period, has been performed here. The majority of cases in mink involved variants with the Y453F substitution and the H69/V70 deletion within the Spike (S) protein; these changes emerged early in the outbreak. However, further introductions of the virus, by variants lacking these changes, from the human population into mink also occurred. Based on phylogenetic analysis of viral genome data, we estimate, using a conservative approach, that about 17 separate examples of mink to human transmission occurred in Denmark but up to 59 such events (90% credible interval: (39-77)) were identified using parsimony to count cross-species jumps on transmission trees inferred using Bayesian methods. Using the latter approach, 136 jumps (90% credible interval: (117-164)) from humans to mink were found, which may underlie the farm-to-farm spread. Thus, transmission of SARS-CoV-2 from humans to mink, mink to mink, from mink to humans and between humans were all observed.


Subject(s)
COVID-19 , Mink , Phylogeny , SARS-CoV-2 , Mink/virology , COVID-19/transmission , COVID-19/virology , COVID-19/epidemiology , COVID-19/veterinary , SARS-CoV-2/genetics , Animals , Denmark/epidemiology , Humans , Pandemics , Farms , Betacoronavirus/genetics , Betacoronavirus/classification , Genome, Viral , Coronavirus Infections/veterinary , Coronavirus Infections/epidemiology , Coronavirus Infections/virology , Coronavirus Infections/transmission , Spike Glycoprotein, Coronavirus/genetics
8.
Trop Anim Health Prod ; 56(6): 211, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39001969

ABSTRACT

Bovine coronavirus (BCoV) is one of the important causes of diarrhoea in cattle. The virus is responsible for the high fatality rate associated with acute diarrhoea in calves. Rapid and accurate tests need to be conducted to detect the virus and minimise economic losses associated with the disease. Nucleic acid-based detection assays including PCR is an accurate test for detecting pathogens. However, these tests need skilled personnel, time and expensive devices. In this study, we developed a novel assay for the detection of BCoV in clinical cases. This novel assay combined reverse transcription-recombinase polymerase amplification with CRISPR/Cas13 and conducted a rapid visualisation of cleavage activity using a Lateral Flow Device. A conserved sequence of the BCV M gene was used as a target gene and the assays were tested in terms of specificity, sensitivity and time consumption. The result showed the specificity of the assay as 100% with no false positives being detected. Ten copies of the input RNA were enough to detect the virus and perform the assay. It took up to forty minutes for reading the results. Conducted together, the assay should be used as a rapid test to clinically diagnose infectious pathogens including bovine coronavirus. However, the assay needed the RNA to be extracted from the clinical sample in order to detect the virus. Therefore, more studies are needed to optimise the assay to be able to detect the virus in the clinical sample without extracting the RNA.


Subject(s)
CRISPR-Cas Systems , Cattle Diseases , Coronavirus, Bovine , Diarrhea , Sensitivity and Specificity , Animals , Cattle , Coronavirus, Bovine/isolation & purification , Coronavirus, Bovine/genetics , Diarrhea/veterinary , Diarrhea/virology , Diarrhea/diagnosis , Cattle Diseases/virology , Cattle Diseases/diagnosis , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Coronavirus Infections/diagnosis
9.
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
10.
Mol Med ; 30(1): 92, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38898389

ABSTRACT

BACKGROUND: COVID-19 is a new infectious disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS CoV-2). Since the outbreak in December 2019, it has caused an unprecedented world pandemic, leading to a global human health crisis. Although SARS CoV-2 mainly affects the lungs, causing interstitial pneumonia and severe acute respiratory distress syndrome, a number of patients often have extensive clinical manifestations, such as gastrointestinal symptoms, cardiovascular damage and renal dysfunction. PURPOSE: This review article discusses the pathogenic mechanisms of cardiovascular damage in COVID-19 patients and provides some useful suggestions for future clinical diagnosis, treatment and prevention. METHODS: An English-language literature search was conducted in PubMed and Web of Science databases up to 12th April, 2024 for the terms "COVID-19", "SARS CoV-2", "cardiovascular damage", "myocardial injury", "myocarditis", "hypertension", "arrhythmia", "heart failure" and "coronary heart disease", especially update articles in 2023 and 2024. Salient medical literatures regarding the cardiovascular damage of COVID-19 were selected, extracted and synthesized. RESULTS: The most common cardiovascular damage was myocarditis and pericarditis, hypertension, arrhythmia, myocardial injury and heart failure, coronary heart disease, stress cardiomyopathy, ischemic stroke, blood coagulation abnormalities, and dyslipidemia. Two important pathogenic mechanisms of the cardiovascular damage may be direct viral cytotoxicity as well as indirect hyperimmune responses of the body to SARS CoV-2 infection. CONCLUSIONS: Cardiovascular damage in COVID-19 patients is common and portends a worse prognosis. Although the underlying pathophysiological mechanisms of cardiovascular damage related to COVID-19 are not completely clear, two important pathogenic mechanisms of cardiovascular damage may be the direct damage of the SARSCoV-2 infection and the indirect hyperimmune responses.


Subject(s)
COVID-19 , Cardiovascular Diseases , Pandemics , SARS-CoV-2 , Humans , COVID-19/complications , Cardiovascular Diseases/etiology , Pneumonia, Viral/complications , Pneumonia, Viral/immunology , Pneumonia, Viral/virology , Pneumonia, Viral/pathology , Coronavirus Infections/complications , Coronavirus Infections/immunology , Coronavirus Infections/virology , Betacoronavirus , Myocarditis/etiology , Myocarditis/virology
11.
Spectrochim Acta A Mol Biomol Spectrosc ; 320: 124670, 2024 Nov 05.
Article in English | MEDLINE | ID: mdl-38908108

ABSTRACT

Porcine epidemic diarrhea virus (PEDV) and rotavirus has posed a significant threat to the pig industry annually across different nations, resulting in huge economic losses. The frequent co-infection of these two viruses in clinical settings complicates the process of differential diagnoses. Rapid and accurate detection of PEDV and rotavirus is in great demand for timely diarrhea disease prevention and control. In this study, tris stabilized AuNPs were prepared and a sensitive lateral flow immunoassay (LFIA) sensor was developed for the simultaneous and rapid detection of PEDV and rotavirus on site. After the system optimization, the established LFIA can simultaneously identify PEDV and rotavirus with limits of detection (LOD) of 1.25 × 103 TCID50 mL-1 and 3.13 × 102 pg mL-1, respectively. When applying for clinical samples, the LFIA show a concordance of 95 % and 100 % to reverse transcript polymerase chain reaction (RT-PCR) for PEDV and rotavirus respectively. Therefore, this LFIA can qualitatively detect PEDV and rotavirus in 18 min with high sensitivity and accuracy without any sophisticated equipment and operation, making it a promising candidate for the early diagnosis of PEDV or/and rotavirus diarrhea on site.


Subject(s)
Chromatography, Affinity , Gold , Metal Nanoparticles , Porcine epidemic diarrhea virus , Rotavirus , Gold/chemistry , Porcine epidemic diarrhea virus/isolation & purification , Rotavirus/isolation & purification , Animals , Metal Nanoparticles/chemistry , Swine , Chromatography, Affinity/methods , Limit of Detection , Rotavirus Infections/diagnosis , Rotavirus Infections/veterinary , Rotavirus Infections/virology , Swine Diseases/diagnosis , Swine Diseases/virology , Immunoassay/methods , Coronavirus Infections/diagnosis , Coronavirus Infections/virology , Coronavirus Infections/veterinary
12.
PLoS Pathog ; 20(6): e1011777, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38913740

ABSTRACT

COVID-associated coagulopathy seemly plays a key role in post-acute sequelae of SARS- CoV-2 infection. However, the underlying pathophysiological mechanisms are poorly understood, largely due to the lack of suitable animal models that recapitulate key clinical and pathological symptoms. Here, we fully characterized AC70 line of human ACE2 transgenic (AC70 hACE2 Tg) mice for SARS-CoV-2 infection. We noted that this model is highly permissive to SARS-CoV-2 with values of 50% lethal dose and infectious dose as ~ 3 and ~ 0.5 TCID50 of SARS-CoV-2, respectively. Mice infected with 105 TCID50 of SARS-CoV-2 rapidly succumbed to infection with 100% mortality within 5 days. Lung and brain were the prime tissues harboring high viral titers, accompanied by histopathology. However, viral RNA and inflammatory mediators could be detectable in other organs, suggesting the nature of a systemic infection. Lethal challenge of AC70 hACE2 Tg mice caused acute onset of leukopenia, lymphopenia, along with an increased neutrophil-to-lymphocyte ratio (NLR). Importantly, infected animals recapitulated key features of COVID-19-associated coagulopathy. SARS-CoV-2 could induce the release of circulating neutrophil extracellular traps (NETs), along with activated platelet/endothelium marker. Immunohistochemical staining with anti-platelet factor-4 (PF4) antibody revealed profound platelet aggregates especially within blocked veins of the lungs. We showed that acute SARS-CoV-2 infection triggered a hypercoagulable state coexisting with ill-regulated fibrinolysis. Finally, we highlighted the potential role of Annexin A2 (ANXA2) in fibrinolytic failure. ANXA2 is a calcium-dependent phospholipid-binding protein that forms a heterotertrameric complexes localized at the extracellular membranes with two S100A10 small molecules acting as a co-receptor for tissue-plasminogen activator (t-PA), tightly involved in cell surface fibrinolysis. Thus, our results revealing elevated IgG type anti-ANXA2 antibody production, downregulated de novo ANXA2/S100A10 synthesis, and reduced ANXA2/S100A10 association in infected mice, this protein might serve as druggable targets for development of antithrombotic and/or anti-fibrinolytic agents to attenuate pathogenesis of COVID-19.


Subject(s)
Angiotensin-Converting Enzyme 2 , COVID-19 , Disease Models, Animal , Mice, Transgenic , SARS-CoV-2 , Animals , COVID-19/pathology , COVID-19/complications , COVID-19/virology , COVID-19/metabolism , Mice , Angiotensin-Converting Enzyme 2/metabolism , Angiotensin-Converting Enzyme 2/genetics , Humans , Blood Coagulation Disorders/virology , Blood Coagulation Disorders/pathology , Pneumonia, Viral/virology , Pneumonia, Viral/pathology , Pneumonia, Viral/metabolism , Betacoronavirus , Lung/virology , Lung/pathology , Lung/metabolism , Coronavirus Infections/virology , Coronavirus Infections/pathology , Coronavirus Infections/complications , Pandemics , Extracellular Traps/metabolism
13.
Viruses ; 16(6)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38932190

ABSTRACT

Human coronavirus 229E (HCoV-229E) is associated with upper respiratory tract infections and generally causes mild respiratory symptoms. HCoV-229E infection can cause cell death, but the molecular pathways that lead to virus-induced cell death as well as the interplay between viral proteins and cellular cell death effectors remain poorly characterized for HCoV-229E. Studying how HCoV-229E and other common cold coronaviruses interact with and affect cell death pathways may help to understand its pathogenesis and compare it to that of highly pathogenic coronaviruses. Here, we report that the main protease (Mpro) of HCoV-229E can cleave gasdermin D (GSDMD) at two different sites (Q29 and Q193) within its active N-terminal domain to generate fragments that are now unable to cause pyroptosis, a form of lytic cell death normally executed by this protein. Despite GSDMD cleavage by HCoV-229E Mpro, we show that HCoV-229E infection still leads to lytic cell death. We demonstrate that during virus infection caspase-3 cleaves and activates gasdermin E (GSDME), another key executioner of pyroptosis. Accordingly, GSDME knockout cells show a significant decrease in lytic cell death upon virus infection. Finally, we show that HCoV-229E infection leads to increased lytic cell death levels in cells expressing a GSDMD mutant uncleavable by Mpro (GSDMD Q29A+Q193A). We conclude that GSDMD is inactivated by Mpro during HCoV-229E infection, preventing GSDMD-mediated cell death, and point to the caspase-3/GSDME axis as an important player in the execution of virus-induced cell death. In the context of similar reported findings for highly pathogenic coronaviruses, our results suggest that these mechanisms do not contribute to differences in pathogenicity among coronaviruses. Nonetheless, understanding the interactions of common cold-associated coronaviruses and their proteins with the programmed cell death machineries may lead to new clues for coronavirus control strategies.


Subject(s)
Cell Death , Coronavirus 229E, Human , Intracellular Signaling Peptides and Proteins , Phosphate-Binding Proteins , Pyroptosis , Humans , Phosphate-Binding Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Coronavirus 229E, Human/physiology , Coronavirus 229E, Human/genetics , Coronavirus Infections/virology , Coronavirus Infections/metabolism , Neoplasm Proteins/metabolism , Neoplasm Proteins/genetics , Viral Proteins/metabolism , Viral Proteins/genetics , Cell Line , Host-Pathogen Interactions , HEK293 Cells , Gasdermins
14.
Viruses ; 16(6)2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38932222

ABSTRACT

Gammacoronavirus infectious bronchitis virus (IBV) causes a highly contagious disease in chickens and seriously endangers the poultry industry. The emergence and co-circulation of diverse IBV serotypes and genotypes with distinct pathogenicity worldwide pose a serious challenge to the development of effective intervention measures. In this study, we report the epidemic trends of IBV in China from 2019 to 2023 and a comparative analysis on the antigenic characteristics and pathogenicity of isolates among major prevalent lineages. Phylogenetic and recombination analyses based on the nucleotide sequences of the spike (S) 1 gene clustered a total of 205 isolates into twelve distinct lineages, with GI-19 as a predominant lineage (61.77 ± 4.56%) exhibiting an overall increasing trend over the past five years, and demonstrated that a majority of the variants were derived from gene recombination events. Further characterization of the growth and pathogenic properties of six representative isolates from different lineages classified four out of the six isolates as nephropathogenic types with mortality rates in one-day-old SPF chickens varying from 20-60%, one as a respiratory type with weak virulence, and one as a naturally occurring avirulent strain. Taken together, our findings illuminate the epidemic trends, prevalence, recombination, and pathogenicity of current IBV strains in China, providing key information for further strengthening the surveillance and pathogenicity studies of IBV.


Subject(s)
Chickens , Coronavirus Infections , Genetic Variation , Genotype , Infectious bronchitis virus , Phylogeny , Poultry Diseases , Animals , Infectious bronchitis virus/genetics , Infectious bronchitis virus/pathogenicity , Infectious bronchitis virus/classification , Infectious bronchitis virus/isolation & purification , China/epidemiology , Poultry Diseases/virology , Poultry Diseases/epidemiology , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Coronavirus Infections/epidemiology , Prevalence , Virulence , Recombination, Genetic , Serogroup
15.
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
16.
Viruses ; 16(6)2024 Jun 16.
Article in English | MEDLINE | ID: mdl-38932257

ABSTRACT

Bovine coronavirus (BCoV) poses a threat to cattle health worldwide, contributing to both respiratory and enteric diseases. However, few contemporary strains have been isolated. In this study, 71 samples (10 nasal and 61 fecal) were collected from one farm in Ohio in 2021 and three farms in Georgia in 2023. They were screened by BCoV-specific real-time reverse transcription-PCR, and 15 BCoV-positive samples were identified. Among them, five BCoV strains from fecal samples were isolated using human rectal tumor-18 (HRT-18) cells. The genomic sequences of five strains were obtained. The phylogenetic analysis illustrated that these new strains clustered with US BCoVs that have been detected since the 1990s. Sequence analyses of the spike proteins of four pairs of BCoVs, with each pair originally collected from the respiratory and enteric sites of one animal, revealed the potential amino acid residue patterns, such as D1180 for all four enteric BCoVs and G1180 for three of four respiratory BCoVs. This project provides new BCoV isolates and sequences and underscores the genetic diversity of BcoVs, the unknown mechanisms of disease types, and the necessity of sustained surveillance and research for BCoVs.


Subject(s)
Cattle Diseases , Coronavirus Infections , Coronavirus, Bovine , Feces , Phylogeny , Cattle , Animals , Coronavirus, Bovine/genetics , Coronavirus, Bovine/isolation & purification , Coronavirus, Bovine/classification , Feces/virology , Cattle Diseases/virology , Coronavirus Infections/virology , Coronavirus Infections/veterinary , Genome, Viral , Spike Glycoprotein, Coronavirus/genetics , Humans , Genetic Variation , Ohio
17.
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
18.
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
19.
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
20.
Sci Rep ; 14(1): 12928, 2024 06 05.
Article in English | MEDLINE | ID: mdl-38839918

ABSTRACT

Coronaviruses have been confirmed to infect a variety of species, but only one case of associated winter dysentery of European bison has been described. The study aimed to analyze the prevalence, and define the impact on the species conservation, the source of coronavirus infection, and the role of the European bison in the transmission of the pathogen in Poland. Molecular and serological screening was performed on 409 European bison from 6 free-ranging and 14 captive herds over the period of 6 years (2017-2023). Presence of coronavirus was confirmed in one nasal swab by pancoronavirus RT-PCR and in 3 nasal swab samples by bovine coronavirus (BCoV) specific real time RT-PCR. The detected virus showed high (> 98%) homology in both RdRp and Spike genes to BCoV strains characterised recently in Polish cattle and strains isolated from wild cervids in Italy. Antibodies specific to BCoV were found in 6.4% of tested samples, all originating from free-ranging animals. Seroprevalence was higher in adult animals over 5 years of age (p = 0.0015) and in females (p = 0.09). Our results suggest that European bison play only a limited role as reservoirs of bovine-like coronaviruses. Although the most probable source of infections in the European bison population in Poland is cattle, other wild ruminants could also be involved. In addition, the zoonotic potential of bovine coronaviruses is quite low.


Subject(s)
Bison , Coronavirus Infections , Animals , Bison/virology , Poland/epidemiology , Female , Male , Coronavirus Infections/epidemiology , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Prevalence , Coronavirus/genetics , Coronavirus/classification , Coronavirus/isolation & purification , Seroepidemiologic Studies , Cattle , Coronavirus, Bovine/genetics , Coronavirus, Bovine/isolation & purification , Phylogeny , Antibodies, Viral/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...