Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.349
Filter
1.
J Med Virol ; 96(9): e29902, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39228345

ABSTRACT

The whole-genome sequence (WGS) analysis of Aichivirus (AiV) identified in Korea was performed in this study. Using Sanger and Nanopore sequencing, the 8228-nucleotide-long genomic sequence of AiV (OQ121963) was determined and confirmed to belong to genotype A. The full-length genome of OQ121963 consisted of a 7296 nt open reading frame (ORF) that encodes a single polyprotein, and 5' UTR (676 nt) and 3' UTR (256 nt) at 5' and 3' ends, respectively. The ORF consisted of leader protein (L), structural protein P1 (VP0, VP1, and VP3), and nonstructural protein P2 (2A, 2B, and 2C) and P3 (3A, 3B, 3C, and 3D). The secondary structure analysis of the 5' UTR identified only stem-loop C (SL-C) and not SL-A and SL-B. The variable region of the AiV genome was analyzed by MegAlign Pro and reconfirmed by SimPlot analysis using 16 AiV whole genomes known to date. Among the entire regions, structural protein region P1 showed the lowest amino acid identity (96.07%) with reference sequence AB040749 (originated in Japan; genotype A), while the highest amino acid identity (98.26%) was confirmed in the 3D region among nonstructural protein region P2 and P3. Moreover, phylogenetic analysis of the WGS of OQ121963 showed the highest homology (96.96%) with JX564249 (originated in Taiwan; genotype A) and lowest homology (90.14%) with DQ028632 (originated in Brazil; genotype B). Therefore, the complete genome characterization of OQ121963 and phylogenetic analysis of the AiV conducted in this study provide useful information allowing to improve diagnostic tools and epidemiological studies of AiVs.


Subject(s)
Genome, Viral , Genotype , Kobuvirus , Open Reading Frames , Phylogeny , Whole Genome Sequencing , Genome, Viral/genetics , Republic of Korea , Humans , Kobuvirus/genetics , Kobuvirus/classification , Kobuvirus/isolation & purification , Picornaviridae Infections/virology , Picornaviridae Infections/epidemiology , 5' Untranslated Regions/genetics , Adult , RNA, Viral/genetics , 3' Untranslated Regions/genetics
2.
J Med Virol ; 96(9): e29833, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39233489

ABSTRACT

Rhinoviruses (RVs) are a leading cause of acute respiratory infections (ARI) in children. The relationship between RV viral loads (VL), RV/viral-co-detections and disease severity, is incompletely understood. We studied children and adolescents ≤21 years with RV-ARI that were identified as inpatients or outpatients using a PCR panel from 2011-2013. RV VL were stratified according to cycle threshold (CT) values in high (≤25), intermediate (26-32) and low (>32). Adjusted analyses were performed to assess the role RV VL and RV/viral codetections on hospital admission, oxygen requirement, PICU care, and length of stay. Of 1,899 children with RV-ARI, 78% had chronic comorbidities and 24% RV/viral co-detections. Single RV vs RV/viral co-detections was associated with higher VL (24.74 vs 26.62 CT; p = 0.001) and older age (14.9 vs 9.5 months; p = 0.0001). Frequency of RV/viral co-detections were inversely proportional to RV loads: 32% with low; 28% with intermediate, and 19% with high VL, p = 0.0001. Underlying conditions were independently associated with all clinical outcomes, high VL with PICU care, and single RV-ARI with higher odds of hospitalization. In summary, single RV vs RV/viral co-detections were associated with higher VL and older age. Underlying diseases, rather than RV loads or RV/viral co-detections, consistently predicted worse clinical outcomes.


Subject(s)
Coinfection , Comorbidity , Picornaviridae Infections , Respiratory Tract Infections , Rhinovirus , Severity of Illness Index , Viral Load , Humans , Rhinovirus/genetics , Rhinovirus/isolation & purification , Male , Female , Child , Child, Preschool , Adolescent , Infant , Picornaviridae Infections/epidemiology , Picornaviridae Infections/virology , Coinfection/virology , Coinfection/epidemiology , Respiratory Tract Infections/virology , Respiratory Tract Infections/epidemiology , Hospitalization/statistics & numerical data , Young Adult
3.
Virol J ; 21(1): 193, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39175061

ABSTRACT

BACKGROUND: Emerging zoonotic diseases arise from cross-species transmission events between wild or domesticated animals and humans, with bats being one of the major reservoirs of zoonotic viruses. Viral metagenomics has led to the discovery of many viruses, but efforts have mainly been focused on some areas of the world and on certain viral families. METHODS: We set out to describe full-length genomes of new picorna-like viruses by collecting feces from hundreds of bats captured in different regions of Spain. Viral sequences were obtained by high-throughput Illumina sequencing and analyzed phylogenetically to classify them in the context of known viruses. Linear discriminant analysis (LDA) was performed to infer likely hosts based on genome composition. RESULTS: We found five complete or nearly complete genomes belonging to the family Picornaviridae, including a new species of the subfamily Ensavirinae. LDA suggested that these were true vertebrate viruses, rather than viruses from the bat diet. Some of these viruses were related to picornaviruses previously found in other bat species from distant geographical regions. We also found a calhevirus genome that most likely belongs to a proposed new family within the order Picornavirales, and for which genome composition analysis suggested a plant host. CONCLUSIONS: Our findings describe new picorna-like viral species and variants circulating in the Iberian Peninsula, illustrate the wide geographical distribution and interspecies transmissibility of picornaviruses, and suggest new hosts for calheviruses.


Subject(s)
Chiroptera , Feces , Genetic Variation , Genome, Viral , Phylogeny , Picornaviridae Infections , Picornaviridae , Chiroptera/virology , Animals , Spain , Picornaviridae/genetics , Picornaviridae/classification , Picornaviridae/isolation & purification , Feces/virology , Picornaviridae Infections/transmission , Picornaviridae Infections/virology , Picornaviridae Infections/veterinary , High-Throughput Nucleotide Sequencing , Metagenomics , Humans , Zoonoses/virology , Zoonoses/transmission
4.
Viruses ; 16(8)2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39205161

ABSTRACT

The SARS-CoV-2 pandemic resulted in a scale-up of viral genomic surveillance globally. However, the wet lab constraints (economic, infrastructural, and personnel) of translating novel virus variant sequence information to meaningful immunological and structural insights that are valuable for the development of broadly acting countermeasures (especially for emerging and re-emerging viruses) remain a challenge in many resource-limited settings. Here, we describe a workflow that couples wastewater surveillance, high-throughput sequencing, phylogenetics, immuno-informatics, and virus capsid structure modeling for the genotype-to-serotype characterization of uncultivated picornavirus sequences identified in wastewater. Specifically, we analyzed canine picornaviruses (CanPVs), which are uncultivated and yet-to-be-assigned members of the family Picornaviridae that cause systemic infections in canines. We analyzed 118 archived (stored at -20 °C) wastewater (WW) samples representing a population of ~700,000 persons in southwest USA between October 2019 to March 2020 and October 2020 to March 2021. Samples were pooled into 12 two-liter volumes by month, partitioned (into filter-trapped solids [FTSs] and filtrates) using 450 nm membrane filters, and subsequently concentrated to 2 mL (1000×) using 10,000 Da MW cutoff centrifugal filters. The 24 concentrates were subjected to RNA extraction, CanPV complete capsid single-contig RT-PCR, Illumina sequencing, phylogenetics, immuno-informatics, and structure prediction. We detected CanPVs in 58.3% (14/24) of the samples generated 13,824,046 trimmed Illumina reads and 27 CanPV contigs. Phylogenetic and pairwise identity analyses showed eight CanPV genotypes (intragenotype divergence <14%) belonging to four clusters, with intracluster divergence of <20%. Similarity analysis, immuno-informatics, and virus protomer and capsid structure prediction suggested that the four clusters were likely distinct serological types, with predicted cluster-distinguishing B-cell epitopes clustered in the northern and southern rims of the canyon surrounding the 5-fold axis of symmetry. Our approach allows forgenotype-to-serotype characterization of uncultivated picornavirus sequences by coupling phylogenetics, immuno-informatics, and virus capsid structure prediction. This consequently bypasses a major wet lab-associated bottleneck, thereby allowing resource-limited settings to leapfrog from wastewater-sourced genomic data to valuable immunological insights necessary for the development of prophylaxis and other mitigation measures.


Subject(s)
High-Throughput Nucleotide Sequencing , Phylogeny , Picornaviridae , Wastewater , Picornaviridae/genetics , Picornaviridae/classification , Picornaviridae/isolation & purification , Animals , Dogs , Wastewater/virology , Capsid Proteins/genetics , Capsid Proteins/chemistry , Genome, Viral , Capsid/immunology , Capsid/chemistry , United States/epidemiology , Picornaviridae Infections/veterinary , Picornaviridae Infections/virology , Picornaviridae Infections/epidemiology , Dog Diseases/virology , Dog Diseases/epidemiology , Genotype , Genetic Variation
5.
Viruses ; 16(8)2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39205256

ABSTRACT

Rhinovirus C (RV-C) infects airway epithelial cells and is an important cause of acute respiratory disease in humans. To interrogate the mechanisms of RV-C-mediated disease, animal models are essential. Towards this, RV-C infection was recently reported in wild-type (WT) mice, yet, titers were not sustained. Therefore, the requirements for RV-C infection in mice remain unclear. Notably, prior work has implicated human cadherin-related family member 3 (CDHR3) and stimulator of interferon genes (STING) as essential host factors for virus uptake and replication, respectively. Here, we report that even though human (h) and murine (m) CDHR3 orthologs have similar tissue distribution, amino acid sequence homology is limited. Further, while RV-C can replicate in mouse lung epithelial type 1 (LET1) cells and produce infectious virus, we observed a significant increase in the frequency and intensity of dsRNA-positive cells following hSTING expression. Based on these findings, we sought to assess the impact of hCDHR3 and hSTING on RV-C infection in mice in vivo. Thus, we developed hCDHR3 transgenic mice, and utilized adeno-associated virus (AAV) to deliver hSTING to the murine airways. Subsequent challenge of these mice with RV-C15 revealed significantly higher titers 24 h post-infection in mice expressing both hCDHR3 and hSTING-compared to either WT mice, or mice with hCDHR3 or hSTING alone, indicating more efficient infection. Ultimately, this mouse model can be further engineered to establish a robust in vivo model, recapitulating viral dynamics and disease.


Subject(s)
Cadherin Related Proteins , Cadherins , Mice, Transgenic , Virus Replication , Animals , Mice , Humans , Cadherins/genetics , Cadherins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Epithelial Cells/virology , Disease Models, Animal , Enterovirus/physiology , Enterovirus/genetics , Cell Line , Picornaviridae Infections/virology , Mice, Inbred C57BL , Lung/virology
6.
Nat Commun ; 15(1): 7452, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39198414

ABSTRACT

The global epidemic of Mpox virus (MPXV) continues, and a local outbreak has occurred in Shenzhen city since June 2023. Herein, the evolutionary trajectory and characteristics of MPXV in 2023 were analyzed using 92 MPXV sequences from the Shenzhen outbreak and the available genomes from GISAID and GenBank databases. Phylogenetic tracing of the 92 MPXVs suggests that MPXVs in Shenzhen may have multiple sources of importation, and two main transmission chains have been established. The combination of phylogenetic relationships, epidemiological features, and mutation characteristics supports the emergence of a new lineage C.1.1. Together with the B.1 lineage diverging from the A.1 lineage, C.1.1 lineage diverging from the C.1 lineage may serve as another significant evolutionary events of MPXV. Moreover, increasing apolipoprotein B mRNA-editing catalytic polypeptide-like 3 (APOBEC3) related mutations, higher rate of missense mutations, and less mutations in the non-coding regions have been shown during MPXV evolution. Host regulation proteins of MPXV have accumulated considerable amino acid mutations since the B.1 lineage, and a lineage-defining APOBEC3-related mutation that disrupts the N2L gene encoding a viral innate immune modulator has been identified in the C.1.1 lineage. In summary, our study provides compelling evidence for the ongoing evolution of MPXV with specific features.


Subject(s)
Evolution, Molecular , Genome, Viral , Phylogeny , China/epidemiology , Humans , Genome, Viral/genetics , Mutation , Picornaviridae Infections/epidemiology , Picornaviridae Infections/virology , Disease Outbreaks , Genomics/methods , APOBEC Deaminases/genetics , Cytidine Deaminase/genetics
7.
Curr Microbiol ; 81(10): 309, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39150576

ABSTRACT

Clarifying the etiology of diarrhea cases of unknown cause is important in the fight against enteric infections. In this study, we aimed to investigate the role of canine kobuvirus (CaKoV), in cases of diarrhea of unknown origin in dogs. A total 121 swab samples from dogs with diarrhea were collected. Molecular analyses of the samples were performed. For this purpose, after the sequence reaction, a phylogenetic tree was created, and bioinformatics analyses were performed. The prevalence rate of CaKoV in the sampled population was determined as 16.5% (20/121). The presence of parvovirus and coronavirus, which are common viral agents in CaKoV-positive dogs, was determined as 35% (7/20) and 10% (2/20), respectively. The rate of dogs with only CaKoV detected was 65% (13/20). Phylogenetic analysis of CaKoV strains clustered together closely related to reference strains. There are very limited studies on the role of CaKoV in the etiology of diarrhea cases of unknown cause in dogs around the world. So far, only one study has been done on CaKoV in Turkey. In this report which includes molecular characterization and epidemiological data on CaKoV determined the importance of CaKoV in cases of diarrhea of unknown origin. More comprehensive studies are needed to better understand the pathogenesis, epidemiology, and biology of CaKoV and to determine effective strategies to combat it.


Subject(s)
Diarrhea , Dog Diseases , Kobuvirus , Phylogeny , Picornaviridae Infections , Dogs , Animals , Kobuvirus/genetics , Kobuvirus/isolation & purification , Kobuvirus/classification , Diarrhea/virology , Diarrhea/veterinary , Diarrhea/epidemiology , Dog Diseases/virology , Picornaviridae Infections/veterinary , Picornaviridae Infections/virology , Picornaviridae Infections/epidemiology , Turkey/epidemiology , Feces/virology
8.
Viruses ; 16(8)2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39205178

ABSTRACT

Equine rhinitis A (ERAV) and B (ERBV) viruses are respiratory pathogens with worldwide distribution. The current study aimed to determine the frequency of infection of ERAV and ERBV among horses and foals at Polish national studs, and to determine genetic variability within the viruses obtained. Virus-specific quantitative RT-PCR assays targeting a 5' untranslated region were used to screen nasal swabs collected from 621 horses at 16 national horse studs from throughout Poland, including 553 healthy horses and 68 horses with respiratory disease. A partial DNA polymerase gene was amplified and sequenced from the qRT-PCR-positive samples. The obtained sequences were analysed using phylogeny and genetic network analysis. None of the nasal swabs were positive for ERAV, whereas ERBV was found in 11/621 (1.78%) samples collected from 10 healthy horses and one foal affected by respiratory disease. Partial DNA polymerase gene sequence variability was correlated with individual horses and studs from which samples were collected when only Polish sequences were analysed, but there was no correlation between country of origin and ERBV sequence when Polish and international sequences were included in the network. The report presents the first detection of ERBV in Poland.


Subject(s)
Horse Diseases , Phylogeny , Picornaviridae Infections , Horses/virology , Animals , Poland/epidemiology , Horse Diseases/virology , Horse Diseases/epidemiology , Picornaviridae Infections/veterinary , Picornaviridae Infections/virology , Picornaviridae Infections/epidemiology , Prevalence , Genetic Variation , Erbovirus/genetics , Erbovirus/isolation & purification , Rhinovirus/genetics , Rhinovirus/isolation & purification , Rhinovirus/classification , Sequence Analysis, DNA
9.
J Med Virol ; 96(8): e29804, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39092809

ABSTRACT

Although rhinoviruses play a major role in exacerbations of childhood asthma, the presence of rhinovirus (RV) RNA in plasma, referred to as viremia, has been investigated in a few studies. The aim of the study was to investigate the presence of rhinovirus viremia at the time of asthma exacerbation and to describe the molecular characteristics of rhinoviruses associated with viremia. We conducted an observational, prospective, multicenter study in eight pediatric hospitals (VIRASTHMA2). Preschool-aged recurrent wheezers (1-5 years) hospitalized for a severe exacerbation were included. Reverse-transcription polymerase chain reaction (RT-PCR) and molecular typing for RV/enteroviruses (EV) were performed on nasal swabs and plasma. Plasma specimens were available for 105 children with positive RT-PCR for RV/EV in respiratory specimens. Thirty-six (34.3%) had positive viremia. In plasma, 28 (82.4%) of the typable specimens were RV-C, five (14.7%) were EV-D68, and one was RV-A (2.9%). In all cases, the RV/EV type was identical in the plasma and respiratory specimens. In conclusion, RV/EV viremia is frequent in severe exacerbations of preschool recurrent wheezers, particularly in RV-C infections.


Subject(s)
Asthma , Picornaviridae Infections , Rhinovirus , Viremia , Humans , Viremia/virology , Child, Preschool , Rhinovirus/genetics , Rhinovirus/isolation & purification , Rhinovirus/classification , Asthma/virology , Male , Female , Prospective Studies , Picornaviridae Infections/virology , Infant , RNA, Viral/genetics , Reverse Transcriptase Polymerase Chain Reaction , Plasma/virology
10.
Nat Commun ; 15(1): 5766, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38982052

ABSTRACT

Respiratory viruses are a major trigger of exacerbations in chronic obstructive pulmonary disease (COPD). Airway neutrophilia is a hallmark feature of stable and exacerbated COPD but roles played by neutrophil extracellular traps (NETS) in driving disease pathogenesis are unclear. Here, using human studies of experimentally-induced and naturally-occurring exacerbations we identify that rhinovirus infection induces airway NET formation which is amplified in COPD and correlates with magnitude of inflammation and clinical exacerbation severity. We show that inhibiting NETosis protects mice from immunopathology in a model of virus-exacerbated COPD. NETs drive inflammation during exacerbations through release of double stranded DNA (dsDNA) and administration of DNAse in mice has similar protective effects. Thus, NETosis, through release of dsDNA, has a functional role in the pathogenesis of COPD exacerbations. These studies open up the potential for therapeutic targeting of NETs or dsDNA as a strategy for treating virus-exacerbated COPD.


Subject(s)
Extracellular Traps , Neutrophils , Pulmonary Disease, Chronic Obstructive , Rhinovirus , Extracellular Traps/immunology , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/virology , Pulmonary Disease, Chronic Obstructive/pathology , Animals , Humans , Rhinovirus/immunology , Mice , Neutrophils/immunology , Male , Female , Picornaviridae Infections/immunology , Picornaviridae Infections/virology , Picornaviridae Infections/complications , Mice, Inbred C57BL , DNA/immunology , Disease Models, Animal , Middle Aged , Inflammation/immunology , Inflammation/virology , Aged
11.
Viruses ; 16(7)2024 Jun 25.
Article in English | MEDLINE | ID: mdl-39066186

ABSTRACT

A diagnostic investigation into an outbreak of fatal respiratory disease among young goats in Iowa, USA revealed bronchitis lesions of unknown etiology and secondary bacterial bronchopneumonia. Hypothesis-free metagenomics identified a previously unreported picornavirus (USA/IA26017/2023), and further phylogenetic analysis classified USA/IA26017/2023 as an aphthovirus related to bovine rhinitis B virus. Viral nucleic acid was localized to lesions of bronchitis using in situ hybridization. This marks the first report of a picornavirus putatively causing respiratory disease in goats and highlights the potential for cross-species transmission of aphthoviruses.


Subject(s)
Bronchitis , Goat Diseases , Goats , Phylogeny , Animals , Goat Diseases/virology , Bronchitis/virology , Bronchitis/veterinary , Picornaviridae Infections/veterinary , Picornaviridae Infections/virology , Iowa , Disease Outbreaks/veterinary , Metagenomics , Cattle
12.
PLoS Pathog ; 20(7): e1012398, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39038050

ABSTRACT

Inflammasomes play pivotal roles in inflammation by processing and promoting the secretion of IL-1ß. Caspase-1 is involved in the maturation of IL-1ß and IL-18, while human caspase-4 specifically processes IL-18. Recent structural studies of caspase-4 bound to Pro-IL-18 reveal the molecular basis of Pro-IL-18 activation by caspase-4. However, the mechanism of caspase-1 processing of pro-IL-1ß and other IL-1ß-converting enzymes remains elusive. Here, we observed that swine Pro-IL-1ß (sPro-IL-1ß) exists as an oligomeric precursor unlike monomeric human Pro-IL-1ß (hPro-IL-1ß). Interestingly, Seneca Valley Virus (SVV) 3C protease cleaves sPro-IL-1ß to produce mature IL-1ß, while it cleaves hPro-IL-1ß but does not produce mature IL-1ß in a specific manner. When the inflammasome is blocked, SVV 3C continues to activate IL-1ß through direct cleavage in porcine alveolar macrophages (PAMs). Through molecular modeling and mutagenesis studies, we discovered that the pro-domain of sPro-IL-1ß serves as an 'exosite' with its hydrophobic residues docking into a positively charged 3C protease pocket, thereby directing the substrate to the active site. The cleavage of sPro-IL-1ß generates a monomeric and active form of IL-1ß, initiating the downstream signaling. Thus, these studies provide IL-1ß is an inflammatory sensor that directly detects viral protease through an independent pathway operating in parallel with host inflammasomes.


Subject(s)
3C Viral Proteases , Inflammasomes , Interleukin-1beta , Picornaviridae , Viral Proteins , Animals , Interleukin-1beta/metabolism , 3C Viral Proteases/metabolism , Swine , Humans , Viral Proteins/metabolism , Inflammasomes/metabolism , Inflammation/metabolism , Picornaviridae Infections/metabolism , Picornaviridae Infections/virology , Cysteine Endopeptidases/metabolism , Species Specificity , Macrophages, Alveolar/virology , Macrophages, Alveolar/metabolism
13.
Vet Microbiol ; 296: 110198, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39067145

ABSTRACT

Senecavirus A (SVA) is a causative agent that can cause vesicular disease in swine, which causes a great threat to the swine husbandry in the world. Therefore, it is necessary to develop a vaccine that can effectively prevent the spread of SVA. In this study, we developed a 24-polymeric nano-scaffold using ß-annulus peptide from tomato bushy effect virus (TBSV) by coupling this antigen to SVA B cell epitope VP121-26 and VP2 proteins via linkers, respectively. The SVA-based nanoparticle protein of the VP1(B)-ß-VP2 was expressed and purified by low-cost prokaryotic system to prepare a SVA nanoparticle vaccine. The immunological protective effect of SVA nanoparticle vaccine was evaluated in mouse and swine models, respectively. The results suggested that both mice and swine could induce high levels SVA neutralizing antibodies and IgG antibodies after two doses immunization. In addition, the swine challenge protection experiment showed that the protection rate of immune SVA nanoparticle vaccine and SVA inactivated vaccine both were 80 %, while the negative control had no protection effect. It demonstrated that SVA nanoparticle vaccine effectively prevented SVA infection in swine. In summary, the preparation of SVA vaccine by using ß-annulus peptide is a promising candidate vaccine for prevent SVA transmission, and provides a new idea for the development of novel SVA vaccines.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Nanovaccines , Picornaviridae Infections , Picornaviridae , Swine Diseases , Viral Vaccines , Animals , Female , Mice , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Capsid Proteins/immunology , Mice, Inbred BALB C , Nanovaccines/administration & dosage , Nanovaccines/immunology , Picornaviridae/immunology , Picornaviridae Infections/veterinary , Picornaviridae Infections/prevention & control , Picornaviridae Infections/immunology , Picornaviridae Infections/virology , Swine , Swine Diseases/prevention & control , Swine Diseases/virology , Swine Diseases/immunology , Viral Structural Proteins/immunology , Viral Vaccines/immunology , Viral Vaccines/administration & dosage
14.
J Med Chem ; 67(15): 12945-12968, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39018526

ABSTRACT

Acute respiratory viral infections, such as pneumovirus and respiratory picornavirus infections, exacerbate disease in COPD and asthma patients. A research program targeting respiratory syncytial virus (RSV) led to the discovery of GS-7682 (1), a novel phosphoramidate prodrug of a 4'-CN-4-aza-7,9-dideazaadenosine C-nucleoside GS-646089 (2) with broad antiviral activity against RSV (EC50 = 3-46 nM), human metapneumovirus (EC50 = 210 nM), human rhinovirus (EC50 = 54-61 nM), and enterovirus (EC50 = 83-90 nM). Prodrug optimization for cellular potency and lung cell metabolism identified 5'-methyl [(S)-hydroxy(phenoxy)phosphoryl]-l-alaninate in combination with 2',3'-diisobutyrate promoieties as being optimal for high levels of intracellular triphosphate formation in vitro and in vivo. 1 demonstrated significant reductions of viral loads in the lower respiratory tract of RSV-infected African green monkeys when administered once daily via intratracheal nebulized aerosol. Together, these findings support additional evaluation of 1 and its analogues as potential therapeutics for pneumo- and picornaviruses.


Subject(s)
Antiviral Agents , Picornaviridae , Prodrugs , Respiratory Syncytial Virus Infections , Animals , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Prodrugs/pharmacology , Prodrugs/chemistry , Prodrugs/chemical synthesis , Chlorocebus aethiops , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Infections/virology , Humans , Picornaviridae/drug effects , Structure-Activity Relationship , Respiratory Syncytial Viruses/drug effects , Drug Discovery , Nucleosides/chemistry , Nucleosides/pharmacology , Picornaviridae Infections/drug therapy , Picornaviridae Infections/virology
15.
Viruses ; 16(6)2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38932208

ABSTRACT

Viruses from Picornaviridae family are known pathogens of poultry, although the information on their occurrence and pathogenicity in pigeons is scarce. In this research, efforts are made to broaden the knowledge on Megrivirus B and Pigeon picornavirus B prevalence, phylogenetic relationship with other avian picornaviruses and their possible connection with enteric disease in racing pigeons. As a result of Oxford Nanopore Sequencing, five Megrivirus and two pigeon picornavirus B-like genome sequences were recovered, among which three recombinant strains were detected. The recombinant fragments represented an average of 10.9% and 25.5% of the genome length of the Pigeon picornavirus B and Megrivirus B reference strains, respectively. The phylogenetic analysis revealed that pigeons are carriers of species-specific picornaviruses. TaqMan qPCR assays revealed 7.8% and 19.0% prevalence of Megrivirus B and 32.2% and 39.7% prevalence of Pigeon picornavirus B in the group of pigeons exhibiting signs of enteropathy and in the group of asymptomatic pigeons, respectively. In turn, digital droplet PCR showed a considerably higher number of genome copies of both viruses in sick than in asymptomatic pigeons. The results of quantitative analysis leave the role of picornaviruses in enteropathies of pigeons unclear.


Subject(s)
Bird Diseases , Columbidae , Genome, Viral , Phylogeny , Picornaviridae Infections , Picornaviridae , Animals , Columbidae/virology , Picornaviridae/genetics , Picornaviridae/classification , Picornaviridae/isolation & purification , Bird Diseases/virology , Picornaviridae Infections/veterinary , Picornaviridae Infections/virology , Recombination, Genetic
16.
Viruses ; 16(6)2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38932217

ABSTRACT

Wheezing children infected with rhinovirus (RV) have a markedly increased risk of subsequently developing recurrencies and asthma. No previous studies have assessed the association between cytokine response and the severity of acute illness in the first wheezing episode in children infected with RV. Forty-seven children treated both as inpatients and as outpatients infected with RV only, aged 3-23 months, with severe first wheezing episodes were recruited. During acute illness, peripheral blood mononuclear cells (PBMCs) were isolated and stimulated with anti-CD3/anti-CD28 in vitro. A multiplex ELISA was used to quantitatively identify 56 different cytokines. The mean age of the children was 17 months, 74% were males, 79% were hospitalized, and 33% were sensitized. In adjusted analyses, the inpatient group was characterized by decreased expressions of interferon gamma (IFN-γ), interleukin 10 (IL-10), macrophage inflammatory protein 1 alpha (MIP-1α), RANTES (CCL5), and tumor necrosis factor-alpha (TNF-α) and an increased expression of ENA-78 (CXCL5) compared to the outpatient group. The cytokine response profiles from the PBMCs were different between the inpatient and outpatient groups. Our results support that firmly controlled interplay between pro-inflammatory and anti-inflammatory responses are required during acute viral infection to absolve the initial infection leading, to less severe illness.


Subject(s)
Cytokines , Leukocytes, Mononuclear , Picornaviridae Infections , Respiratory Sounds , Rhinovirus , Humans , Male , Rhinovirus/immunology , Female , Cytokines/metabolism , Infant , Respiratory Sounds/etiology , Picornaviridae Infections/immunology , Picornaviridae Infections/virology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/virology , Severity of Illness Index
17.
Viruses ; 16(6)2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38932262

ABSTRACT

Hepatitis A virus (HAV), a member of the genus Hepatovirus (Picornaviridae HepV), remains a significant viral pathogen, frequently causing enterically transmitted hepatitis worldwide. In this study, we conducted an epidemiological survey of HepVs carried by small terrestrial mammals in the wild in Yunnan Province, China. Utilizing HepV-specific broad-spectrum RT-PCR, next-generation sequencing (NGS), and QNome nanopore sequencing (QNS) techniques, we identified and characterized two novel HepVs provisionally named EpMa-HAV and EpLe-HAV, discovered in the long-tailed mountain shrew (Episoriculus macrurus) and long-tailed brown-toothed shrew (Episoriculus leucops), respectively. Our sequence and phylogenetic analyses of EpMa-HAV and EpLe-HAV indicated that they belong to the species Hepatovirus I (HepV-I) clade II, also known as the Chinese shrew HepV clade. Notably, the codon usage bias pattern of novel shrew HepVs is consistent with that of previously identified Chinese shrew HepV. Furthermore, our structural analysis demonstrated that shrew HepVs differ from other mammalian HepVs in RNA secondary structure and exhibit variances in key protein sites. Overall, the discovery of two novel HepVs in shrews expands the host range of HepV and underscores the existence of genetically diverse animal homologs of human HAV within the genus HepV.


Subject(s)
Genome, Viral , Phylogeny , Shrews , Animals , Shrews/virology , China/epidemiology , RNA, Viral/genetics , Genomics/methods , High-Throughput Nucleotide Sequencing , Picornaviridae Infections/veterinary , Picornaviridae Infections/virology , Picornaviridae Infections/epidemiology
18.
Mol Biol Rep ; 51(1): 777, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38904698

ABSTRACT

BACKGROUND: Senecavirus A (SV-A) is an RNA virus that belongs to the genus Senecavirus within the family Picornaviridae. This study aimed to analyze factors that can influence the molecular diagnosis of Senecavirus A, such as oligonucleotides, RNA extraction methods, and RT-qPCR kits. METHODS: Samples from suspected cases of vesicular disease in Brazilian pigs were analyzed for foot-and-mouth disease, swine vesicular disease, and vesicular stomatitis. All tested negative for these diseases but positive for SV-A. RT-qPCR tests were used, comparing different reagent kits and RNA extraction methods. Sensitivity and repeatability were evaluated, demonstrating efficacy in detecting SV-A in clinical samples. RESULTS: In RNA extraction, significant reduction in Cq values was observed with initial dilutions, particularly with larger supernatant volumes. Trizol and Maxwell showed greater sensitivity in automated equipment protocols, though results varied in tissue tests. RT-qPCR kit comparison revealed differences in amplification using viral RNA but minimal differences with plasmid DNA. Sensitivity among methods was comparable, with slight variations in non-amplified samples. Repeatability tests showed consistent results among RT-qPCRs, demonstrating similarity between methods despite minor discrepancies in Cq values. CONCLUSIONS: Trizol, silica columns, and semi-automated extraction were compared, as well as different RT-qPCR kits. The study found significant variations that could impact the final diagnosis.


Subject(s)
Picornaviridae Infections , Picornaviridae , RNA, Viral , Swine Diseases , Animals , Picornaviridae/genetics , Picornaviridae/isolation & purification , Swine , Picornaviridae Infections/diagnosis , Picornaviridae Infections/veterinary , Picornaviridae Infections/virology , RNA, Viral/genetics , Swine Diseases/virology , Swine Diseases/diagnosis , Sensitivity and Specificity , Real-Time Polymerase Chain Reaction/methods , Swine Vesicular Disease/diagnosis , Swine Vesicular Disease/virology , Foot-and-Mouth Disease/diagnosis , Foot-and-Mouth Disease/virology , Brazil , Reproducibility of Results
19.
J Med Virol ; 96(6): e29755, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38922896

ABSTRACT

Throughout the COVID-19 pandemic, rhinovirus (RV) remained notable persistence, maintaining its presence while other seasonal respiratory viruses were largely suppressed by pandemic restrictions during national lockdowns. This research explores the epidemiological dynamics of RV infections among pediatric populations on Hainan Island, China, specifically focusing on the impact before and after the zero-COVID policy was lifted. From January 2021 to December 2023, 19 680 samples were collected from pediatric patients hospitalized with acute lower respiratory tract infections (ARTIs) at the Hainan Maternal and Child Health Hospital. The infection of RV was detected by tNGS. RV species and subtypes were identified in 32 RV-positive samples representing diverse time points by analyzing the VP4/VP2 partial regions. Among the 19 680 pediatric inpatients with ARTIs analyzed, 21.55% were found to be positive for RV infection, with notable peaks observed in April 2021 and November 2022. A gradual annual decline in RV infections was observed, alongside a seasonal pattern of higher prevalence during the colder months. The highest proportion of RV infections was observed in the 0-1-year age group. Phylogenetic analysis on 32 samples indicated a trend from RV-A to RV-C in 2022. This observation suggests potential evolving dynamics within the RV species although further studies are needed due to the limited sample size. The research emphasizes the necessity for ongoing surveillance and targeted management, particularly for populations highly susceptible to severe illnesses caused by RV infections.


Subject(s)
COVID-19 , Genetic Variation , Phylogeny , Picornaviridae Infections , Respiratory Tract Infections , Rhinovirus , Humans , Rhinovirus/genetics , Rhinovirus/classification , Rhinovirus/isolation & purification , China/epidemiology , Infant , Child, Preschool , Picornaviridae Infections/epidemiology , Picornaviridae Infections/virology , Child , Female , Male , COVID-19/epidemiology , COVID-19/virology , Respiratory Tract Infections/virology , Respiratory Tract Infections/epidemiology , Infant, Newborn , Seasons , Adolescent , Prevalence , Child, Hospitalized/statistics & numerical data , SARS-CoV-2/genetics , Hospitalization/statistics & numerical data
20.
J Virol ; 98(7): e0058524, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38869319

ABSTRACT

Senecavirus A (SVA), a picornavirus, causes vesicular diseases and epidemic transient neonatal losses in swine, resulting in a multifaceted economic impact on the swine industry. SVA counteracts host antiviral response through multiple strategies facilitatng viral infection and transmission. However, the mechanism of how SVA modulates interferon (IFN) response remains elusive. Here, we demonstrate that SVA 3C protease (3Cpro) blocks the transduction of Janus kinase-signal transducer and activator of transcription (JAK-STAT) signaling pathway to antagonize type I IFN response. Mechanistically, 3Cpro selectively cleaves and degrades STAT1 and STAT2 while does not target JAK1, JAK2, and IRF9, through its protease activity. Notably, SVA 3Cpro cleaves human and porcine STAT1 on a Leucine (L)-Aspartic acid (D) motif, specifically L693/D694. In the case of STAT2, two cleavage sites were identified: glutamine (Q) 707 was identified in both human and porcine, while the second cleavage pattern differed, with residues 754-757 (Valine-Leucine-Glutamine-Serine motifs) in human STAT2 and Q758 in porcine STAT2. These cleavage patterns by SVA 3Cpro partially differ from previously reported classical motifs recognized by other picornaviral 3Cpro, highlighting the distinct characteristics of SVA 3Cpro. Together, these results reveal a mechanism by which SVA 3Cpro antagonizes IFN-induced antiviral response but also expands our knowledge about the substrate recognition patterns for picornaviral 3Cpro.IMPORTANCESenecavirus A (SVA), the only member in the Senecavirus genus within the Picornaviridae family, causes vesicular diseases in pigs that are clinically indistinguishable from foot-and-mouth disease (FMD), a highly contagious viral disease listed by the World Organization for Animal Health (WOAH). Interferon (IFN)-mediated antiviral response plays a pivotal role in restricting and controlling viral infection. Picornaviruses evolved numerous strategies to antagonize host antiviral response. However, how SVA modulates the JAK-STAT signaling pathway, influencing the type I IFN response, remains elusive. Here, we identify that 3Cpro, a protease of SVA, functions as an antagonist for the IFN response. 3Cpro utilizes its protease activity to cleave STAT1 and STAT2, thereby diminishing the host IFN response to promote SVA infection. Our findings underscore the significance of 3Cpro as a key virulence factor in the antagonism of the type I signaling pathway during SVA infection.


Subject(s)
Cysteine Endopeptidases , Picornaviridae Infections , Picornaviridae , STAT1 Transcription Factor , STAT2 Transcription Factor , Signal Transduction , Viral Proteins , Animals , Swine , STAT2 Transcription Factor/metabolism , Humans , Picornaviridae Infections/virology , Picornaviridae Infections/immunology , Picornaviridae Infections/metabolism , STAT1 Transcription Factor/metabolism , Cysteine Endopeptidases/metabolism , Viral Proteins/metabolism , 3C Viral Proteases , Swine Diseases/virology , Swine Diseases/immunology , HEK293 Cells , Interferon Type I/antagonists & inhibitors , Interferon Type I/metabolism , Cell Line , Janus Kinases/metabolism , Janus Kinases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL