Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.757
Filtrer
1.
Vet Med Sci ; 10(4): e1523, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38958584

RÉSUMÉ

BACKGROUND: Canine parvovirus type 2 (CPV-2) is the most common enteric virus that infects canids. CPV is the causative agent of a contagious disease defined mostly by clinical gastrointestinal signs in dogs. During the late 1970s, CPV-2 emerged as a new virus capable of infecting domestic dogs and growing across the world. The VP2 gene stands out as a key determinant in the pathogenicity, antigenicity, and host interactions of CPV-2. AIMS: The molecular characterization of the VP2 gene is crucial for understanding CPV evolution and epidemiology. MATERIALS & METHODS: Genes encoding the VP2 protein were sequenced and compared to reference strains worldwide. The maximum likelihood method was used to build a phylogenetic tree using CPV VP2 gene nucleotide sequences. RESULTS: Our phylogenetic analysis of the VP2 gene revealed that five strains were very similar and clustered together, and three strains were in the 2b clade, whereas the other two were in the 2a/2b clade. DISCUSSION: This paper reports the molecular characterization of two novel CPV-2a/2b subtypes in dogs with gastrointestinal symptoms. Genetic analysis was conducted on a CPV genomic region encompassing one of the open reading frames (ORFs) encoding the structural protein VP2. Sequence analysis indicates new and unreported sequence changes, mainly affecting the VP2 gene, which includes the mutations Ser297Ala and Leu87Met. This study represents the first evidence of a new CPV-2a/2b subtype in Türkiye. Due to VP2's crucial role in encoding the capsid protein of CPV-2 and its significant involvement in the host-virus interaction, it is critical to closely monitor its evolutionary changes and be cautious while searching for novel or pre-existing subtypes. CONCLUSION: This study highlights the significance of continuous molecular research for acquiring more insights on the circulation of novel CPV mutants.


Sujet(s)
Variation génétique , Parvovirus canin , Parvovirus canin/classification , Parvovirus canin/génétique , Animaux , Chiens , Phylogenèse , Protéines de capside/composition chimique , Protéines de capside/génétique , Maladies gastro-intestinales/médecine vétérinaire , Maladies gastro-intestinales/virologie , Infections à Parvoviridae/médecine vétérinaire , Infections à Parvoviridae/virologie , Turquie , Spécificité d'espèce , Génotype
2.
Virulence ; 15(1): 2366874, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38869140

RÉSUMÉ

Recombinant Muscovy duck parvovirus (rMDPV) is a product of genetic recombination between classical Muscovy duck parvovirus (MDPV) and goose parvovirus (GPV). The recombination event took place within a 1.1-kb DNA segment located in the middle of the VP3 gene, and a 187-bp sequence extending from the P9 promoter to the 5' initiation region of the Rep1 ORF. This resulted in the alteration of five amino acids within VP3. Despite these genetic changes, the precise influence of recombination and amino acid mutations on the pathogenicity of rMDPV remains ambiguous. In this study, based on the rMDPV strain ZW and the classical MDPV strain YY, three chimeric viruses (rZW-mP9, rZW-mPR187, and rYY-rVP3) and the five amino acid mutations-introduced mutants (rZW-g5aa and rYY-5aa(ZW)) were generated using reverse genetic technology. When compared to the parental virus rZW, rZW-g5aa exhibited a prolonged mean death time (MDT) and a decreased median lethal dose (ELD50) in embryonated duck eggs. In contrast, rYY-5aa(ZW) did not display significant differences in MDT and ELD50 compared to rYY. In 2-day-old Muscovy ducklings, infection with rZW-g5aa and rYY-5aa(ZW) resulted in mortality rates of only 20% and 10%, respectively, while infections with the three chimeric viruses (rZW-mP9, rZW-mPR187, rYY-rVP3) and rZW still led to 100% mortality. Notably, rYY-rVP3, containing the VP3 region from strain ZW, exhibited 50% mortality in 6-day-old Muscovy ducklings and demonstrated significant horizontal transmission. Collectively, our findings indicate that recombination and consequent amino acid changes in VP3 have a synergistic impact on the heightened virulence of rMDPV in Muscovy ducklings.


Sujet(s)
Protéines de capside , Canards , Infections à Parvoviridae , Mutation ponctuelle , Maladies de la volaille , Recombinaison génétique , Animaux , Virulence , Infections à Parvoviridae/virologie , Infections à Parvoviridae/médecine vétérinaire , Maladies de la volaille/virologie , Protéines de capside/génétique , Parvovirinae/génétique , Parvovirinae/pathogénicité
3.
Proc Natl Acad Sci U S A ; 121(25): e2320782121, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38875150

RÉSUMÉ

Human bocavirus 1 (HBoV1) is a human parvovirus that causes lower respiratory tract infections in young children. It contains a single-stranded (ss) DNA genome of ~5.5 kb that encodes a small noncoding RNA of 140 nucleotides known as bocavirus-encoded small RNA (BocaSR), in addition to viral proteins. Here, we determined the secondary structure of BocaSR in vivo by using DMS-MaPseq. Our findings reveal that BocaSR undergoes N6-methyladenosine (m6A) modification at multiple sites, which is critical for viral DNA replication in both dividing HEK293 cells and nondividing cells of the human airway epithelium. Mechanistically, we found that m6A-modified BocaSR serves as a mediator for recruiting Y-family DNA repair DNA polymerase (Pol) η and Pol κ likely through a direct interaction between BocaSR and the viral DNA replication origin at the right terminus of the viral genome. Thus, this report represents direct involvement of a viral small noncoding RNA in viral DNA replication through m6A modification.


Sujet(s)
Adénosine , Réplication de l'ADN , ADN viral , DNA-directed DNA polymerase , ARN viral , Réplication virale , Humains , Adénosine/analogues et dérivés , Adénosine/métabolisme , Réplication virale/génétique , DNA-directed DNA polymerase/métabolisme , DNA-directed DNA polymerase/génétique , ADN viral/génétique , ADN viral/métabolisme , Cellules HEK293 , ARN viral/génétique , ARN viral/métabolisme , Bocavirus humain/génétique , Bocavirus humain/métabolisme , Génome viral/génétique , Infections à Parvoviridae/virologie
4.
Viruses ; 16(6)2024 Jun 08.
Article de Anglais | MEDLINE | ID: mdl-38932224

RÉSUMÉ

Porcine parvoviruses (PPVs) are among the most important agents of reproductive failure in swine worldwide. PPVs comprise eight genetically different species ascribed to four genera: Protoparvovirus (PPV1, PPV8), Tetraparvovirus (PPV2-3), Copiparvovirus (PPV4-6), and Chaphamaparvovirus (PPV7). In 2016, PPV7 was firstly detected in the USA and afterwards in Europe, Asia, and South America. Recently, it was also identified in Italy in pig farms with reproductive failure. This study aimed to evaluate the circulation of PPV7 in domestic and wild pigs in Sardinia, Italy. In addition, its coinfection with Porcine Circovirus 2 (PCV2) and 3 (PCV3) was analysed, and PPV7 Italian strains were molecularly characterised. PPV7 was detected in domestic pigs and, for the first time, wild pigs in Italy. The PPV7 viral genome was detected in 20.59% of domestic and wild pig samples. PPV7 detection was significantly lower in domestic pigs, with higher PCV2/PCV3 co-infection rates observed in PPV7-positive than in PPV7-negative domestic pigs. Molecular characterisation of the NS1 gene showed a very high frequency of recombination that could presumably promote virus spreading.


Sujet(s)
Co-infection , Infections à Parvoviridae , Parvovirus porcin , Phylogenèse , Maladies des porcs , Animaux , Parvovirus porcin/génétique , Parvovirus porcin/classification , Parvovirus porcin/isolement et purification , Italie/épidémiologie , Infections à Parvoviridae/médecine vétérinaire , Infections à Parvoviridae/épidémiologie , Infections à Parvoviridae/virologie , Suidae , Maladies des porcs/virologie , Maladies des porcs/épidémiologie , Co-infection/virologie , Co-infection/médecine vétérinaire , Co-infection/épidémiologie , Génome viral , Circovirus/génétique , Circovirus/classification , Circovirus/isolement et purification , Infections à Circoviridae/médecine vétérinaire , Infections à Circoviridae/virologie , Infections à Circoviridae/épidémiologie , ADN viral/génétique
5.
Arch Virol ; 169(7): 139, 2024 Jun 08.
Article de Anglais | MEDLINE | ID: mdl-38849620

RÉSUMÉ

Amdoparvoviruses infect various carnivores, including mustelids, canids, skunks, and felids. Aleutian mink disease virus (AMDV) belongs to the prototypical species Amdoparvovirus carnivoran1. Here, we identified a novel amdoparvovirus in farmed Asian badgers (Meles meles), and we named this virus "Meles meles amdoparvovirus" (MMADV). A total of 146 clinical samples were collected from 134 individual badgers, and 30.6% (41/134) of the sampled badgers tested positive for amdoparvovirus by PCR. Viral DNA was detected in feces, blood, spleen, liver, lung, and adipose tissue from these animals. Viral sequences from eight samples were determined, five of which represented nearly full-length genome sequences (4,237-4,265 nt). Six serum samples tested positive by PCR, CIEP, and IAT, four of which had high antibody titers (> 512) against AMDV-G. Twenty-six of the 41 amdoparvovirus-positive badgers showed signs of illness, and necropsy revealed lesions in their organs. Sequence comparisons and phylogenetic analysis of the viral NS1 and VP2 genes of these badger amdoparvoviruses showed that their NS1 proteins shared 62.6%-88.8% sequence identity with known amdoparvoviruses, and they clustered phylogenetically into two related clades. The VP2 proteins shared 76.6%-97.2% identity and clustered into two clades, one of which included raccoon dog and arctic fox amdoparvovirus (RFAV), and the other of which did not include other known amdoparvoviruses. According to the NS1-protein-based criterion for parvovirus species demarcation, the MMADV isolate from farm YS should be classified as a member of a new species of the genus Amdoparvovirus. In summary, we have discovered a novel MMADV and other badger amdoparvoviruses that naturally infect Asian badgers and are possibly pathogenic in badgers.


Sujet(s)
Virus de la maladie aléoutienne du vison , Mustelidae , Phylogenèse , Animaux , Mustelidae/virologie , Virus de la maladie aléoutienne du vison/génétique , Virus de la maladie aléoutienne du vison/isolement et purification , Virus de la maladie aléoutienne du vison/classification , ADN viral/génétique , Génome viral/génétique , Infections à Parvoviridae/médecine vétérinaire , Infections à Parvoviridae/virologie , Maladie aléoutienne du vison/virologie , Maladie aléoutienne du vison/épidémiologie , Anticorps antiviraux/sang
6.
Virol J ; 21(1): 132, 2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38844968

RÉSUMÉ

Tetraparvovirus is an emerging parvovirus infecting a variety of mammals and humans, and associated with human diseases including severe acute respiratory infection and acute encephalitis syndrome. In the present study, a Tetraparvovirus ungulate 1 (formerly known as bovine hokovirus) strain HNU-CBY-2023 was identified and characterized from diseased Chinese Simmental from Hunan province, China. The nearly complete genome of HNU-CBY-2023 is 5346 nt in size and showed genomic identities of 85-95.5% to the known Tetraparvovirus ungulate 1 strains from GenBank, indicating a rather genetic variation. Phylogenetic and genetic divergence analyses indicated that Tetraparvovirus ungulate 1 could be divided into two genotypes (I and II), and HNU-CBY-2023 was clustered into genotype II. This study, for the first time, identified Tetraparvovirus ungulate 1 from domestic cattle from mainland China, which will be helpful to understand the prevalence and genetic diversity of Tetraparvovirus ungulate 1.


Sujet(s)
Maladies des bovins , Variation génétique , Génome viral , Génotype , Infections à Parvoviridae , Phylogenèse , Animaux , Bovins , Chine , Maladies des bovins/virologie , Maladies des bovins/épidémiologie , Infections à Parvoviridae/médecine vétérinaire , Infections à Parvoviridae/virologie , Infections à Parvoviridae/épidémiologie , Génome viral/génétique , Parvovirinae/génétique , Parvovirinae/isolement et purification , Parvovirinae/classification , Analyse de séquence d'ADN , ADN viral/génétique , Peuples d'Asie de l'Est
7.
J Med Virol ; 96(6): e29706, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38888111

RÉSUMÉ

The diversity and evolution of the genomes of human bocavirus (HBoV), which causes respiratory diseases, have been scarcely studied. Here, we aimed to obtain and characterize HBoV genomes from patients's nasopharyngeal samples collected between 2017 and 2022 period (5 years and 7 months). Next-generation sequencing (NGS) used Illumina technology after having implemented using GEMI an in-house multiplex PCR amplification strategy. Genomes were assembled and analyzed with CLC Genomics, Mafft, BioEdit, MeV, Nextclade, MEGA, and iTol. A total of 213 genomes were obtained. Phylogeny classified them all as of Bocavirus 1 (HBoV1) species. Five HBoV1 genotypic clusters determined by hierarchical clustering analysis of 27 variable genome positions were scattered over the study period although with differences in yearly prevalence. A total of 167 amino acid substitutions were detected. Besides, coinfection was observed for 52% of the samples, rhinoviruses then adenoviruses (HAdVs) being the most common viruses. Principal component analysis showed that HBoV1 genotypic cluster α tended to be correlated with HAdV co-infection. Subsequent HAdV typing for HBoV1-positive samples and negative controls demonstrated that HAdVC species predominated but HAdVB was that significantly HBoV1-associated. Overall, we described here the first HBoV1 genomes sequenced for France. HBoV1 and HAdVB association deserves further investigation.


Sujet(s)
Co-infection , Génome viral , Génotype , Séquençage nucléotidique à haut débit , Bocavirus humain , Infections à Parvoviridae , Phylogenèse , Humains , Bocavirus humain/génétique , Bocavirus humain/classification , Bocavirus humain/isolement et purification , Génome viral/génétique , France/épidémiologie , Infections à Parvoviridae/virologie , Infections à Parvoviridae/épidémiologie , Femelle , Enfant d'âge préscolaire , Mâle , Enfant , Adulte , Nourrisson , Adulte d'âge moyen , Co-infection/virologie , Co-infection/épidémiologie , Adolescent , Partie nasale du pharynx/virologie , Jeune adulte , Sujet âgé , Analyse de séquence d'ADN , Variation génétique , ADN viral/génétique
8.
J Infect Dev Ctries ; 18(5): 809-816, 2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38865411

RÉSUMÉ

INTRODUCTION: The objective of the present study was to report, for the first time, the presence of canine parvovirus type 2c (CPV-2c) in domesticated dogs with acute gastroenteritis and to characterize the antigenic variants circulating in Palestine. METHODOLOGY: A veterinary clinical-based epidemiological study was carried out between December 2022 and April 2023. Fifty fecal samples were collected from dogs with gastroenteritis and screened for CPV-2 infection by polymerase chain reaction. The distribution of positive cases according to various epidemiological factors was studied. Partial sequencing of the viral protein 2 (VP2) gene was performed for the analysis of CPV-2 variants. RESULTS: Most of the investigated samples (60%; n = 50) during the study period were found positive for CPV-2 infection. There was no difference in the distribution of positive cases of CPV-2 infection based on age group, gender, location, and vaccination status. The analysis of nucleotide and amino acid sequences from amplified products, as well as phylogenetic analysis, revealed the presence of CPV-2c clustered with Asian CPV-2c variants. CONCLUSIONS: In summary, this study represents the initial genetic analysis of CPV-2 present in Palestinian dogs with gastroenteritis and provides evidence that confirms the existence of the CPV-2c variants. To determine the prevailing CPV-2 variant associated with the infection, it is crucial to conduct further sequence analysis using large populations of both domestic and wild canines.


Sujet(s)
Maladies des chiens , Fèces , Gastroentérite , Infections à Parvoviridae , Parvovirus canin , Phylogenèse , Chiens , Animaux , Parvovirus canin/génétique , Parvovirus canin/isolement et purification , Parvovirus canin/classification , Infections à Parvoviridae/médecine vétérinaire , Infections à Parvoviridae/épidémiologie , Infections à Parvoviridae/virologie , Maladies des chiens/virologie , Maladies des chiens/épidémiologie , Fèces/virologie , Gastroentérite/virologie , Gastroentérite/épidémiologie , Gastroentérite/médecine vétérinaire , Moyen Orient/épidémiologie , Femelle , Mâle , Réaction de polymérisation en chaîne
9.
Viruses ; 16(6)2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38932156

RÉSUMÉ

Reports of newly discovered equine hepatotropic flavi- and parvoviruses have emerged throughout the last decade in many countries, the discovery of which has stimulated a great deal of interest and clinical research. Although commonly detected in horses without signs of disease, equine parvovirus hepatitis (EqPV-H) and equine hepacivirus (EqHV) have been associated with liver disease, including following the administration of contaminated anti-toxin. Our aim was to determine whether EqPV-H and EqHV are present in Australian horses and whether EqPV-H was present in French horses and to examine sequence diversity between strains of both viruses amongst infected horses on either side of the globe. Sera from 188 Australian horses and 256 French horses from horses with and without clinical signs of disease were collected. Twelve out of 256 (4.7%) and 6 out of 188 (3.2%) French and Australian horses, respectively, were positive for the molecular detection of EqPV-H. Five out of 256 (1.9%) and 21 out of 188 (11.2%) French and Australian horses, respectively, were positive for the molecular detection of EqHV. Australian strains for both viruses were genomically clustered, in contrast to strains from French horses, which were more broadly distributed. The findings of this preliminary survey, with the molecular detection of EqHV and EqPV-H in Australia and the latter in France, adds to the growing body of awareness regarding these recently discovered hepatotropic viruses. It has provided valuable information not just in terms of geographic endemicity but will guide equine clinicians, carers, and authorities regarding infectious agents and potential impacts of allogenic tissue contamination. Although we have filled many gaps in the world map regarding equine hepatotropic viruses, further prospective studies in this emerging field may be useful in terms of elucidating risk factors and pathogenesis of these pathogens and management of cases in terms of prevention and diagnosis.


Sujet(s)
Hepacivirus , Hépatite virale animale , Maladies des chevaux , Infections à Parvoviridae , Parvovirus , Phylogenèse , Animaux , Equus caballus , Maladies des chevaux/virologie , Maladies des chevaux/épidémiologie , Maladies des chevaux/sang , Australie/épidémiologie , Infections à Parvoviridae/médecine vétérinaire , Infections à Parvoviridae/épidémiologie , Infections à Parvoviridae/virologie , Infections à Parvoviridae/sang , France/épidémiologie , Hépatite virale animale/virologie , Hépatite virale animale/épidémiologie , Hépatite virale animale/sang , Parvovirus/génétique , Parvovirus/isolement et purification , Parvovirus/classification , Parvovirus/immunologie , Hepacivirus/génétique , Hepacivirus/isolement et purification , Hepacivirus/immunologie , Hépatite C/médecine vétérinaire , Hépatite C/virologie , Hépatite C/épidémiologie
10.
Poult Sci ; 103(7): 103853, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38795515

RÉSUMÉ

Short-beak and dwarf syndrome (SBDS) is caused by infection with novel goose parvovirus (NGPV), which leads to intestinal dysbiosis, developmental delay, short beak, lameness, and paralysis in ducks and is the cause of skeletal health problems. NGPV infection can cause intestinal microbial disturbances, but it is still unclear whether the intestinal microbiota affects the pathogenicity of NGPV. Here, the effects of intestinal microbiota on NGPV-induced SBDS in Cherry Valley ducks were assessed by establishing a duck model for gut microflora depletion/reestablishment through antibiotics (ABX) treatment/fecal microbiota transplanted (FMT). By measuring body weight, beak length, beak width and tarsal length, we found that SBDS clinical symptoms were alleviated in ducks treated with ABX, but not in FMT ducks. Next, we conducted a comprehensive analysis of bone metabolism, gut barrier integrity, and inflammation levels using quantitative real-time PCR (qPCR), enzyme linked immunosorbent assay (ELISA), biochemical analysis and histological analysis. The results showed that ABX treatment improved bone quality reduced bone resorption, mitigated tissue lesions, protected intestinal barrier integrity, and inhibited systemic inflammation in NGPV-infected ducks. Moreover, cecal microflora composition and short-chain fatty acids (SCFAs) production were examined by bacterial 16S rRNA sequencing and gas chromatography. The results revealed that ABX treatment mitigated the decreased abundance of Firmicutes and Bacteroidota in NGPV-infected ducks, as well as increased SCFAs production. Furthermore, ABX treatment reduced the mucosa-associated lymphoid tissue lymphoma translocation protein 1 (Malt1) and nuclear factor κB (NF-κB) expression, which are correlated with systemic inflammation in SBDS ducks. These findings suggested that intestinal microflora depletion alleviated NGPV-induced SBDS by maintaining intestinal homeostasis, inhibiting inflammatory response and alleviating bone resorption. These results provide evidence for the pivotal role of intestinal microbiota in the process of SBDS and contribute a theoretical basis for the feasibility of microecological preparation as a method to control SBDS.


Sujet(s)
Canards , Microbiome gastro-intestinal , Infections à Parvoviridae , Parvovirinae , Maladies de la volaille , Animaux , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Maladies de la volaille/virologie , Maladies de la volaille/microbiologie , Infections à Parvoviridae/médecine vétérinaire , Infections à Parvoviridae/virologie , Parvovirinae/génétique , Parvovirinae/physiologie , Antibactériens/pharmacologie , Antibactériens/administration et posologie , Transplantation de microbiote fécal/médecine vétérinaire
11.
J Virol ; 98(6): e0063324, 2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38775479

RÉSUMÉ

Adeno-associated viruses (AAVs) package a single-stranded (ss) DNA genome of 4.7 kb in their capsid of ~20 nm in diameter. AAV replication requires co-infection of a helper virus, such as adenovirus. During the optimization of recombinant AAV production, a small viral nonstructural protein, membrane-associated accessory protein (MAAP), was identified. However, the function of the MAAP in the context of AAV infection remains unknown. Here, we investigated the expression strategy and function of the MAAP during infection of both AAV2 and AAV5 in human embryonic kidney (HEK)293 cells. We found that AAV2 MAAP2 and AAV5 MAAP5 are expressed from the capsid gene (cap)-transcribing mRNA spliced from the donor to the second splice site that encodes VP2 and VP3. Thus, this AAV cap gene transcribes a multicistronic mRNA that can be translated to four viral proteins, MAAP, VP2, AAP, and VP3 in order. In AAV2 infection, MAAP2 predominantly localized in the cytoplasm, alongside the capsid, near the nuclear and plasma membranes, but a fraction of MAAP2 exhibited nuclear localization. In AAV5 infection, MAAP5 revealed a distinct pattern, predominantly localizing within the nucleus. In the cells infected with an MAAP knockout mutant of AAV2 or AAV5, both viral DNA replication and virus replication increased, whereas virus egress decreased, and the decrease in virus egress can be restored by providing MAAP in trans. In summary, MAAP, a novel AAV nonstructural protein translated from a multicistronic viral cap mRNA, not only facilitates cellular egress of AAV but also likely negatively affects viral DNA replication during infection. IMPORTANCE: Recombinant adeno-associated virus (rAAV) has been used as a gene delivery vector in clinical gene therapy. In current gene therapies employing rAAV, a high dose of the vector is required. Consequently, there is a high demand for efficient and high-purity vector production systems. In this study, we demonstrated that membrane-associated accessory protein (MAAP), a small viral nonstructural protein, is translated from the same viral mRNA transcript encoding VP2 and VP3. In AAV-infected cells, apart from its prevalent expression in the cytoplasm with localization near the plasma and nuclear membranes, the MAAP also exhibits notable localization within the nucleus. During AAV infection, MAAP expression increases the cellular egress of progeny virions and decreases viral DNA replication and progeny virion production. Thus, the choice of MAAP expression has pros and cons during AAV infection, which could provide a guide to rAAV production.


Sujet(s)
Protéines de capside , Dependovirus , Réplication virale , Humains , Dependovirus/génétique , Dependovirus/métabolisme , Cellules HEK293 , Protéines de capside/métabolisme , Protéines de capside/génétique , Protéines virales non structurales/métabolisme , Protéines virales non structurales/génétique , Infections à Parvoviridae/virologie , Infections à Parvoviridae/métabolisme , Capside/métabolisme
12.
Diagn Microbiol Infect Dis ; 109(4): 116300, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38759541

RÉSUMÉ

BACKGROUND: Parvovirus B19(B19) is a DNA virus. The most common B19 disease is erythema infectiosum (fifth-disease). PCR and ELISA are sensitive for detecting of acute disease. However, it is not clear which test better and the relationship between laboratory tests and clinical findings. OBJECTIVE: To discuss the clinical and laboratory characteristics of pediatric patients infected with B19. STUDY DESIGN: 236 children were examined. Children with at least one positive molecular or serological test were included. Positive serum B19-DNA and/or B19-IgM was considered an acute B19 infection. RESULTS: B19DNA was detected in 80.8 % of acute cases. Serological tests were less positive. Acute B19 infection was observed in 24 patients. Only 17 patients were positive for B19 DNA, 3 for IgM and 4 for both. The sensitivity of B19 DNA is 87.5 %. However, this rate is 29.2 % for B19 IgM. CONCLUSION: B19-DNA and IgM together provide a better, highly accurate diagnosis.


Sujet(s)
Anticorps antiviraux , ADN viral , Érythème infectieux , Immunoglobuline M , Parvovirus humain B19 , Humains , Parvovirus humain B19/génétique , Parvovirus humain B19/immunologie , Parvovirus humain B19/isolement et purification , Enfant , Immunoglobuline M/sang , Enfant d'âge préscolaire , Femelle , Mâle , ADN viral/sang , Anticorps antiviraux/sang , Érythème infectieux/diagnostic , Érythème infectieux/sang , Érythème infectieux/immunologie , Nourrisson , Sensibilité et spécificité , Adolescent , Réaction de polymérisation en chaîne/méthodes , Infections à Parvoviridae/diagnostic , Infections à Parvoviridae/sang , Infections à Parvoviridae/immunologie , Infections à Parvoviridae/virologie , Test ELISA , Tests sérologiques/méthodes
13.
Comp Immunol Microbiol Infect Dis ; 110: 102190, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38815398

RÉSUMÉ

Canine parvovirus type 2 (CPV-2) is a major cause of fatal gastroenteritis and myocarditis in puppies of domestic and wild carnivores. CPV-2 has accumulated changes over time lead to the emergence of three antigenic variants CPV-2a, CPV-2b, and CPV-2c. VP2 is the major capsid protein that determines virus antigenicity, and host range. Although the three CPV-2 variants were previously identified in Egypt, most reports covered a restricted geographic region and/or time period, and only analyzed partial fragments of VP2 gene. Therefore, this study was designed to test 100 rectal swabs collected from 7 Egyptian governorates between 2019 and 2021 for CPV-2 using PCR. A total of 65 positive samples were identified, mostly in pure dog breeds of young age. The three variants co-circulated in 2019, while CPV-2b was not detected in 2020 and 2021. The frequency of CPV-2b and CPV-2c was higher in 2019 and 2021, respectively. Analysis of CPV-2 full-length VP2 gene sequence from 19/65 positive samples has identified four common amino acid substitutions F267Y, S297A, A300G, Y324I, which are characteristic for the new CPV-2 variants currently circulating worldwide. Unique substitutions including A5G, G36R, V38E, Q370R, and G392V were recognized in certain samples, and appears to have distinct effect on receptor binding, nuclear translocation, and inter-species transmission. Phylogenetic analysis showed separation of CPV-2 strains into two clades. All strains of this study were classified in clade I with Asian strains. In conclusion, this study provides updated comprehensive molecular analysis of CPV-2 variants in Egypt.


Sujet(s)
Protéines de capside , Maladies des chiens , Infections à Parvoviridae , Parvovirus canin , Phylogenèse , Animaux , Égypte/épidémiologie , Chiens , Parvovirus canin/génétique , Parvovirus canin/classification , Parvovirus canin/isolement et purification , Protéines de capside/génétique , Infections à Parvoviridae/médecine vétérinaire , Infections à Parvoviridae/virologie , Infections à Parvoviridae/épidémiologie , Maladies des chiens/virologie , Maladies des chiens/épidémiologie , Substitution d'acide aminé
14.
Microbiol Spectr ; 12(6): e0391423, 2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38742903

RÉSUMÉ

Porcine parvovirus (PPV) is one of the most important pathogens that cause reproductive failure in pigs. However, the pathogenesis of PPV infection remains unclear. Proteomics is a powerful tool to understand the interaction between virus and host cells. In the present study, we analyzed the proteomics of PPV-infected PK-15 cells. A total of 32 and 345 proteins were differentially expressed at the early and replication stages, respectively. Subsequent gene ontology annotation and Kyoto Encyclopedia of Genes and Genomes enrichment analysis showed these differentially expressed proteins were significantly enriched in pathways including toll-like receptor signaling pathway, tumor necrosis factor signaling pathway, and viral carcinogenesis. The expression of poly (rC) binding protein 1 (PCBP1) was observed to decrease after PPV infection. Overexpressed or silenced PCBP1 expression inhibited or promoted PPV infection. Our studies established a foundation for further exploration of the multiplication mechanism of PPV. IMPORTANCE: Porcine parvovirus (PPV) is a cause of reproductive failure in the swine industry. Our knowledge of PPV remains limited, and there is no effective treatment for PPV infection. Proteomics of PPV-infected PK-15 cells was conducted to identify differentially expressed proteins at 6 hours post-infection (hpi) and 36 hpi. Gene ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analysis showed that various pathways participate in PPV infection. Poly (rC) binding protein 1 was confirmed to inhibit PPV replication, which provided potential targets for anti-PPV infection. Our findings improve the understanding of PPV infection and pave the way for future research in this area.


Sujet(s)
Infections à Parvoviridae , Parvovirus porcin , Protéomique , Protéines de liaison à l'ARN , Maladies des porcs , Réplication virale , Parvovirus porcin/génétique , Parvovirus porcin/physiologie , Animaux , Suidae , Lignée cellulaire , Protéines de liaison à l'ARN/génétique , Protéines de liaison à l'ARN/métabolisme , Infections à Parvoviridae/virologie , Infections à Parvoviridae/métabolisme , Infections à Parvoviridae/médecine vétérinaire , Maladies des porcs/virologie , Maladies des porcs/métabolisme , Maladies des porcs/génétique , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme
15.
Virol J ; 21(1): 108, 2024 05 10.
Article de Anglais | MEDLINE | ID: mdl-38730285

RÉSUMÉ

BACKGROUND: The immature and suppressed immune response makes transplanted children a special susceptible group to Parvovirus B19 (PVB19). However, the clinical features of transplanted children with PVB19 infection haven't been comprehensively described. METHODS: We searched the medical records of all the transplant recipients who attended the Children's Hospital of Fudan University from 1 Oct 2020 to 31 May 2023, and reviewed the medical literature for PVB19 infection cases among transplanted children. RESULTS: A total of 10 cases of PVB19 infection were identified in 201 transplanted children at our hospital, and the medical records of each of these cases were shown. Also, we retrieved 40 cases of PVB19 infection among transplanted children from the literature, thus summarizing a total of 50 unique cases of PVB19 infection. The median time to the first positive PVB19 DNA detection was 14 weeks post-transplantation. PVB19 IgM and IgG were detected in merely 26% and 24% of the children, respectively. The incidence of graft loss/dysfunction was as high as 36%. Hematopoietic stem cell transplant (HSCT) recipients showed higher PVB19 load, lower HGB level, greater platelet damage, lower PVB19 IgM/IgG positive rates, and more graft dysfunction than solid-organ transplant (SOT) recipients, indicating a more incompetent immune system. CONCLUSIONS: Compared with the published data of transplanted adults, transplanted children displayed distinct clinical features upon PVB19 infection, including lower PVB19 IgM/IgG positive rates, more graft dysfunction, and broader damage on hematopoietic cell lines, which was even more prominent in HSCT recipients, thus should be of greater concern.


Sujet(s)
Anticorps antiviraux , Transplantation de cellules souches hématopoïétiques , Infections à Parvoviridae , Parvovirus humain B19 , Humains , Parvovirus humain B19/immunologie , Parvovirus humain B19/génétique , Enfant , Femelle , Mâle , Enfant d'âge préscolaire , Infections à Parvoviridae/virologie , Infections à Parvoviridae/immunologie , Transplantation de cellules souches hématopoïétiques/effets indésirables , Anticorps antiviraux/sang , Nourrisson , Adolescent , Immunoglobuline M/sang , Immunoglobuline G/sang , Receveurs de transplantation , ADN viral/sang , Charge virale , Transplantation d'organe/effets indésirables
16.
Viruses ; 16(5)2024 04 29.
Article de Anglais | MEDLINE | ID: mdl-38793587

RÉSUMÉ

A massive mortality event concerning farmed Chinese tongue soles occurred in Tianjin, China, and the causative agent remains unknown. Here, a novel Cynoglossus semilaevis papillomavirus (CsPaV) and parvovirus (CsPV) were simultaneously isolated and identified from diseased fish via electron microscopy, virus isolation, genome sequencing, experimental challenges, and fluorescence in situ hybridization (FISH). Electron microscopy showed large numbers of virus particles present in the tissues of diseased fish. Viruses that were isolated and propagated in flounder gill cells (FG) induced typical cytopathic effects (CPE). The cumulative mortality of fish given intraperitoneal injections reached 100% at 7 dpi. The complete genomes of CsPaV and CsPV comprised 5939 bp and 3663 bp, respectively, and the genomes shared no nucleotide sequence similarities with other viruses. Phylogenetic analysis based on the L1 and NS1 protein sequences revealed that CsPaV and CsPV were novel members of the Papillomaviridae and Parvoviridae families. The FISH results showed positive signals in the spleen tissues of infected fish, and both viruses could co-infect single cells. This study represents the first report where novel papillomavirus and parvovirus are identified in farmed marine cultured fish, and it provides a basis for further studies on the prevention and treatment of emerging viral diseases.


Sujet(s)
Maladies des poissons , Poissons plats , Génome viral , Papillomaviridae , Infections à Parvoviridae , Parvovirus , Phylogenèse , Animaux , Maladies des poissons/virologie , Maladies des poissons/mortalité , Chine , Poissons plats/virologie , Infections à Parvoviridae/médecine vétérinaire , Infections à Parvoviridae/virologie , Parvovirus/génétique , Parvovirus/isolement et purification , Parvovirus/classification , Papillomaviridae/génétique , Papillomaviridae/isolement et purification , Papillomaviridae/classification , Infections à papillomavirus/virologie , Infections à papillomavirus/médecine vétérinaire , Hybridation fluorescente in situ
17.
Viruses ; 16(5)2024 05 13.
Article de Anglais | MEDLINE | ID: mdl-38793654

RÉSUMÉ

Based on several clinical observations it was hypothesized that herpesviruses may influence the replication of human bocaviruses, the second known parvoviruses that have been confirmed as human pathogens. While several cell lines support the growth of HSV-1, HBoV-1 was exclusively cultivated on air-liquid interface cultures, the latter being a rather complicated, slow, and low throughput system. One of the cell lines are T84 cells, which are derived from the lung metastasis of a colorectal tumor. In this study, we provide evidence that T84 also supports HBoV replication when cultivated as monolayers, while simultaneously being permissive for HSV-1. The cell culture model thus would enable co-infection studies of both viruses and is worth being optimized for high throughput studies with HBoV-1. Additionally, the study provides evidence for a supporting effect of HSV-1 on the replication and packaging of HBoV-1 progeny DNA into DNase-resistant viral particles.


Sujet(s)
Co-infection , Herpèsvirus humain de type 1 , Bocavirus humain , Réplication virale , Herpèsvirus humain de type 1/physiologie , Humains , Co-infection/virologie , Bocavirus humain/physiologie , Bocavirus humain/génétique , Lignée cellulaire , Lignée cellulaire tumorale , Techniques de culture cellulaire/méthodes , Herpès/virologie , Infections à Parvoviridae/virologie , Chlorocebus aethiops , Culture virale/méthodes
18.
Mol Microbiol ; 121(6): 1127-1147, 2024 06.
Article de Anglais | MEDLINE | ID: mdl-38629786

RÉSUMÉ

Minute virus of canines (MVC) belongs to the genus Bocaparvovirus (formerly Bocavirus) within the Parvoviridae family and causes serious respiratory and gastrointestinal symptoms in neonatal canines worldwide. A productive viral infection relies on the successful recruitment of host factors for various stages of the viral life cycle. However, little is known about the MVC-host cell interactions. In this study, we identified that two cellular proteins (Hsc70 and Hsp70) interacted with NS1 and VP2 proteins of MVC, and both two domains of Hsc70/Hsp70 were mediated for their interactions. Functional studies revealed that Hsp70 was induced by MVC infection, knockdown of Hsc70 considerably suppressed MVC replication, whereas the replication was dramatically promoted by Hsp70 knockdown. It is interesting that low amounts of overexpressed Hsp70 enhanced viral protein expression and virus production, but high amounts of Hsp70 overexpression weakened them. Upon Hsp70 overexpressing, we observed that the ubiquitination of viral proteins changed with Hsp70 overexpression, and proteasome inhibitor (MG132) restored an accumulation of viral proteins. In addition, we verified that Hsp70 family inhibitors remarkably decreased MVC replication. Overall, we identified Hsc70 and Hsp70 as interactors of MVC NS1 and VP2 proteins and were involved in MVC replication, which may provide novel targets for anti-MVC approach.


Sujet(s)
Protéines du choc thermique HSC70 , Protéines du choc thermique HSP70 , Réplication virale , Protéines du choc thermique HSC70/métabolisme , Protéines du choc thermique HSC70/génétique , Animaux , Protéines du choc thermique HSP70/métabolisme , Protéines du choc thermique HSP70/génétique , Chiens , Bocavirus/génétique , Bocavirus/métabolisme , Bocavirus/physiologie , Protéines virales non structurales/métabolisme , Protéines virales non structurales/génétique , Humains , Infections à Parvoviridae/virologie , Infections à Parvoviridae/métabolisme , Ubiquitination , Protéines virales/métabolisme , Protéines virales/génétique , Cellules HEK293 , Interactions hôte-pathogène , Lignée cellulaire , Protéines de capside/métabolisme , Protéines de capside/génétique , Maladies des chiens/virologie
19.
J Virol ; 98(5): e0009324, 2024 May 14.
Article de Anglais | MEDLINE | ID: mdl-38591899

RÉSUMÉ

Feline parvovirus (FPV) infection is highly fatal in felines. NS1, which is a key nonstructural protein of FPV, can inhibit host innate immunity and promote viral replication, which is the main reason for the severe pathogenicity of FPV. However, the mechanism by which the NS1 protein disrupts host immunity and regulates viral replication is still unclear. Here, we identified an FPV M1 strain that is regulated by the NS1 protein and has more pronounced suppression of innate immunity, resulting in robust replication. We found that the neutralization titer of the FPV M1 strain was significantly lower than that of the other strains. Moreover, FPV M1 had powerful replication ability, and the FPV M1-NS1 protein had heightened efficacy in repressing interferon-stimulated genes (ISGs) expression. Subsequently, we constructed an FPV reverse genetic system, which confirmed that the N588 residue of FPV M1-NS1 protein is a key amino acid that bolsters viral proliferation. Recombinant virus containing N588 also had stronger ability to inhibit ISGs, and lower ISGs levels promoted viral replication and reduced the neutralization titer of the positive control serum. Finally, we confirmed that the difference in viral replication was abolished in type I IFN receptor knockout cell lines. In conclusion, our results demonstrate that the N588 residue of the NS1 protein is a critical amino acid that promotes viral proliferation by increasing the inhibition of ISGs expression. These insights provide a reference for studying the relationship between parvovirus-mediated inhibition of host innate immunity and viral replication while facilitating improved FPV vaccine production.IMPORTANCEFPV infection is a viral infectious disease with the highest mortality rate in felines. A universal feature of parvovirus is its ability to inhibit host innate immunity, and its ability to suppress innate immunity is mainly accomplished by the NS1 protein. In the present study, FPV was used as a viral model to explore the mechanism by which the NS1 protein inhibits innate immunity and regulates viral replication. Studies have shown that the FPV-NS1 protein containing the N588 residue strongly inhibits the expression of host ISGs, thereby increasing the viral proliferation titer. In addition, the presence of the N588 residue can increase the proliferation titer of the strain 5- to 10-fold without affecting its virulence and immunogenicity. In conclusion, our findings provide new insights and guidance for studying the mechanisms by which parvoviruses suppress innate immunity and for developing high-yielding FPV vaccines.


Sujet(s)
Virus de la panleucopénie féline , Protéines virales non structurales , Réplication virale , Animaux , Chats , Lignée cellulaire , Virus de la panleucopénie féline/génétique , Virus de la panleucopénie féline/immunologie , Immunité innée , Mutation , Infections à Parvoviridae/virologie , Infections à Parvoviridae/immunologie , Protéines virales non structurales/génétique , Protéines virales non structurales/métabolisme , Protéines virales non structurales/immunologie
20.
Virology ; 595: 110081, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38599030

RÉSUMÉ

Parvoviruses are known to be significant viral pathogens that infect a wide range of species globally. However, little is known about the parvoviruses circulating in Australian birds, including yellow canaries. Here, we present four parvoviral sequences including three novel parvoviruses detected from 10 yellow canaries (Crithagra flaviventris), named canary chaphamaparvovirus 1 and -2 (CaChPV1 and CaChPV2), canary dependoparvovirus 1 and -2 (CaDePV1 and CaDePV2). The whole genome sequences of CaChPV1, CaChPV2, CaDePV1, and CaDePV2 showed the highest identity with other parvoviruses at 76.4%, 75.9%, 84.0%, and 59.1%, respectively. Phylogenetic analysis demonstrated that CaChPV1 and CaChPV2 were clustered within the genus Chaphamaparvovirus. Meanwhile, CaDePV1 and CaDePV2 fall within the genus Dependoparvovirus and have the closest evolutionary relationship to the bird-associated dependoparvoviruses. Overall, this study enriched our understanding of the genetic diversity among avian parvoviruses within the Parvoviridae family.


Sujet(s)
Variation génétique , Génome viral , Infections à Parvoviridae , Phylogenèse , Animaux , Infections à Parvoviridae/médecine vétérinaire , Infections à Parvoviridae/virologie , Australie , Parvovirus/génétique , Parvovirus/classification , Parvovirus/isolement et purification , Maladies des oiseaux/virologie , ADN viral/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...