Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 374
Filter
1.
Front Immunol ; 15: 1277447, 2024.
Article in English | MEDLINE | ID: mdl-38633245

ABSTRACT

Modified vaccinia virus Ankara (MVA) has been widely tested in clinical trials as recombinant vector vaccine against infectious diseases and cancers in humans and animals. However, one biosafety concern about the use of MVA vectored vaccine is the potential for MVA to recombine with naturally occurring orthopoxviruses in cells and hosts in which it multiplies poorly and, therefore, producing viruses with mosaic genomes with altered genetic and phenotypic properties. We previously conducted co-infection and superinfection experiments with MVA vectored influenza vaccine (MVA-HANP) and a feline Cowpox virus (CPXV-No-F1) in Vero cells (that were semi-permissive to MVA infection) and showed that recombination occurred in both co-infected and superinfected cells. In this study, we selected the putative recombinant viruses and performed genomic characterization of these viruses. Some putative recombinant viruses displayed plaque morphology distinct of that of the parental viruses. Our analysis demonstrated that they had mosaic genomes of different lengths. The recombinant viruses, with a genome more similar to MVA-HANP (>50%), rescued deleted and/or fragmented genes in MVA and gained new host ranges genes. Our analysis also revealed that some MVA-HANP contained a partially deleted transgene expression cassette and one recombinant virus contained part of the transgene expression cassette similar to that incomplete MVA-HANP. The recombination in co-infected and superinfected Vero cells resulted in recombinant viruses with unpredictable biological and genetic properties as well as recovery of delete/fragmented genes in MVA and transfer of the transgene into replication competent CPXV. These results are relevant to hazard characterization and risk assessment of MVA vectored biologicals.


Subject(s)
Coinfection , Influenza Vaccines , Superinfection , Chlorocebus aethiops , Animals , Cats , Humans , Influenza Vaccines/genetics , Cowpox virus/genetics , Vero Cells , Vaccinia virus , Vaccines, Synthetic/genetics , Whole Genome Sequencing
2.
Antiviral Res ; 216: 105651, 2023 08.
Article in English | MEDLINE | ID: mdl-37270160

ABSTRACT

Many poxviruses are significant human and animal pathogens, including viruses that cause smallpox and mpox (formerly monkeypox). Identifying novel and potent antiviral compounds is critical to successful drug development targeting poxviruses. Here we tested two compounds, nucleoside trifluridine, and nucleotide adefovir dipivoxil, for antiviral activities against vaccinia virus (VACV), mpox virus (MPXV), and cowpox virus (CPXV) in physiologically relevant primary human fibroblasts. Both compounds potently inhibited the replication of VACV, CPXV, and MPXV (MA001 2022 isolate) in plaque assays. In our recently developed assay based on a recombinant VACV expressing secreted Gaussia luciferase, they both exhibited high potency in inhibiting VACV replication with EC50s in the low nanomolar range. In addition, both trifluridine and adefovir dipivoxil inhibited VACV DNA replication and downstream viral gene expression. Our results characterized trifluridine and adefovir dipivoxil as strong poxvirus antiviral compounds and further validate the VACV Gaussia luciferase assay as a highly efficient and reliable reporter tool for identifying poxvirus inhibitors. Given that both compounds are FDA-approved drugs, and trifluridine is already used to treat ocular vaccinia, further development of trifluridine and adefovir dipivoxil holds great promise in treating poxvirus infections, including mpox.


Subject(s)
Mpox (monkeypox) , Poxviridae , Vaccinia , Animals , Humans , Vaccinia virus/physiology , Vaccinia/drug therapy , Cowpox virus , Antiviral Agents/pharmacology , Antiviral Agents/metabolism , Trifluridine/metabolism , Cell Line , Poxviridae/metabolism
3.
J Comp Pathol ; 204: 39-46, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37352825

ABSTRACT

Cowpox virus is considered to be a re-emerging zoonotic pathogen and a public health threat due to increasing numbers of cases in humans and animals in Europe over the past decade, including within the United Kingdom (UK). We present epidemiological data and diagnostic features of 27 recent, naturally occurring cowpox cases in zoo and wild animals across the UK, including the first reports of cowpox in two snow leopards (Panthera uncia), a Bengal tiger (Panthera tigris tigris), three Chilean pudus (Pudu puda), a Malayan tapir (Tapirus indicus) and a Eurasian otter (Lutra lutra), and the first reports of Orthopoxvirus infection in a lar gibbon (Hylobates lar), a Southern tamandua (Tamandua tetradactyla) and an aardvark (Orycteropus afer). This study provides a detailed overview of cowpox infections in a wide range of non-domestic animal species, presents a range of methods for diagnosis and demonstrates the value of retrospective analysis of pathology surveillance in revealing epidemiological links.


Subject(s)
Cowpox , Deer , Otters , Panthera , Tigers , Humans , Animals , Animals, Wild , Cowpox/epidemiology , Cowpox/veterinary , Retrospective Studies , Vermilingua , Cowpox virus , United Kingdom/epidemiology , Animals, Zoo
4.
mSphere ; 8(2): e0006223, 2023 04 20.
Article in English | MEDLINE | ID: mdl-36920219

ABSTRACT

The ongoing worldwide monkeypox outbreak is caused by viral lineages (globally referred to as hMPXV1) that are related to but distinct from clade IIb MPXV viruses transmitted within Nigeria. Analysis of the genetic differences has indicated that APOBEC-mediated editing might be responsible for the unexpectedly high number of mutations observed in hMPXV1 genomes. Here, using 1,624 publicly available hMPXV1 sequences, we analyzed the mutations that accrued between 2017 and the emergence of the current predominant variant (B.1), as well as those that that have been accumulating during the 2022 outbreak. We confirmed an overwhelming prevalence of C-to-T and G-to-A mutations, with a sequence context (5'-TC-3') consistent with the preferences of several human APOBEC3 enzymes. We also found that mutations preferentially occur in highly expressed viral genes, although no transcriptional asymmetry was observed. A comparison of the mutation spectrum and context was also performed against the human-specific variola virus (VARV) and the zoonotic cowpox virus (CPXV), as well as fowlpox virus (FWPV). The results indicated that in VARV genomes, C-to-T and G-to-A changes were more common than the opposite substitutions, although the effect was less marked than for hMPXV1. Conversely, no preference toward C-to-T and G-to-A changes was observed in CPXV and FWPV. Consistently, the sequence context of C-to-T changes confirmed a preference for a T in the -1 position for VARV, but not for CPXV or FWPV. Overall, our results strongly support the view that, irrespective of the transmission route, orthopoxviruses infecting humans are edited by the host APOBEC3 enzymes. IMPORTANCE Analysis of the viral lineages responsible for the 2022 monkeypox outbreak suggested that APOBEC enzymes are driving hMPXV1 evolution. Using 1,624 public sequences, we analyzed the mutations that accumulated between 2017 and the emergence of the predominant variant and those that characterize the last outbreak. We found that the mutation spectrum of hMPXV1 has been dominated by TC-to-TT and GA-to-AA changes, consistent with the editing activity of human APOBEC3 proteins. We also found that mutations preferentially affect highly expressed viral genes, possibly because transcription exposes single-stranded DNA (ssDNA), a target of APOBEC3 editing. Notably, analysis of the human-specific variola virus (VARV) and the zoonotic cowpox virus (CPXV) indicated that in VARV genomes, TC-to-TT and GA-to-AA changes are likewise extremely frequent. Conversely, no preference toward TC-to-TT and GA-to-AA changes is observed in CPXV. These results suggest that APOBEC3 proteins have an impact on the evolution of different human-infecting orthopoxviruses.


Subject(s)
Mpox (monkeypox) , Orthopoxvirus , Smallpox , Variola virus , Animals , Humans , Orthopoxvirus/genetics , Cowpox virus/genetics , Cowpox virus/metabolism , Mutation , APOBEC Deaminases/genetics , APOBEC Deaminases/metabolism
5.
Viruses ; 15(2)2023 01 26.
Article in English | MEDLINE | ID: mdl-36851570

ABSTRACT

The ongoing global Monkeypox outbreak that started in the spring of 2022 has reinforced the importance of protecting the population using live virus vaccines based on the vaccinia virus (VACV). Smallpox also remains a biothreat and although some U.S. military personnel are immunized with VACV, safety concerns limit its use in other vulnerable groups. Consequently, there is a need for an effective and safer, single dose, live replicating vaccine against both viruses. One potential approach is to use the horsepox virus (HPXV) as a vaccine. Contemporary VACV shares a common ancestor with HPXV, which from the time of Edward Jenner and through the 19th century, was extensively used to vaccinate against smallpox. However, it is unknown if early HPXV-based vaccines exhibited different safety and efficacy profiles compared to modern VACV. A deeper understanding of HPXV as a vaccine platform may allow the construction of safer and more effective vaccines against the poxvirus family. In a proof-of-concept study, we vaccinated cynomolgus macaques with TNX-801, a recombinant chimeric horsepox virus (rcHPXV), and showed that the vaccine elicited protective immune responses against a lethal challenge with monkeypox virus (MPXV), strain Zaire. The vaccine was well tolerated and protected animals from the development of lesions and severe disease. These encouraging data support the further development of TNX-801.


Subject(s)
Mpox (monkeypox) , Orthopoxvirus , Poxviridae Infections , Smallpox , Variola virus , Animals , Orthopoxvirus/genetics , Mpox (monkeypox)/prevention & control , Smallpox/prevention & control , Cowpox virus , Poxviridae Infections/prevention & control , Poxviridae Infections/veterinary , Vaccination , Vaccinia virus , Macaca fascicularis , Vaccines, Attenuated
6.
Biomolecules ; 13(2)2023 02 08.
Article in English | MEDLINE | ID: mdl-36830694

ABSTRACT

Cowpox viruses (CPXVs) exhibit the broadest known host range among the Poxviridae family and have caused lethal outbreaks in various zoo animals and pets across 12 Eurasian countries, as well as an increasing number of human cases. Herein, we review the history of how the cowpox name has evolved since the 1700s up to modern times. Despite early documentation of the different properties of CPXV isolates, only modern genetic analyses and phylogenies have revealed the existence of multiple Orthopoxvirus species that are currently constrained under the CPXV designation. We further chronicle modern outbreaks in zoos, domesticated animals, and humans, and describe animal models of experimental CPXV infections and how these can help shaping CPXV species distinctions. We also describe the pathogenesis of modern CPXV infections in animals and humans, the geographic range of CPXVs, and discuss CPXV-host interactions at the molecular level and their effects on pathogenicity and host range. Finally, we discuss the potential threat of these viruses and the future of CPXV research to provide a comprehensive review of CPXVs.


Subject(s)
Cowpox virus , Cowpox , Animals , Humans , Cowpox virus/genetics , Cowpox/epidemiology , Phylogeny , Disease Outbreaks
7.
Int J Mol Sci ; 24(2)2023 Jan 16.
Article in English | MEDLINE | ID: mdl-36675269

ABSTRACT

Cowpox is caused by a DNA virus known as the cowpox virus (CPXV) belonging to the Orthopoxvirus genus in the family Poxviridae. Cowpox is a zoonotic disease with the broadest host range among the known poxviruses. The natural reservoir hosts of CPXV are wild rodents. Recently, the cases of orthopoxviral infections have been increasing worldwide, and cowpox is considered the most common orthopoxviral infection in Europe. Cowpox is often a self-limiting disease, although cidofovir or anti-vaccinia gammaglobulin can be used in severe and disseminated cases of human cowpox. In this computational study, a molecular docking analysis of thymine- and arabinofuranosyl-thymine-related structures (1-21) on two cowpox-encoded proteins was performed with respect to the cidofovir standard and a 3D ligand-based pharmacophore model was generated. Three chemical structures (PubChem IDs: 123370001, 154137224, and 90413364) were identified as potential candidates for anti-cowpox agents. Further studies combining in vitro and in silico molecular dynamics simulations to test the stability of these promising compounds could effectively improve the future design of cowpox virus inhibitors, as molecular docking studies are not sufficient to consider a ligand a potential drug.


Subject(s)
Cowpox virus , Cowpox , Animals , Humans , Cowpox virus/genetics , Cowpox virus/metabolism , Thymine/metabolism , Cidofovir/pharmacology , Ligands , Molecular Docking Simulation , Rodentia
9.
Acta Clin Belg ; 78(2): 180-184, 2023 Apr.
Article in English | MEDLINE | ID: mdl-35731612

ABSTRACT

BACKGROUND: Human cowpox virus infection is a rare zoonotic disease. Cowpox virus is a member of the Orthopoxvirus genus, like smallpox. Over the last years records of cowpox virus transmission from pet cats and pet rats to humans in Europe have increased. This observation may result from the loss of cross-immunity against orthopoxviruses after discontinuation of routine smallpox vaccination in the 1980s. CASE PRESENTATION: We report the first case of a human cowpox infection in an unvaccinated Belgian citizen. This 19-year-old student presented with multiple necrotic skin lesions on the chin, the scalp and the pubic region, and with cervical lymphadenopathy and flu-like symptoms. The diagnosis of human cowpox was based on electron microscopic findings and PCR examination performed on a skin biopsy of the pubic lesion. Close contact with cats (her domestic cats or cats from a local shelter) was probably the source of transmission. Spreading of the lesions was likely the result of autoinoculation. After six months all lesions spontaneously healed with atrophic scars. DISCUSSION: To enhance awareness of this rare viral zoonosis and to verify the suspected increase in incidence and symptom severity after cessation of smallpox vaccination, one could argue whether human cowpox should become a notifiable disease.


Subject(s)
Cowpox , Smallpox , Female , Humans , Animals , Cats , Rats , Young Adult , Adult , Cowpox/pathology , Cowpox/veterinary , Smallpox/prevention & control , Belgium , Cowpox virus , Vaccination
10.
J Gen Virol ; 103(11)2022 11.
Article in English | MEDLINE | ID: mdl-36409607

ABSTRACT

The vaccine effectiveness (VE) of the A/H1N1pdm09 component of the 2017-18 quadrivalent live attenuated influenza vaccine (QLAIV) was improved by performing rational haemagglutinin (HA) mutagenesis. Introducing N125D, D127E, D222G and R223Q substitutions into the HA protein of A/Slovenia/2903/2015 (A/SLOV15) enhanced replicative fitness in primary human nasal epithelial cells (hNECs). This allowed A/SLOV15 to overcome inter-strain competition in QLAIV, resulting in improved VE.During strain development for the 2021-22 QLAIV formulation, A/H1N1pdm09 LAIV viruses containing wild-type (WT) HA and neuraminidase (NA) sequences were found to replicate poorly in embryonated eggs and hNECs. We aimed to enhance replicative fitness via the HA mutagenesis approach that was performed previously for A/SLOV15. Therefore, combinations of these four mutations were introduced into the HA protein of representative 6B.1A-5a.2 viruses, A/Victoria/2570/2019 and A/Victoria/1/2020 (A/VIC1). Replicative fitness of A/VIC1 V7 was improved ~30-fold in eggs and ~300-fold in hNECs relative to its parent, without compromising other critical LAIV characteristics.


Subject(s)
Influenza Vaccines , Influenza, Human , Humans , Influenza Vaccines/genetics , Cowpox virus , Seasons , Vaccines, Attenuated/genetics , Hemagglutinins
11.
Viruses ; 14(11)2022 10 29.
Article in English | MEDLINE | ID: mdl-36366493

ABSTRACT

During the last decade, endemic swine H1 influenza A viruses (IAV) from six different genetic clades of the hemagglutinin gene caused zoonotic infections in humans. The majority of zoonotic events with swine IAV were restricted to a single case with no subsequent transmission. However, repeated introduction of human-seasonal H1N1, continual reassortment between endemic swine IAV, and subsequent drift in the swine host resulted in highly diverse swine IAV with human-origin genes that may become a risk to the human population. To prepare for the potential of a future swine-origin IAV pandemic in humans, public health laboratories selected candidate vaccine viruses (CVV) for use as vaccine seed strains. To assess the pandemic risk of contemporary US swine H1N1 or H1N2 strains, we quantified the genetic diversity of swine H1 HA genes, and identified representative strains from each circulating clade. We then characterized the representative swine IAV against human seasonal vaccine and CVV strains using ferret antisera in hemagglutination inhibition assays (HI). HI assays revealed that 1A.3.3.2 (pdm09) and 1B.2.1 (delta-2) demonstrated strong cross reactivity to human seasonal vaccines or CVVs. However, swine IAV from three clades that represent more than 50% of the detected swine IAVs in the USA showed significant reduction in cross-reactivity compared to the closest CVV virus: 1A.1.1.3 (alpha-deletion), 1A.3.3.3-clade 3 (gamma), and 1B.2.2.1 (delta-1a). Representative viruses from these three clades were further characterized in a pig-to-ferret transmission model and shown to exhibit variable transmission efficiency. Our data prioritize specific genotypes of swine H1N1 and H1N2 to further investigate in the risk they pose to the human population.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza A virus , Orthomyxoviridae Infections , Swine Diseases , Animals , Swine , Humans , Ferrets , Influenza A Virus, H1N1 Subtype/genetics , Orthomyxoviridae Infections/epidemiology , Cowpox virus , Immune Sera , Swine Diseases/epidemiology
12.
Viruses ; 14(10)2022 09 28.
Article in English | MEDLINE | ID: mdl-36298689

ABSTRACT

Cowpox virus (CPXV; genus Orthopoxvirus; family Poxviridae) is the causative agent of cowpox, a self-limiting zoonotic infection. CPXV is endemic in Eurasia, and human CPXV infections are associated with exposure to infected animals. In the Fennoscandian region, five CPXVs isolated from cats and humans were collected and used in this study. We report the complete sequence of their genomes, which ranged in size from 220-222 kbp, containing between 215 and 219 open reading frames. The phylogenetic analysis of 87 orthopoxvirus strains, including the Fennoscandian CPXV isolates, confirmed the division of CPXV strains into at least five distinct major clusters (CPXV-like 1, CPXV-like 2, VACV-like, VARV-like and ECTV-Abatino-like) and can be further divided into eighteen sub-species based on the genetic and patristic distances. Bayesian time-scaled evolutionary history of CPXV was reconstructed employing concatenated 62 non-recombinant conserved genes of 55 CPXV. The CPXV evolution rate was calculated to be 1.65 × 10-5 substitution/site/year. Our findings confirmed that CPXV is not a single species but a polyphyletic assemblage of several species and thus, a reclassification is warranted.


Subject(s)
Cowpox , Orthopoxvirus , Animals , Humans , Cowpox virus , Phylogeny , Bayes Theorem , Cowpox/veterinary , Orthopoxvirus/genetics , Genomics
13.
PLoS Pathog ; 18(9): e1010783, 2022 09.
Article in English | MEDLINE | ID: mdl-36121874

ABSTRACT

The genome of cowpoxvirus (CPXV) could be considered prototypical for orthopoxviridae (OXPV) since it contains many open reading frames (ORFs) absent or lost in other OPXV, including vaccinia virus (VACV). These additional ORFs are non-essential for growth in vitro but are expected to contribute to the broad host range, virulence and immune evasion characteristics of CPXV. For instance, unlike VACV, CPXV encodes proteins that interfere with T cell stimulation, either directly or by preventing antigen presentation or co-stimulation. When studying the priming of naïve T cells, we discovered that CPXV, but not VACV, encodes a secreted factor that interferes with activation and proliferation of naïve CD8+ and CD4+ T cells, respectively, in response to anti-CD3 antibodies, but not to other stimuli. Deletion mapping revealed that the inhibitory protein is encoded by CPXV14, a small secreted glycoprotein belonging to the poxvirus immune evasion (PIE) family and containing a smallpoxvirus encoded chemokine receptor (SECRET) domain that mediates binding to chemokines. We demonstrate that CPXV14 inhibition of antibody-mediated T cell activation depends on the presence of Fc-gamma receptors (FcγRs) on bystander cells. In vitro, CPXV14 inhibits FcγR-activation by antigen/antibody complexes by binding to FcγRs with high affinity and immobilized CPXV14 can trigger signaling through FcγRs, particularly the inhibitory FcγRIIB. In vivo, CPXV14-deleted virus showed reduced viremia and virulence resulting in reduced weight loss and death compared to wildtype virus whereas both antibody and CD8+ T cell responses were increased in the absence of CPXV14. Furthermore, no impact of CPXV14-deletion on virulence was observed in mice lacking the inhibitory FcγRIIB. Taken together our results suggest that CPXV14 contributes to virulence and immune evasion by binding to host FcγRs.


Subject(s)
Cowpox virus , Immune Evasion , Animals , Cowpox virus/genetics , Glycoproteins , Mice , Receptors, Chemokine , Receptors, IgG , Vaccinia virus , Virulence
14.
Viruses ; 14(9)2022 08 27.
Article in English | MEDLINE | ID: mdl-36146705

ABSTRACT

Among the Poxviridae family, orthopoxvirus is the most notorious genus. Several DNA viruses belonging to this group are known to produce human disease from the life-threatening variola virus (VARV) (the causative agent of smallpox), monkeypox virus (MPXV), cowpox virus (CPXV), and vaccinia virus (VACV). These orthopoxviruses still remain a public health concern as VACV or CPXV still cause emerging endemic threads, especially in developing countries. MPXV is able to cause sporadic human outbreaks of a smallpox-like zoonotic disease and, in May 2022, hundreds of cases related to MPXV have been reported from more than 30 countries around the globe. At the end of July, monkeypox (MPX) outbreak was even declared a global health emergency by the World Health Organization (WHO). Many aspects remain unclear regarding this outbreak and a deep understanding of orthopoxvirus might have crucial and evident implications. During the era in which people under 45 years old are not protected against VACV, the potential use of orthopoxviruses as a biological weapon raises global concern considering the rapid spreading of the current MPX outbreak in vulnerable populations. Hence, we review the most recent evidence about phylogenesis, pathogenesis, prevention, and treatment for this concerning disease.


Subject(s)
Mpox (monkeypox) , Orthopoxvirus , Smallpox , Variola virus , Biological Warfare Agents , Cowpox virus , Humans , Middle Aged , Mpox (monkeypox)/epidemiology , Monkeypox virus/genetics , Orthopoxvirus/genetics , Vaccinia virus , Variola virus/genetics
16.
Viruses ; 13(8)2021 08 09.
Article in English | MEDLINE | ID: mdl-34452435

ABSTRACT

Hemorrhagic smallpox, caused by variola virus (VARV), was a rare but nearly 100% lethal human disease manifestation. Hemorrhagic smallpox is frequently characterized by secondary bacterial infection, coagulopathy, and myocardial and subendocardial hemorrhages. Previous experiments have demonstrated that intravenous (IV) cowpox virus (CPXV) exposure of macaques mimics human hemorrhagic smallpox. The goal of this experiment was to further understand the onset, nature, and severity of cardiac pathology and how it may contribute to disease. The findings support an acute late-stage myocarditis with lymphohistiocytic infiltrates in the CPXV model of hemorrhagic smallpox.


Subject(s)
Cowpox virus/pathogenicity , Hemorrhage/virology , Myocarditis/virology , Smallpox/physiopathology , Smallpox/virology , Acute Disease , Animals , Disease Models, Animal , Female , Macaca fascicularis/virology , Male , Myocarditis/veterinary , Smallpox/complications
17.
Viruses ; 13(8)2021 08 17.
Article in English | MEDLINE | ID: mdl-34452494

ABSTRACT

Mass vaccination has played a critical role in the global eradication of smallpox. Various vaccinia virus (VACV) strains, whose origin has not been clearly documented in most cases, have been used as live vaccines in different countries. These VACV strains differed in pathogenicity towards various laboratory animals and in reactogenicity exhibited upon vaccination of humans. In this work, we studied the development of humoral and cellular immune responses in BALB/c mice inoculated intranasally (i.n.) or intradermally (i.d.) with the VACV LIVP strain at a dose of 105 PFU/mouse, which was used in Russia as the first generation smallpox vaccine. Active synthesis of VACV-specific IgM in the mice occurred on day 7 after inoculation, reached a maximum on day 14, and decreased by day 29. Synthesis of virus-specific IgG was detected only from day 14, and the level increased significantly by day 29 after infection of the mice. Immunization (i.n.) resulted in significantly higher production of VACV-specific antibodies compared to that upon i.d. inoculation of LIVP. There were no significant differences in the levels of the T cell response in mice after i.n. or i.d. VACV administration at any time point. The maximum level of VACV-specific T-cells was detected on day 14. By day 29 of the experiment, the level of VACV-specific T-lymphocytes in the spleen of mice significantly decreased for both immunization procedures. On day 30 after immunization with LIVP, mice were infected with the cowpox virus at a dose of 46 LD50. The i.n. immunized mice were resistant to this infection, while 33% of i.d. immunized mice died. Our findings indicate that the level of the humoral immune response to vaccination may play a decisive role in protection of animals from orthopoxvirus reinfection.


Subject(s)
Adaptive Immunity , Cowpox virus/physiology , Cowpox/prevention & control , Reinfection/prevention & control , Vaccinia virus/immunology , Vaccinia/immunology , Viral Vaccines/administration & dosage , Animals , Antibodies, Viral/immunology , Cowpox/immunology , Cowpox/virology , Cowpox virus/genetics , Cowpox virus/immunology , Humans , Mice , Mice, Inbred BALB C , Reinfection/immunology , Reinfection/virology , T-Lymphocytes/immunology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Vaccinia/virology , Vaccinia virus/genetics , Vaccinia virus/physiology , Viral Vaccines/immunology
18.
Emerg Infect Dis ; 27(10): 2570-2577, 2021 10.
Article in English | MEDLINE | ID: mdl-34352194

ABSTRACT

Cowpox virus (CPXV) has an animal reservoir and is typically transmitted to humans by contact with infected animals. In 2017, CPXV infection of a pregnant woman in France led to the death of her fetus. Fetal death after maternal orthopoxvirus (smallpox) vaccination has been reported; however, this patient had not been vaccinated. Investigation of the patient's domestic animals failed to demonstrate prevalence of CPXV infection among them. The patient's diagnosis was confirmed by identifying CPXV DNA in all fetal and maternal biopsy samples and infectious CPXV in biopsy but not plasma samples. This case of fetal death highlights the risk for complications of orthopoxvirus infection during pregnancy. Among orthopoxviruses, fetal infection has been reported for variola virus and vaccinia virus; our findings suggest that CPXV poses the same threats for infection complications as vaccinia virus.


Subject(s)
Cowpox , Orthopoxvirus , Animals , Cowpox/diagnosis , Cowpox/epidemiology , Cowpox/veterinary , Cowpox virus/genetics , Female , Fetal Death , Fetus , France/epidemiology , Humans , Young Adult
19.
Virol J ; 18(1): 173, 2021 08 23.
Article in English | MEDLINE | ID: mdl-34425838

ABSTRACT

Zoonotic orthopoxvirus infections continue to represent a threat to human health. The disease caused by distinct orthopoxviruses differs in terms of symptoms and severity, which may be explained by the unique repertoire of virus factors that modulate the host's immune response and cellular machinery. We report here on the construction of recombinant cowpox viruses (CPXV) which either lack the host range factor p28 completely or express truncated variants of p28. We show that p28 is essential for CPXV replication in macrophages of human or mouse origin and that the C-terminal RING finger domain of p28 is necessary to allow CPXV replication in macrophages.


Subject(s)
Cowpox virus , Host Specificity , Macrophages/virology , Viral Proteins/genetics , Virus Replication , Animals , Cowpox virus/genetics , Cowpox virus/physiology , Mice
20.
Antiviral Res ; 191: 105086, 2021 07.
Article in English | MEDLINE | ID: mdl-33992710

ABSTRACT

Decades after the eradication of smallpox and the discontinuation of routine smallpox vaccination, over half of the world's population is immunologically naïve to variola virus and other orthopoxviruses (OPXVs). Even in those previously vaccinated against smallpox, protective immunity wanes over time. As such, there is a concomitant increase in the incidence of human OPXV infections worldwide. To identify novel antiviral compounds with potent anti-OPXV potential, we characterized the inhibitory activity of PAV-866 and other methylene blue derivatives against the prototypic poxvirus, vaccinia virus (VACV). These compounds inactivated virions prior to infection and consequently inhibited viral binding, fusion and entry. The compounds exhibited strong virucidal activity at non-cytotoxic concentrations, and inhibited VACV infection when added before, during or after viral adsorption. The compounds were effective against other OPXVs including monkeypox virus, cowpox virus and the newly identified Akhmeta virus. Altogether, these findings reveal a novel mode of inhibition that has not previously been demonstrated for small molecule compounds against VACV. Additional studies are in progress to determine the in vivo efficacy of these compounds against OPXVs and further characterize the anti-viral effects of these derivatives.


Subject(s)
Antiviral Agents/pharmacology , Methylene Blue/chemistry , Methylene Blue/pharmacology , Orthopoxvirus/drug effects , Antiviral Agents/chemistry , Cell Line , Cowpox virus/drug effects , HeLa Cells , Humans , Monkeypox virus/drug effects , Orthopoxvirus/classification , Vaccinia virus/drug effects , Virus Attachment/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...