Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
1.
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
2.
PLoS Pathog ; 17(4): e1009171, 2021 04.
Article in English | MEDLINE | ID: mdl-33872324

ABSTRACT

Virions are a common antigen source for many viral vaccines. One limitation to using virions is that the antigen abundance is determined by the content of each protein in the virus. This caveat especially applies to viral-based influenza vaccines where the low abundance of the neuraminidase (NA) surface antigen remains a bottleneck for improving the NA antibody response. Our systematic analysis using recent H1N1 vaccine antigens demonstrates that the NA to hemagglutinin (HA) ratio in virions can be improved by exchanging the viral backbone internal genes, especially the segment encoding the polymerase PB1 subunit. The purified inactivated virions with higher NA content show a more spherical morphology, a shift in the balance between the HA receptor binding and NA receptor release functions, and induce a better NA inhibitory antibody response in mice. These results indicate that influenza viruses support a range of ratios for a given NA and HA pair which can be used to produce viral-based influenza vaccines with higher NA content that can elicit more balanced neutralizing antibody responses to NA and HA.


Subject(s)
Antibodies, Viral/immunology , Hemagglutinins/immunology , Influenza Vaccines/immunology , Influenza, Human/virology , Neuraminidase/genetics , Animals , Antibodies, Neutralizing/blood , Cowpox virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Influenza A Virus, H1N1 Subtype/immunology , Mice
3.
Viruses ; 12(8)2020 07 24.
Article in English | MEDLINE | ID: mdl-32722032

ABSTRACT

The mass smallpox vaccination campaign has played a crucial role in smallpox eradication. Various strains of the vaccinia virus (VACV) were used as a live smallpox vaccine in different countries, their origin being unknown in most cases. The VACV strains differ in terms of pathogenicity exhibited upon inoculation of laboratory animals and reactogenicity exhibited upon vaccination of humans. Therefore, each generated strain or clonal variant of VACV needs to be thoroughly studied in in vivo systems. The clonal variant 14 of LIVP strain (LIVP-14) was the study object in this work. A comparative analysis of the virulence and immunogenicity of LIVP-14 inoculated intranasally (i.n.), intradermally (i.d.), or subcutaneously (s.c.) to BALB/c mice at doses of 108, 107, and 106 pfu was carried out. Adult mice exhibited the highest sensitivity to the i.n. administered LIVP-14 strain, although the infection was not lethal. The i.n. inoculated LIVP-14 replicated efficiently in the lungs. Furthermore, this virus was accumulated in the brain at relatively high concentrations. Significantly lower levels of LIVP-14 were detected in the liver, kidneys, and spleen of experimental animals. No clinical manifestations of the disease were observed after i.d. or s.c. injection of LIVP-14 to mice. After s.c. inoculation, the virus was detected only at the injection site, while it could disseminate to the liver and lungs when delivered via i.d. administration. A comparative analysis of the production of virus-specific antibodies by ELISA and PRNT revealed that the highest level of antibodies was induced in i.n. inoculated mice; a lower level of antibodies was observed after i.d. administration of the virus and the lowest level after s.c. injection. Even at the lowest studied dose (106 pfu), i.n. or i.d. administered LIVP-14 completely protected mice against infection with the cowpox virus at the lethal dose. Our findings imply that, according to the ratio between such characteristics as pathogenicity/immunogenicity/protectivity, i.d. injection is the optimal method of inoculation with the VACV LIVP-14 strain to ensure the safe formation of immune defense after vaccination against orthopoxviral infections.


Subject(s)
Antibodies, Viral/blood , Vaccinia virus/immunology , Vaccinia virus/pathogenicity , Administration, Intranasal , Animals , Antibodies, Neutralizing/blood , Cowpox virus/immunology , Female , Immunogenicity, Vaccine , Injections, Intradermal , Injections, Subcutaneous , Male , Mice , Mice, Inbred BALB C , Smallpox Vaccine , Vaccinia/prevention & control , Vaccinia/virology , Vaccinia virus/classification , Virulence
4.
Proc Natl Acad Sci U S A ; 116(42): 21113-21119, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31575740

ABSTRACT

Costimulation is required for optimal T cell activation, yet it is unclear whether poxviruses dedicatedly subvert costimulation during infection. Here, we report that the secreted M2 protein encoded by cowpox virus (CPXV) specifically interacts with human and murine B7.1 (CD80) and B7.2 (CD86). We also show that M2 competes with CD28 and CTLA4 for binding to cell surface B7 ligands, with stronger efficacy against CD28. Functionally, recombinant M2 and culture supernatants from wild-type (WT) but not M2-deficient (∆M2) CPXV-infected cells can potently suppress B7 ligand-mediated T cell proliferation and interleukin-2 (IL-2) production. Furthermore, we observed increased antiviral CD4 and CD8 T cell responses in C57BL/6 mice challenged by ∆M2 CPXV compared with WT virus. These differences in immune responses to ∆M2 and WT CPXV were not observed in CD28-deficient mice. Taken together, our findings define a mechanism of viral sabotage of T cell activation that highlights the role of CD28 costimulation in host defense against poxvirus infections.


Subject(s)
B7-1 Antigen/immunology , B7-2 Antigen/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cowpox virus/immunology , Lymphocyte Activation/immunology , Viral Proteins/immunology , Animals , Antigens, CD/immunology , CHO Cells , Cell Line , Cell Line, Tumor , Cell Proliferation/physiology , Cowpox/immunology , Cowpox/virology , Cricetulus , Humans , Interleukin-2/immunology , Jurkat Cells , Mice , Mice, Inbred C57BL , THP-1 Cells , U937 Cells
5.
Emerg Infect Dis ; 25(2): 212-219, 2019 02.
Article in English | MEDLINE | ID: mdl-30666929

ABSTRACT

We report a case of atypical cowpox virus infection in France in 2016. The patient sought care for thoracic lesions after injury from the sharp end of a metallic guardrail previously stored in the ground. We isolated a cowpox virus from the lesions and sequenced its whole genome. The patient reported that he had been previously vaccinated against smallpox. We describe an alternative route of cowpox virus infection and raise questions about the immunological status of smallpox-vaccinated patients for circulating orthopoxviruses.


Subject(s)
Cowpox virus/immunology , Smallpox/epidemiology , Smallpox/virology , Animals , Cell Line , Computational Biology/methods , Cowpox/immunology , Cowpox/pathology , Cowpox/virology , Cowpox virus/classification , Cowpox virus/genetics , Cowpox virus/isolation & purification , France/epidemiology , Genome, Viral , High-Throughput Nucleotide Sequencing , Humans , Phylogeny , Smallpox/prevention & control , Smallpox Vaccine/immunology , Vaccination , Virus Replication
6.
Viruses ; 10(12)2018 12 05.
Article in English | MEDLINE | ID: mdl-30563103

ABSTRACT

The poxviruses are large, linear, double-stranded DNA viruses about 130 to 230 kbp, that have an animal origin and evolved to infect a wide host range. Variola virus (VARV), the causative agent of smallpox, is a poxvirus that infects only humans, but other poxviruses such as monkey poxvirus and cowpox virus (CPXV) have crossed over from animals to infect humans. Therefore understanding the biology of poxviruses can devise antiviral strategies to prevent these human infections. In this study we used a system-based approach to examine the host responses to three orthopoxviruses, CPXV, vaccinia virus (VACV), and ectromelia virus (ECTV) in the murine macrophage RAW 264.7 cell line. Overall, we observed a significant down-regulation of gene expressions for pro-inflammatory cytokines, chemokines, and related receptors. There were also common and virus-specific changes in the immune-regulated gene expressions for each poxvirus-infected RAW cells. Collectively our results showed that the murine macrophage RAW 264.7 cell line is a suitable cell-based model system to study poxvirus host response.


Subject(s)
Cowpox virus/immunology , Cytokines/immunology , Ectromelia virus/immunology , Macrophages/immunology , Vaccinia virus/immunology , Animals , Chemokines/genetics , Chemokines/immunology , Cytokines/genetics , Down-Regulation , Gene Expression , Macrophages/virology , Mice , Microarray Analysis , Polymerase Chain Reaction , RAW 264.7 Cells , Up-Regulation
8.
Lancet Infect Dis ; 18(2): e55-e63, 2018 02.
Article in English | MEDLINE | ID: mdl-28827144

ABSTRACT

In 1796, Edward Jenner developed the smallpox vaccine consisting of pustular material obtained from lesions on cows affected by so-called cow-pox. The disease, caused by cowpox virus, confers crossprotection against smallpox. However, historical evidence suggests that Jenner might have used vaccinia virus or even horsepox virus instead of cowpox virus. Mysteries surrounding the origin and nature of the smallpox vaccine persisted during the 19th century, a period of intense exchange of vaccine strains, including the Beaugency lymph. This lymph was obtained from spontaneous cases of cow-pox in France in 1866 and then distributed worldwide. A detailed Historical Review of the distribution of the Beaugency lymph supports recent genetic analyses of extant vaccine strains, suggesting the lymph was probably a vaccinia strain or a horsepox-like virus. This Review is a historical investigation that revisits the mysteries of the smallpox vaccine and reveals an intricate evolutionary relationship of extant vaccinia strains.


Subject(s)
Cowpox virus/immunology , Cowpox virus/isolation & purification , Smallpox Vaccine/history , Smallpox Vaccine/isolation & purification , Smallpox/prevention & control , Vaccination/history , Animals , Cattle , Cowpox virus/classification , Cowpox virus/genetics , France , History, 16th Century , History, 17th Century , History, 18th Century , History, 19th Century , History, 20th Century , History, 21st Century , Humans
9.
Viruses ; 9(11)2017 11 18.
Article in English | MEDLINE | ID: mdl-29156539

ABSTRACT

Four cowpox virus (CPXV) outbreaks occurred in unrelated alpaca herds in Eastern Germany during 2012-2017. All incidents were initially noticed due to severe, generalized, and finally lethal CPXV infections, which were confirmed by testing of tissue and serum samples. As CPXV-infection has been described in South American camelids (SACs) only three times, all four herds were investigated to gain a deeper understanding of CPXV epidemiology in alpacas. The different herds were investigated twice, and various samples (serum, swab samples, and crusts of suspicious pox lesions, feces) were taken to identify additionally infected animals. Serum was used to detect CPXV-specific antibodies by performing an indirect immunofluorescence assay (iIFA); swab samples, crusts, and feces were used for detection of CPXV-specific DNA in a real-time PCR. In total, 28 out of 107 animals could be identified as affected by CPXV, by iIFA and/or PCR. Herd seroprevalence ranged from 16.1% to 81.2%. To investigate the potential source of infection, wild small mammals were trapped around all alpaca herds. In two herds, CPXV-specific antibodies were found in the local rodent population. In the third herd, CPXV could be isolated from a common vole (Microtus arvalis) found drowned in a water bucket used to water the alpacas. Full genome sequencing and comparison with the genome of a CPXV from an alpaca from the same herd reveal 99.997% identity, providing further evidence that the common vole is a reservoir host and infection source of CPXV. Only in the remaining fourth herd, none of the trapped rodents were found to be CPXV-infected. Rodents, as ubiquitous reservoir hosts, in combination with increasingly popular alpacas, as susceptible species, suggest an enhanced risk of future zoonotic infections.


Subject(s)
Camelids, New World/virology , Cowpox/epidemiology , Disease Outbreaks , Zoonoses/epidemiology , Animals , Antibodies, Viral/blood , Arvicolinae/virology , Cowpox/immunology , Cowpox/virology , Cowpox virus/genetics , Cowpox virus/immunology , Cowpox virus/physiology , Disease Reservoirs/virology , Germany/epidemiology , Phylogeny , Polymerase Chain Reaction , Seroepidemiologic Studies , Zoonoses/immunology , Zoonoses/virology
10.
PLoS One ; 12(11): e0187089, 2017.
Article in English | MEDLINE | ID: mdl-29121668

ABSTRACT

Cowpox virus infections in captive cheetahs (Acinonyx jubatus) with high morbidity and mortality have already been reported in the UK and Russia in the 1970s. However, most of the reported cases have been singular events. Here, we report a total of five cowpox virus outbreaks in cheetahs in the same safari park in Denmark between 2010 and 2014. Nine cheetahs showed varying severity of clinical disease; two of them died (22%). All episodes occurred between August and October of the respective year. No other carnivores kept at the same institution nor the keepers taking care of the animals were clinically affected. The clinical picture of cowpox was confirmed by extensive laboratory investigations including histopathological and molecular analyses as well as cell culture isolation of a cowpox virus. High anti-orthopoxvirus antibody titers were detected in all 9 diseased cheetahs compared to seven contact cheetahs without clinical signs and 13 cheetahs not in direct contact. Additionally, whole genome sequencing from one sample of each cluster with subsequent phylogenetic analysis showed that the viruses from different outbreaks have individual sequences but clearly form a clade distinct from other cowpox viruses. However, the intra-clade distances are still larger than those usually observed within clades of one event. These findings indicate multiple and separate introductions of cowpox virus, probably from wild rodent populations, where the virus keeps circulating naturally and is only sporadically introduced into the cheetahs. Sero-positivity of voles (Arvicola amphibious) caught in zoo grounds strengthens this hypothesis. As a consequence, recommendations are given for medical and physical management of diseased cheetahs, for hygienic measures as well as for pre-shipment isolation before cheetah export from zoo grounds.


Subject(s)
Acinonyx/virology , Cowpox virus/physiology , Cowpox/epidemiology , Cowpox/veterinary , Disease Outbreaks/statistics & numerical data , Seasons , Animals , Animals, Zoo/virology , Antibodies, Viral/immunology , Cowpox/immunology , Cowpox/virology , Cowpox virus/immunology , Denmark/epidemiology , Phylogeny , Real-Time Polymerase Chain Reaction
11.
Viruses ; 9(10)2017 10 13.
Article in English | MEDLINE | ID: mdl-29027916

ABSTRACT

Transcripts are known to be incorporated in particles of DNA viruses belonging to the families of Herpesviridae and Mimiviridae, but the presence of transcripts in other DNA viruses, such as poxviruses, has not been analyzed yet. Therefore, we first established a next-generation-sequencing (NGS)-based protocol, enabling the unbiased identification of transcripts in virus particles. Subsequently, we applied our protocol to analyze RNA in an emerging zoonotic member of the Poxviridae family, namely Cowpox virus. Our results revealed the incorporation of 19 viral transcripts, while host identifications were restricted to ribosomal and mitochondrial RNA. Most viral transcripts had an unknown and immunomodulatory function, suggesting that transcript incorporation may be beneficial for poxvirus immune evasion. Notably, the most abundant transcript originated from the D5L/I1R gene that encodes a viral inhibitor of the host cytoplasmic DNA sensing machinery.


Subject(s)
Cowpox virus/genetics , High-Throughput Nucleotide Sequencing/methods , Virion/genetics , Animals , Cowpox virus/immunology , Host-Pathogen Interactions , Humans , Immunomodulation , Phylogeny , RNA/genetics , RNA, Mitochondrial , Transcription, Genetic
12.
J Immunol ; 199(2): 403-407, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28615415

ABSTRACT

The vertebrate immune system uses multiple, sometimes redundant, mechanisms to contain pathogenic microorganisms that are always evolving to evade host defenses. Thus, the cowpox virus (CPXV) uses genes encoding CPXV12 and CPXV203 to prevent direct MHC class I presentation of viral peptides by infected cells. However, CD8 T cells are effectively primed against CPXV by cross-presentation of viral Ags in young mice. Old mice accumulate defects in both CD8 T cell activation and cross-presentation. Using a double-deletion mutant (∆12∆203) of CPXV, we show that direct priming of CD8 T cells in old mice yields superior recall responses, establishing a key contribution of this mechanism to host antipoxvirus responses and enhancing our fundamental understanding of how viral manipulation of direct presentation impacts pathogenesis. This also provides a proof of principle that suboptimal CD8 T cell in old organisms can be optimized by manipulating Ag presentation, with implications for vaccine design.


Subject(s)
Aging/immunology , Antigen Presentation , CD8-Positive T-Lymphocytes/immunology , Animals , Antigens, Viral/immunology , Cowpox virus/genetics , Cowpox virus/immunology , Cowpox virus/pathogenicity , Cross-Priming , Histocompatibility Antigens Class I/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Viral Proteins/genetics , Viral Proteins/immunology
13.
Cell ; 167(3): 684-694.e9, 2016 Oct 20.
Article in English | MEDLINE | ID: mdl-27768891

ABSTRACT

Monkeypox (MPXV) and cowpox (CPXV) are emerging agents that cause severe human infections on an intermittent basis, and variola virus (VARV) has potential for use as an agent of bioterror. Vaccinia immune globulin (VIG) has been used therapeutically to treat severe orthopoxvirus infections but is in short supply. We generated a large panel of orthopoxvirus-specific human monoclonal antibodies (Abs) from immune subjects to investigate the molecular basis of broadly neutralizing antibody responses for diverse orthopoxviruses. Detailed analysis revealed the principal neutralizing antibody specificities that are cross-reactive for VACV, CPXV, MPXV, and VARV and that are determinants of protection in murine challenge models. Optimal protection following respiratory or systemic infection required a mixture of Abs that targeted several membrane proteins, including proteins on enveloped and mature virion forms of virus. This work reveals orthopoxvirus targets for human Abs that mediate cross-protective immunity and identifies new candidate Ab therapeutic mixtures to replace VIG.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antibody Specificity , Poxviridae Infections/immunology , Cowpox/immunology , Cowpox virus/immunology , Cross Reactions , Humans , Leukocytes, Mononuclear/immunology , Mpox (monkeypox)/immunology , Monkeypox virus/immunology , Smallpox/immunology , Vaccinia/immunology , Vaccinia virus/immunology , Variola virus/immunology
14.
Vector Borne Zoonotic Dis ; 15(1): 65-72, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25629782

ABSTRACT

In the Mekong Delta in southern Vietnam, rats are commonly traded in wet markets and sold live for food consumption. We investigated seroprevalence to selected groups of rodent-borne viruses among human populations with high levels of animal exposure and among co-located rodent populations. The indirect fluorescence antibody test (IFAT) was used to determine seropositivity to representative reference strains of hantaviruses (Dobrava virus [DOBV], Seoul virus [SEOV]), cowpox virus, arenaviruses (lymphocytic choriomeningitis virus [LCMV]), flaviviruses (tick-borne encephalitis virus [TBEV]), and rodent parechoviruses (Ljungan virus), using sera from 245 humans living in Dong Thap Province and 275 rodents representing the five common rodent species sold in wet markets and present in peridomestic and farm settings. Combined seropositivity to DOBV and SEOV among the rodents and humans was 6.9% (19/275) and 3.7% (9/245), respectively; 1.1% (3/275) and 4.5% (11/245) to cowpox virus; 5.4% (15/275) and 47.3% (116/245) for TBEV; and exposure to Ljungan virus was 18.8% (46/245) in humans, but 0% in rodents. Very little seroreactivity was observed to LCMV in either rodents (1/275, 0.4%) or humans (2/245, 0.8%). Molecular screening of rodent liver tissues using consensus primers for flaviviruses did not yield any amplicons, whereas molecular screening of rodent lung tissues for hantavirus yielded one hantavirus sequence (SEOV). In summary, these results indicate low to moderate levels of endemic hantavirus circulation, possible circulation of a flavivirus in rodent reservoirs, and the first available data on human exposures to parechoviruses in Vietnam. Although the current evidence suggests only limited exposure of humans to known rodent-borne diseases, further research is warranted to assess public health implications of the rodent trade.


Subject(s)
Disease Vectors , Meat/virology , Rodentia/virology , Seroepidemiologic Studies , Animals , Arenavirus/immunology , Arenavirus/isolation & purification , Cowpox virus/immunology , Cowpox virus/isolation & purification , Flavivirus/immunology , Flavivirus/isolation & purification , Orthohantavirus/immunology , Orthohantavirus/isolation & purification , Humans , Parechovirus/immunology , Parechovirus/isolation & purification , Rodentia/immunology , Vietnam/epidemiology , Zoonoses
15.
J Immunol ; 193(4): 1578-89, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25024387

ABSTRACT

CD8(+) CTLs detect virus-infected cells through recognition of virus-derived peptides presented at the cell surface by MHC class I molecules. The cowpox virus protein CPXV012 deprives the endoplasmic reticulum (ER) lumen of peptides for loading onto newly synthesized MHC class I molecules by inhibiting the transporter associated with Ag processing (TAP). This evasion strategy allows the virus to avoid detection by the immune system. In this article, we show that CPXV012, a 9-kDa type II transmembrane protein, prevents peptide transport by inhibiting ATP binding to TAP. We identified a segment within the ER-luminal domain of CPXV012 that imposes the block in peptide transport by TAP. Biophysical studies show that this domain has a strong affinity for phospholipids that are also abundant in the ER membrane. We discuss these findings in an evolutionary context and show that a frameshift deletion in the CPXV012 gene in an ancestral cowpox virus created the current form of CPXV012 that is capable of inhibiting TAP. In conclusion, our findings indicate that the ER-luminal domain of CPXV012 inserts into the ER membrane, where it interacts with TAP. CPXV012 presumably induces a conformational arrest that precludes ATP binding to TAP and, thus, activity of TAP, thereby preventing the presentation of viral peptides to CTLs.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Adenosine Triphosphate/metabolism , Cowpox virus/immunology , Immune Evasion/immunology , T-Lymphocytes, Cytotoxic/immunology , Viral Proteins/immunology , ATP-Binding Cassette Transporters/antagonists & inhibitors , Antigen Presentation/genetics , Antigen Presentation/immunology , Cell Line, Tumor , Cell Membrane/metabolism , Cowpox virus/genetics , Endoplasmic Reticulum/immunology , Frameshift Mutation , HEK293 Cells , Histocompatibility Antigens Class I/immunology , Humans , Protein Binding/immunology , Protein Transport/immunology , Viral Proteins/genetics
16.
Bing Du Xue Bao ; 29(4): 437-41, 2013 Jun.
Article in Chinese | MEDLINE | ID: mdl-23895011

ABSTRACT

Orthopoxvirus vector has a broad prospect in recombinant vaccine research, but the rarely severe side-effect impedes its development. Vaccinia virus and Cowpox virus of Orthopoxvirus have broad host range, and they have typical host range genes as K1L, CP77 and C7L. These three genes affect host range of Vaccinia virus, disturb the cell signaling pathways, suppress immune response and are related to virulence.


Subject(s)
Host Specificity/genetics , Orthopoxvirus/physiology , Viral Proteins/metabolism , Viral Vaccines/immunology , Cell Line , Cowpox virus/genetics , Cowpox virus/immunology , Cowpox virus/pathogenicity , Cowpox virus/physiology , Genetic Vectors , Orthopoxvirus/genetics , Orthopoxvirus/immunology , Orthopoxvirus/pathogenicity , Signal Transduction , Vaccines, Synthetic/immunology , Vaccinia virus/genetics , Vaccinia virus/immunology , Vaccinia virus/pathogenicity , Vaccinia virus/physiology , Viral Proteins/genetics , Virulence
17.
PLoS One ; 8(7): e68685, 2013.
Article in English | MEDLINE | ID: mdl-23874722

ABSTRACT

An adenovirus 5 vector encoding for mouse interferon alpha, subtype 5 (mDEF201) was evaluated for efficacy against lethal cowpox (Brighton strain) and vaccinia (WR strain) virus respiratory and systemic infections in mice. Two routes of mDEF201 administration were used, nasal sinus (5-µl) and pulmonary (50-µl), to compare differences in efficacy, since the preferred treatment of humans would be in a relatively small volume delivered intranasally. Lower respiratory infections (LRI), upper respiratory infections (URI), and systemic infections were induced by 50-µl intranasal, 10-µl intranasal, and 100-µl intraperitoneal virus challenges, respectively. mDEF201 treatments were given prophylactically either 24 h (short term) or 56d (long-term) prior to virus challenge. Single nasal sinus treatments of 10(6) and 10(7) PFU/mouse of mDEF201 protected all mice from vaccinia-induced LRI mortality (comparable to published studies with pulmonary delivered mDEF201). Systemic vaccinia infections responded significantly better to nasal sinus delivered mDEF201 than to pulmonary treatments. Cowpox LRI infections responded to 10(7) mDEF201 treatments, but a 10(6) dose was only weakly protective. Cowpox URI infections were equally treatable by nasal sinus and pulmonary delivered mDEF201 at 10(7) PFU/mouse. Dose-responsive prophylaxis with mDEF201, given one time only 56 d prior to initiating a vaccinia virus LRI infection, was 100% protective from 10(5) to 10(7) PFU/mouse. Improvements in lung hemorrhage score and lung weight were evident, as were decreases in liver, lung, and spleen virus titers. Thus, mDEF201 was able to treat different vaccinia and cowpox virus infections using both nasal sinus and pulmonary treatment regimens, supporting its development for humans.


Subject(s)
Cowpox virus/immunology , Cowpox/prevention & control , Genetic Vectors/genetics , Interferons/genetics , Vaccinia virus/immunology , Vaccinia/prevention & control , Adenoviridae/genetics , Administration, Intranasal , Animals , Cowpox/mortality , Disease Models, Animal , Female , Genetic Vectors/administration & dosage , Humans , Injections, Intraperitoneal , Mice , Vaccinia/mortality
18.
Virol J ; 10: 229, 2013 Jul 10.
Article in English | MEDLINE | ID: mdl-23842430

ABSTRACT

Vaccinia virus protein A33 (A33VACV) plays an important role in protection against orthopoxviruses, and hence is included in experimental multi-subunit smallpox vaccines. In this study we show that single-dose vaccination with recombinant Sindbis virus expressing A33VACV, is sufficient to protect mice against lethal challenge with vaccinia virus WR (VACV-WR) and ectromelia virus (ECTV) but not against cowpox virus (CPXV), a closely related orthopoxvirus. Moreover, a subunit vaccine based on the cowpox virus A33 ortholog (A33CPXV) failed to protect against cowpox and only partially protected mice against VACV-WR challenge. We mapped regions of sequence variation between A33VACV and A33CPXVand analyzed the role of such variations in protection. We identified a single protective region located between residues 104-120 that harbors a putative H-2Kd T cell epitope as well as a B cell epitope - a target for the neutralizing antibody MAb-1G10 that blocks spreading of extracellular virions. Both epitopes in A33CPXV are mutated and predicted to be non-functional. Whereas vaccination with A33VACV did not induce in-vivo CTL activity to the predicted epitope, inhibition of virus spread in-vitro, and protection from lethal VACV challenge pointed to the B cell epitope highlighting the critical role of residue L118 and of adjacent compensatory residues in protection. This epitope's critical role in protection, as well as its modifications within the orthopoxvirus genus should be taken in context with the failure of A33 to protect against CPXV as demonstrated here. These findings should be considered when developing new subunit vaccines and monoclonal antibody based therapeutics against orthopoxviruses, especially variola virus, the etiologic agent of smallpox.


Subject(s)
Cowpox virus/immunology , Ectromelia virus/immunology , Ectromelia, Infectious/prevention & control , Membrane Glycoproteins/immunology , Vaccinia virus/immunology , Vaccinia/prevention & control , Viral Envelope Proteins/immunology , Viral Vaccines/immunology , Adaptive Immunity , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Disease Models, Animal , Drug Carriers , Epitopes, B-Lymphocyte/genetics , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Female , Genetic Variation , Genetic Vectors , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Sindbis Virus/genetics , Viral Envelope Proteins/genetics , Viral Vaccines/administration & dosage
19.
Virol J ; 10: 61, 2013 Feb 20.
Article in English | MEDLINE | ID: mdl-23425254

ABSTRACT

BACKGROUND: Animal-borne orthopoxviruses, like monkeypox, vaccinia and the closely related cowpox virus, are all capable of causing zoonotic infections in humans, representing a potential threat to human health. The disease caused by each virus differs in terms of symptoms and severity, but little is yet know about the reasons for these varying phenotypes. They may be explained by the unique repertoire of immune and host cell modulating factors encoded by each virus. In this study, we analysed the specific modulation of the host cell's gene expression profile by cowpox, monkeypox and vaccinia virus infection. We aimed to identify mechanisms that are either common to orthopoxvirus infection or specific to certain orthopoxvirus species, allowing a more detailed description of differences in virus-host cell interactions between individual orthopoxviruses. To this end, we analysed changes in host cell gene expression of HeLa cells in response to infection with cowpox, monkeypox and vaccinia virus, using whole-genome gene expression microarrays, and compared these to each other and to non-infected cells. RESULTS: Despite a dominating non-responsiveness of cellular transcription towards orthopoxvirus infection, we could identify several clusters of infection-modulated genes. These clusters are either commonly regulated by orthopoxvirus infection or are uniquely regulated by infection with a specific orthopoxvirus, with major differences being observed in immune response genes. Most noticeable was an induction of genes involved in leukocyte migration and activation in cowpox and monkeypox virus-infected cells, which was not observed following vaccinia virus infection. CONCLUSION: Despite their close genetic relationship, the expression profiles induced by infection with different orthopoxviruses vary significantly. It may be speculated that these differences at the cellular level contribute to the individual characteristics of cowpox, monkeypox and vaccinia virus infections in certain host species.


Subject(s)
Cowpox virus/immunology , Gene Expression Regulation , Genes, MHC Class II , Host-Pathogen Interactions , Monkeypox virus/immunology , Vaccinia virus/immunology , Epithelial Cells/virology , Gene Expression Profiling , HeLa Cells , Humans , Microarray Analysis
20.
Mol Immunol ; 55(2): 156-8, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23312338

ABSTRACT

Smallpox decimated humanity for thousands of years before being eradicated by vaccination, a success facilitated by the fact that humans are the only host of variola virus. In contrast, other orthopoxviruses such as cowpox virus can infect a variety of mammalian species, although its dominant reservoir appears to be rodents. This difference in host specificity suggests that cowpox may have developed promiscuous immune evasion strategies to facilitate zoonosis. Recent experiments have established that cowpox can disrupt MHCI antigen presentation during viral infection of both human and murine cells, a process enabled by two unique proteins, CPXV012 and CPXV203. While CPXV012 inhibits antigenic peptide transport from the cytosol to the ER, CPXV203 blocks MHCI trafficking to the cell surface by exploiting the KDEL-receptor recycling pathway. Our recent investigations of CPXV203 reveal that it binds a diverse array of classical and non-classical MHCI proteins with dramatically increased affinities at the lower pH of the Golgi relative to the ER, thereby providing mechanistic insight into how it works synergistically with KDEL receptors to block MHCI surface expression. The strategy used by cowpox to both limit peptide supply and disrupt trafficking of fully assembled MHCI acts as a dual-edged sword that effectively disables adaptive immune surveillance of infected cells.


Subject(s)
Antigen Presentation , Cowpox virus/immunology , Cowpox/immunology , Immune Evasion , Animals , Golgi Apparatus/metabolism , Histocompatibility Antigens Class I/immunology , Humans , Mice , Protein Structure, Secondary , Protein Transport , Receptors, Peptide/metabolism , Viral Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...