Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Virol ; 96(8): e0020922, 2022 04 27.
Article in English | MEDLINE | ID: mdl-35343792

ABSTRACT

Several highly pathogenic mammarenaviruses cause severe hemorrhagic and neurologic disease in humans for which vaccines and antivirals are limited or unavailable. New World (NW) mammarenavirus Machupo virus (MACV) infection causes Bolivian hemorrhagic fever in humans. We previously reported that the disruption of specific N-linked glycan sites on the glycoprotein (GPC) partially attenuates MACV in an interferon alpha/beta and gamma (IFN-α/ß and -γ) receptor knockout (R-/-) mouse model. However, some capability to induce neurological pathology still remained. The highly pathogenic Junin virus (JUNV) is another NW arenavirus closely related to MACV. An F427I substitution in the GPC transmembrane domain (TMD) rendered JUNV attenuated in a lethal mouse model after intracranial inoculation. In this study, we rationally designed and rescued a MACV containing mutations at two glycosylation sites and the corresponding F438I substitution in the GPC TMD. The MACV mutant is fully attenuated in IFN-α/ß and -γ R-/- mice and outbred guinea pigs. Furthermore, inoculation with this mutant MACV completely protected guinea pigs from wild-type MACV lethal challenge. Last, we found the GPC TMD F438I substitution greatly impaired MACV growth in neuronal cell lines of mouse and human origins. Our results highlight the critical roles of the glycans and the TMD on the GPC in arenavirus virulence, which provide insight into the rational design of potential vaccine candidates for highly pathogenic arenaviruses. IMPORTANCE For arenaviruses, the only vaccine available is the live attenuated Candid#1 vaccine, a JUNV vaccine approved in Argentina. We and others have found that the glycans on GPC and the F427 residue in the GPC TMD are important for virulence of JUNV. Nevertheless, mutating either of them is not sufficient for full and stable attenuation of JUNV. Using reverse genetics, we disrupted specific glycosylation sites on MACV GPC and also introduced the corresponding F438I substitution in the GPC TMD. This MACV mutant is fully attenuated in two animal models and protects animals from lethal infection. Thus, our studies highlight the feasibility of rational attenuation of highly pathogenic arenaviruses for vaccine development. Another important finding from this study is that the F438I substitution in GPC TMD could substantially affect MACV replication in neurons. Future studies are warranted to elucidate the underlying mechanism and the implication of this mutation in arenavirus neural tropism.


Subject(s)
Arenaviruses, New World , Hemorrhagic Fever, American , Viral Vaccines , Animals , Arenaviruses, New World/genetics , Arenaviruses, New World/immunology , Disease Models, Animal , Glycoproteins/metabolism , Glycosylation , Guinea Pigs , Hemorrhagic Fever, American/immunology , Hemorrhagic Fever, American/virology , Junin virus/genetics , Junin virus/immunology , Mutation , Vaccines, Attenuated/immunology , Viral Vaccines/immunology
2.
PLoS Pathog ; 17(3): e1009356, 2021 03.
Article in English | MEDLINE | ID: mdl-33647064

ABSTRACT

Several arenaviruses cause hemorrhagic fevers in humans with high case fatality rates. A vaccine named Candid#1 is available only against Junin virus (JUNV) in Argentina. Specific N-linked glycans on the arenavirus surface glycoprotein (GP) mask important epitopes and help the virus evade antibody responses. However the role of GPC glycans in arenavirus pathogenicity is largely unclear. In a lethal animal model of hemorrhagic fever-causing Machupo virus (MACV) infection, we found that a chimeric MACV with the ectodomain of GPC from Candid#1 vaccine was partially attenuated. Interestingly, mutations resulting in acquisition of N-linked glycans at GPC N83 and N166 frequently occurred in late stages of the infection. These glycosylation sites are conserved in the GPC of wild-type MACV, indicating that this is a phenotypic reversion for the chimeric MACV to gain those glycans crucial for infection in vivo. Further studies indicated that the GPC mutant viruses with additional glycans became more resistant to neutralizing antibodies and more virulent in animals. On the other hand, disruption of these glycosylation sites on wild-type MACV GPC rendered the virus substantially attenuated in vivo and also more susceptible to antibody neutralization, while loss of these glycans did not affect virus growth in cultured cells. We also found that MACV lacking specific GPC glycans elicited higher levels of neutralizing antibodies against wild-type MACV. Our findings revealed the critical role of specific glycans on GPC in arenavirus pathogenicity and have important implications for rational design of vaccines against this group of hemorrhagic fever-causing viruses.


Subject(s)
Antibodies, Viral/immunology , Arenavirus/immunology , Hemorrhagic Fever, American/virology , Junin virus/pathogenicity , Animals , Antibodies, Neutralizing/immunology , Arenaviruses, New World/genetics , Arenaviruses, New World/immunology , Arenaviruses, New World/pathogenicity , Hemorrhagic Fever, American/immunology , Hemorrhagic Fever, American/prevention & control , Humans , Junin virus/immunology , Viral Vaccines/immunology
3.
Sci Rep ; 8(1): 1882, 2018 01 30.
Article in English | MEDLINE | ID: mdl-29382894

ABSTRACT

Zika virus (ZIKV) causes mostly asymptomatic infection or mild febrile illness. However, with an increasing number of patients, various clinical features such as microcephaly, Guillain-Barré syndrome and thrombocytopenia have also been reported. To determine which host factors are related to pathogenesis, the E protein of ZIKV was analyzed with the Informational Spectrum Method, which identifies common information encoded by primary structures of the virus and the respective host protein. The data showed that the ZIKV E protein and the complement component C1q cross-spectra are characterized by a single dominant peak at the frequency F = 0.338, suggesting similar biological properties. Indeed, C1q-specific antibodies were detected in sera obtained from mice and monkeys infected with ZIKV. As C1q has been known to be involved not only in immunity, but also in synaptic organization and different autoimmune diseases, a ZIKV-induced anti-C1q antibody response may contribute to the neurological complications. These findings might also be exploited for the design of safe and efficacious vaccines in the future.


Subject(s)
Antibodies, Viral/immunology , Autoantibodies/immunology , Complement C1q/immunology , Zika Virus Infection/immunology , Zika Virus/immunology , Animals , Antibodies, Neutralizing/immunology , Guillain-Barre Syndrome/immunology , Guillain-Barre Syndrome/virology , Macaca fascicularis , Mice , Microcephaly/immunology , Microcephaly/virology
4.
PLoS Negl Trop Dis ; 10(8): e0004969, 2016 08.
Article in English | MEDLINE | ID: mdl-27580122

ABSTRACT

Machupo virus (MACV), a New World arenavirus, is the etiological agent of Bolivian hemorrhagic fever (BHF). Junin virus (JUNV), a close relative, causes Argentine hemorrhagic fever (AHF). Previously, we reported that a recombinant, chimeric MACV (rMACV/Cd#1-GPC) expressing glycoprotein from the Candid#1 (Cd#1) vaccine strain of JUNV is completely attenuated in a murine model and protects animals from lethal challenge with MACV. A rMACV with a single F438I substitution in the transmembrane domain (TMD) of GPC, which is equivalent to the F427I attenuating mutation in Cd#1 GPC, was attenuated in a murine model but genetically unstable. In addition, the TMD mutation alone was not sufficient to fully attenuate JUNV, indicating that other domains of the GPC may also contribute to the attenuation. To investigate the requirement of different domains of Cd#1 GPC for successful attenuation of MACV, we rescued several rMACVs expressing the ectodomain of GPC from Cd#1 either alone (MCg1), along with the TMD F438I substitution (MCg2), or with the TMD of Cd#1 (MCg3). All rMACVs exhibited similar growth curves in cultured cells. In mice, the MCg1 displayed significant reduction in lethality as compared with rMACV. The MCg1 was detected in brains and spleens of MCg1-infected mice and the infection was associated with tissue inflammation. On the other hand, all animals survived MCg2 and MCg3 infection without detectable levels of virus in various organs while producing neutralizing antibody against Cd#1. Overall our data suggest the indispensable role of each GPC domain in the full attenuation and immunogenicity of rMACV/Cd#1 GPC.


Subject(s)
Junin virus/immunology , Membrane Glycoproteins/immunology , Receptors, Interferon/deficiency , Viral Envelope Proteins/immunology , Viral Vaccines/immunology , A549 Cells , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Cricetinae , Disease Models, Animal , Haplorhini , Hemorrhagic Fever, American/prevention & control , Junin virus/pathogenicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutralization Tests , Receptors, Interferon/genetics , Recombinant Proteins/immunology , Vaccines, Attenuated/immunology
5.
J Virol ; 90(3): 1290-7, 2016 02 01.
Article in English | MEDLINE | ID: mdl-26581982

ABSTRACT

UNLABELLED: Machupo virus (MACV) is the causative agent of Bolivian hemorrhagic fever. Our previous study demonstrated that a MACV strain with a single amino acid substitution (F438I) in the transmembrane domain of glycoprotein is attenuated but genetically unstable in mice. MACV is closely related to Junin virus (JUNV), the causative agent of Argentine hemorrhagic fever. Others and our group have identified the glycoprotein to be the major viral factor determining JUNV attenuation. In this study, we tested the compatibility of the glycoprotein of the Candid#1 live-attenuated vaccine strain of JUNV in MACV replication and its ability to attenuate MACV in vivo. Recombinant MACV with the Candid#1 glycoprotein (rMACV/Cd#1-GPC) exhibited growth properties similar to those of Candid#1 and was genetically stable in vitro. In a mouse model of lethal infection, rMACV/Cd#1-GPC was fully attenuated, more immunogenic than Candid#1, and fully protective against MACV infection. Therefore, the MACV strain expressing the glycoprotein of Candid#1 is safe, genetically stable, and highly protective against MACV infection in a mouse model. IMPORTANCE: Currently, there are no FDA-approved vaccines and/or treatments for Bolivian hemorrhagic fever, which is a fatal human disease caused by MACV. The development of antiviral strategies to combat viral hemorrhagic fevers, including Bolivian hemorrhagic fever, is one of the top priorities of the Implementation Plan of the U.S. Department of Health and Human Services Public Health Emergency Medical Countermeasures Enterprise. Here, we demonstrate for the first time that MACV expressing glycoprotein of Candid#1 is a safe, genetically stable, highly immunogenic, and protective vaccine candidate against Bolivian hemorrhagic fever.


Subject(s)
Arenaviruses, New World/genetics , Arenaviruses, New World/immunology , Membrane Glycoproteins/genetics , Recombination, Genetic , Viral Envelope Proteins/genetics , Viral Vaccines/administration & dosage , Viral Vaccines/immunology , Animal Structures/pathology , Animals , Arenaviruses, New World/pathogenicity , Body Weight , Disease Models, Animal , Genomic Instability , Hemorrhagic Fever, American/pathology , Hemorrhagic Fever, American/prevention & control , Histocytochemistry , Mice, Inbred C57BL , Molecular Sequence Data , Sequence Analysis, DNA , Survival Analysis , Temperature , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Viral Vaccines/genetics , Virulence
6.
J Virol ; 89(11): 5949-56, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25810546

ABSTRACT

UNLABELLED: The New World arenavirus Junin virus (JUNV) is the causative agent of Argentine hemorrhagic fever (AHF), a potentially deadly disease endemic to central regions of Argentina. The live-attenuated Candid #1 (Can) strain of JUNV is currently used to vaccinate the human population at risk. However, the mechanism of attenuation of this strain is still largely unknown. Therefore, the identification and functional characterization of viral genetic determinants dictating JUNV virulence or attenuation would significantly improve the understanding of the mechanisms underlying AHF and facilitate the development of novel, more effective, and safer vaccines. Here, we utilized a reverse genetics approach to generate recombinant JUNV (rJUNV) strains encoding different gene combinations of the pathogenic Romero (Rom) and attenuated Can strains of JUNV. All strains of rJUNV exhibited in vitro growth kinetics similar to those of their parental counterparts. Analysis of virulence of the rJUNV in a guinea pig model of lethal infection that closely reproduces the features of AHF identified the envelope glycoproteins (GPs) as the major determinants of pathogenesis and attenuation of JUNV. Accordingly, rJUNV strains expressing the full-length GPs of Rom and Can exhibited virulent and attenuated phenotypes, respectively, in guinea pigs. Mutation F427I in the transmembrane region of JUNV envelope glycoprotein GP2 has been shown to attenuate the neurovirulence of JUNV in suckling mice. We document that in the guinea pig model of AHF, mutation F427I in GP2 is also highly attenuating but insufficient to prevent virus dissemination and development of mild clinical and pathological symptoms, indicating that complete attenuation of JUNV requires additional mutations present in Can glycoprotein precursor (GPC). IMPORTANCE: Development of antiviral strategies against viral hemorrhagic fevers, including AHF, is one of the top priorities within the Implementation Plan of the U.S. Department of Health and Human Services Public Health Emergency Medical Countermeasures Enterprise. Live-attenuated Candid #1 strain, derived from the 44th mouse brain passage of the prototype XJ strain of JUNV, has been demonstrated to be safe, immunogenic, and highly protective and is currently licensed for human use in Argentina. However, the bases for the attenuated phenotype of Candid #1 have not been established. Therefore, the identification and functional characterization of viral genetic factors implicated in JUNV pathogenesis and attenuation would significantly improve the understanding of the molecular mechanisms underlying AHF and facilitate the development of novel antiviral strategies.


Subject(s)
Glycoproteins/metabolism , Hemorrhagic Fever, American/virology , Junin virus/physiology , Viral Envelope Proteins/metabolism , Animals , Disease Models, Animal , Glycoproteins/genetics , Guinea Pigs , Hemorrhagic Fever, American/pathology , Junin virus/genetics , Reverse Genetics , Viral Envelope Proteins/genetics , Virulence , Virulence Factors
7.
J Virol ; 90(6): 2920-7, 2015 Dec 30.
Article in English | MEDLINE | ID: mdl-26719273

ABSTRACT

UNLABELLED: Approximately one-third of Lassa virus (LASV)-infected patients develop sensorineural hearing loss (SNHL) in the late stages of acute disease or in early convalescence. With 500,000 annual cases of Lassa fever (LF), LASV is a major cause of hearing loss in regions of West Africa where LF is endemic. To date, no animal models exist that depict the human pathology of LF with associated hearing loss. Here, we aimed to develop an animal model to study LASV-induced hearing loss using human isolates from a 2012 Sierra Leone outbreak. We have recently established a murine model for LF that closely mimics many features of human disease. In this model, LASV isolated from a lethal human case was highly virulent, while the virus isolated from a nonlethal case elicited mostly mild disease with moderate mortality. More importantly, both viruses were able to induce SNHL in surviving animals. However, utilization of the nonlethal, human LASV isolate allowed us to consistently produce large numbers of survivors with hearing loss. Surviving mice developed permanent hearing loss associated with mild damage to the cochlear hair cells and, strikingly, significant degeneration of the spiral ganglion cells of the auditory nerve. Therefore, the pathological changes in the inner ear of the mice with SNHL supported the phenotypic loss of hearing and provided further insights into the mechanistic cause of LF-associated hearing loss. IMPORTANCE: Sensorineural hearing loss is a major complication for LF survivors. The development of a small-animal model of LASV infection that replicates hearing loss and the clinical and pathological features of LF will significantly increase knowledge of pathogenesis and vaccine studies. In addition, such a model will permit detailed characterization of the hearing loss mechanism and allow for the development of appropriate diagnostic approaches and medical care for LF patients with hearing impairment.


Subject(s)
Disease Models, Animal , Hearing Loss, Sensorineural/pathology , Lassa Fever/complications , Animals , Cochlear Nerve/pathology , Disease Outbreaks , Ear, Inner/pathology , Hearing Loss, Sensorineural/epidemiology , Histocytochemistry , Humans , Lassa Fever/epidemiology , Lassa virus/isolation & purification , Mice , Microscopy , Sierra Leone/epidemiology , Virulence
8.
J Virol ; 87(19): 10908-11, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23903830

ABSTRACT

Lassa fever (LF) is a potentially lethal human disease that is caused by the arenavirus Lassa virus (LASV). Annually, around 300,000 infections with up to 10,000 deaths occur in regions of Lassa fever endemicity in West Africa. Here we demonstrate that mice lacking a functional STAT1 pathway are highly susceptible to infection with LASV and develop lethal disease with pathology similar to that reported in humans.


Subject(s)
Lassa Fever/virology , Lassa virus/pathogenicity , STAT1 Transcription Factor/physiology , Africa, Western , Animals , Cells, Cultured , Chlorocebus aethiops , Humans , Kidney/metabolism , Kidney/virology , Lassa Fever/genetics , Lassa Fever/mortality , Mice , Mice, Knockout , Receptor, Interferon alpha-beta/physiology , Survival Rate , Vero Cells
9.
J Virol ; 86(6): 3389-92, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22238311

ABSTRACT

Lassa virus (LASV) is the causative agent of Lassa hemorrhagic fever (LF) in humans, a deadly disease endemic to West Africa that results in 5,000 to 10,000 deaths annually. Here we present results demonstrating that functional type I and type II interferon (IFN) signaling is required for efficient control of LASV dissemination and clearance.


Subject(s)
Interferons/immunology , Lassa Fever/immunology , Lassa virus/immunology , Animals , Female , Humans , Lassa Fever/virology , Lassa virus/physiology , Male , Mice , Mice, Knockout , Receptors, Interferon/genetics , Receptors, Interferon/immunology
10.
J Virol ; 84(24): 13063-7, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20926559

ABSTRACT

Junin virus (JUNV) causes a highly lethal human disease, Argentine hemorrhagic fever. Previous work has demonstrated the requirement for human transferrin receptor 1 for virus entry, and the absence of the receptor was proposed to be a major cause for the resistance of laboratory mice to JUNV infection. In this study, we present for the first time in vivo evidence that the disruption of interferon signaling is sufficient to generate a disease-susceptible mouse model for JUNV infection. After peripheral inoculation with virulent JUNV, adult mice lacking alpha/beta and gamma interferon receptors developed disseminated infection and severe disease.


Subject(s)
Arenaviridae Infections/pathology , Arenaviridae Infections/virology , Disease Susceptibility , Junin virus/pathogenicity , Receptor, Interferon alpha-beta/physiology , Receptors, Interferon/physiology , Animals , Arenaviridae Infections/genetics , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Virus Replication , Interferon gamma Receptor
11.
Vaccine ; 28(30): 4713-8, 2010 Jul 05.
Article in English | MEDLINE | ID: mdl-20452431

ABSTRACT

Junin virus (JUNV) is the etiological agent of the potentially lethal, reemerging human disease, Argentine hemorrhagic fever (AHF). The mechanism of the disease development is not well understood and no antiviral therapy is available. Candid 1, a live-attenuated vaccine, has been developed by the US Army and is being used in the endemic area to prevent AHF. This vaccine is only approved for use in Argentina. In this study we have used the alphavirus-based approach to engineer a replicon system based on a human (United States Food and Drug Administration Investigational New Drug status) vaccine TC83 that express heterologous viral antigens, such as glycoproteins (GPC) of Junin virus (JUNV). Preclinical studies testing the immunogenicity and efficacy of TC83/GPC were performed in guinea pigs. A single dose of the live-attenuated alphavirus based vaccine expressing only GPC was immunogenic and provided partial protection, while a double dose of the same vaccine provided a complete protection against JUNV. This is the first scientific report to our knowledge that the immune response against GPC alone is sufficient to prevent lethal disease against JUNV in an animal model.


Subject(s)
Arenaviridae Infections/immunology , Arenaviridae Infections/prevention & control , Junin virus/genetics , Junin virus/immunology , Replicon/immunology , Viral Vaccines/immunology , Animals , Antibodies, Viral/analysis , Antibodies, Viral/biosynthesis , Arenaviridae Infections/virology , Cell Line , Chlorocebus aethiops , Cricetinae , DNA, Viral/genetics , DNA, Viral/immunology , Female , Genetic Vectors , Glycoproteins/chemistry , Glycoproteins/immunology , Guinea Pigs , Neutralization Tests , Plasmids/immunology , Survival Analysis , Telemetry , Vaccines, DNA/immunology , Vero Cells , Viral Load , Viral Plaque Assay
12.
Vaccine ; 27(42): 5875-84, 2009 Sep 25.
Article in English | MEDLINE | ID: mdl-19654064

ABSTRACT

Transmission of highly pathogenic avian influenza (HPAI) between birds and humans is an ongoing threat that holds potential for the emergence of a pandemic influenza strain. A major barrier to an effective vaccine against avian influenza has been the generally poor immunopotency of many of the HPAI strains coupled with the manufacturing constraints employing conventional methodologies. Fusion of flagellin, a toll-like receptor-5 ligand, to vaccine antigens has been shown to enhance the immune response to the fused antigen in preclinical studies. Here, we have evaluated the immunogenicity and efficacy of a panel of flagellin-based hemagglutinin (HA) globular head fusion vaccines in inbred mice. The HA globular head of these vaccines is derived from the A/Vietnam/1203/04 (VN04; H5N1) HA molecule. We find that replacement of domain D3 of flagellin with the VN04 HA globular head creates a highly effective vaccine that elicits protective HAI titers which protect mice against disease and death in a lethal challenge model.


Subject(s)
Flagellin/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Animals , Cell Line , Female , Flagellin/metabolism , Hemagglutination Inhibition Tests , Humans , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Recombinant Proteins/immunology , Toll-Like Receptor 5/metabolism
13.
Vaccine ; 27(30): 4064-73, 2009 Jun 19.
Article in English | MEDLINE | ID: mdl-19446933

ABSTRACT

Studying the mechanisms of host survival resulting from viral encephalitis is critical to the development of vaccines. Here we have shown in several independent studies that high dose treatment with neutralizing antibody prior to intranasal infection with Venezuelan equine encephalitis virus had an antiviral effect in the visceral organs and prolonged survival time of infected mice, even in the absence of alphabeta T cells. Nevertheless, antibody treatment did not prevent the development of lethal encephalitis. On the contrary, the adoptive transfer of primed CD4(+) T cells was necessary to prevent lethal encephalitis in mice lacking alphabeta T cell receptor.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Encephalitis Virus, Venezuelan Equine/immunology , Encephalomyelitis, Venezuelan Equine/immunology , Adoptive Transfer , Animals , Antibodies, Viral/administration & dosage , Antibodies, Viral/immunology , Encephalomyelitis, Venezuelan Equine/prevention & control , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutralization Tests , Receptors, Antigen, T-Cell/deficiency , Survival Analysis
14.
Am J Trop Med Hyg ; 79(2): 275-82, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18689636

ABSTRACT

Argentine hemorrhagic fever (AHF), a systemic infectious disease caused by infection with Junin virus, affects several organs, and patients can show hematologic, cardiovascular, renal, or neurologic symptoms. We compared the virulence of two Junin virus strains in inbred and outbred guinea pigs with the aim of characterizing this animal model better for future vaccine/antiviral efficacy studies. Our data indicate that this passage of the XJ strain is attenuated in guinea pigs. In contrast, the Romero strain is highly virulent in Strain 13 as well as in Hartley guinea pigs, resulting in systemic infection, thrombocytopenia, elevated aspartate aminotransferase levels, and ultimately, uniformly lethal disease. We detected viral antigen in formalin-fixed, paraffin-embedded tissues. Thus, both guinea pig strains are useful animal models for lethal Junin virus (Romero strain) infection and potentially can be used for preclinical trials in vaccine or antiviral drug development.


Subject(s)
Hemorrhagic Fever, American/virology , Junin virus/classification , Junin virus/pathogenicity , Animals , Antigens, Viral/analysis , Chlorocebus aethiops , Female , Guinea Pigs , Liver/virology , Spleen/virology , Vero Cells , Virus Replication
15.
Hum Vaccin ; 4(6): 410-9, 2008.
Article in English | MEDLINE | ID: mdl-18443425

ABSTRACT

Vaccine development for possible influenza pandemics has been challenging. Conventional vaccines such as inactivated and live attenuated virus preparations are limited in terms of production speed and capacity. DNA vaccination has emerged as a potential alternative to conventional vaccines against influenza pandemics. In this study, we use a novel, cell-free DNA manufacturing process (synDNA) to produce prototype linear DNA vaccines against the influenza virus type A/H5N1. This synDNA process does not require bacterial fermentation, so it avoids the use of antibiotic resistance genes and other nucleic acid sequences unrelated to the antigen gene expression in the actual therapeutic DNA construct. The efficacy of various vaccines expressing the hemagglutinin and neuraminidase proteins (H5N1 synDNA), hemagglutinin alone (H5 synDNA) or neuraminidase alone (N1 synDNA) was evaluated in mice. Two of the constructs (H5 synDNA and H5N1 synDNA) induced a robust protective immune response with up to 93% of treated mice surviving a lethal challenge of a virulent influenza A/Vietnam/1203/04 H5N1 isolate. In combination with a potent biological activity and simplified production footprint, these characteristics make DNA vaccines prepared with our synDNA process highly suitable as alternatives to other vaccine preparations.


Subject(s)
Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/chemical synthesis , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Vaccines, DNA/chemical synthesis , Vaccines, DNA/immunology , Animals , Antibodies, Viral/blood , Body Temperature , Body Weight , Enzyme-Linked Immunosorbent Assay , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Mice , Neuraminidase/immunology , Orthomyxoviridae Infections/immunology , Survival Analysis , Viral Proteins/immunology
16.
Virology ; 374(1): 198-209, 2008 Apr 25.
Article in English | MEDLINE | ID: mdl-18234269

ABSTRACT

The post-exposure therapeutic efficacy of injectable peramivir against highly pathogenic avian influenza type A H5N1 was evaluated in mice and in ferrets. Seventy to eighty percent of the H5N1-infected peramivir-treated mice, and 70% in the oseltamivir treated mice survived the 15-day study period, as compared to 36% in control (vehicle) group. Ferrets were infected intranasally with H5N1 followed by treatment with multiple doses of peramivir. In two of three trials, a statistically significant increase in survival over a 16-18 day period resulted from peramivir treatment, with improved survival of 40-64% in comparison to mock-treated or untreated animals. Injected peramivir mitigates virus-induced disease, reduces infectious virus titers in the lungs and brains and promotes survival in ferrets infected intranasally with this highly neurovirulent isolate. A single intramuscular peramivir injection protected mice against severe disease outcomes following infection with highly pathogenic avian influenza and multi-dose treatment was efficacious in ferrets.


Subject(s)
Antiviral Agents/therapeutic use , Cyclopentanes/therapeutic use , Guanidines/therapeutic use , Influenza A Virus, H5N1 Subtype/drug effects , Orthomyxoviridae Infections/drug therapy , Acids, Carbocyclic , Animals , Cyclopentanes/administration & dosage , Ferrets , Guanidines/administration & dosage , Injections, Intramuscular , Lung/virology , Mice , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/physiopathology , Orthomyxoviridae Infections/virology , Oseltamivir/therapeutic use , Survival Analysis , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL