Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters

Publication year range
1.
J Infect Dis ; 209(12): 1891-9, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24403555

ABSTRACT

Chikungunya virus (CHIKV) is a mosquito-borne alphavirus that causes major epidemics of rash, fever, and debilitating arthritis. Currently, there are no vaccines or antivirals available for prevention or treatment. We therefore generated 2 live-attenuated vaccine candidates based on the insertion of a picornavirus internal ribosome entry site (IRES) sequence into the genome of CHIKV. Vaccination of cynomolgus macaques with a single dose of either vaccine produced no signs of disease but was highly immunogenic. After challenge with a subcutaneous inoculation of wild-type CHIKV, both vaccine candidates prevented the development of detectable viremia. Protected animals also exhibited no significant changes in core body temperature or cardiovascular rhythm, whereas sham-vaccinated animals showed hyperthermia, followed by sustained hypothermia, as well as significant changes in heart rate. These CHIKV/IRES vaccine candidates appear to be safe and efficacious, supporting their strong potential as human vaccines to protect against CHIKV infection and reduce transmission and further spread.


Subject(s)
Alphavirus Infections/prevention & control , Chikungunya virus/isolation & purification , Macaca fascicularis/immunology , Viral Vaccines/administration & dosage , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Chikungunya Fever , Chikungunya virus/genetics , Chikungunya virus/immunology , Disease Models, Animal , Dose-Response Relationship, Drug , Macaca fascicularis/virology , Telemetry , Vaccination , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Viral Vaccines/immunology
2.
Proc Natl Acad Sci U S A ; 108(38): 15978-83, 2011 Sep 20.
Article in English | MEDLINE | ID: mdl-21930925

ABSTRACT

Antiviral compounds that increase the resistance of host tissues represent an attractive class of therapeutic. Here, we show that squalamine, a compound previously isolated from the tissues of the dogfish shark (Squalus acanthias) and the sea lamprey (Petromyzon marinus), exhibits broad-spectrum antiviral activity against human pathogens, which were studied in vitro as well as in vivo. Both RNA- and DNA-enveloped viruses are shown to be susceptible. The proposed mechanism involves the capacity of squalamine, a cationic amphipathic sterol, to neutralize the negative electrostatic surface charge of intracellular membranes in a way that renders the cell less effective in supporting viral replication. Because squalamine can be readily synthesized and has a known safety profile in man, we believe its potential as a broad-spectrum human antiviral agent should be explored.


Subject(s)
Antiviral Agents/pharmacology , Virus Diseases/drug therapy , Virus Replication/drug effects , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Antiviral Agents/chemistry , Cell Line , Cell Membrane/chemistry , Cell Membrane/drug effects , Cells, Cultured , Cholestanols/chemistry , Cholestanols/pharmacology , Cricetinae , Female , Hepatitis B virus/drug effects , Hepatitis B virus/growth & development , Hepatitis Delta Virus/drug effects , Hepatitis Delta Virus/growth & development , Hepatocytes/drug effects , Hepatocytes/virology , Humans , Male , Mesocricetus , Mice , Mice, Inbred BALB C , Molecular Structure , Muromegalovirus/drug effects , Muromegalovirus/growth & development , Scattering, Small Angle , Virus Diseases/virology , X-Ray Diffraction , rac1 GTP-Binding Protein/chemistry
3.
J Virol ; 86(11): 6084-96, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22457519

ABSTRACT

Chikungunya virus (CHIKV) is the mosquito-borne alphavirus that is the etiologic agent of massive outbreaks of arthralgic febrile illness that recently affected millions of people in Africa and Asia. The only CHIKV vaccine that has been tested in humans, strain 181/clone 25, is a live-attenuated derivative of Southeast Asian human isolate strain AF15561. The vaccine was immunogenic in phase I and II clinical trials; however, it induced transient arthralgia in 8% of the vaccinees. There are five amino acid differences between the vaccine and its parent, as well as five synonymous mutations, none of which involves cis-acting genome regions known to be responsible for replication or packaging. To identify the determinants of attenuation, we therefore tested the five nonsynonymous mutations by cloning them individually or in different combinations into infectious clones derived from two wild-type (WT) CHIKV strains, La Reunion and AF15561. Levels of virulence were compared with those of the WT strains and the vaccine strain in two different murine models: infant CD1 and adult A129 mice. An attenuated phenotype indistinguishable from that of the 181/clone 25 vaccine strain was obtained by the simultaneous expression of two E2 glycoprotein substitutions, with intermediate levels of attenuation obtained with the single E2 mutations. The other three amino acid mutations, in nsP1, 6K, and E1, did not have a detectable effect on CHIKV virulence. These results indicate that the attenuation of strain 181/clone 25 is mediated by two point mutations, explaining the phenotypic instability observed in human vaccinees and also in our studies.


Subject(s)
Amino Acid Substitution , Chikungunya virus/immunology , Chikungunya virus/pathogenicity , Viral Envelope Proteins/genetics , Viral Vaccines/genetics , Viral Vaccines/immunology , Alphavirus Infections/pathology , Alphavirus Infections/virology , Animals , Chikungunya Fever , Disease Models, Animal , Female , Glycoproteins/genetics , Glycoproteins/immunology , Mice , Pregnancy , Vaccines, Attenuated/adverse effects , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Viral Envelope Proteins/immunology , Viral Vaccines/adverse effects , Virulence
4.
J Virol ; 84(2): 1014-25, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19889755

ABSTRACT

The eastern equine encephalitis (EEE) complex consists of four distinct genetic lineages: one that circulates in North America (NA EEEV) and the Caribbean and three that circulate in Central and South America (SA EEEV). Differences in their geographic, pathogenic, and epidemiologic profiles prompted evaluation of their genetic diversity and evolutionary histories. The structural polyprotein open reading frames of all available SA EEEV and recent NA EEEV isolates were sequenced and used in evolutionary and phylogenetic analyses. The nucleotide substitution rate per year for SA EEEV (1.2 x 10(-4)) was lower and more consistent than that for NA EEEV (2.7 x 10(-4)), which exhibited considerable rate variation among constituent clades. Estimates of time since divergence varied widely depending upon the sequences used, with NA and SA EEEV diverging ca. 922 to 4,856 years ago and the two main SA EEEV lineages diverging ca. 577 to 2,927 years ago. The single, monophyletic NA EEEV lineage exhibited mainly temporally associated relationships and was highly conserved throughout its geographic range. In contrast, SA EEEV comprised three divergent lineages, two consisting of highly conserved geographic groupings that completely lacked temporal associations. A phylogenetic comparison of SA EEEV and Venezuelan equine encephalitis viruses (VEEV) demonstrated similar genetic and evolutionary patterns, consistent with the well-documented use of mammalian reservoir hosts by VEEV. Our results emphasize the evolutionary and genetic divergences between members of the NA and SA EEEV lineages, consistent with major differences in pathogenicity and ecology, and propose that NA and SA EEEV be reclassified as distinct species in the EEE complex.


Subject(s)
Encephalomyelitis, Eastern Equine , Evolution, Molecular , Genetic Variation , Animals , Bayes Theorem , Cricetinae , Encephalitis Virus, Eastern Equine/classification , Encephalitis Virus, Eastern Equine/genetics , Encephalitis Virus, Eastern Equine/pathogenicity , Encephalitis Virus, Eastern Equine/physiology , Encephalomyelitis, Eastern Equine/epidemiology , Encephalomyelitis, Eastern Equine/virology , Horse Diseases/epidemiology , Horse Diseases/virology , Horses/virology , Humans , North America/epidemiology , Open Reading Frames , Phylogeny , RNA, Viral/genetics , RNA, Viral/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , South America/epidemiology , Species Specificity , Viral Structural Proteins/genetics
5.
J Virol ; 84(13): 6497-504, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20410280

ABSTRACT

Chikungunya virus (CHIKV), a mosquito-borne alphavirus, has traditionally circulated in Africa and Asia, causing human febrile illness accompanied by severe, chronic joint pain. In Africa, epidemic emergence of CHIKV involves the transition from an enzootic, sylvatic cycle involving arboreal mosquito vectors and nonhuman primates, into an urban cycle where peridomestic mosquitoes transmit among humans. In Asia, however, CHIKV appears to circulate only in the endemic, urban cycle. Recently, CHIKV emerged into the Indian Ocean and the Indian subcontinent to cause major epidemics. To examine patterns of CHIKV evolution and the origins of these outbreaks, as well as to examine whether evolutionary rates that vary between enzootic and epidemic transmission, we sequenced the genomes of 40 CHIKV strains and performed a phylogenetic analysis representing the most comprehensive study of its kind to date. We inferred that extant CHIKV strains evolved from an ancestor that existed within the last 500 years and that some geographic overlap exists between two main enzootic lineages previously thought to be geographically separated within Africa. We estimated that CHIKV was introduced from Africa into Asia 70 to 90 years ago. The recent Indian Ocean and Indian subcontinent epidemics appear to have emerged independently from the mainland of East Africa. This finding underscores the importance of surveillance to rapidly detect and control African outbreaks before exportation can occur. Significantly higher rates of nucleotide substitution appear to occur during urban than during enzootic transmission. These results suggest fundamental differences in transmission modes and/or dynamics in these two transmission cycles.


Subject(s)
Alphavirus Infections/epidemiology , Chikungunya virus/classification , Chikungunya virus/genetics , Disease Outbreaks , Genome, Viral , Phylogeny , RNA, Viral/genetics , Alphavirus Infections/virology , Animals , Chikungunya virus/isolation & purification , Cluster Analysis , Evolution, Molecular , Genotype , Geography , Humans , Molecular Epidemiology , Sequence Analysis, DNA
6.
Reproduction ; 141(6): 849-56, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21389079

ABSTRACT

The invasive and fully antigenic trophoblast of the chorionic girdle portion of the equine fetal membranes has the capacity to survive and differentiate after transplantation to ectopic sites. The objectives of this study were to determine i) the survival time of ectopically transplanted allogeneic trophoblast cells in non-pregnant recipient mares, ii) whether equine chorionic gonadotropin (eCG) can be delivered systemically by transplanted chorionic girdle cells, and iii) whether eCG delivered by the transplanted cells is biologically active and can suppress behavioral signs associated with estrus. Ectopically transplanted chorionic girdle survived for up to 105 days with a mean lifespan of 75 days (95% confidence interval 55-94) and secreted sufficient eCG for the hormone to be measurable in the recipients' circulation. Immunohistochemical labeling of serial biopsies of the transplant sites and measurement of eCG profiles demonstrated that graft survival was similar to the lifespan of equine endometrial cups in normal horse pregnancy. The eCG secreted by the transplanted cells induced corpora lutea formation and sustained systemic progesterone levels in the recipient mares, effects that are also observed during pregnancy. This in turn caused suppression of estrus behavior in the recipients for up to 3 months. Thus, ectopically transplanted equine trophoblast provides an unusual example of sustained viability and function of an immunogenic transplant in a recipient with an intact immune system. This model highlights the importance of innate immunoregulatory capabilities of invasive trophoblast cells and describes a new method to deliver sustained circulating concentrations of eCG in non-pregnant mares.


Subject(s)
Graft Survival , Trophoblasts/transplantation , Vulva/surgery , Analysis of Variance , Animals , Biopsy , Cell Survival , Chorionic Gonadotropin/biosynthesis , Chorionic Gonadotropin/blood , Estrus/metabolism , Female , Horses , Immunohistochemistry , Sexual Behavior, Animal , Time Factors , Transplantation, Homologous , Trophoblasts/immunology , Trophoblasts/metabolism , Vulva/immunology , Vulva/metabolism
7.
Emerg Infect Dis ; 16(9): 1373-80, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20735920

ABSTRACT

Eastern equine encephalitis virus (EEEV; family Togaviridae, genus Alphavirus) is an arbovirus that causes severe disease in humans in North America and in equids throughout the Americas. The enzootic transmission cycle of EEEV in North America involves passerine birds and the ornithophilic mosquito vector, Culiseta melanura, in freshwater swamp habitats. However, the ecology of EEEV in South America is not well understood. Culex (Melanoconion) spp. mosquitoes are considered the principal vectors in Central and South America; however, a primary vertebrate host for EEEV in South America has not yet been identified. Therefore, to further assess the reservoir host potential of wild rodents and wild birds, we compared the infection dynamics of North American and South American EEEV in cotton rats (Sigmodon hispidus) and house sparrows (Passer domesticus). Our findings suggested that each species has the potential to serve as amplification hosts for North and South America EEEVs.


Subject(s)
Disease Vectors , Encephalitis Virus, Eastern Equine , Encephalomyelitis, Eastern Equine/veterinary , Horse Diseases/transmission , Sigmodontinae/virology , Sparrows/virology , Animals , Antibodies, Viral/blood , Communicable Diseases, Emerging/transmission , Communicable Diseases, Emerging/veterinary , Communicable Diseases, Emerging/virology , Encephalitis Virus, Eastern Equine/classification , Encephalitis Virus, Eastern Equine/immunology , Encephalitis Virus, Eastern Equine/isolation & purification , Encephalomyelitis, Eastern Equine/transmission , Encephalomyelitis, Eastern Equine/virology , Horse Diseases/virology , Horses , North America , South America , Species Specificity
8.
J Virol ; 82(10): 4920-30, 2008 May.
Article in English | MEDLINE | ID: mdl-18353963

ABSTRACT

Eastern equine encephalitis virus (EEEV) causes sporadic epidemics of human and equine disease in North America, but South American strains have seldom been associated with human neurologic disease or mortality, despite serological evidence of infection. In mice, most North American and South American strains of EEEV produce neurologic disease that resembles that associated with human and equine infections. We identified a South American strain that is unable to replicate efficiently in the brain or cause fatal disease in mice yet produces 10-fold higher viremia than virulent EEEV strains. The avirulent South American strain was also sensitive to human interferon (IFN)-alpha, -beta, and -gamma, like most South American strains, in contrast to North American strains that were highly resistant. To identify genes associated with IFN sensitivity and virulence, infectious cDNA clones of a virulent North American strain and the avirulent South American strain were constructed. Two reciprocal chimeric viruses containing swapped structural and nonstructural protein gene regions of the North American and South American strains were also constructed and found to replicate efficiently in vitro. Both chimeras produced fatal disease in mice, similar to that caused by the virulent North American strain. Both chimeric viruses also exhibited intermediate sensitivity to human IFN-alpha, -beta, and -gamma compared to that of the North American and South American strains. Virulence 50% lethal dose assays and serial sacrifice experiments further demonstrated that both structural and nonstructural proteins are important contributors to neurovirulence and viral tissue tropism. Together, the results of this study emphasize the complex and important influences of structural and nonstructural protein gene regions on EEEV virulence.


Subject(s)
Encephalitis Virus, Eastern Equine/immunology , Encephalitis Virus, Eastern Equine/pathogenicity , Interferons/immunology , Viral Nonstructural Proteins/immunology , Viral Nonstructural Proteins/physiology , Viral Structural Proteins/immunology , Viral Structural Proteins/physiology , Animals , Encephalitis Virus, Eastern Equine/genetics , Encephalitis Virus, Eastern Equine/growth & development , Encephalomyelitis, Equine/virology , Lethal Dose 50 , Mice , Survival Analysis , Viral Plaque Assay , Viremia , Virulence
9.
J Virol ; 82(18): 9035-42, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18614636

ABSTRACT

Eastern equine encephalitis virus (EEEV) produces the most severe human arboviral disease in North America (NA) and is a potential biological weapon. However, genetically and antigenically distinct strains from South America (SA) have seldom been associated with human disease or mortality despite serological evidence of infection. Because mice and other small rodents do not respond differently to the NA versus SA viruses like humans, we tested common marmosets (Callithrix jacchus) by using intranasal infection and monitoring for weight loss, fever, anorexia, depression, and neurologic signs. The NA EEEV-infected animals either died or were euthanized on day 4 or 5 after infection due to anorexia and neurologic signs, but the SA EEEV-infected animals remained healthy and survived. The SA EEEV-infected animals developed peak viremia titers of 2.8 to 3.1 log(10) PFU/ml on day 2 or 4 after infection, but there was no detectable viremia in the NA EEEV-infected animals. In contrast, virus was detected in the brain, liver, and muscle of the NA EEEV-infected animals at the time of euthanasia or death. Similar to the brain lesions described for human EEE, the NA EEEV-infected animals developed meningoencephalitis in the cerebral cortex with some perivascular hemorrhages. The findings of this study identify the common marmoset as a useful model of human EEE for testing antiviral drugs and vaccine candidates and highlight their potential for corroborating epidemiological evidence that some, if not all, SA EEEV strains are attenuated for humans.


Subject(s)
Callithrix , Disease Models, Animal , Encephalitis Virus, Eastern Equine/pathogenicity , Encephalomyelitis, Equine/pathology , Encephalomyelitis, Equine/physiopathology , Animals , Callithrix/virology , Encephalomyelitis, Equine/mortality , Encephalomyelitis, Equine/virology , Humans , Immunohistochemistry , North America , South America , Viremia/mortality , Viremia/pathology , Viremia/physiopathology , Viremia/virology , Virulence
10.
PLoS Negl Trop Dis ; 11(8): e0005693, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28771475

ABSTRACT

Venezuelan equine encephalitis (VEE) complex alphaviruses are important re-emerging arboviruses that cause life-threatening disease in equids during epizootics as well as spillover human infections. We conducted a comprehensive analysis of VEE complex alphaviruses by sequencing the genomes of 94 strains and performing phylogenetic analyses of 130 isolates using complete open reading frames for the nonstructural and structural polyproteins. Our analyses confirmed purifying selection as a major mechanism influencing the evolution of these viruses as well as a confounding factor in molecular clock dating of ancestors. Times to most recent common ancestors (tMRCAs) could be robustly estimated only for the more recently diverged subtypes; the tMRCA of the ID/IAB/IC/II and IE clades of VEE virus (VEEV) were estimated at ca. 149-973 years ago. Evolution of the IE subtype has been characterized by a significant evolutionary shift from the rest of the VEEV complex, with an increase in structural protein substitutions that are unique to this group, possibly reflecting adaptation to its unique enzootic mosquito vector Culex (Melanoconion) taeniopus. Our inferred tree topologies suggest that VEEV is maintained primarily in situ, with only occasional spread to neighboring countries, probably reflecting the limited mobility of rodent hosts and mosquito vectors.


Subject(s)
Encephalitis Virus, Venezuelan Equine/genetics , Encephalomyelitis, Venezuelan Equine/epidemiology , Evolution, Molecular , Horse Diseases/virology , Americas , Amino Acid Sequence , Animals , Culex/virology , Encephalitis Virus, Venezuelan Equine/isolation & purification , Encephalomyelitis, Venezuelan Equine/virology , Horse Diseases/epidemiology , Horses/virology , Humans , Insect Vectors/virology , Phylogeny
11.
PLoS Negl Trop Dis ; 9(6): e0003800, 2015.
Article in English | MEDLINE | ID: mdl-26115459

ABSTRACT

Chikungunya virus (CHIKV) is a positive sense, single stranded RNA virus in the genus Alphavirus, and the etiologic agent of epidemics of severe arthralgia in Africa, Asia, Europe and, most recently, the Americas. CHIKV causes chikungunya fever (CHIK), a syndrome characterized by rash, fever, and debilitating, often chronic arthritis. In recent outbreaks, CHIKV has been recognized to manifest more neurologic signs of illness in the elderly and those with co-morbidities. The syndrome caused by CHIKV is often self-limited; however, many patients develop persistent arthralgia that can last for months or years. These characteristics make CHIKV not only important from a human health standpoint, but also from an economic standpoint. Despite its importance as a reemerging disease, there is no licensed vaccine or specific treatment to prevent CHIK. Many studies have begun to elucidate the pathogenesis of CHIKF and the mechanism of persistent arthralgia, including the role of the adaptive immune response, which is still poorly understood. In addition, the lack of an animal model for chronic infection has limited studies of CHIKV pathogenesis as well as the ability to assess the safety of vaccine candidates currently under development. To address this deficiency, we used recombination activating gene 1 (RAG1-/-) knockout mice, which are deficient in both T and B lymphocytes, to develop a chronic CHIKV infection model. Here, we describe this model as well as its use in evaluating the safety of a live-attenuated vaccine candidate.


Subject(s)
Adaptive Immunity/immunology , Arthralgia/physiopathology , Chikungunya Fever/immunology , Chikungunya Fever/physiopathology , Chikungunya virus/genetics , Disease Models, Animal , Viral Vaccines/immunology , Analysis of Variance , Animals , Base Sequence , Homeodomain Proteins/genetics , Mice , Mice, Knockout , Molecular Sequence Data , Sequence Analysis, RNA , Viral Load , Viral Plaque Assay
12.
PLoS Negl Trop Dis ; 8(8): e2969, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25101995

ABSTRACT

Mayaro virus (MAYV) is an emerging, mosquito-borne alphavirus that causes a dengue-like illness in many regions of South America, and which has the potential to urbanize. Because no specific treatment or vaccine is available for MAYV infection, we capitalized on an IRES-based approach to develop a live-attenuated MAYV vaccine candidate. Testing in infant, immunocompetent as well as interferon receptor-deficient mice demonstrated a high degree of attenuation, strong induction of neutralizing antibodies, and efficacy against lethal challenge. This vaccine strain was also unable to infect mosquito cells, a major safety feature for a live vaccine derived from a mosquito-borne virus. Further preclinical development of this vaccine candidate is warranted to protect against this important emerging disease.


Subject(s)
Alphavirus/immunology , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Cells, Cultured , Mice , South America , Vaccines, Attenuated/immunology , Virus Replication
13.
Am J Trop Med Hyg ; 88(3): 455-60, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23358636

ABSTRACT

The long-term persistence of Modoc virus (MODV) infection was investigated in a hamster model. Golden hamsters (Mesocricetus auratus) were infected by subcutaneous inoculation with MODV, in which fatal encephalitis developed in 12.5% (2 of 16). Surviving hamsters shed infectious MODV in their urine during the first five months after infection, and infectious MODV was recovered by co-cultivation of kidney tissue up to eight months after infection. There were no histopathologic changes observed in the kidneys despite detection of viral antigen for 250 days after infection. Mild inflammation and neuronal degeneration in the central nervous system were the primary lesions observed during early infection. These findings confirm previous reports of persistent flavivirus infection in animals and suggest a mechanism for the maintenance of MODV in nature.


Subject(s)
Flavivirus Infections/pathology , Flavivirus/classification , Rodent Diseases/virology , Animals , Antigens, Viral , Cricetinae , Immunohistochemistry , Kidney/virology , Rodent Diseases/pathology
14.
Vaccine ; 31(11): 1464-70, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23333212

ABSTRACT

Eastern equine encephalitis virus (EEEV) is a mosquito-borne alphavirus that causes sporadic, often fatal disease outbreaks in humans and equids, and is also a biological threat agent. Two chimeric vaccine candidates were constructed using a cDNA clone with a Sindbis virus (SINV) backbone and structural protein genes from either a North (SIN/NAEEEV) or South American (SIN/SAEEEV) strain of EEEV. The vaccine candidates were tested in a nonhuman primate (NHP) model of eastern equine encephalitis (EEE). Cynomolgus macaques were either sham-vaccinated, or vaccinated with a single dose of either SIN/NAEEEV or SIN/SAEEEV. After vaccination, animals were challenged by aerosol with a virulent North American strain of EEEV (NA EEEV). The SIN/NAEEEV vaccine provided significant protection, and most vaccinated animals survived EEEV challenge (82%) with little evidence of disease, whereas most SIN/SAEEEV-vaccinated (83%) and control (100%) animals died. Protected animals exhibited minimal changes in temperature and cardiovascular rhythm, whereas unprotected animals showed profound hyperthermia and changes in heart rate postexposure. Acute inflammation and neuronal necrosis were consistent with EEEV-induced encephalitis in unprotected animals, whereas no encephalitis-related histopathologic changes were observed in the SIN/NAEEEV-vaccinated animals. These results demonstrate that the chimeric SIN/NAEEEV vaccine candidate protects against an aerosol EEEV exposure.


Subject(s)
Encephalitis Virus, Eastern Equine/immunology , Encephalomyelitis, Equine/prevention & control , Sindbis Virus/genetics , Viral Vaccines/immunology , Aerosols , Animals , Disease Models, Animal , Drug Carriers , Encephalitis Virus, Eastern Equine/genetics , Encephalomyelitis, Equine/immunology , Encephalomyelitis, Equine/mortality , Encephalomyelitis, Equine/pathology , Female , Fever/prevention & control , Genetic Vectors , Macaca , Male , Survival Analysis , Tachycardia/prevention & control , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
15.
PLoS Negl Trop Dis ; 6(4): e1606, 2012.
Article in English | MEDLINE | ID: mdl-22509419

ABSTRACT

Venezuelan equine encephalitis (VEE) is a re-emerging, mosquito-borne viral disease with the potential to cause fatal encephalitis in both humans and equids. Recently, detection of endemic VEE caused by enzootic strains has escalated in Mexico, Peru, Bolivia, Colombia and Ecuador, emphasizing the importance of understanding the enzootic transmission cycle of the etiologic agent, VEE virus (VEEV). The majority of work examining the viral determinants of vector infection has been performed in the epizootic mosquito vector, Aedes (Ochlerotatus) taeniorhynchus. Based on the fundamental differences between the epizootic and enzootic cycles, we hypothesized that the virus-vector interaction of the enzootic cycle is fundamentally different from that of the epizootic model. We therefore examined the determinants for VEEV IE infection in the enzootic vector, Culex (Melanoconion) taeniopus, and determined the number and susceptibility of midgut epithelial cells initially infected and their distribution compared to the epizootic virus-vector interaction. Using chimeric viruses, we demonstrated that the determinants of infection for the enzootic vector are different than those observed for the epizootic vector. Similarly, we showed that, unlike A. taeniorhynchus infection with subtype IC VEEV, C. taeniopus does not have a limited subpopulation of midgut cells susceptible to subtype IE VEEV. These findings support the hypothesis that the enzootic VEEV relationship with C. taeniopus differs from the epizootic virus-vector interaction in that the determinants appear to be found in both the nonstructural and structural regions, and initial midgut infection is not limited to a small population of susceptible cells.


Subject(s)
Culex/virology , Disease Vectors , Encephalitis Virus, Venezuelan Equine/growth & development , Host-Pathogen Interactions , Animals , Encephalitis Virus, Venezuelan Equine/pathogenicity , Epithelial Cells/virology , Female , Gastrointestinal Tract/virology
16.
PLoS Negl Trop Dis ; 6(11): e1875, 2012.
Article in English | MEDLINE | ID: mdl-23133685

ABSTRACT

Venezuelan equine encephalitis virus (VEEV) has been the causative agent for sporadic epidemics and equine epizootics throughout the Americas since the 1930s. In 1969, an outbreak of Venezuelan equine encephalitis (VEE) spread rapidly from Guatemala and through the Gulf Coast region of Mexico, reaching Texas in 1971. Since this outbreak, there have been very few studies to determine the northward extent of endemic VEEV in this region. This study reports the findings of serologic surveillance in the Gulf Coast region of Mexico from 2003-2010. Phylogenetic analysis was also performed on viral isolates from this region to determine whether there have been substantial genetic changes in VEEV since the 1960s. Based on the findings of this study, the Gulf Coast lineage of subtype IE VEEV continues to actively circulate in this region of Mexico and appears to be responsible for infection of humans and animals throughout this region, including the northern State of Tamaulipas, which borders Texas.


Subject(s)
Encephalitis Virus, Venezuelan Equine/isolation & purification , Encephalomyelitis, Venezuelan Equine/epidemiology , Endemic Diseases , Adolescent , Adult , Aged , Child , Child, Preschool , Cluster Analysis , Female , Humans , Infant , Infant, Newborn , Male , Mexico/epidemiology , Middle Aged , Molecular Epidemiology , Molecular Sequence Data , Phylogeny , RNA, Viral/genetics , Sequence Analysis, DNA , Seroepidemiologic Studies , Young Adult
17.
Am J Trop Med Hyg ; 82(2): 354-9, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20134017

ABSTRACT

Western equine encephalitis virus (WEEV) is a zoonotic alphavirus that circulates in western North America between passerine birds and mosquitoes, primarily Culex tarsalis. Since it was isolated in 1930, WEEV has caused tens of thousands of equine deaths in addition to thousands of human cases. In addition because WEEV is a virus of agricultural importance in addition to a public health threat, we developed two live-attenuated chimeric vaccine candidates that have been shown to be immunogenic and efficacious in mouse models. Vaccine candidate strains were developed by inserting the structural protein genes of WEEV strain McMillan (McM) or CO92-1356 into a Sindbis virus (SINV) strain AR339 backbone. The SIN/McM chimera also derived the N-terminal half of its capsid gene from a North American eastern equine encephalitis virus (EEEV) strain FL39-939 (henceforth referred to as SIN/EEE/McM). Although these vaccines do not generate viremia in mice, we further assessed their safety by exposing Cx. tarsalis to artificial blood meals containing high viral titers of each vaccine candidate. Both viruses exhibited a decreased rate of infection, dissemination, and transmission potential compared with the parental alphaviruses. Specifically, SIN/CO92 infected 37% of mosquitoes and disseminated in 8%, but failed to reach the saliva of the mosquitoes. In contrast, the SIN/EEE/McM virus was unable to infect, disseminate, or be transmitted in the saliva of any mosquitoes. These findings suggest that both vaccine candidates are less competent than the parental strains to be transmitted by the primary mosquito vector, Cx. tarsalis, and are unlikely to be reintroduced into a natural WEEV transmission cycle.


Subject(s)
Alphavirus Infections/immunology , Culex/physiology , Encephalitis Virus, Western Equine/genetics , Encephalitis Virus, Western Equine/immunology , Viral Vaccines/administration & dosage , Animals , Host-Pathogen Interactions , Insect Vectors/virology , Mice , Recombination, Genetic , Vaccines, Synthetic , Viral Vaccines/immunology
18.
Am J Trop Med Hyg ; 83(6): 1262-5, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21118932

ABSTRACT

Mosquito surveillance was carried out in three forested regions of Trinidad during July 2007-March 2009. A total of 185,397 mosquitoes representing at least 46 species was collected, divided into pools of 1-50 mosquitoes according to species and sex, and screened for arboviruses using cytopathic effect assays on Vero cell monolayers. Eighty-five viruses were isolated, including members of the genera Alphavirus (Mucambo virus; MUCV) and Orthobunyavirus (Caraparu, Oriboca, Bimiti, and Wyeomyia viruses). Species of the Culex subgenus Melanoconion accounted for 56% of the total number of mosquitoes collected and 97% of the viruses isolated; Cx. (Mel.) portesi accounted for 92% of virus isolations. Our results also implicate for the first time Aedes (Ochlerotatus) hortator as a potential vector of MUCV. Phylogenetic analyses of 43 MUCV strains suggest population subdivision within Trinidad, consistent with the hypothesis of enzootic maintenance in localized rodent populations.


Subject(s)
Alphavirus/classification , Culicidae/virology , Insect Vectors , Orthobunyavirus/classification , Alphavirus/genetics , Alphavirus/isolation & purification , Alphavirus/physiology , Animals , Orthobunyavirus/genetics , Orthobunyavirus/isolation & purification , Orthobunyavirus/physiology , Phylogeny , Population Dynamics , Time Factors , Trees , Trinidad and Tobago
19.
Vaccine ; 27(32): 4309-19, 2009 Jul 09.
Article in English | MEDLINE | ID: mdl-19446595

ABSTRACT

We developed two types of chimeric Sindbis virus (SINV)/western equine encephalitis virus (WEEV) alphaviruses to investigate their potential use as live virus vaccines against WEE. The first-generation vaccine candidate, SIN/CO92, was derived from structural protein genes of WEEV strain CO92-1356, and two second-generation candidates were derived from WEEV strain McMillan. For both first- and second-generation vaccine candidates, the nonstructural protein genes were derived from SINV strain AR339. Second-generation vaccine candidates SIN/SIN/McM and SIN/EEE/McM included the envelope glycoprotein genes from WEEV strain McMillan; however, the amino-terminal half of the capsid, which encodes the RNA-binding domain, was derived from either SINV or eastern equine encephalitis virus (EEEV) strain FL93-939. All chimeric viruses replicated efficiently in mammalian and mosquito cell cultures and were highly attenuated in 6-week-old mice. Vaccinated mice developed little or no detectable disease and showed little or no evidence of challenge virus replication; however, all developed high titers of neutralizing antibodies. Upon intranasal challenge with high doses of virulent WEEV strains, mice vaccinated with >or=10(5)PFU of SIN/CO92 or >or=10(4)PFU of SIN/SIN/McM or SIN/EEE/McM were completely protected from disease. These findings support the potential use of these live-attenuated vaccine candidates as safe and effective vaccines against WEE.


Subject(s)
Encephalitis Virus, Western Equine/immunology , Encephalomyelitis, Equine/prevention & control , Genetic Vectors , Sindbis Virus/genetics , Viral Vaccines/immunology , Animals , Antibodies, Viral/blood , Cell Line , Cricetinae , Encephalitis Virus, Western Equine/genetics , Female , Genome, Viral , Mice , Molecular Sequence Data , Neutralization Tests , Pregnancy , Recombination, Genetic , Sequence Analysis, DNA , Survival Analysis , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/immunology , Viral Structural Proteins/genetics , Viral Structural Proteins/immunology , Viral Vaccines/genetics
20.
PLoS Negl Trop Dis ; 3(9): e514, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19753102

ABSTRACT

Venezuelan equine encephalitis virus (VEEV) has been responsible for hundreds of thousands of human and equine cases of severe disease in the Americas. A passive surveillance study was conducted in Peru, Bolivia and Ecuador to determine the arboviral etiology of febrile illness. Patients with suspected viral-associated, acute, undifferentiated febrile illness of <7 days duration were enrolled in the study and blood samples were obtained from each patient and assayed by virus isolation. Demographic and clinical information from each patient was also obtained at the time of voluntary enrollment. In 2005-2007, cases of Venezuelan equine encephalitis (VEE) were diagnosed for the first time in residents of Bolivia; the patients did not report traveling, suggesting endemic circulation of VEEV in Bolivia. In 2001 and 2003, VEE cases were also identified in Ecuador. Since 1993, VEEV has been continuously isolated from patients in Loreto, Peru, and more recently (2005), in Madre de Dios, Peru. We performed phylogenetic analyses with VEEV from Bolivia, Ecuador and Peru and compared their relationships to strains from other parts of South America. We found that VEEV subtype ID Panama/Peru genotype is the predominant one circulating in Peru. We also demonstrated that VEEV subtype ID strains circulating in Ecuador belong to the Colombia/Venezuela genotype and VEEV from Madre de Dios, Peru and Cochabamba, Bolivia belong to a new ID genotype. In summary, we identified a new major lineage of enzootic VEEV subtype ID, information that could aid in the understanding of the emergence and evolution of VEEV in South America.

SELECTION OF CITATIONS
SEARCH DETAIL