Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
J Infect Dis ; 228(Suppl 6): S414-S426, 2023 10 18.
Article in English | MEDLINE | ID: mdl-37849399

ABSTRACT

The Togaviridae family, genus, Alphavirus, includes several mosquito-borne human pathogens with the potential to spread to near pandemic proportions. Most of these are zoonotic, with spillover infections of humans and domestic animals, but a few such as chikungunya virus (CHIKV) have the ability to use humans as amplification hosts for transmission in urban settings and explosive outbreaks. Most alphaviruses cause nonspecific acute febrile illness, with pathogenesis sometimes leading to either encephalitis or arthralgic manifestations with severe and chronic morbidity and occasional mortality. The development of countermeasures, especially against CHIKV and Venezuelan equine encephalitis virus that are major threats, has included vaccines and antibody-based therapeutics that are likely to also be successful for rapid responses with other members of the family. However, further work with these prototypes and other alphavirus pathogens should target better understanding of human tropism and pathogenesis, more comprehensive identification of cellular receptors and entry, and better understanding of structural mechanisms of neutralization.


Subject(s)
Chikungunya virus , Culicidae , Animals , Horses , Humans , Research
2.
Arch Virol ; 165(7): 1715-1717, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32417973

ABSTRACT

Venezuelan equine encephalitis virus (VEEV) is an important pathogen of medical and veterinary importance in the Americas. In this report, we present the complete genome sequences of five VEEV isolates obtained from pools of Culex (Melanoconion) gnomatos (4) or Culex (Melanoconion) pedroi (1) from Iquitos, Peru. Genetic and phylogenetic analyses showed that all five isolates grouped within the VEEV complex sister to VEEV IIIC and are members of subtype IIID. This is the first report of full-length genomic sequences of VEEV IIID.


Subject(s)
Culex/virology , Encephalitis Virus, Venezuelan Equine/isolation & purification , Encephalomyelitis, Venezuelan Equine/virology , Genome, Viral , Mosquito Vectors/virology , Animals , Base Sequence , Encephalitis Virus, Venezuelan Equine/classification , Encephalitis Virus, Venezuelan Equine/genetics , Encephalomyelitis, Venezuelan Equine/transmission , Genomics , Horses , Peru , Phylogeny
3.
J Virol ; 92(4)2018 02 15.
Article in English | MEDLINE | ID: mdl-29187545

ABSTRACT

Most alphaviruses are mosquito borne and exhibit a broad host range, infecting many different vertebrates, including birds, rodents, equids, humans, and nonhuman primates. Recently, a host-restricted, mosquito-borne alphavirus, Eilat virus (EILV), was described with an inability to infect vertebrate cells based on defective attachment and/or entry, as well as a lack of genomic RNA replication. We investigated the utilization of EILV recombinant technology as a vaccine platform against eastern (EEEV) and Venezuelan equine encephalitis viruses (VEEV), two important pathogens of humans and domesticated animals. EILV chimeras containing structural proteins of EEEV or VEEV were engineered and successfully rescued in Aedes albopictus cells. Cryo-electron microscopy reconstructions at 8 and 11 Å of EILV/VEEV and EILV/EEEV, respectively, showed virion and glycoprotein spike structures similar to those of VEEV-TC83 and other alphaviruses. The chimeras were unable to replicate in vertebrate cell lines or in brains of newborn mice when injected intracranially. Histopathologic examinations of the brain tissues showed no evidence of pathological lesions and were indistinguishable from those of mock-infected animals. A single-dose immunization of either monovalent or multivalent EILV chimera(s) generated neutralizing antibody responses and protected animals against lethal challenge 70 days later. Lastly, a single dose of monovalent EILV chimeras generated protective responses as early as day 1 postvaccination and partial or complete protection by day 6. These data demonstrate the safety, immunogenicity, and efficacy of novel insect-specific EILV-based chimeras as potential EEEV and VEEV vaccines.IMPORTANCE Mostly in the last decade, insect-specific viruses have been discovered in several arbovirus families. However, most of these viruses are not well studied and largely have been ignored. We explored the use of the mosquito-specific alphavirus EILV as an alphavirus vaccine platform in well-established disease models for eastern (EEE) and Venezuelan equine encephalitis (VEE). EILV-based chimeras replicated to high titers in a mosquito cell line yet retained their host range restriction in vertebrates both in vitro and in vivo In addition, the chimeras generated immune responses that were higher than those of other human and/or equine vaccines. These findings indicate the feasibility of producing a safe, efficacious, mono- or multivalent vaccine against the encephalitic alphaviruses VEEV and EEEV. Lastly, these data demonstrate how host-restricted, insect-specific viruses can be engineered to develop vaccines against related pathogenic arboviruses that cause severe disease in humans and domesticated animals.


Subject(s)
Alphavirus Infections/immunology , Alphavirus/growth & development , Encephalitis Virus, Venezuelan Equine/immunology , Viral Vaccines/immunology , Alphavirus/immunology , Alphavirus/isolation & purification , Alphavirus Infections/virology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cryoelectron Microscopy , Encephalitis Virus, Venezuelan Equine/genetics , Genetic Engineering , HEK293 Cells , Host Specificity , Humans , Mice , Virus Replication
4.
J Virol ; 92(14)2018 07 15.
Article in English | MEDLINE | ID: mdl-29743363

ABSTRACT

Recent advances in mass spectrometry methods and instrumentation now allow for more accurate identification of proteins in low abundance. This technology was applied to Sindbis virus, the prototypical alphavirus, to investigate the viral proteome. To determine if host proteins are specifically packaged into alphavirus virions, Sindbis virus (SINV) was grown in multiple host cells representing vertebrate and mosquito hosts, and total protein content of purified virions was determined. This analysis identified host factors not previously associated with alphavirus entry, replication, or egress. One host protein, sorting nexin 5 (SNX5), was shown to be critical for the replication of three different alphaviruses, Sindbis, Mayaro, and Chikungunya viruses. The most significant finding was that in addition to the host proteins, SINV nonstructural protein 2 (nsP2) was detected within virions grown in all host cells examined. The protein and RNA-interacting capabilities of nsP2 coupled with its presence in the virion support a role for nsP2 during packaging and/or entry of progeny virus. This function has not been identified for this protein. Taken together, this strategy identified at least one host factor integrally involved in alphavirus replication. Identification of other host proteins provides insight into alphavirus-host interactions during viral replication in both vertebrate and invertebrate hosts. This method of virus proteome analysis may also be useful for the identification of protein candidates for host-based therapeutics.IMPORTANCE Pathogenic alphaviruses, such as Chikungunya and Mayaro viruses, continue to plague public health in developing and developed countries alike. Alphaviruses belong to a group of viruses vectored in nature by hematophagous (blood-feeding) insects and are termed arboviruses (arthropod-borne viruses). This group of viruses contains many human pathogens, such as dengue fever, West Nile, and Yellow fever viruses. With few exceptions, there are no vaccines or prophylactics for these agents, leaving one-third of the world population at risk of infection. Identifying effective antivirals has been a long-term goal for combating these diseases not only because of the lack of vaccines but also because they are effective during an ongoing epidemic. Mass spectrometry-based analysis of the Sindbis virus proteome can be effective in identifying host genes involved in virus replication and novel functions for virus proteins. Identification of these factors is invaluable for the prophylaxis of this group of viruses.


Subject(s)
Alphavirus Infections/metabolism , Culicidae/metabolism , Cysteine Endopeptidases/metabolism , Proteome/metabolism , Sindbis Virus/physiology , Sorting Nexins/metabolism , Virion , Alphavirus Infections/virology , Amino Acid Sequence , Animals , Cricetinae , Culicidae/virology , HEK293 Cells , Humans , Sequence Homology , Virus Replication
5.
J Gen Virol ; 99(6): 761-762, 2018 06.
Article in English | MEDLINE | ID: mdl-29745869

ABSTRACT

The Togaviridae is a family of small, enveloped viruses with single-stranded, positive-sense RNA genomes of 10-12 kb. Within the family, the genus Alphavirus includes a large number of diverse species, while the genus Rubivirus includes the single species Rubella virus. Most alphaviruses are mosquito-borne and are pathogenic in their vertebrate hosts. Many are important human and veterinary pathogens (e.g. chikungunya virus and eastern equine encephalitis virus). Rubella virus is transmitted by respiratory routes among humans. This is a summary of the International Committee on Taxonomy of Viruses (ICTV) Report on the taxonomy of the Togaviridae, which is available at www.ictv.global/report/togaviridae.


Subject(s)
Togaviridae/classification , Togaviridae/genetics , Animals , Chikungunya virus/genetics , Genome, Viral , Humans , Rubella virus/genetics , Togaviridae/pathogenicity
6.
J Virol ; 91(21)2017 11 01.
Article in English | MEDLINE | ID: mdl-28794043

ABSTRACT

There is an urgent need for therapeutic development to combat infections caused by Rift Valley fever virus (RVFV), which causes devastating disease in both humans and animals. In an effort to repurpose drugs for RVFV treatment, our previous studies screened a library of FDA-approved drugs. The most promising candidate identified was the hepatocellular and renal cell carcinoma drug sorafenib. Mechanism-of-action studies indicated that sorafenib targeted a late stage in virus infection and caused a buildup of virions within cells. In addition, small interfering RNA (siRNA) knockdown studies suggested that nonclassical targets of sorafenib are important for the propagation of RVFV. Here we extend our previous findings to identify the mechanism by which sorafenib inhibits the release of RVFV virions from the cell. Confocal microscopy imaging revealed that glycoprotein Gn colocalizes and accumulates within the endoplasmic reticulum (ER) and the transport of Gn from the Golgi complex to the host cell membrane is reduced. Transmission electron microscopy demonstrated that sorafenib caused virions to be present inside large vacuoles inside the cells. p97/valosin-containing protein (VCP), which is involved in membrane remodeling in the secretory pathway and a known target of sorafenib, was found to be important for RVFV egress. Knockdown of VCP resulted in decreased RVFV replication, reduced Gn Golgi complex localization, and increased Gn ER accumulation. The intracellular accumulation of RVFV virions was also observed in cells transfected with siRNA targeting VCP. Collectively, these data indicate that sorafenib causes a disruption in viral egress by targeting VCP and the secretory pathway, resulting in a buildup of virions within dilated ER vesicles.IMPORTANCE In humans, symptoms of RVFV infection mainly include a self-limiting febrile illness. However, in some cases, infected individuals can also experience hemorrhagic fever, neurological disorders, liver failure, and blindness, which could collectively be lethal. The ability of RVFV to expand geographically outside sub-Saharan Africa is of concern, particularly to the Americas, where native mosquito species are capable of virus transmission. Currently, there are no FDA-approved therapeutics to treat RVFV infection, and thus, there is an urgent need to understand the mechanisms by which the virus hijacks the host cell machinery to replicate. The significance of our research is in identifying the cellular target of sorafenib that inhibits RVFV propagation, so that this information can be used as a tool for the further development of therapeutics used to treat RVFV infection.


Subject(s)
Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/metabolism , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Rift Valley Fever/drug therapy , Rift Valley fever virus/physiology , Secretory Pathway/drug effects , Virus Release/drug effects , Adenosine Triphosphatases/genetics , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/virology , Cell Cycle Proteins/genetics , Chlorocebus aethiops , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/virology , Niacinamide/pharmacology , Rift Valley Fever/metabolism , Rift Valley Fever/virology , Rift Valley fever virus/drug effects , Sorafenib , Tumor Cells, Cultured , Valosin Containing Protein , Vero Cells , Virion/drug effects , Virus Replication/drug effects
7.
J Virol ; 91(8)2017 04 15.
Article in English | MEDLINE | ID: mdl-28148802

ABSTRACT

The demonstrated clinical efficacy of a recombinant vesicular stomatitis virus (rVSV) vaccine vector has stimulated the investigation of additional serologically distinct Vesiculovirus vectors as therapeutic and/or prophylactic vaccine vectors to combat emerging viral diseases. Among these viral threats are the encephalitic alphaviruses Venezuelan equine encephalitis virus (VEEV) and Eastern equine encephalitis virus (EEEV), which have demonstrated potential for natural disease outbreaks, yet no licensed vaccines are available in the event of an epidemic. Here we report the rescue of recombinant Isfahan virus (rISFV) from genomic cDNA as a potential new vaccine vector platform. The rISFV genome was modified to attenuate virulence and express the VEEV and EEEV E2/E1 surface glycoproteins as vaccine antigens. A single dose of the rISFV vaccine vectors elicited neutralizing antibody responses and protected mice from lethal VEEV and EEEV challenges at 1 month postvaccination as well as lethal VEEV challenge at 8 months postvaccination. A mixture of rISFV vectors expressing the VEEV and EEEV E2/E1 glycoproteins also provided durable, single-dose protection from lethal VEEV and EEEV challenges, demonstrating the potential for a multivalent vaccine formulation. These findings were paralleled in studies with an attenuated form of rVSV expressing the VEEV E2/E1 glycoproteins. Both the rVSV and rISFV vectors were attenuated by using an approach that has demonstrated safety in human trials of an rVSV/HIV-1 vaccine. Vaccines based on either of these vaccine vector platforms may present a safe and effective approach to prevent alphavirus-induced disease in humans.IMPORTANCE This work introduces rISFV as a novel vaccine vector platform that is serologically distinct and phylogenetically distant from VSV. The rISFV vector has been attenuated by an approach used for an rVSV vector that has demonstrated safety in clinical studies. The vaccine potential of the rISFV vector was investigated in a well-established alphavirus disease model. The findings indicate the feasibility of producing a safe, efficacious, multivalent vaccine against the encephalitic alphaviruses VEEV and EEEV, both of which can cause fatal disease. This work also demonstrates the efficacy of an attenuated rVSV vector that has already demonstrated safety and immunogenicity in multiple HIV-1 phase I clinical studies. The absence of serological cross-reactivity between rVSV and rISFV and their phylogenetic divergence within the Vesiculovirus genus indicate potential for two stand-alone vaccine vector platforms that could be used to target multiple bacterial and/or viral agents in successive immunization campaigns or as heterologous prime-boost agents.


Subject(s)
Drug Carriers , Encephalitis Virus, Eastern Equine/immunology , Encephalitis Virus, Venezuelan Equine/immunology , Encephalomyelitis, Equine/prevention & control , Vesiculovirus/genetics , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Disease Models, Animal , Encephalitis Virus, Eastern Equine/genetics , Encephalitis Virus, Venezuelan Equine/genetics , Glycoproteins/genetics , Glycoproteins/immunology , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Survival Analysis , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Vaccines/genetics
8.
Adv Exp Med Biol ; 1062: 303-318, 2018.
Article in English | MEDLINE | ID: mdl-29845541

ABSTRACT

The United States Army Medical Research Institute of Infectious Diseases (USAMRIID) possesses an array of expertise in diverse capabilities for the characterization of emerging infectious diseases from the pathogen itself to human or animal infection models. The recent Zika virus (ZIKV) outbreak was a challenge and an opportunity to put these capabilities to work as a cohesive unit to quickly respond to a rapidly developing threat. Next-generation sequencing was used to characterize virus stocks and to understand the introduction and spread of ZIKV in the United States. High Content Imaging was used to establish a High Content Screening process to evaluate antiviral therapies. Functional genomics was used to identify critical host factors for ZIKV infection. An animal model using the temporal blockade of IFN-I in immunocompetent laboratory mice was investigated in conjunction with Positron Emission Tomography to study ZIKV. Correlative light and electron microscopy was used to examine ZIKV interaction with host cells in culture and infected animals. A quantitative mass spectrometry approach was used to examine the protein and metabolite type or concentration changes that occur during ZIKV infection in blood, cells, and tissues. Multiplex fluorescence in situ hybridization was used to confirm ZIKV replication in mouse and NHP tissues. The integrated rapid response approach developed at USAMRIID presented in this review was successfully applied and provides a new template pathway to follow if a new biological threat emerges. This streamlined approach will increase the likelihood that novel medical countermeasures could be rapidly developed, evaluated, and translated into the clinic.


Subject(s)
Academies and Institutes , Zika Virus Infection/virology , Zika Virus/physiology , Academies and Institutes/trends , Animals , Biomedical Research , Humans , Zika Virus/genetics
9.
Emerg Infect Dis ; 23(8): 1274-1281, 2017 08.
Article in English | MEDLINE | ID: mdl-28548637

ABSTRACT

Unprotected sexual intercourse between persons residing in or traveling from regions with Zika virus transmission is a risk factor for infection. To model risk for infection after sexual intercourse, we inoculated rhesus and cynomolgus macaques with Zika virus by intravaginal or intrarectal routes. In macaques inoculated intravaginally, we detected viremia and virus RNA in 50% of macaques, followed by seroconversion. In macaques inoculated intrarectally, we detected viremia, virus RNA, or both, in 100% of both species, followed by seroconversion. The magnitude and duration of infectious virus in the blood of macaques suggest humans infected with Zika virus through sexual transmission will likely generate viremias sufficient to infect competent mosquito vectors. Our results indicate that transmission of Zika virus by sexual intercourse might serve as a virus maintenance mechanism in the absence of mosquito-to-human transmission and could increase the probability of establishment and spread of Zika virus in regions where this virus is not present.


Subject(s)
Macaca fascicularis , Macaca mulatta , Zika Virus Infection/virology , Zika Virus/physiology , Animals , Female , Male , Vagina , Virus Replication , Virus Shedding , Zika Virus Infection/transmission
10.
J Virol ; 89(2): 1404-18, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25392227

ABSTRACT

UNLABELLED: Most alphaviruses are mosquito-borne and exhibit a broad host range, infecting many different vertebrates, including birds, rodents, equids, humans, and nonhuman primates. This ability of most alphaviruses to infect arthropods and vertebrates is essential for their maintenance in nature. Recently, a new alphavirus, Eilat virus (EILV), was described, and in contrast to all other mosquito-borne viruses, it is unable to replicate in vertebrate cell lines. Investigations into the nature of its host range restriction showed the inability of genomic EILV RNA to replicate in vertebrate cells. Here, we investigated whether the EILV host range restriction is present at the entry level and further explored the viral factors responsible for the lack of genomic RNA replication. Utilizing Sindbis virus (SINV) and EILV chimeras, we show that the EILV vertebrate host range restriction is also manifested at the entry level. Furthermore, the EILV RNA replication restriction is independent of the 3' untranslated genome region (UTR). Complementation experiments with SINV suggested that RNA replication is restricted by the inability of the EILV nonstructural proteins to form functional replicative complexes. These data demonstrate that the EILV host range restriction is multigenic, involving at least one gene from both nonstructural protein (nsP) and structural protein (sP) open reading frames (ORFs). As EILV groups phylogenetically within the mosquito-borne virus clade of pathogenic alphaviruses, our findings have important evolutionary implications for arboviruses. IMPORTANCE: Our work explores the nature of host range restriction of the first "mosquito-only alphavirus," EILV. EILV is related to pathogenic mosquito-borne viruses (Eastern equine encephalitis virus [EEEV], Western equine encephalitis virus [WEEV], Venezuelan equine encephalitis virus [VEEV], and Chikungunya virus [CHIKV]) that cause severe disease in humans. Our data demonstrate that EILV is restricted both at entry and genomic RNA replication levels in vertebrate cells. These findings have important implications for arbovirus evolution and will help elucidate the viral factors responsible for the broad host range of pathogenic mosquito-borne alphaviruses, facilitate vaccine development, and inform potential strategies to reduce/prevent alphavirus transmission.


Subject(s)
Alphavirus/immunology , Alphavirus/physiology , Host Specificity , Virus Internalization , Virus Replication , Alphavirus/genetics , Animals , Culicidae , Genetic Complementation Test , Sindbis Virus/genetics , Vertebrates
11.
J Virol ; 88(12): 6690-701, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24696472

ABSTRACT

UNLABELLED: In previous work, a prototypic recombinant vesicular stomatitis virus Indiana serotype (rVSIV) vector expressing simian immunodeficiency virus (SIV) gag and human immunodeficiency virus type 1 (HIV-1) env antigens protected nonhuman primates (NHPs) from disease following challenge with an HIV-1/SIV recombinant (SHIV). However, when tested in a stringent NHP neurovirulence (NV) model, this vector was not adequately attenuated for clinical evaluation. For the work described here, the prototypic rVSIV vector was attenuated by combining specific G protein truncations with either N gene translocations or mutations (M33A and M51A) that ablate expression of subgenic M polypeptides, by incorporation of temperature-sensitive mutations in the N and L genes, and by deletion of the VSIV G gene to generate a replicon that is dependent on trans expression of G protein for in vitro propagation. When evaluated in a series of NHP NV studies, these attenuated rVSIV variants caused no clinical disease and demonstrated a very significant reduction in neuropathology compared to wild-type VSIV and the prototypic rVSIV vaccine vector. In spite of greatly increased in vivo attenuation, some of the rVSIV vectors elicited cell-mediated immune responses that were similar in magnitude to those induced by the much more virulent prototypic vector. These data demonstrate novel approaches to the rational attenuation of VSIV NV while retaining vector immunogenicity and have led to identification of an rVSIV N4CT1gag1 vaccine vector that has now successfully completed phase I clinical evaluation. IMPORTANCE: The work described in this article demonstrates a rational approach to the attenuation of vesicular stomatitis virus neurovirulence. The major attenuation strategy described here will be most likely applicable to other members of the Rhabdoviridae and possibly other families of nonsegmented negative-strand RNA viruses. These studies have also enabled the identification of an attenuated, replication-competent rVSIV vector that has successfully undergone its first clinical evaluation in humans. Therefore, these studies represent a major milestone in the development of attenuated rVSIV, and likely other vesiculoviruses, as a new vaccine platform(s) for use in humans.


Subject(s)
AIDS Vaccines/immunology , Central Nervous System/virology , Genetic Vectors/immunology , HIV Infections/immunology , HIV-1/immunology , Macaca fascicularis , Vesicular stomatitis Indiana virus/immunology , AIDS Vaccines/administration & dosage , AIDS Vaccines/genetics , Animals , Antibodies, Viral/immunology , Central Nervous System/immunology , Disease Models, Animal , Genetic Vectors/genetics , HIV Infections/prevention & control , HIV Infections/virology , HIV-1/genetics , Humans , Macaca fascicularis/genetics , Macaca fascicularis/immunology , Macaca fascicularis/virology , Male , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vesicular stomatitis Indiana virus/genetics , gag Gene Products, Human Immunodeficiency Virus/administration & dosage , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/immunology
12.
Proc Natl Acad Sci U S A ; 109(36): 14622-7, 2012 Sep 04.
Article in English | MEDLINE | ID: mdl-22908261

ABSTRACT

Most alphaviruses and many other arboviruses are mosquito-borne and exhibit a broad host range, infecting many different vertebrates including birds, rodents, equids, humans, and nonhuman primates. Consequently, they can be propagated in most vertebrate and insect cell cultures. This ability of arboviruses to infect arthropods and vertebrates is usually essential for their maintenance in nature. However, several flaviviruses have recently been described that infect mosquitoes but not vertebrates, although the mechanism of their host restriction has not been determined. Here we describe a unique alphavirus, Eilat virus (EILV), isolated from a pool of Anopheles coustani mosquitoes from the Negev desert of Israel. Phylogenetic analyses placed EILV as a sister to the Western equine encephalitis antigenic complex within the main clade of mosquito-borne alphaviruses. Electron microscopy revealed that, like other alphaviruses, EILV virions were spherical, 70 nm in diameter, and budded from the plasma membrane of mosquito cells in culture. EILV readily infected a variety of insect cells with little overt cytopathic effect. However, in contrast to typical mosquito-borne alphaviruses, EILV could not infect mammalian or avian cell lines, and viral as well as RNA replication could not be detected at 37 °C or 28 °C. Evolutionarily, these findings suggest that EILV lost its ability to infect vertebrate cells. Thus, EILV seems to be mosquito-specific and represents a previously undescribed complex within the genus Alphavirus. Reverse genetic studies of EILV may facilitate the discovery of determinants of alphavirus host range that mediate disease emergence.


Subject(s)
Alphavirus/genetics , Alphavirus/physiology , Anopheles/virology , Biological Evolution , Host-Pathogen Interactions/physiology , Phylogeny , Virus Replication/physiology , Alphavirus/ultrastructure , Animals , Base Sequence , Bayes Theorem , Cloning, Molecular , Cluster Analysis , Electrophoresis, Agar Gel , Israel , Likelihood Functions , Microscopy, Electron, Transmission , Models, Genetic , Molecular Sequence Data , RNA, Viral/genetics , Sequence Analysis, DNA , Species Specificity
13.
J Gen Virol ; 95(Pt 9): 2071-2074, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24878640

ABSTRACT

The genus Negevirus consists of insect-only viruses isolated from mosquitoes and sandflies. Here, we report the successful construction of a full-length infectious cDNA clone of Negev virus (NEGV) strain M30957. Viral RNA was transcribed in vitro and virus was readily rescued with or without the use of a cap analogue. These results strongly suggest that NEGV, and likely other members within the genus, is a non-segmented, single-stranded, positive-sense RNA virus.


Subject(s)
DNA, Complementary/genetics , Insect Viruses/genetics , Insecta/virology , RNA Viruses/classification , RNA Viruses/genetics , Animals , Cells, Cultured , Cloning, Molecular , Genome, Viral , RNA, Viral/genetics
14.
J Virol ; 87(5): 2475-88, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23255793

ABSTRACT

Six novel insect-specific viruses, isolated from mosquitoes and phlebotomine sand flies collected in Brazil, Peru, the United States, Ivory Coast, Israel, and Indonesia, are described. Their genomes consist of single-stranded, positive-sense RNAs with poly(A) tails. By electron microscopy, the virions appear as spherical particles with diameters of ∼45 to 55 nm. Based on their genome organization and phylogenetic relationship, the six viruses, designated Negev, Ngewotan, Piura, Loreto, Dezidougou, and Santana, appear to form a new taxon, tentatively designated Negevirus. Their closest but still distant relatives are citrus leposis virus C (CiLV-C) and viruses in the genus Cilevirus, which are mite-transmitted plant viruses. The negeviruses replicate rapidly and to high titer (up to 10(10) PFU/ml) in mosquito cells, producing extensive cytopathic effect and plaques, but they do not appear to replicate in mammalian cells or mice. A discussion follows on their possible biological significance and effect on mosquito vector competence for arboviruses.


Subject(s)
Anopheles/virology , Culex/virology , Insect Viruses/classification , Phlebotomus/virology , RNA Viruses/classification , Animals , Base Sequence , Cell Line , Chlorocebus aethiops/virology , Cricetinae , Drosophila melanogaster/virology , Insect Viruses/genetics , Insect Viruses/isolation & purification , Phylogeny , RNA Viruses/genetics , RNA Viruses/isolation & purification , RNA, Viral , Sequence Analysis, RNA , Vero Cells , Virus Replication
15.
Am J Trop Med Hyg ; 110(6): 1178-1179, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38653230

ABSTRACT

We report strong Zika virus (ZIKV) neutralizing antibody responses in African green monkeys (Chlorocebus sabaeus) up to 1,427 days after ZIKV exposure via the subcutaneous, intravaginal, or intrarectal routes. Our results suggest that immunocompetent African green monkeys previously infected with ZIKV are likely protected from reinfection for years, possibly life, and would not contribute to virus amplification during ZIKV epizootics.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Zika Virus Infection , Zika Virus , Animals , Chlorocebus aethiops , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Zika Virus/immunology , Zika Virus Infection/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Female
16.
Am J Trop Med Hyg ; 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38917780

ABSTRACT

Mayaro virus (MAYV) is an alphavirus endemic in many parts of Central and South America transmitted to humans by Aedes aegypti. Currently, there is no vaccine or treatment of Mayaro infection, and therefore it is essential to control transmission by reducing populations of Ae. aegypti. Unfortunately, Ae. aegypti are extremely difficult to control with traditional integrated vector management (IVM) because of factors such as growing resistance to a dwindling list of registered insecticides and cryptic immature and adult habitats. The sterile insect technique (SIT) by irradiation is gaining traction as a novel supplemental tool to IVM. The SIT is being used operationally to release large numbers of sterilized colony-reared male mosquitoes in an intervention area to overwhelm females in the natural population, eventually causing population decline because of high frequencies of unfertilized eggs. However, little is known about the effect of irradiation on vector competence for mosquito-borne viruses such as MAYV in females that may be accidentally reared, irradiated, and released alongside males. In this investigation, we exposed female Ae. aegypti pupae to radiation and evaluated vector competence after inoculation with MAYV. Infection and dissemination rates of irradiated (10 and 40 Gy) Ae. aegypti were higher than those of non-irradiated cohorts at 7 and 14 days after infection. Although these results indicate a need to maintain effective sex sorting prior to irradiation and release of Ae. aegypti, our results are consistent with several previous observations that vectorial capacity and vector competence are likely lower in irradiated than in nonirradiated females.

17.
Viruses ; 15(10)2023 10 21.
Article in English | MEDLINE | ID: mdl-37896908

ABSTRACT

TNX-1800 is a preclinical stage synthetic-derived live attenuated chimeric horsepox virus vaccine engineered to express the SARS-CoV-2 spike (S) gene. The objectives of this study were to assess the safety, tolerability, and immunogenicity of TNX-1800 administration in Syrian golden hamsters and New Zealand white rabbits. Animals were vaccinated at three doses via percutaneous inoculation. The data showed that the single percutaneous administration of three TNX-1800 vaccine dose levels was well tolerated in both hamsters and rabbits. At all dose levels, rabbits were more decerning regarding vaccine site reaction than hamsters. Lastly, no TNX-1800 genomes could be detected at the site of vaccination. Post-vaccination, all animals had anti-SARS-CoV-2 spike protein IgG specific antibody responses. These data demonstrate that TNX-1800 infection was limited, asymptomatic, and cleared by the end of this study, and a single dose was able to generate immune responses.


Subject(s)
COVID-19 , Poxviridae , Cricetinae , Rabbits , Animals , Mesocricetus , SARS-CoV-2/genetics , Vaccines, Attenuated/adverse effects , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/genetics , Antibodies, Viral , Immunoglobulin G , Spike Glycoprotein, Coronavirus/genetics , Antibodies, Neutralizing
18.
Antiviral Res ; 210: 105513, 2023 02.
Article in English | MEDLINE | ID: mdl-36592670

ABSTRACT

Antibody-based therapy is emerging as a critical therapeutic countermeasure to treat acute viral infections by offering rapid protection against clinical disease. The advancements in structural biology made it feasible to rationalize monoclonal antibodies (mAbs) by identifying key and, possibly, neutralizing epitopes of viral proteins for therapeutic purposes. A critical component in assessing mAbs during pandemics requires the development of rapid but detailed methods to detect and quantitate the neutralization activity. In this study, we developed and optimized two high-content image (HCI)-based assays: one to detect viral proteins by staining and the second to quantify cytopathic viral effects by a label-free phenotypic assay. These assays were employed to screen for therapeutic antibodies against the monkeypox virus (MPXV) using surrogate poxviruses such as vaccinia virus (VACV). Plaque-based neutralization results confirmed the HCI data. The phenotypic assay found pox virus-induced syncytia formation in various cells, and we were able to quantitate and use this phenotype to screen mAbs. The HCI identified several potent VACV-neutralizing antibodies that showed in vitro efficacy against both clades of MPXV. In addition, a combination study of ST-246/tecovirimat/TPOXX a single neutralizing antibody Ab-40, showed synergistic activity against VACV in an in-vitro neutralization assay. This rapid high-content method utilizing state-of-the-art technologies enabled the evaluation of hundreds of mAbs quickly to identify several potent anti-MPXV neutralizing mAbs for further development.


Subject(s)
Antibodies, Viral , Monkeypox virus , Antibodies, Neutralizing , Vaccinia virus/genetics , Viral Proteins , Antibodies, Monoclonal/pharmacology , Neutralization Tests
19.
Vaccines (Basel) ; 11(11)2023 Nov 02.
Article in English | MEDLINE | ID: mdl-38006014

ABSTRACT

TNX-1800 is a synthetically derived live recombinant chimeric horsepox virus (rcHPXV) vaccine candidate expressing Wuhan SARS-CoV-2 spike (S) protein. The primary objective of this study was to evaluate the immunogenicity and efficacy of TNX-1800 in two nonhuman primate species challenged with USA-WA1/2020 SARS-CoV-2. TNX-1800 vaccination was well tolerated with no serious adverse events or significant changes in clinical parameters. A single dose of TNX-1800 generated humoral responses in African Green Monkeys and Cynomolgus Macaques, as measured by the total binding of anti-SARS-CoV-2 S IgG and neutralizing antibody titers against the USA-WA1/2020 strain. In addition, a single dose of TNX-1800 induced an interferon-gamma (IFN-γ)-mediated T-cell response in Cynomolgus Macaques. Following challenge with SARS-CoV-2, African Green and Cynomolgus Macaques exhibited rapid clearance of virus in the upper and lower respiratory tract. Future studies will assess the efficacy of TNX-1800 against newly emerging variants and demonstrate its safety in humans.

20.
PLoS Negl Trop Dis ; 16(5): e0010081, 2022 05.
Article in English | MEDLINE | ID: mdl-35533188

ABSTRACT

Eastern equine encephalitis virus (EEEV) is mosquito-borne virus that produces fatal encephalitis in humans. We recently conducted a first of its kind study to investigate EEEV clinical disease course following aerosol challenge in a cynomolgus macaque model utilizing the state-of-the-art telemetry to measure critical physiological parameters. Here, we report the results of a comprehensive pathology study of NHP tissues collected at euthanasia to gain insights into EEEV pathogenesis. Viral RNA and proteins as well as microscopic lesions were absent in the visceral organs. In contrast, viral RNA and proteins were readily detected throughout the brain including autonomic nervous system (ANS) control centers and spinal cord. However, despite presence of viral RNA and proteins, majority of the brain and spinal cord tissues exhibited minimal or no microscopic lesions. The virus tropism was restricted primarily to neurons, and virus particles (~61-68 nm) were present within axons of neurons and throughout the extracellular spaces. However, active virus replication was absent or minimal in majority of the brain and was limited to regions proximal to the olfactory tract. These data suggest that EEEV initially replicates in/near the olfactory bulb following aerosol challenge and is rapidly transported to distal regions of the brain by exploiting the neuronal axonal transport system to facilitate neuron-to-neuron spread. Once within the brain, the virus gains access to the ANS control centers likely leading to disruption and/or dysregulation of critical physiological parameters to produce severe disease. Moreover, the absence of microscopic lesions strongly suggests that the underlying mechanism of EEEV pathogenesis is due to neuronal dysfunction rather than neuronal death. This study is the first comprehensive investigation into EEEV pathology in a NHP model and will provide significant insights into the evaluation of countermeasure.


Subject(s)
Encephalitis Virus, Eastern Equine , Encephalomyelitis, Equine , Aerosols , Animals , Brain , Disease Models, Animal , Encephalomyelitis, Equine/pathology , Horses , Macaca fascicularis , RNA, Viral , Spinal Cord/pathology
SELECTION OF CITATIONS
SEARCH DETAIL