Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 20(2): e1012047, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38412195

ABSTRACT

Variability in how individuals respond to pathogens is a hallmark of infectious disease, yet the basis for individual variation in host response is often poorly understood. The titer of infectious virus among individual mosquitoes infected with arboviruses is frequently observed to vary by several orders of magnitude in a single experiment, even when the mosquitoes are highly inbred. To better understand the basis for this titer variation, we sequenced populations of Sindbis virus (SINV) obtained from individual infected Aedes aegypti mosquitoes that, despite being from a highly inbred laboratory colony, differed in their titers of infectious virus by approximately 10,000-fold. We observed genetic differences between these virus populations that indicated the virus present in the midguts of low titer mosquitoes was less fit than that of high titer mosquitoes, possibly due to founder effects that occurred during midgut infection. Furthermore, we found dramatic differences in the specific infectivity or SI (the ratio of infectious units/viral genome equivalents) between these virus populations, with the SI of low titer mosquitoes being up to 10,000-fold lower than that of high titer mosquitoes. Despite having similar amounts of viral genomes, low titer mosquitoes appeared to contain less viral particles, suggesting that viral genomes were packaged into virions less efficiently than in high titer mosquitoes. Finally, antibiotic treatment, which has been shown to suppress mosquito antiviral immunity, caused an increase in SI. Our results indicate that the extreme variation that is observed in SINV infectious titer between individual Ae. aegypti mosquitoes is due to both genetic differences between virus populations and to differences in the proportion of genomes that are packaged into infectious particles.


Subject(s)
Aedes , Alphavirus Infections , Humans , Animals , Sindbis Virus/genetics , Base Sequence , Mosquito Vectors
2.
Pathogens ; 12(2)2023 Jan 31.
Article in English | MEDLINE | ID: mdl-36839492

ABSTRACT

Arboviral diseases spread by mosquitoes cause significant morbidity and mortality throughout much of the world. The treatment and prevention of these diseases through medication and vaccination is often limited, which makes controlling arboviruses at the level of the vector ideal. One way to prevent the spread of an arbovirus would be to stop its vector from developing a disseminated infection, which is required for the virus to make its way to the saliva of the mosquito to be potentially transmitted to a new host. The midgut of the mosquito provides one such opportunity to stop an arbovirus in its tracks. It has been known for many years that in certain arbovirus-vector combinations, or under certain circumstances, an arbovirus can infect and replicate in the midgut but is unable to escape from the tissue to cause disseminated infection. This situation is known as a midgut escape barrier. If we better understand why this barrier occurs, it might aid in the development of more informed control strategies. In this review, we discuss how the midgut escape barrier contributes to virus-vector specificity and possible mechanisms that may allow this barrier to be overcome in successful virus-vector combinations. We also discuss several of the known factors that either increase or decrease the likelihood of midgut escape.

3.
Viruses ; 14(9)2022 09 13.
Article in English | MEDLINE | ID: mdl-36146841

ABSTRACT

Arboviruses continue to threaten a significant portion of the human population, and a better understanding is needed of the determinants of successful arbovirus infection of arthropod vectors. Avoiding apoptosis has been shown to be one such determinant. Previous work showed that a Sindbis virus (SINV) construct called MRE/rpr that expresses the Drosophila pro-apoptotic protein Reaper via a duplicated subgenomic promoter had a reduced ability to orally infect Aedes aegypti mosquitoes at 3 days post-blood meal (PBM), but this difference diminished over time as virus variants containing deletions in the inserted reaper gene rapidly predominated. In order to further clarify the effect of midgut apoptosis on disseminated infection in Ae. aegypti, we constructed MRE/rprORF, a version of SINV containing reaper inserted into the structural open reading frame (ORF) as an in-frame fusion. MRE/rprORF successfully expressed Reaper, replicated similarly to MRE/rpr in cell lines, induced apoptosis in cultured cells, and caused increased effector caspase activity in mosquito midgut tissue. Mosquitoes that fed on blood containing MRE/rprORF developed significantly less midgut and disseminated infection when compared to MRE/rpr or a control virus up to at least 7 days PBM, when less than 50% of mosquitoes that ingested MRE/rprORF had detectable disseminated infection, compared with around 80% or more of mosquitoes fed with MRE/rpr or control virus. However, virus titer in the minority of mosquitoes that became infected with MRE/rprORF was not significantly different from control virus. Deep sequencing of virus populations from ten mosquitoes infected with MRE/rprORF indicated that the reaper insert was stable, with only a small number of point mutations and no deletions being observed at frequencies greater than 1%. Our results indicate that expression of Reaper by this method significantly reduces infection prevalence, but if infection is established then Reaper expression has limited ability to continue to suppress replication.


Subject(s)
Aedes , Sindbis Virus , Animals , Apoptosis Regulatory Proteins/genetics , Caspases, Effector/genetics , Humans , Mosquito Vectors , Open Reading Frames , Sindbis Virus/genetics
4.
Viruses ; 14(1)2022 01 12.
Article in English | MEDLINE | ID: mdl-35062335

ABSTRACT

The mechanisms involved in determining arbovirus vector competence, or the ability of an arbovirus to infect and be transmitted by an arthropod vector, are still incompletely understood. It is well known that vector competence for a particular arbovirus can vary widely among different populations of a mosquito species, which is generally attributed to genetic differences between populations. What is less understood is the considerable variability (up to several logs) that is routinely observed in the virus titer between individual mosquitoes in a single experiment, even in mosquitoes from highly inbred lines. This extreme degree of variation in the virus titer between individual mosquitoes has been largely ignored in past studies. We investigated which biological factors can affect titer variation between individual mosquitoes of a laboratory strain of Aedes aegypti, the Orlando strain, after Sindbis virus infection. Greater titer variation was observed after oral versus intrathoracic infection, suggesting that the midgut barrier contributes to titer variability. Among the other factors tested, only the length of the incubation period affected the degree of titer variability, while virus strain, mosquito strain, mosquito age, mosquito weight, amount of blood ingested, and virus concentration in the blood meal had no discernible effect. We also observed differences in culture adaptability and in the ability to orally infect mosquitoes between virus populations obtained from low and high titer mosquitoes, suggesting that founder effects may affect the virus titer in individual mosquitoes, although other explanations also remain possible.


Subject(s)
Aedes/virology , Biological Factors , Saliva/virology , Viral Load , Animals , Arboviruses , Cell Line , Communicable Diseases/epidemiology , Culicidae , Disease Vectors , Female , Mosquito Vectors/virology , Prevalence , Sindbis Virus , Virus Replication
5.
J Virol ; 95(13): e0013621, 2021 06 10.
Article in English | MEDLINE | ID: mdl-33853958

ABSTRACT

Arboviruses are transmitted by specific vectors, and the reasons for this specificity are not fully understood. One contributing factor is the existence of tissue barriers within the vector such as the midgut escape barrier. We used microRNA (miRNA) targeting of Sindbis virus (SINV) to study how replication in midgut cells contributes to overcoming this barrier in the mosquito Aedes aegypti. SINV constructs were designed to be attenuated specifically in midgut cells by inserting binding sites for midgut-specific miRNAs into either the 3' untranslated region (MRE3'miRT) or the structural open reading frame (MRE-ORFmiRT) of the SINV genome. Both miRNA-targeted viruses replicated less efficiently than control viruses in the presence of these miRNAs. When mosquitoes were given infectious blood meals containing miRNA-targeted viruses, only around 20% (MRE3'miRT) or 40% (MRE-ORFmiRT) of mosquitoes developed disseminated infection. In contrast, dissemination occurred in almost all mosquitoes fed control viruses. Deep sequencing of virus populations from individual mosquitoes ruled out selection for mutations in the inserted target sequences as the cause for dissemination in these mosquitoes. In mosquitoes that became infected with miRNA-targeted viruses, titers were equivalent to those of mosquitoes infected with control virus in both the midgut and the carcass, and there was no evidence of a threshold titer necessary for dissemination. Instead, it appeared that if infection was successfully established in the midgut, replication and dissemination were largely normal. Our results support the hypothesis that replication is an important factor in allowing SINV to overcome the midgut escape barrier but hint that other factors are also likely involved. IMPORTANCE When a mosquito ingests an arbovirus during a blood meal, the arbovirus must escape from the midgut of the vector and infect the salivary glands in order to be transmitted to a new host. We used tissue-specific miRNA targeting to examine the requirement for Sindbis virus (SINV) to replicate in midgut epithelium in order to cause disseminated infection in the mosquito Aedes aegypti. Our results indicate that specifically reducing the ability of SINV to replicate in the mosquito midgut reduces its overall ability to establish infection in the mosquito, but if infection is established, replication and dissemination occur normally. These results are consistent with an importance for replication in the midgut epithelium in aiding arboviruses in crossing the midgut barrier.


Subject(s)
Aedes/virology , Gastrointestinal Tract/virology , MicroRNAs/genetics , Sindbis Virus/growth & development , Virus Replication/genetics , Animals , Cell Line , Cricetinae , Mosquito Vectors/virology , Organ Specificity , Salivary Glands/virology , Sindbis Virus/genetics , Sindbis Virus/metabolism
6.
Viruses ; 12(9)2020 08 26.
Article in English | MEDLINE | ID: mdl-32858937

ABSTRACT

Fibroblast growth factors (FGFs) are conserved among vertebrate and invertebrate animals and function in cell proliferation, cell differentiation, tissue repair, and embryonic development. A viral fibroblast growth factor (vFGF) homolog encoded by baculoviruses, a group of insect viruses, is involved in escape of baculoviruses from the insect midgut by stimulating basal lamina remodeling. This led us to investigate whether cellular FGF is involved in the escape of an arbovirus from mosquito midgut. In this study, the effects of manipulating FGF expression on Sindbis virus (SINV) replication and escape from the midgut of the mosquito vector Aedes aegypti were examined. RNAi-mediated silencing of either Ae. aegypti FGF (AeFGF) or FGF receptor (AeFGFR) expression reduced SINV replication following oral infection of Ae. aegypti mosquitoes. However, overexpression of baculovirus vFGF using recombinant SINV constructs had no effect on replication of these viruses in cultured mosquito or vertebrate cells, or in orally infected Ae. aegypti mosquitoes. We conclude that reducing FGF signaling decreases the ability of SINV to replicate in mosquitoes, but that overexpression of vFGF has no effect, possibly because endogenous FGF levels are already sufficient for optimal virus replication. These results support the hypothesis that FGF signaling, possibly by inducing remodeling of midgut basal lamina, is involved in arbovirus midgut escape following virus acquisition from a blood meal.


Subject(s)
Aedes/virology , Fibroblast Growth Factors/metabolism , Insect Proteins/metabolism , Mosquito Vectors/virology , Sindbis Virus/physiology , Animals , Caspases/metabolism , Cell Movement , Fibroblast Growth Factors/genetics , Gastrointestinal Tract/virology , Insect Proteins/genetics , RNA Interference , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction , Virus Replication
7.
Sci Rep ; 9(1): 14494, 2019 10 10.
Article in English | MEDLINE | ID: mdl-31601846

ABSTRACT

Prior studies have suggested that insect DNA viruses are negatively affected by dicer-2-mediated RNA interference (RNAi). To examine this further, we utilized an in vitro assay to measure dicer activity in lepidopteran and dipteran cells, combined with baculoviruses expressing the RNAi suppressor B2 from Flock House virus or Aedes aegypti dicer-2 (Aedicer-2) using a constitutive heat shock promoter. Addition of cell lysates containing baculovirus-expressed B2 to lysates from dipteran (S2, Aag2) or lepidopteran (Sf9) cells inhibited endogenous dicer activity in a dose-dependent manner, while expression of Aedicer-2 restored siRNA production in Ae. albopictus C6/36 cells, which are dicer-2 defective. However, B2 expression from the constitutive heat shock promoter had no impact on baculovirus replication or virulence in cell lines or larvae that were either highly permissive (Trichoplusia ni) or less susceptible (Spodoptera frugiperda) to infection. We determined that this constitutive level of B2 expression had little to no ability to suppress dicer activity in cell lysates, but higher expression of B2, following heat shock treatment, inhibited dicer activity in all cells tested. Thus, we cannot rule out the possibility that optimized expression of B2 or other RNAi suppressors may increase baculovirus replication and expression of heterologous proteins by baculoviruses.


Subject(s)
Baculoviridae/genetics , Nodaviridae/genetics , Ribonuclease III/genetics , Animals , Diptera/enzymology , Gene Expression Regulation, Viral/genetics , Insect Viruses/genetics , Lepidoptera/enzymology , RNA, Small Interfering
8.
Sci Rep ; 6: 28072, 2016 06 20.
Article in English | MEDLINE | ID: mdl-27321273

ABSTRACT

As baculoviruses usually have a narrow insecticidal spectrum, knowing the mechanisms by which they control the host-range is prerequisite for improvement of their applications as pesticides. In this study, from supernatant of culture cells transfected with DNAs of an Autographa californica multiple nucleopolyhedrovirus (AcMNPV) mutant lacking the antiapoptotic gene p35 (vAc(∆P35)) and a cosmid representing a fragment of Spodoptera exigua nucleopolyhedrovirus (SeMNPV), a viral strain was plaque-purified and named vAcRev. vAcRev had a broader host range than either vAc(∆P35) or SeMNPV parental virus, being able to infect not only the permissive hosts of its parental viruses but also a nonpermissive host (Spodoptera litura). Genome sequencing indicated that vAcRev comprises a mixture of two viruses with different circular dsDNA genomes. One virus contains a genome similar to vAc(∆P35), while in the other viral genome, a 24.4 kbp-fragment containing 10 essential genesis replaced with a 4 kbp-fragment containing three SeMNPV genes including a truncated Se-iap3 gene. RNA interference and ectopic expression assays found that Se-iap3 is responsible for the host range expansion of vAcRev, suggesting that Se-iap3 inhibits the progression of apoptosis initiated by viral infection and promotes viral propagation in hosts both permissive and non-permissive for AcMNPV and SeMNPV.


Subject(s)
Host Specificity/physiology , Nucleopolyhedroviruses/genetics , Spodoptera/virology , Animals , Cosmids/genetics , Cosmids/metabolism , DNA, Viral/chemistry , DNA, Viral/metabolism , Genome, Viral/genetics , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Lepidoptera/virology , Nucleopolyhedroviruses/growth & development , Nucleopolyhedroviruses/physiology , RNA Interference , Sequence Analysis, DNA , Sf9 Cells/cytology , Sf9 Cells/virology , Viral Proteins/antagonists & inhibitors , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication
9.
Sci Rep ; 6: 24612, 2016 06 07.
Article in English | MEDLINE | ID: mdl-27273152

ABSTRACT

The genome of a novel group II alphabaculovirus, Perigonia lusca single nucleopolyhedrovirus (PeluSNPV), was sequenced and shown to contain 132,831 bp with 145 putative ORFs (open reading frames) of at least 50 amino acids. An interesting feature of this novel genome was the presence of a putative nucleotide metabolism enzyme-encoding gene (pelu112). The pelu112 gene was predicted to encode a fusion of thymidylate kinase (tmk) and dUTP diphosphatase (dut). Phylogenetic analysis indicated that baculoviruses have independently acquired tmk and dut several times during their evolution. Two homologs of the tmk-dut fusion gene were separately introduced into the Autographa californica multiple nucleopolyhedrovirus (AcMNPV) genome, which lacks tmk and dut. The recombinant baculoviruses produced viral DNA, virus progeny, and some viral proteins earlier during in vitro infection and the yields of viral occlusion bodies were increased 2.5-fold when compared to the parental virus. Interestingly, both enzymes appear to retain their active sites, based on separate modeling using previously solved crystal structures. We suggest that the retention of these tmk-dut fusion genes by certain baculoviruses could be related to accelerating virus replication and to protecting the virus genome from deleterious mutation.


Subject(s)
Genome, Viral , Nucleopolyhedroviruses/genetics , Nucleoside-Phosphate Kinase/metabolism , Pyrophosphatases/metabolism , Viral Proteins/metabolism , Animals , Base Sequence , Binding Sites , DNA, Viral/chemistry , DNA, Viral/isolation & purification , DNA, Viral/metabolism , Genetic Vectors/genetics , Genetic Vectors/metabolism , Microscopy, Confocal , Microscopy, Electron, Scanning , Nucleopolyhedroviruses/classification , Nucleopolyhedroviruses/physiology , Nucleoside-Phosphate Kinase/chemistry , Nucleoside-Phosphate Kinase/genetics , Nucleotides/biosynthesis , Nucleotides/chemistry , Open Reading Frames/genetics , Phylogeny , Protein Structure, Tertiary , Pyrophosphatases/chemistry , Pyrophosphatases/genetics , Sequence Alignment , Sequence Analysis, DNA , Sf9 Cells , Spodoptera , Tandem Repeat Sequences/genetics , Viral Proteins/genetics , Virus Replication
10.
Sci Rep ; 6: 24729, 2016 04 22.
Article in English | MEDLINE | ID: mdl-27102548

ABSTRACT

Chikungunya virus (CHIKV) is an emerging mosquito-borne virus belonging to the Togaviridae, which is transmitted to humans by Aedes aegypti and Ae. albopictus. We describe the infection pattern of CHIKV in two New World Ae. aegypti strains, HWE and ORL. Both mosquito strains were susceptible to the virus but showed different infection patterns in midguts and salivary glands. Even though acquisition of a bloodmeal showed moderate levels of apoptosis in midgut tissue, there was no obvious additional CHIKV-induced apoptosis detectable during midgut infection. Analysis of expression of apoptosis-related genes suggested that CHIKV infection dampens rather than promotes apoptosis in the mosquito midgut. In both mosquito strains, the virus was present in saliva within two days post-oral infection. HWE and ORL mosquitoes exhibited no salivary gland infection barrier; however, only 60% (HWE) to 65% (ORL) of the females had released the virus in their saliva at one week post-oral acquisition, suggesting a salivary gland escape barrier. CHIKV induced an apoptotic response in salivary glands of HWE and ORL mosquitoes, demonstrating that the virus caused pathology in its natural vector.


Subject(s)
Aedes/virology , Chikungunya virus/growth & development , Mosquito Vectors , Animals , Apoptosis , Gastrointestinal Tract/pathology , Gastrointestinal Tract/virology , Host-Pathogen Interactions , Saliva/virology , Salivary Glands/pathology , Salivary Glands/virology
11.
J Gen Virol ; 97(5): 1033-1036, 2016 05.
Article in English | MEDLINE | ID: mdl-26872460

ABSTRACT

A relatively small number of mosquito species transmit arboviruses such as dengue, yellow fever, chikungunya and West Nile viruses to hundreds of millions of people each year, yet we still lack a thorough understanding of the molecular factors that determine vector competence. Apoptosis has been shown to be an important factor in determining the outcome of virus infection for many viruses. However, until recently, it was not clear whether apoptosis plays a role in determining the outcome of arbovirus infections in mosquitoes. Recent work has begun to shed light on the roles of apoptosis in this important process.


Subject(s)
Apoptosis , Arboviruses/physiology , Culicidae/virology , Insect Vectors/virology , Animals , Female , Virus Replication/physiology
12.
J Virol ; 90(3): 1668-72, 2016 02 01.
Article in English | MEDLINE | ID: mdl-26537678

ABSTRACT

The GP64 envelope fusion protein is a hallmark of group I alphabaculoviruses. However, the Diatraea saccharalis granulovirus genome sequence revealed the first betabaculovirus species harboring a gp64 homolog (disa118). In this work, we have shown that this homolog encodes a functional envelope fusion protein and could enable the infection and fusogenic abilities of a gp64-null prototype baculovirus. Therefore, GP64 may complement or may be in the process of replacing F protein activity in this virus lineage.


Subject(s)
Granulovirus/physiology , Viral Fusion Proteins/genetics , Viral Fusion Proteins/metabolism , Virus Internalization , Animals , Genetic Complementation Test , Granulovirus/genetics , Lepidoptera/virology
13.
Viruses ; 7(7): 3741-67, 2015 Jul 08.
Article in English | MEDLINE | ID: mdl-26184281

ABSTRACT

Arthropod-borne viruses (arboviruses) circulate in nature between arthropod vectors and vertebrate hosts. Arboviruses often cause devastating diseases in vertebrate hosts, but they typically do not cause significant pathology in their arthropod vectors. Following oral acquisition of a viremic bloodmeal from a vertebrate host, the arbovirus disease cycle requires replication in the cellular environment of the arthropod vector. Once the vector has become systemically and persistently infected, the vector is able to transmit the virus to an uninfected vertebrate host. In order to systemically infect the vector, the virus must cope with innate immune responses and overcome several tissue barriers associated with the midgut and the salivary glands. In this review we describe, in detail, the typical arbovirus infection route in competent mosquito vectors. Based on what is known from the literature, we explain the nature of the tissue barriers that arboviruses are confronted with in a mosquito vector and how arboviruses might surmount these barriers. We also point out controversial findings to highlight particular areas that are not well understood and require further research efforts.


Subject(s)
Arbovirus Infections/transmission , Arboviruses/physiology , Culicidae/virology , Insect Vectors/virology , Animals , Arbovirus Infections/virology , Humans , Salivary Glands/virology
14.
Insect Biochem Mol Biol ; 62: 64-74, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25858029

ABSTRACT

Signal transduction pathways and their coordination are critically important for proper functioning of animal immune systems. Our knowledge of the constituents of the intracellular signaling network in insects mainly comes from genetic analyses in Drosophila melanogaster. To facilitate future studies of similar systems in the tobacco hornworm and other lepidopteran insects, we have identified and examined the homologous genes in the genome of Manduca sexta. Based on 1:1 orthologous relationships in most cases, we hypothesize that the Toll, Imd, MAPK-JNK-p38 and JAK-STAT pathways are intact and operative in this species, as are most of the regulatory mechanisms. Similarly, cellular processes such as autophagy, apoptosis and RNA interference probably function in similar ways, because their mediators and modulators are mostly conserved in this lepidopteran species. We have annotated a total of 186 genes encoding 199 proteins, studied their domain structures and evolution, and examined their mRNA levels in tissues at different life stages. Such information provides a genomic perspective of the intricate signaling system in a non-drosophiline insect.


Subject(s)
Insect Proteins/genetics , Manduca/immunology , Amino Acid Sequence , Animals , Gene Expression Profiling , Genome, Insect , Immunity, Innate , Insect Proteins/metabolism , Manduca/genetics , Manduca/metabolism , Molecular Sequence Annotation , Molecular Sequence Data , RNA, Messenger/metabolism , Sequence Analysis, RNA , Signal Transduction
15.
PLoS One ; 10(3): e0122353, 2015.
Article in English | MEDLINE | ID: mdl-25815482

ABSTRACT

In vivo targeted gene disruption is a powerful tool to study gene function. Thus far, two tools for genome editing in Aedes aegypti have been applied, zinc-finger nucleases (ZFN) and transcription activator-like effector nucleases (TALEN). As a promising alternative to ZFN and TALEN, which are difficult to produce and validate using standard molecular biological techniques, the clustered regularly interspaced short palindromic repeats/CRISPR-associated sequence 9 (CRISPR/Cas9) system has recently been discovered as a "do-it-yourself" genome editing tool. Here, we describe the use of CRISPR/Cas9 in the mosquito vector, Aedes aegypti. In a transgenic mosquito line expressing both Dsred and enhanced cyan fluorescent protein (ECFP) from the eye tissue-specific 3xP3 promoter in separated but tightly linked expression cassettes, we targeted the ECFP nucleotide sequence for disruption. When supplying the Cas9 enzyme and two sgRNAs targeting different regions of the ECFP gene as in vitro transcribed mRNAs for germline transformation, we recovered four different G1 pools (5.5% knockout efficiency) where individuals still expressed DsRed but no longer ECFP. PCR amplification, cloning, and sequencing of PCR amplicons revealed indels in the ECFP target gene ranging from 2-27 nucleotides. These results show for the first time that CRISPR/Cas9 mediated gene editing is achievable in Ae. aegypti, paving the way for further functional genomics related studies in this mosquito species.


Subject(s)
Aedes/genetics , CRISPR-Cas Systems/genetics , Genome, Insect , Yellow Fever/genetics , Aedes/pathogenicity , Animals , Base Sequence , Humans , INDEL Mutation , RNA Editing/genetics , Yellow Fever/transmission , Zinc Fingers/genetics
16.
Virology ; 479-480: 637-49, 2015 May.
Article in English | MEDLINE | ID: mdl-25724418

ABSTRACT

Baculovirus infection of a host insect involves several steps, beginning with initiation of virus infection in the midgut, followed by dissemination of infection from the midgut to other tissues in the insect, and finally culminating in "melting" or liquefaction of the host, which allows for horizontal spread of infection to other insects. While all of the viral gene products are involved in ultimately reaching this dramatic infection endpoint, this review focuses on two particular types of baculovirus-encoded proteins: degradative enzymes and protease inhibitors. Neither of these types of proteins is commonly found in other virus families, but they both play important roles in baculovirus infection. The types of degradative enzymes and protease inhibitors encoded by baculoviruses are discussed, as are the roles of these proteins in the infection process.


Subject(s)
Baculoviridae/enzymology , Baculoviridae/growth & development , Chitinases/metabolism , Insecta/virology , Peptide Hydrolases/metabolism , Protease Inhibitors/metabolism , Animals
17.
Proc Natl Acad Sci U S A ; 112(10): E1152-61, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25713358

ABSTRACT

Millions of people are infected each year by arboviruses (arthropod-borne viruses) such as chikungunya, dengue, and West Nile viruses, yet for reasons that are largely unknown, only a relatively small number of mosquito species are able to transmit arboviruses. Understanding the complex factors that determine vector competence could facilitate strategies for controlling arbovirus infections. Apoptosis is a potential antiviral defense response that has been shown to be important in other virus-host systems. However, apoptosis is rarely seen in arbovirus-infected mosquito cells, raising questions about its importance as an antiviral defense in mosquitoes. We tested the effect of stimulating apoptosis during arbovirus infection by infecting Aedes aegypti mosquitoes with a Sindbis virus (SINV) clone called MRE/Rpr, in which the MRE-16 strain of SINV was engineered to express the proapoptotic gene reaper from Drosophila. MRE/Rpr exhibited an impaired infection phenotype that included delayed midgut infection, delayed virus replication, and reduced virus accumulation in saliva. Nucleotide sequencing of the reaper insert in virus populations isolated from individual mosquitoes revealed evidence of rapid and strong selection against maintenance of Reaper expression in MRE/Rpr-infected mosquitoes. The impaired phenotype of MRE/Rpr, coupled with the observed negative selection against Reaper expression, indicates that apoptosis is a powerful defense against arbovirus infection in mosquitoes and suggests that arboviruses have evolved mechanisms to avoid stimulating apoptosis in mosquitoes that serve as vectors.


Subject(s)
Aedes/virology , Apoptosis/physiology , Insect Vectors/virology , Selection, Genetic , Sindbis Virus/physiology , Aedes/genetics , Animals , Insect Vectors/genetics , Saliva/virology , Virus Replication
18.
Semin Cell Dev Biol ; 39: 72-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25652775

ABSTRACT

The identification, now more than 20 years ago, of the first iap genes in baculoviruses subsequently led to many important discoveries concerning the regulation of apoptosis and other important biological processes in insects and mammals. Currently there are more than 200 known viral IAP homologs in baculoviruses and other families of invertebrate DNA viruses. This review begins with a personal account of the events leading up to the discovery of the first iap genes, followed by a summary of what is currently known about the different types of viral IAPs and their functions in regulating apoptosis, and possibly other cellular processes.


Subject(s)
Inhibitor of Apoptosis Proteins/history , Inhibitor of Apoptosis Proteins/metabolism , Viral Proteins/history , Viral Proteins/metabolism , Animals , Apoptosis , History, 20th Century , Inhibitor of Apoptosis Proteins/chemistry , Inhibitor of Apoptosis Proteins/genetics , Phylogeny , Viral Proteins/chemistry , Viral Proteins/genetics
19.
J Gen Virol ; 96(Pt 5): 1150-1160, 2015 May.
Article in English | MEDLINE | ID: mdl-25573886

ABSTRACT

The serpin family of serine proteinase inhibitors plays key roles in a variety of biochemical pathways. In insects, one of the important functions carried out by serpins is regulation of the phenoloxidase (PO) cascade - a pathway that produces melanin and other compounds that are important in insect humoral immunity. Recent sequencing of the baculovirus Hemileuca sp. nucleopolyhedrovirus (HespNPV) genome revealed the presence of a gene, hesp018, with homology to insect serpins. To our knowledge, hesp018 is the first viral serpin homologue to be characterized outside of the chordopoxviruses. The Hesp018 protein was found to be a functional serpin with inhibitory activity against a subset of serine proteinases. Hesp018 also inhibited PO activation when mixed with lepidopteran haemolymph. The Hesp018 protein was secreted when expressed in lepidopteran cells and a baculovirus expressing Hesp018 exhibited accelerated production of viral progeny during in vitro infection. Expression of Hesp018 also reduced caspase activity induced by baculovirus infection, but caused increased cathepsin activity. In infected insect larvae, expression of Hesp018 resulted in faster larval melanization, consistent with increased activity of viral cathepsin. Finally, expression of Hesp018 increased the virulence of a prototype baculovirus by fourfold in orally infected neonate Trichoplusia ni larvae. Based on our observations, we hypothesize that hesp018 may have been retained in HespNPV due to its ability to inhibit the activity of select host proteinases, possibly including proteinases involved in the PO response, during infection of host insects.


Subject(s)
Baculoviridae/physiology , Serpins/metabolism , Viral Proteins/metabolism , Animals , Baculoviridae/genetics , Baculoviridae/growth & development , Host-Pathogen Interactions , Insecta , Monophenol Monooxygenase/antagonists & inhibitors , Serine Proteases/metabolism , Serpins/genetics , Viral Proteins/genetics
20.
Curr Trop Med Rep ; 1(1): 21-31, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24693489

ABSTRACT

Aedes aegypti is the principal vector of dengue virus (DENV) throughout the tropical world. This anthropophilic mosquito species needs to be persistently infected with DENV before it can transmit the virus through its saliva to a new vertebrate host. In the mosquito, DENV is confronted with several innate immune pathways, among which RNA interference is considered the most important. The Ae. aegypti genome project opened the doors for advanced molecular studies on pathogen-vector interactions including genetic manipulation of the vector for basic research and vector control purposes. Thus, Ae. aegypti has become the primary model for studying vector competence for arboviruses at the molecular level. Here, we present recent findings regarding DENV-mosquito interactions, emphasizing how innate immune responses modulate DENV infections in Ae. aegypti. We also describe the latest advancements in genetic manipulation of Ae. aegypti and discuss how this technology can be used to investigate vector transmission of DENV at the molecular level and to control transmission of the virus in the field.

SELECTION OF CITATIONS
SEARCH DETAIL
...