Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
3.
Nat Biotechnol ; 38(4): 482-492, 2020 04.
Article in English | MEDLINE | ID: mdl-32265562

ABSTRACT

The range of the mosquito Aedes aegypti continues to expand, putting more than two billion people at risk of arboviral infection. The sterile insect technique (SIT) has been used to successfully combat agricultural pests at large scale, but not mosquitoes, mainly because of challenges with consistent production and distribution of high-quality male mosquitoes. We describe automated processes to rear and release millions of competitive, sterile male Wolbachia-infected mosquitoes, and use of these males in a large-scale suppression trial in Fresno County, California. In 2018, we released 14.4 million males across three replicate neighborhoods encompassing 293 hectares. At peak mosquito season, the number of female mosquitoes was 95.5% lower (95% CI, 93.6-96.9) in release areas compared to non-release areas, with the most geographically isolated neighborhood reaching a 99% reduction. This work demonstrates the high efficacy of mosquito SIT in an area ninefold larger than in previous similar trials, supporting the potential of this approach in public health and nuisance-mosquito eradication programs.


Subject(s)
Aedes/microbiology , Aedes/physiology , Mosquito Control/methods , Mosquito Vectors/microbiology , Mosquito Vectors/physiology , Wolbachia/physiology , Aedes/growth & development , Animal Migration , Animals , California , Female , Larva/growth & development , Larva/microbiology , Larva/physiology , Male , Mosquito Control/statistics & numerical data , Mosquito Vectors/growth & development , Population Dynamics , Sex Characteristics
4.
Genes Dev ; 30(14): 1658-70, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27474443

ABSTRACT

RNA degradation is tightly regulated to selectively target aberrant RNAs, including viral RNA, but this regulation is incompletely understood. Through RNAi screening in Drosophila cells, we identified the 3'-to-5' RNA exosome and two components of the exosome cofactor TRAMP (Trf4/5-Air1/2-Mtr4 polyadenylation) complex, dMtr4 and dZcchc7, as antiviral against a panel of RNA viruses. We extended our studies to human orthologs and found that the exosome as well as TRAMP components hMTR4 and hZCCHC7 are antiviral. While hMTR4 and hZCCHC7 are normally nuclear, infection by cytoplasmic RNA viruses induces their export, forming a cytoplasmic complex that specifically recognizes and induces degradation of viral mRNAs. Furthermore, the 3' untranslated region (UTR) of bunyaviral mRNA is sufficient to confer virus-induced exosomal degradation. Altogether, our results reveal that signals from viral infection repurpose TRAMP components to a cytoplasmic surveillance role where they selectively engage viral RNAs for degradation to restrict a broad range of viruses.


Subject(s)
Exosomes/metabolism , RNA Stability/physiology , RNA, Viral/metabolism , Animals , Cell Line , Cytoplasm/metabolism , Drosophila/virology , Humans , Multiprotein Complexes/genetics , Polyadenylation , Protein Binding , Protein Transport , RNA Interference , RNA Virus Infections/metabolism , RNA Virus Infections/virology , RNA Viruses/physiology , Transcription Factors/metabolism
5.
Proc Natl Acad Sci U S A ; 112(25): 7797-802, 2015 Jun 23.
Article in English | MEDLINE | ID: mdl-26056282

ABSTRACT

Viruses must gain entry into cells to establish infection. In general, viruses enter either at the plasma membrane or from intracellular endosomal compartments. Viruses that use endosomal pathways are dependent on the cellular factors that control this process; however, these genes have proven to be essential for endogenous cargo uptake, and thus are of limited value for therapeutic intervention. The identification of genes that are selectively required for viral uptake would make appealing drug targets, as their inhibition would block an early step in the life cycle of diverse viruses. At this time, we lack pan-antiviral therapeutics, in part because of our lack of knowledge of such cellular factors. RNAi screening has begun to reveal previously unknown genes that play roles in viral infection. We identified dRNASEK in two genome-wide RNAi screens performed in Drosophila cells against West Nile and Rift Valley Fever viruses. Here we found that ribonuclease kappa (RNASEK) is essential for the infection of human cells by divergent and unrelated positive- and negative-strand-enveloped viruses from the Flaviviridae, Togaviridae, Bunyaviridae, and Orthomyxoviridae families that all enter cells from endosomal compartments. In contrast, RNASEK was dispensable for viruses, including parainfluenza virus 5 and Coxsackie B virus, that enter at the plasma membrane. RNASEK is dispensable for attachment but is required for uptake of these acid-dependent viruses. Furthermore, this requirement appears specific, as general endocytic uptake of transferrin is unaffected in RNASEK-depleted cells. Therefore, RNASEK is a potential host cell Achilles' heel for viral infection.


Subject(s)
Endocytosis , Membrane Fusion , Ribonucleases/metabolism , Animals , Drosophila , Hydrogen-Ion Concentration , Rift Valley fever virus/physiology , West Nile virus/physiology
6.
Proc Natl Acad Sci U S A ; 112(22): E2920-9, 2015 Jun 02.
Article in English | MEDLINE | ID: mdl-26038567

ABSTRACT

The mosquito-transmitted bunyavirus, Rift Valley fever virus (RVFV), is a highly successful pathogen for which there are no vaccines or therapeutics. Translational arrest is a common antiviral strategy used by hosts. In response, RVFV inhibits two well-known antiviral pathways that attenuate translation during infection, PKR and type I IFN signaling. Despite this, translational arrest occurs during RVFV infection by unknown mechanisms. Here, we find that RVFV infection triggers the decay of core translation machinery mRNAs that possess a 5'-terminal oligopyrimidine (5'-TOP) motif in their 5'-UTR, including mRNAs encoding ribosomal proteins, which leads to a decrease in overall ribosomal protein levels. We find that the RNA decapping enzyme NUDT16 selectively degrades 5'-TOP mRNAs during RVFV infection and this decay is triggered in response to mTOR attenuation via the translational repressor 4EBP1/2 axis. Translational arrest of 5'-TOPs via 4EBP1/2 restricts RVFV replication, and this increased RNA decay results in the loss of visible RNA granules, including P bodies and stress granules. Because RVFV cap-snatches in RNA granules, the increased level of 5'-TOP mRNAs in this compartment leads to snatching of these targets, which are translationally suppressed during infection. Therefore, translation of RVFV mRNAs is compromised by multiple mechanisms during infection. Together, these data present a previously unknown mechanism for translational shutdown in response to viral infection and identify mTOR attenuation as a potential therapeutic avenue against bunyaviral infection.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Phosphoproteins/metabolism , Protein Biosynthesis/physiology , Pyrophosphatases/metabolism , RNA 5' Terminal Oligopyrimidine Sequence/physiology , Rift Valley Fever/metabolism , Rift Valley fever virus/metabolism , TOR Serine-Threonine Kinases/metabolism , Cell Cycle Proteins , Cell Line , DNA Primers/genetics , Electrophoresis, Polyacrylamide Gel , Fluorescent Antibody Technique , Humans , Immunoblotting , Linear Models , Luciferases , RNA 5' Terminal Oligopyrimidine Sequence/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction
7.
Proc Natl Acad Sci U S A ; 111(37): E3890-9, 2014 Sep 16.
Article in English | MEDLINE | ID: mdl-25197089

ABSTRACT

In response to infection, the innate immune system rapidly activates an elaborate and tightly orchestrated gene expression program to induce critical antimicrobial genes. While many key players in this program have been identified in disparate biological systems, it is clear that there are additional uncharacterized mechanisms at play. Our previous studies revealed that a rapidly-induced antiviral gene expression program is active against disparate human arthropod-borne viruses in Drosophila. Moreover, one-half of this program is regulated at the level of transcriptional pausing. Here we found that Nup98, a virus-induced gene, was antiviral against a panel of viruses both in cells and adult flies since its depletion significantly enhanced viral infection. Mechanistically, we found that Nup98 promotes antiviral gene expression in Drosophila at the level of transcription. Expression profiling revealed that the virus-induced activation of 36 genes was abrogated upon loss of Nup98; and we found that a subset of these Nup98-dependent genes were antiviral. These Nup98-dependent virus-induced genes are Cdk9-dependent and translation-independent suggesting that these are rapidly induced primary response genes. Biochemically, we demonstrate that Nup98 is directly bound to the promoters of virus-induced genes, and that it promotes occupancy of the initiating form of RNA polymerase II at these promoters, which are rapidly induced on viral infection to restrict human arboviruses in insects.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/virology , Gene Expression Regulation , Nuclear Pore Complex Proteins/metabolism , RNA Virus Infections/genetics , RNA Virus Infections/virology , RNA Viruses/physiology , Aging/pathology , Animals , Cell Nucleus/metabolism , Genes, Insect , Humans , Nuclear Pore/metabolism , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Protein Transport , RNA Polymerase II/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sindbis Virus/physiology
8.
Cell Rep ; 5(6): 1737-48, 2013 Dec 26.
Article in English | MEDLINE | ID: mdl-24332855

ABSTRACT

Alphaviruses are a large class of insect-borne human pathogens and little is known about the host-factor requirements for infection. To identify such factors, we performed a genome-wide RNAi screen using model Drosophila cells and validated 94 genes that impacted infection of Sindbis virus (SINV), the prototypical alphavirus. We identified a conserved role for SEC61A and valosin-containing protein (VCP) in facilitating SINV entry in insects and mammals. SEC61A and VCP selectively regulate trafficking of the entry receptor NRAMP2, and loss or pharmacological inhibition of these proteins leads to altered NRAMP2 trafficking to lysosomal compartments and proteolytic digestion within lysosomes. NRAMP2 is the major iron transporter in cells, and loss of NRAMP2 attenuates intracellular iron transport. Thus, this study reveals genes and pathways involved in both infection and iron homeostasis that may serve as targets for antiviral therapeutics or for iron-imbalance disorders.


Subject(s)
Adenosine Triphosphatases/metabolism , Drosophila Proteins/metabolism , Genome, Insect , Membrane Proteins/metabolism , Sindbis Virus/pathogenicity , Virus Internalization , Adenosine Triphosphatases/genetics , Aedes/genetics , Aedes/metabolism , Aedes/virology , Animals , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Line, Tumor , Drosophila/genetics , Drosophila/metabolism , Drosophila/virology , Drosophila Proteins/genetics , Humans , Iron/metabolism , Membrane Proteins/genetics , Protein Transport , RNA Interference , RNA, Small Interfering/genetics , SEC Translocation Channels , Valosin Containing Protein
9.
Genes Dev ; 27(13): 1511-25, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23824541

ABSTRACT

Bunyaviruses are an emerging group of medically important viruses, many of which are transmitted from insects to mammals. To identify host factors that impact infection, we performed a genome-wide RNAi screen in Drosophila and identified 131 genes that impacted infection of the mosquito-transmitted bunyavirus Rift Valley fever virus (RVFV). Dcp2, the catalytic component of the mRNA decapping machinery, and two decapping activators, DDX6 and LSM7, were antiviral against disparate bunyaviruses in both insect cells and adult flies. Bunyaviruses 5' cap their mRNAs by "cap-snatching" the 5' ends of poorly defined host mRNAs. We found that RVFV cap-snatches the 5' ends of Dcp2 targeted mRNAs, including cell cycle-related genes. Loss of Dcp2 allows increased viral transcription without impacting viral mRNA stability, while ectopic expression of Dcp2 impedes viral transcription. Furthermore, arresting cells in late S/early G2 led to increased Dcp2 mRNA targets and increased RVFV replication. Therefore, RVFV competes for the Dcp2-accessible mRNA pool, which is dynamically regulated and can present a bottleneck for viral replication.


Subject(s)
Drosophila melanogaster/genetics , Drosophila melanogaster/virology , Genome, Insect/genetics , Orthobunyavirus/physiology , RNA Caps/metabolism , Transcription Factors , Virus Replication/physiology , Aedes/virology , Animals , Cell Cycle Checkpoints , Cell Line , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Gene Expression Regulation , RNA Caps/genetics , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...