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










Publication year range
1.
Elife ; 122024 Jan 30.
Article in English | MEDLINE | ID: mdl-38289340

ABSTRACT

Each year, hundreds of millions of people are infected with arboviruses such as dengue, yellow fever, chikungunya, and Zika, which are all primarily spread by the notorious mosquito Aedes aegypti. Traditional control measures have proven insufficient, necessitating innovations. In response, here we generate a next-generation CRISPR-based precision-guided sterile insect technique (pgSIT) for Ae. aegypti that disrupts genes essential for sex determination and fertility, producing predominantly sterile males that can be deployed at any life stage. Using mathematical models and empirical testing, we demonstrate that released pgSIT males can effectively compete with, suppress, and eliminate caged mosquito populations. This versatile species-specific platform has the potential for field deployment to effectively control wild populations of disease vectors.


Subject(s)
Aedes , Infertility, Male , Zika Virus Infection , Zika Virus , Humans , Male , Animals , Mosquito Vectors/genetics , Aedes/genetics , Disease Vectors , Species Specificity , Zika Virus Infection/prevention & control
2.
Sci Rep ; 13(1): 19866, 2023 11 14.
Article in English | MEDLINE | ID: mdl-37964160

ABSTRACT

Tephritid fruit fly pests pose an increasing threat to the agricultural industry due to their global dispersion and a highly invasive nature. Here we showcase the feasibility of an early-detection SEPARATOR sex sorting approach through using the non-model Tephritid pest, Ceratitis capitata. This system relies on female-only fluorescent marker expression, accomplished through the use of a sex-specific intron of the highly-conserved transformer gene from C. capitata and Anastrepha ludens. The herein characterized strains have 100% desired phenotype outcomes, allowing accurate male-female separation during early development. Overall, we describe an antibiotic and temperature-independent sex-sorting system in C. capitata, which, moving forward, may be implemented in other non-model Tephritid pest species. This strategy can facilitate the establishment of genetic sexing systems with endogenous elements exclusively, which, on a wider scale, can improve pest population control strategies like sterile insect technique.


Subject(s)
Ceratitis capitata , Tephritidae , Animals , Male , Female , Ceratitis capitata/genetics , Ceratitis capitata/metabolism , Phenotype , Pest Control, Biological/methods
3.
bioRxiv ; 2023 Nov 15.
Article in English | MEDLINE | ID: mdl-37131747

ABSTRACT

Each year, hundreds of millions of people are infected with arboviruses such as dengue, yellow fever, chikungunya, and Zika, which are all primarily spread by the notorious mosquito Aedes aegypti. Traditional control measures have proven insufficient, necessitating innovations. In response, here we generate a next generation CRISPR-based precision-guided sterile insect technique (pgSIT) for Aedes aegypti that disrupts genes essential for sex determination and fertility, producing predominantly sterile males that can be deployed at any life stage. Using mathematical models and empirical testing, we demonstrate that released pgSIT males can effectively compete with, suppress, and eliminate caged mosquito populations. This versatile species-specific platform has the potential for field deployment to effectively control wild populations of disease vectors.

4.
Nat Commun ; 13(1): 7145, 2022 11 21.
Article in English | MEDLINE | ID: mdl-36414618

ABSTRACT

CRISPR/Cas gene drives can bias transgene inheritance through different mechanisms. Homing drives are designed to replace a wild-type allele with a copy of a drive element on the homologous chromosome. In Aedes aegypti, the sex-determining locus is closely linked to the white gene, which was previously used as a target for a homing drive element (wGDe). Here, through an analysis using this linkage we show that in males inheritance bias of wGDe did not occur by homing, rather through increased propagation of the donor drive element. We test the same wGDe drive element with transgenes expressing Cas9 with germline regulatory elements sds3, bgcn, and nup50. We only find inheritance bias through homing, even with the identical nup50-Cas9 transgene. We propose that DNA repair outcomes may be more context dependent than anticipated and that other previously reported homing drives may, in fact, bias their inheritance through other mechanisms.


Subject(s)
Aedes , Gene Drive Technology , Male , CRISPR-Cas Systems/genetics , Endonucleases/genetics , Germ Cells , Inheritance Patterns/genetics , Aedes/genetics , Animals , Transgenes
5.
Nat Commun ; 12(1): 7202, 2021 12 10.
Article in English | MEDLINE | ID: mdl-34893590

ABSTRACT

CRISPR-based genetic engineering tools aimed to bias sex ratios, or drive effector genes into animal populations, often integrate the transgenes into autosomal chromosomes. However, in species with heterogametic sex chromsomes (e.g. XY, ZW), sex linkage of endonucleases could be beneficial to drive the expression in a sex-specific manner to produce genetic sexing systems, sex ratio distorters, or even sex-specific gene drives, for example. To explore this possibility, here we develop a transgenic line of Drosophila melanogaster expressing Cas9 from the Y chromosome. We functionally characterize the utility of this strain for both sex selection and gene drive finding it to be quite effective. To explore its utility for population control, we built mathematical models illustrating its dynamics as compared to other state-of-the-art systems designed for both population modification and suppression. Taken together, our results contribute to the development of current CRISPR genetic control tools and demonstrate the utility of using sex-linked Cas9 strains for genetic control of animals.


Subject(s)
CRISPR-Cas Systems , Gene Drive Technology/methods , Genes, Y-Linked , Sex Preselection/methods , Y Chromosome , Animals , Animals, Genetically Modified , Drosophila melanogaster/genetics , Endonucleases/genetics , Female , Gene Editing/methods , Male , Sex Ratio , Synthetic Biology/methods , Transgenes
6.
CRISPR J ; 4(6): 822-835, 2021 12.
Article in English | MEDLINE | ID: mdl-34735288

ABSTRACT

Releases of sterile males are the gold standard for many insect population control programs, and precise sex sorting to remove females prior to male releases is essential to the success of these operations. To advance traditional methods for scaling the generation of sterile males, we previously described a CRISPR-mediated precision-guided sterile insect technique (pgSIT), in which Cas9 and gRNA strains are genetically crossed to generate sterile males for mass release. While effective at generating F1 sterile males, pgSIT requires a genetic cross between the two parental strains, which requires maintenance and sexing of two strains in a factory. Therefore, to advance pgSIT further by removing this crossing step, here we describe a next-generation temperature-inducible pgSIT (TI-pgSIT) technology and demonstrate its proof-of-concept in Drosophila melanogaster. Importantly, we were able to develop a true breeding strain for TI-pgSIT that eliminates the requirement for sex sorting-a feature that may help further automate production at scale.


Subject(s)
Drosophila melanogaster , Gene Editing , Animals , CRISPR-Cas Systems/genetics , Drosophila melanogaster/genetics , Female , Male , RNA, Guide, Kinetoplastida/genetics , Temperature
7.
CRISPR J ; 4(5): 739-751, 2021 10.
Article in English | MEDLINE | ID: mdl-34661429

ABSTRACT

Originally from Asia, Drosophila suzukii Matsumura is a global pest of economically important soft-skinned fruits. Also commonly known as spotted wing drosophila, it is largely controlled through repeated applications of broad-spectrum insecticides by which resistance has been observed in the field. There is a pressing need for a better understanding of D. suzukii biology and for developing alternative environmentally friendly methods of control. The RNA-guided Cas9 nuclease has revolutionized functional genomics and is an integral component of several recently developed genetic strategies for population control of insects. Here, we describe genetically modified strains that encode three different terminators and four different promoters to express Cas9 robustly in both the soma and/or germline of D. suzukii. The Cas9 strains were rigorously evaluated through genetic crossing to transgenic strains that encode single-guide RNAs targeting the conserved X-linked yellow body and white eye genes. We find that several Cas9/gRNA strains display remarkably high editing capacity. Going forward, these tools will be instrumental for evaluating gene function in D. suzukii and may even provide tools useful for the development of new genetic strategies for control of this invasive species.


Subject(s)
CRISPR-Cas Systems , Drosophila/genetics , Gene Editing/methods , Pest Control, Biological/methods , Animals , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/metabolism , Drosophila/pathogenicity , Drosophila Proteins/genetics , Fruit/parasitology , Introduced Species
8.
Nat Commun ; 12(1): 5374, 2021 09 10.
Article in English | MEDLINE | ID: mdl-34508072

ABSTRACT

The mosquito Aedes aegypti is the principal vector for arboviruses including dengue/yellow fever, chikungunya, and Zika virus, infecting hundreds of millions of people annually. Unfortunately, traditional control methodologies are insufficient, so innovative control methods are needed. To complement existing measures, here we develop a molecular genetic control system termed precision-guided sterile insect technique (pgSIT) in Aedes aegypti. PgSIT uses a simple CRISPR-based approach to generate flightless females and sterile males that are deployable at any life stage. Supported by mathematical models, we empirically demonstrate that released pgSIT males can compete, suppress, and even eliminate mosquito populations. This platform technology could be used in the field, and adapted to many vectors, for controlling wild populations to curtail disease in a safe, confinable, and reversible manner.


Subject(s)
Aedes/virology , Infertility, Male/veterinary , Mosquito Control/methods , Mosquito Vectors/virology , Aedes/genetics , Animals , Animals, Genetically Modified , Arboviruses , Chikungunya Fever/prevention & control , Chikungunya Fever/transmission , Chikungunya Fever/virology , Dengue/prevention & control , Dengue/transmission , Dengue/virology , Female , Humans , Infertility, Male/genetics , Male , Models, Biological , Mosquito Vectors/genetics , Yellow Fever/prevention & control , Yellow Fever/transmission , Yellow Fever/virology , Zika Virus , Zika Virus Infection/prevention & control , Zika Virus Infection/transmission , Zika Virus Infection/virology
9.
Elife ; 102021 03 05.
Article in English | MEDLINE | ID: mdl-33666174

ABSTRACT

Homing-based gene drives, engineered using CRISPR/Cas9, have been proposed to spread desirable genes throughout populations. However, invasion of such drives can be hindered by the accumulation of resistant alleles. To limit this obstacle, we engineer a confinable population modification home-and-rescue (HomeR) drive in Drosophila targeting an essential gene. In our experiments, resistant alleles that disrupt the target gene function were recessive lethal and therefore disadvantaged. We demonstrate that HomeR can achieve an increase in frequency in population cage experiments, but that fitness costs due to the Cas9 insertion limit drive efficacy. Finally, we conduct mathematical modeling comparing HomeR to contemporary gene drive architectures for population modification over wide ranges of fitness costs, transmission rates, and release regimens. HomeR could potentially be adapted to other species, as a means for safe, confinable, modification of wild populations.


Subject(s)
CRISPR-Cas Systems , Drosophila melanogaster/genetics , Gene Drive Technology/methods , Animals , Genetic Engineering/methods , Genetic Fitness , Genetics, Population , Models, Theoretical
10.
Nat Commun ; 11(1): 2106, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32355156

ABSTRACT

Here, we describe a drug-inducible genetic system for insect sex-separation that demonstrates proof-of-principle for positive sex selection in D. melanogaster. The system exploits the toxicity of commonly used broad-spectrum antibiotics geneticin and puromycin to kill the non-rescued sex. Sex-specific rescue is achieved by inserting sex-specific introns into the coding sequences of antibiotic-resistance genes. When raised on geneticin-supplemented food, the sex-sorter line establishes 100% positive selection for female progeny, while the food supplemented with puromycin positively selects 100% male progeny. Since the described system exploits conserved sex-specific splicing mechanisms and reagents, it has the potential to be adaptable to other insect species of medical and agricultural importance.


Subject(s)
Drosophila melanogaster/drug effects , Genetic Engineering/methods , Gentamicins/pharmacology , Puromycin/pharmacology , Animals , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drug Resistance , Exons , Female , Genetics, Population , Homozygote , Introns , Male , Pest Control , RNA Splicing , Sex Determination Analysis
11.
Elife ; 92020 01 21.
Article in English | MEDLINE | ID: mdl-31960794

ABSTRACT

Aedes aegypti is the principal mosquito vector for many arboviruses that increasingly infect millions of people every year. With an escalating burden of infections and the relative failure of traditional control methods, the development of innovative control measures has become of paramount importance. The use of gene drives has sparked significant enthusiasm for genetic control of mosquitoes; however, no such system has been developed in Ae. aegypti. To fill this void, here we develop several CRISPR-based split gene drives for use in this vector. With cleavage rates up to 100% and transmission rates as high as 94%, mathematical models predict that these systems could spread anti-pathogen effector genes into wild populations in a safe, confinable and reversible manner appropriate for field trials and effective for controlling disease. These findings could expedite the development of effector-linked gene drives that could safely control wild populations of Ae. aegypti to combat local pathogen transmission.


Subject(s)
Aedes/genetics , Gene Drive Technology , Mosquito Vectors/genetics , Aedes/physiology , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/physiology , CRISPR-Cas Systems/genetics , Female , Male , Mosquito Vectors/physiology , RNA, Guide, Kinetoplastida/genetics
12.
G3 (Bethesda) ; 10(2): 827-837, 2020 02 06.
Article in English | MEDLINE | ID: mdl-31882406

ABSTRACT

Homing based gene drives (HGD) possess the potential to spread linked cargo genes into natural populations and are poised to revolutionize population control of animals. Given that host encoded genes have been identified that are important for pathogen transmission, targeting these genes using guide RNAs as cargo genes linked to drives may provide a robust method to prevent disease transmission. However, effectiveness of the inclusion of additional guide RNAs that target separate genes has not been thoroughly explored. To test this approach, we generated a split-HGD in Drosophila melanogaster that encoded a drive linked effector consisting of a second gRNA engineered to target a separate host-encoded gene, which we term a gRNA-mediated effector (GME). This design enabled us to assess homing and knockout efficiencies of two target genes simultaneously, and also explore the timing and tissue specificity of Cas9 expression on cleavage/homing rates. We demonstrate that inclusion of a GME can result in high efficiency of disruption of both genes during super-Mendelian propagation of split-HGD. Furthermore, both genes were knocked out one generation earlier than expected indicating the robust somatic expression of Cas9 driven by Drosophila germline-limited promoters. We also assess the efficiency of 'shadow drive' generated by maternally deposited Cas9 protein and accumulation of drive-induced resistance alleles along multiple generations, and discuss design principles of HGD that could mitigate the accumulation of resistance alleles while incorporating a GME.


Subject(s)
Gene Drive Technology , Gene Knockout Techniques , Gene Targeting , CRISPR-Cas Systems , Gene Editing , Gene Order , Gene Targeting/methods , Genetic Vectors/genetics , Genotyping Techniques , Models, Genetic , Mutation , RNA, Guide, Kinetoplastida , Zygote/metabolism
13.
Front Genet ; 10: 1072, 2019.
Article in English | MEDLINE | ID: mdl-31737050

ABSTRACT

While efforts to control malaria with available tools have stagnated, and arbovirus outbreaks persist around the globe, the advent of clustered regularly interspaced short palindromic repeat (CRISPR)-based gene editing has provided exciting new opportunities for genetics-based strategies to control these diseases. In one such strategy, called "population replacement", mosquitoes, and other disease vectors are engineered with effector genes that render them unable to transmit pathogens. These effector genes can be linked to "gene drive" systems that can bias inheritance in their favor, providing novel opportunities to replace disease-susceptible vector populations with disease-refractory ones over the course of several generations. While promising for the control of vector-borne diseases on a wide scale, this sets up an evolutionary tug-of-war between the introduced effector genes and the pathogen. Here, we review the disease-refractory genes designed to date to target Plasmodium falciparum malaria transmitted by Anopheles gambiae, and arboviruses transmitted by Aedes aegypti, including dengue serotypes 2 and 3, chikungunya, and Zika viruses. We discuss resistance concerns for these effector genes, and genetic approaches to prevent parasite and viral escape variants. One general approach is to increase the evolutionary hurdle required for the pathogen to evolve resistance by attacking it at multiple sites in its genome and/or multiple stages of development. Another is to reduce the size of the pathogen population by other means, such as with vector control and antimalarial drugs. We discuss lessons learned from the evolution of resistance to antimalarial and antiviral drugs and implications for the management of resistance after its emergence. Finally, we discuss the target product profile for population replacement strategies for vector-borne disease control. This differs between early phase field trials and wide-scale disease control. In the latter case, the demands on effector gene efficacy are great; however, with new possibilities ushered in by CRISPR-based gene editing, and when combined with surveillance, monitoring, and rapid management of pathogen resistance, the odds are increasingly favoring effector genes in the upcoming evolutionary tug-of-war.

15.
Nat Commun ; 10(1): 84, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30622266

ABSTRACT

The sterile insect technique (SIT) is an environmentally safe and proven technology to suppress wild populations. To further advance its utility, a novel CRISPR-based technology termed precision guided SIT (pgSIT) is described. PgSIT mechanistically relies on a dominant genetic technology that enables simultaneous sexing and sterilization, facilitating the release of eggs into the environment ensuring only sterile adult males emerge. Importantly, for field applications, the release of eggs will eliminate burdens of manually sexing and sterilizing males, thereby reducing overall effort and increasing scalability. Here, to demonstrate efficacy, we systematically engineer multiple pgSIT systems in Drosophila which consistently give rise to 100% sterile males. Importantly, we demonstrate that pgSIT-generated sterile males are fit and competitive. Using mathematical models, we predict pgSIT will induce substantially greater population suppression than can be achieved by currently-available self-limiting suppression technologies. Taken together, pgSIT offers to potentially transform our ability to control insect agricultural pests and disease vectors.


Subject(s)
Drosophila/genetics , Gene Editing/methods , Insect Vectors/genetics , Pest Control, Biological/methods , Sexual Behavior, Animal , Animals , CRISPR-Cas Systems/genetics , Drosophila/physiology , Female , Genome, Insect/genetics , Insect Vectors/physiology , Male , Models, Biological , Population Control/methods , RNA, Guide, Kinetoplastida/genetics
16.
Nat Commun ; 7: 13100, 2016 11 14.
Article in English | MEDLINE | ID: mdl-27841259

ABSTRACT

Mitochondrial DNA (mtDNA) often exists in a state of heteroplasmy, in which mutant mtDNA co-exists in cells with wild-type mtDNA. High frequencies of pathogenic mtDNA result in maternally inherited diseases; maternally and somatically acquired mutations also accumulate over time and contribute to diseases of ageing. Reducing heteroplasmy is therefore a therapeutic goal and in vivo models in post-mitotic tissues are needed to facilitate these studies. Here we describe a transgene-based model of a heteroplasmic lethal mtDNA deletion (mtDNAΔ) in adult Drosophila muscle. Stimulation of autophagy, activation of the PINK1/parkin pathway or decreased levels of mitofusin result in a selective decrease in mtDNAΔ. Decreased levels of mitofusin and increased levels of ATPIF1, an inhibitor of ATP synthase reversal-dependent mitochondrial repolarization, result in a further decrease in mtDNAΔ levels. These results show that an adult post-mitotic tissue can be cleansed of a deleterious genome, suggesting that therapeutic removal of mutant mtDNA can be achieved.


Subject(s)
DNA, Mitochondrial/genetics , Drosophila melanogaster/genetics , Mitochondrial Dynamics/genetics , Sequence Deletion , Animals , Animals, Genetically Modified , Autophagy/genetics , DNA, Mitochondrial/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Microscopy, Electron, Transmission , Mitochondrial Proton-Translocating ATPases/genetics , Mitochondrial Proton-Translocating ATPases/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , Mutation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proteins/genetics , Proteins/metabolism , ATPase Inhibitory Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...