Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 114
Filtrar
1.
BMC Biol ; 22(1): 201, 2024 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-39256812

RESUMO

BACKGROUND: Methods to suppress pest insect populations using genetic constructs and repeated releases of male homozygotes have recently been shown to be an attractive alternative to older sterile insect techniques based on radiation. Female-specific lethal alleles have substantially increased power, but still require large, sustained transgenic insect releases. Gene drive alleles bias their own inheritance to spread throughout populations, potentially allowing population suppression with a single, small-size release. However, suppression drives often suffer from efficiency issues, and the most well-studied type, homing drives, tend to spread without limit. RESULTS: In this study, we show that coupling female-specific lethal alleles with homing gene drive allowed substantial improvement in efficiency while still retaining the self-limiting nature (and thus confinement) of a lethal allele strategy. Using a mosquito model, we show the required release sizes for population elimination in a variety of scenarios, including different density growth curves, with comparisons to other systems. Resistance alleles reduced the power of this method, but these could be overcome by targeting an essential gene with the drive while also providing rescue. A proof-of-principle demonstration of this system in Drosophila melanogaster was effective in both biasing its inheritance and achieving high lethality among females that inherit the construct in the absence of antibiotic. CONCLUSIONS: Overall, our study shows that substantial improvements can be achieved in female-specific lethal systems for population suppression by combining them with various types of gene drive.


Assuntos
Alelos , Drosophila melanogaster , Tecnologia de Impulso Genético , Animais , Feminino , Tecnologia de Impulso Genético/métodos , Drosophila melanogaster/genética , Masculino , Genes Letais , Controle Biológico de Vetores/métodos , Controle de Mosquitos/métodos , Animais Geneticamente Modificados/genética , Genes Dominantes
2.
Cell Genom ; 4(9): 100653, 2024 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-39265528

RESUMO

Drug resistance is a major hurdle prohibiting effective treatment of many diseases, including cancer. Using model-guided designs, Leighow et al.1 engineered a dual-switch selection gene drive system custom designed to combat drug-resistant tumors. By demonstrating remarkable killing efficacies in preclinical models using human non-small lung cancer cells in vitro and in mice, this work describes an attractive mindset to develop next-generation anticancer therapies.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Resistencia a Medicamentos Antineoplásicos/genética , Humanos , Animais , Camundongos , Tecnologia de Impulso Genético/métodos , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/patologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Neoplasias/genética , Neoplasias/tratamento farmacológico
3.
Nat Commun ; 15(1): 8161, 2024 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-39289368

RESUMO

Gene drives are genetic modifications designed to propagate efficiently through a population. Most applications rely on homologous recombination during sexual reproduction in diploid organisms such as insects, but we recently developed a gene drive in herpesviruses that relies on co-infection of cells by wild-type and engineered viruses. Here, we report on a viral gene drive against human herpes simplex virus 1 (HSV-1) and show that it propagates efficiently in cell culture and during HSV-1 infection in mice. We describe high levels of co-infection and gene drive-mediated recombination in neuronal tissues during herpes encephalitis as the infection progresses from the site of inoculation to the peripheral and central nervous systems. In addition, we show evidence that a superinfecting gene drive virus could recombine with wild-type viruses during latent infection. These findings indicate that HSV-1 achieves high rates of co-infection and recombination during viral infection, a phenomenon that is currently underappreciated. Overall, this study shows that a viral gene drive could spread in vivo during HSV-1 infection, paving the way toward therapeutic applications.


Assuntos
Herpes Simples , Herpesvirus Humano 1 , Animais , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiologia , Camundongos , Herpes Simples/virologia , Herpes Simples/genética , Humanos , Coinfecção/virologia , Tecnologia de Impulso Genético/métodos , Feminino , Células Vero , Chlorocebus aethiops , Encefalite por Herpes Simples/genética , Encefalite por Herpes Simples/virologia , Camundongos Endogâmicos C57BL , Recombinação Genética/genética , Genes Virais/genética
4.
Nat Commun ; 15(1): 8053, 2024 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-39277611

RESUMO

CRISPR homing gene drives can suppress pest populations by targeting female fertility genes, converting wild-type alleles into drive alleles in the germline of drive heterozygotes. fsRIDL (female-specific Release of Insects carrying a Dominant Lethal) is a self-limiting population suppression strategy involving continual release of transgenic males carrying female lethal alleles. Here, we propose an improved pest suppression system called "Release of Insects carrying a Dominant-sterile Drive" (RIDD), combining performance characteristics of homing drive and fsRIDL. We construct a split RIDD system in Drosophila melanogaster by creating a 3-gRNA drive disrupting the doublesex female exon. Drive alleles bias their inheritance in males, while drive alleles and resistance alleles formed by end-joining cause dominant female sterility. Weekly releases of RIDD males progressively suppressed and eventually eliminated cage populations. Modeling shows that RIDD is substantially stronger than SIT and fsRIDL. RIDD is also self-limiting, potentially allowing targeted population suppression.


Assuntos
Animais Geneticamente Modificados , Proteínas de Drosophila , Drosophila melanogaster , Tecnologia de Impulso Genético , Animais , Feminino , Masculino , Drosophila melanogaster/genética , Tecnologia de Impulso Genético/métodos , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Alelos , Sistemas CRISPR-Cas , Genes Dominantes , Controle Biológico de Vetores/métodos , Infertilidade/genética , Infertilidade/terapia , RNA Guia de Sistemas CRISPR-Cas/genética , Proteínas de Ligação a DNA
5.
J Insect Sci ; 24(4)2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-39162172

RESUMO

CRISPR/Cas9 manipulations are possible in many insects and ever expanding. Nonetheless, success in one species and techniques developed for it are not necessarily applicable to other species. As such, the development and expansion of CRISPR-based (clustered regularly interspaced short palindromic repeats) genome-editing tools and methodologies are dependent upon direct experimentation. One useful technique is Cas9-dependent homologous recombination, which is a critical tool for studying gene function but also for developing pest related applications like gene drive. Here, we report our attempts to induce Cas9 homology directed repair (HDR) and subsequent gene drive in Tribolium castaneum (Herbst; Insecta: Coleoptera: Tenebrionidae). Utilizing constructs containing 1 or 2 target gRNAs in combination with Cas9 under 2 different promoters and corresponding homology arms, we found a high incidence of CRISPR/Cas9 induced mutations but no evidence of homologous recombination. Even though the generated constructs provide new resources for CRISPR/Cas9 modification of the Tribolium genome, our results suggest that additional modifications and increased sample sizes will be necessary to increase the potential and detection for HDR of the Tribolium genome.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes , Tribolium , Tribolium/genética , Animais , Edição de Genes/métodos , Reparo de DNA por Recombinação , Tecnologia de Impulso Genético/métodos
6.
Nat Commun ; 15(1): 6846, 2024 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-39122734

RESUMO

CRISPR-Cas9 homing gene drives are designed to induce a targeted double-stranded DNA break at a wild type allele ('recipient'), which, when repaired by the host cell, is converted to the drive allele from the homologous ('donor') chromosome. Germline localisation of this process leads to super-Mendelian inheritance of the drive and the rapid spread of linked traits, offering a novel strategy for population control through the deliberate release of drive individuals. During the homology-based DNA repair, additional segments of the recipient chromosome may convert to match the donor, potentially impacting carrier fitness and strategy success. Using Anopheles gambiae strains with variations around the drive target site, here we assess the extent and nature of chromosomal conversion. We show both homing and meiotic drive contribute as mechanisms of inheritance bias. Additionally, over 80% of homing events resolve within 50 bp of the chromosomal break, enabling rapid gene drive transfer into locally-adapted genetic backgrounds.


Assuntos
Anopheles , Sistemas CRISPR-Cas , Tecnologia de Impulso Genético , Anopheles/genética , Animais , Tecnologia de Impulso Genético/métodos , Feminino , Alelos , Conversão Gênica , Meiose/genética , Masculino , Quebras de DNA de Cadeia Dupla , Cromossomos de Insetos/genética
7.
Gates Open Res ; 8: 14, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39035848

RESUMO

Gene drive technology has been recognized for its potential to provide durable and cost-effective solutions for previously intractable problems in public health, conservation, and agriculture. In recognition of the rapid advances in this field, in 2016 the U.S. National Academies of Sciences, Engineering, and Medicine issued a report making several recommendations aimed at researchers, funders, and policymakers for the safe and responsible research and development of gene drive technology. Subsequently, in 2017 sixteen global organizations self-identifying as sponsors and supporters of gene drive research became public signatories committed to the 'Principles for Gene Drive Research' which were inspired by the report's recommendations. Herein we reflect on the progress of gene drive research in relation to the ethical principles laid out and committed to by the signatories to the Principles. Our analysis indicates high levels of alignment with the Principles in the field of gene drive research. The manuscript also discusses the Gene Drive Research Forum, which had its genesis in the publication of the Principles. Discussions between participants at the latest meeting of the Forum point to the work that lies ahead for gene drive research in line with the Principles. Going forward the gene drive research community can productively focus on: i) safety and efficacy criteria for open release, ii) risk assessment frameworks and methods, iii) more downstream technical, regulatory and policy considerations for field evaluations and implementation, iv) continued transparency and developing mechanisms of accountability, and v) strengthening capacity in locales of potential release and expected drive spread.


Assuntos
Tecnologia de Impulso Genético , Humanos , Tecnologia de Impulso Genético/métodos , Pesquisa Biomédica/ética
9.
Malar J ; 23(1): 192, 2024 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-38898518

RESUMO

Elimination of malaria has become a United Nations member states target: Target 3.3 of the sustainable development goal no. 3 (SDG3). Despite the measures taken, the attainment of this goal is jeopardized by an alarming trend of increasing malaria case incidence. Globally, there were an estimated 241 million malaria cases in 2020 in 85 malaria-endemic countries, increasing from 227 million in 2019. Malaria case incidence was 59, which means effectively no changes in the numbers occurred, compared with the baseline 2015. Jennifer Doudna-co-inventor of CRISPR/Cas9 technology-claims that CRISPR holds the potential to lessen or even eradicate problems lying in the centre of SDGs. On the same note, CRISPR/Cas9-mediated mosquito-targeting gene drives (MGD) are perceived as a potential means to turn this trend back and put momentum into the malaria elimination effort. This paper assessed two of the critical elements of the World Health Organization Genetically modified mosquitoes (WHO GMM) Critical Pathway framework: the community and stakeholders' engagement (inability to employ widely used frameworks, segmentation of the public, 'bystander' status, and guidelines operationalization) and the regulatory landscape (lex generali, 'goldilocks dilemma', and mode of regulation) concerning mosquito-oriented gene drives (MGD) advances. Based on the assessment findings, the author believes that CRISPR/Cas-9-mediated MGD will not contribute to the attainment of SDG3 (Target 3.3), despite the undisputable technology's potential. This research pertains to the state of knowledge, legal frameworks, and legislature, as of November 2022.


Assuntos
Sistemas CRISPR-Cas , Malária , Malária/prevenção & controle , Animais , Erradicação de Doenças , Humanos , Desenvolvimento Sustentável , Participação da Comunidade , Mosquitos Vetores/genética , Tecnologia de Impulso Genético/métodos , Controle de Mosquitos , Edição de Genes
10.
Am J Trop Med Hyg ; 111(2): 341-360, 2024 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-38889708

RESUMO

Engagement is an important component in the advancement of gene-drive vector control research programs as developers look to transition the technology from the laboratory to the field. As research advances and engagement surrounding this novel technology is put into practice, knowledge can be gained from practical experiences and applications in the field. A relationship-based model (RBM) provides a framework for end-user development of engagement programs and strategies. The model places end users at the center of the engagement decision-making processes rather than as recipients of predetermined strategies, methods, and definitions. Successful RBM application for healthcare delivery has previously been demonstrated, and the University of California Malaria Initiative (UCMI) has applied this model to its gene-drive program in the Democratic Republic of São Tomé and Príncipe. The model emphasizes the importance of local leadership in the planning, development, and implementation of all phases of project engagement. The primary aim of this paper is to translate the model from paper to practice and provide a transparent description, using practical examples, of the UCMI program implementation of RBM at its field site. End-user development of the UCMI engagement program provides a unique approach to the development of ethical, transparent, and effective engagement strategies for malaria control programs. This paper may also serve as a reference and example for projects looking to establish an engagement program model that integrates end-user groups in the decision-making processes surrounding engagement.


Assuntos
Tecnologia de Impulso Genético , Malária , Humanos , Malária/prevenção & controle , Tecnologia de Impulso Genético/métodos , Animais , Controle de Mosquitos/métodos , Mosquitos Vetores/genética
11.
Nat Plants ; 10(6): 936-953, 2024 06.
Artigo em Inglês | MEDLINE | ID: mdl-38886522

RESUMO

Gene drive elements promote the spread of linked traits and can be used to change the composition or fate of wild populations. Cleave and Rescue (ClvR) drive elements sit at a fixed chromosomal position and include a DNA sequence-modifying enzyme such as Cas9/gRNAs that disrupts endogenous versions of an essential gene and a recoded version of the essential gene resistant to cleavage. ClvR spreads by creating conditions in which those lacking ClvR die because they lack functional versions of the essential gene. Here we demonstrate the essential features of the ClvR gene drive in the plant Arabidopsis thaliana through killing of gametes that fail to inherit a ClvR that targets the essential gene YKT61. Resistant alleles, which can slow or prevent drive, were not observed. Modelling shows plant ClvRs are robust to certain failure modes and can be used to rapidly drive population modification or suppression. Possible applications are discussed.


Assuntos
Arabidopsis , Tecnologia de Impulso Genético , Arabidopsis/genética , Tecnologia de Impulso Genético/métodos , Células Germinativas Vegetais , Genes de Plantas , Sistemas CRISPR-Cas , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Alelos
12.
Nat Commun ; 15(1): 4983, 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38862555

RESUMO

Engineered sex ratio distorters (SRDs) have been proposed as a powerful component of genetic control strategies designed to suppress harmful insect pests. Two types of CRISPR-based SRD mechanisms have been proposed: X-shredding, which eliminates X-bearing sperm, and X-poisoning, which eliminates females inheriting disrupted X-chromosomes. These differences can have a profound impact on the population dynamics of SRDs when linked to the Y-chromosome: an X-shredder is invasive, constituting a classical meiotic Y-drive, whereas X-poisoning is self-limiting, unable to invade but also insulated from selection. Here, we establish X-poisoning strains in the malaria vector Anopheles gambiae targeting three X-linked genes during spermatogenesis, resulting in male bias. We find that sex distortion is primarily driven by a loss of X-bearing sperm, with limited evidence for postzygotic lethality of female progeny. By leveraging a Drosophila melanogaster model, we show unambiguously that engineered SRD traits can operate differently in these two insects. Unlike X-shredding, X-poisoning could theoretically operate at early stages of spermatogenesis. We therefore explore premeiotic Cas9 expression to target the mosquito X-chromosome. We find that, by pre-empting the onset of meiotic sex chromosome inactivation, this approach may enable the development of Y-linked SRDs if mutagenesis of spermatogenesis-essential genes is functionally balanced.


Assuntos
Anopheles , Drosophila melanogaster , Tecnologia de Impulso Genético , Razão de Masculinidade , Espermatogênese , Cromossomo X , Animais , Masculino , Feminino , Anopheles/genética , Cromossomo X/genética , Drosophila melanogaster/genética , Tecnologia de Impulso Genético/métodos , Espermatogênese/genética , Mosquitos Vetores/genética , Genes Ligados ao Cromossomo X , Sistemas CRISPR-Cas , Espermatozoides/metabolismo , Animais Geneticamente Modificados
13.
Malar J ; 23(1): 156, 2024 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-38773487

RESUMO

Sustainable reductions in African malaria transmission require innovative tools for mosquito control. One proposal involves the use of low-threshold gene drive in Anopheles vector species, where a 'causal pathway' would be initiated by (i) the release of a gene drive system in target mosquito vector species, leading to (ii) its transmission to subsequent generations, (iii) its increase in frequency and spread in target mosquito populations, (iv) its simultaneous propagation of a linked genetic trait aimed at reducing vectorial capacity for Plasmodium, and (v) reduced vectorial capacity for parasites in target mosquito populations as the gene drive system reaches fixation in target mosquito populations, causing (vi) decreased malaria incidence and prevalence. Here the scope, objectives, trial design elements, and approaches to monitoring for initial field releases of such gene dive systems are considered, informed by the successful implementation of field trials of biological control agents, as well as other vector control tools, including insecticides, Wolbachia, larvicides, and attractive-toxic sugar bait systems. Specific research questions to be addressed in initial gene drive field trials are identified, and adaptive trial design is explored as a potentially constructive and flexible approach to facilitate testing of the causal pathway. A fundamental question for decision-makers for the first field trials will be whether there should be a selective focus on earlier points of the pathway, such as genetic efficacy via measurement of the increase in frequency and spread of the gene drive system in target populations, or on wider interrogation of the entire pathway including entomological and epidemiological efficacy. How and when epidemiological efficacy will eventually be assessed will be an essential consideration before decisions on any field trial protocols are finalized and implemented, regardless of whether initial field trials focus exclusively on the measurement of genetic efficacy, or on broader aspects of the causal pathway. Statistical and modelling tools are currently under active development and will inform such decisions on initial trial design, locations, and endpoints. Collectively, the considerations here advance the realization of developer ambitions for the first field trials of low-threshold gene drive for malaria vector control within the next 5 years.


Assuntos
Anopheles , Tecnologia de Impulso Genético , Malária , Controle de Mosquitos , Mosquitos Vetores , Controle de Mosquitos/métodos , Mosquitos Vetores/genética , Malária/prevenção & controle , Malária/transmissão , Animais , Anopheles/genética , Tecnologia de Impulso Genético/métodos
14.
PLoS Comput Biol ; 20(5): e1012046, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38709820

RESUMO

Genetic surveillance of mosquito populations is becoming increasingly relevant as genetics-based mosquito control strategies advance from laboratory to field testing. Especially applicable are mosquito gene drive projects, the potential scale of which leads monitoring to be a significant cost driver. For these projects, monitoring will be required to detect unintended spread of gene drive mosquitoes beyond field sites, and the emergence of alternative alleles, such as drive-resistant alleles or non-functional effector genes, within intervention sites. This entails the need to distribute mosquito traps efficiently such that an allele of interest is detected as quickly as possible-ideally when remediation is still viable. Additionally, insecticide-based tools such as bednets are compromised by insecticide-resistance alleles for which there is also a need to detect as quickly as possible. To this end, we present MGSurvE (Mosquito Gene SurveillancE): a computational framework that optimizes trap placement for genetic surveillance of mosquito populations such that the time to detection of an allele of interest is minimized. A key strength of MGSurvE is that it allows important biological features of mosquitoes and the landscapes they inhabit to be accounted for, namely: i) resources required by mosquitoes (e.g., food sources and aquatic breeding sites) can be explicitly distributed through a landscape, ii) movement of mosquitoes may depend on their sex, the current state of their gonotrophic cycle (if female) and resource attractiveness, and iii) traps may differ in their attractiveness profile. Example MGSurvE analyses are presented to demonstrate optimal trap placement for: i) an Aedes aegypti population in a suburban landscape in Queensland, Australia, and ii) an Anopheles gambiae population on the island of São Tomé, São Tomé and Príncipe. Further documentation and use examples are provided in project's documentation. MGSurvE is intended as a resource for both field and computational researchers interested in mosquito gene surveillance.


Assuntos
Controle de Mosquitos , Animais , Controle de Mosquitos/métodos , Culicidae/genética , Culicidae/fisiologia , Biologia Computacional/métodos , Tecnologia de Impulso Genético/métodos , Mosquitos Vetores/genética , Aedes/genética , Resistência a Inseticidas/genética , Feminino
15.
PLoS Comput Biol ; 20(5): e1012133, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38805562

RESUMO

Novel mosquito genetic control tools, such as CRISPR-based gene drives, hold great promise in reducing the global burden of vector-borne diseases. As these technologies advance through the research and development pipeline, there is a growing need for modeling frameworks incorporating increasing levels of entomological and epidemiological detail in order to address questions regarding logistics and biosafety. Epidemiological predictions are becoming increasingly relevant to the development of target product profiles and the design of field trials and interventions, while entomological surveillance is becoming increasingly important to regulation and biosafety. We present MGDrivE 3 (Mosquito Gene Drive Explorer 3), a new version of a previously-developed framework, MGDrivE 2, that investigates the spatial population dynamics of mosquito genetic control systems and their epidemiological implications. The new framework incorporates three major developments: i) a decoupled sampling algorithm allowing the vector portion of the MGDrivE framework to be paired with a more detailed epidemiological framework, ii) a version of the Imperial College London malaria transmission model, which incorporates age structure, various forms of immunity, and human and vector interventions, and iii) a surveillance module that tracks mosquitoes captured by traps throughout the simulation. Example MGDrivE 3 simulations are presented demonstrating the application of the framework to a CRISPR-based homing gene drive linked to dual disease-refractory genes and their potential to interrupt local malaria transmission. Simulations are also presented demonstrating surveillance of such a system by a network of mosquito traps. MGDrivE 3 is freely available as an open-source R package on CRAN (https://cran.r-project.org/package=MGDrivE2) (version 2.1.0), and extensive examples and vignettes are provided. We intend the software to aid in understanding of human health impacts and biosafety of mosquito genetic control tools, and continue to iterate per feedback from the genetic control community.


Assuntos
Simulação por Computador , Tecnologia de Impulso Genético , Malária , Controle de Mosquitos , Mosquitos Vetores , Animais , Humanos , Mosquitos Vetores/genética , Controle de Mosquitos/métodos , Malária/epidemiologia , Malária/transmissão , Malária/prevenção & controle , Tecnologia de Impulso Genético/métodos , Biologia Computacional/métodos , Culicidae/genética , Algoritmos , Doenças Transmitidas por Vetores/transmissão , Doenças Transmitidas por Vetores/epidemiologia , Doenças Transmitidas por Vetores/prevenção & controle , Dinâmica Populacional
16.
Nat Commun ; 15(1): 4560, 2024 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-38811556

RESUMO

Gene drive systems could be a viable strategy to prevent pathogen transmission or suppress vector populations by propagating drive alleles with super-Mendelian inheritance. CRISPR-based homing gene drives convert wild type alleles into drive alleles in heterozygotes with Cas9 and gRNA. It is thus desirable to identify Cas9 promoters that yield high drive conversion rates, minimize the formation rate of resistance alleles in both the germline and the early embryo, and limit somatic Cas9 expression. In Drosophila, the nanos promoter avoids leaky somatic expression, but at the cost of high embryo resistance from maternally deposited Cas9. To improve drive efficiency, we test eleven Drosophila melanogaster germline promoters. Some achieve higher drive conversion efficiency with minimal embryo resistance, but none completely avoid somatic expression. However, such somatic expression often does not carry detectable fitness costs for a rescue homing drive targeting a haplolethal gene, suggesting somatic drive conversion. Supporting a 4-gRNA suppression drive, one promoter leads to a low drive equilibrium frequency due to fitness costs from somatic expression, but the other outperforms nanos, resulting in successful suppression of the cage population. Overall, these Cas9 promoters hold advantages for homing drives in Drosophila species and may possess valuable homologs in other organisms.


Assuntos
Sistemas CRISPR-Cas , Proteínas de Drosophila , Drosophila melanogaster , Tecnologia de Impulso Genético , Células Germinativas , Regiões Promotoras Genéticas , RNA Guia de Sistemas CRISPR-Cas , Animais , Regiões Promotoras Genéticas/genética , Drosophila melanogaster/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Tecnologia de Impulso Genético/métodos , Células Germinativas/metabolismo , RNA Guia de Sistemas CRISPR-Cas/genética , Animais Geneticamente Modificados , Proteína 9 Associada à CRISPR/metabolismo , Proteína 9 Associada à CRISPR/genética , Alelos , Feminino , Masculino , Proteínas de Ligação a RNA
17.
PLoS Genet ; 20(5): e1011262, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38753875

RESUMO

Engineered gene-drive techniques for population modification and/or suppression have the potential for tackling complex challenges, including reducing the spread of diseases and invasive species. Gene-drive systems with low threshold frequencies for invasion, such as homing-based gene drive, require initially few transgenic individuals to spread and are therefore easy to introduce. The self-propelled behavior of such drives presents a double-edged sword, however, as the low threshold can allow transgenic elements to expand beyond a target population. By contrast, systems where a high threshold frequency must be reached before alleles can spread-above a fitness valley-are less susceptible to spillover but require introduction at a high frequency. We model a proposed drive system, called "daisy quorum drive," that transitions over time from a low-threshold daisy-chain system (involving homing-based gene drive such as CRISPR-Cas9) to a high-threshold fitness-valley system (requiring a high frequency-a "quorum"-to spread). The daisy-chain construct temporarily lowers the high thresholds required for spread of the fitness-valley construct, facilitating use in a wide variety of species that are challenging to breed and release in large numbers. Because elements in the daisy chain only drive subsequent elements in the chain and not themselves and also carry deleterious alleles ("drive load"), the daisy chain is expected to exhaust itself, removing all CRISPR elements and leaving only the high-threshold fitness-valley construct, whose spread is more spatially restricted. Developing and analyzing both discrete patch and continuous space models, we explore how various attributes of daisy quorum drive affect the chance of modifying local population characteristics and the risk that transgenic elements expand beyond a target area. We also briefly explore daisy quorum drive when population suppression is the goal. We find that daisy quorum drive can provide a promising bridge between gene-drive and fitness-valley constructs, allowing spread from a low frequency in the short term and better containment in the long term, without requiring repeated introductions or persistence of CRISPR elements.


Assuntos
Sistemas CRISPR-Cas , Tecnologia de Impulso Genético , Tecnologia de Impulso Genético/métodos , Modelos Genéticos , Aptidão Genética , Alelos , Engenharia Genética/métodos , Animais
18.
J Genet Genomics ; 51(8): 836-843, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38599514

RESUMO

CRISPR homing gene drives have considerable potential for managing populations of medically and agriculturally significant insects. They operate by Cas9 cleavage followed by homology-directed repair, copying the drive allele to the wild-type chromosome and thus increasing in frequency and spreading throughout a population. However, resistance alleles formed by end-joining repair pose a significant obstacle. To address this, we create a homing drive targeting the essential hairy gene in Drosophila melanogaster. Nonfunctional resistance alleles are recessive lethal, while drive carriers have a recoded "rescue" version of hairy. The drive inheritance rate is moderate, and multigenerational cage studies show drive spread to 96%-97% of the population. However, the drive does not reach 100% due to the formation of functional resistance alleles despite using four gRNAs. These alleles have a large deletion but likely utilize an alternate start codon. Thus, revised designs targeting more essential regions of a gene may be necessary to avoid such functional resistance. Replacement of the rescue element's native 3' UTR with a homolog from another species increases drive inheritance by 13%-24%. This was possibly because of reduced homology between the rescue element and surrounding genomic DNA, which could also be an important design consideration for rescue gene drives.


Assuntos
Sistemas CRISPR-Cas , Drosophila melanogaster , Tecnologia de Impulso Genético , RNA Guia de Sistemas CRISPR-Cas , Animais , Tecnologia de Impulso Genético/métodos , Drosophila melanogaster/genética , RNA Guia de Sistemas CRISPR-Cas/genética , Sistemas CRISPR-Cas/genética , Alelos , Proteínas de Drosophila/genética , Edição de Genes
19.
Heredity (Edinb) ; 132(5): 232-246, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38494530

RESUMO

Indoor insecticide applications are the primary tool for reducing malaria transmission in the Solomon Archipelago, a region where Anopheles farauti is the only common malaria vector. Due to the evolution of behavioural resistance in some An. farauti populations, these applications have become less effective. New malaria control interventions are therefore needed in this region, and gene-drives provide a promising new technology. In considering developing a population-specific (local) gene-drive in An. farauti, we detail the species' population genetic structure using microsatellites and whole mitogenomes, finding many spatially confined populations both within and between landmasses. This strong population structure suggests that An. farauti would be a useful system for developing a population-specific, confinable gene-drive for field release, where private alleles can be used as Cas9 targets. Previous work on Anopheles gambiae has used the Cardinal gene for the development of a global population replacement gene-drive. We therefore also analyse the Cardinal gene to assess whether it may be a suitable target to engineer a gene-drive for the modification of local An. farauti populations. Despite the extensive population structure observed in An. farauti for microsatellites, only one remote island population from Vanuatu contained fixed and private alleles at the Cardinal locus. Nonetheless, this study provides an initial framework for further population genomic investigations to discover high-frequency private allele targets in localized An. farauti populations. This would enable the development of gene-drive strains for modifying localised populations with minimal chance of escape and may provide a low-risk route to field trial evaluations.


Assuntos
Anopheles , Tecnologia de Impulso Genético , Genética Populacional , Malária , Repetições de Microssatélites , Mosquitos Vetores , Anopheles/genética , Animais , Mosquitos Vetores/genética , Malária/transmissão , Tecnologia de Impulso Genético/métodos , Melanesia , Alelos
20.
Public Underst Sci ; 33(7): 884-901, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-38469856

RESUMO

Gene drive could be a powerful tool for addressing problems of conservation, agriculture, and human health caused by insect and animal pests but is likely to be controversial as it involves the release of genetically modified organisms. This study examined the social determinants of opinion of gene drive. We asked a representative sample of the U.S. public to respond to a description of a hypothetical application of a gene-drive mosquito to the problem of malaria and examined the relationship of these responses with demographic and ideological beliefs. We found strong general approval for the use of gene-drive mosquitos to address malaria, coinciding with the concern about a possible environmental impact of modified mosquitos and that gene drives represent "too much power over nature." Among the determinants we measured, respondent acceptance of scientism and trust that scientists are advancing the public's interest were the greatest predictors of views of gene drive.


Assuntos
Tecnologia de Impulso Genético , Opinião Pública , Confiança , Animais , Tecnologia de Impulso Genético/métodos , Estados Unidos , Culicidae/genética , Humanos , Malária
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA