51
|
Oberhofer G, Ivy T, Hay BA. Split versions of Cleave and Rescue selfish genetic elements for measured self limiting gene drive. PLoS Genet 2021; 17:e1009385. [PMID: 33600432 PMCID: PMC7951863 DOI: 10.1371/journal.pgen.1009385] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 03/11/2021] [Accepted: 01/28/2021] [Indexed: 12/26/2022] Open
Abstract
Gene drive elements promote the spread of linked traits, providing methods for changing the composition or fate of wild populations. Drive mechanisms that are self-limiting are attractive because they allow control over the duration and extent of trait spread in time and space, and are reversible through natural selection as drive wanes. Self-sustaining Cleave and Rescue (ClvR) elements include a DNA sequence-modifying enzyme such as Cas9/gRNAs that disrupts endogenous versions of an essential gene, a tightly linked recoded version of the essential gene resistant to cleavage (the Rescue), and a Cargo. ClvR spreads by creating loss-of-function (LOF) conditions in which those without ClvR die because they lack functional copies of the essential gene. We use modeling to show that when the Rescue-Cargo and one or both components required for LOF allele creation (Cas9 and gRNA) reside at different locations (split ClvR), drive of Rescue-Cargo is self-limiting due to a progressive decrease in Cas9 frequency, and thus opportunities for creation of LOF alleles, as spread occurs. Importantly, drive strength and duration can be extended in a measured manner-which is still self-limiting-by moving the two components close enough to each other that they experience some degree of linkage. With linkage, Cas9 transiently experiences drive by hitchhiking with Rescue-Cargo until linkage disequilibrium between the two disappears, a function of recombination frequency and number of generations, creating a novel point of control. We implement split ClvR in Drosophila, with key elements on different chromosomes. Cargo/Rescue/gRNAs spreads to high frequency in a Cas9-dependent manner, while the frequency of Cas9 decreases. These observations show that measured, transient drive, coupled with a loss of future drive potential, can be achieved using the simple toolkit that make up ClvR elements-Cas9 and gRNAs and a Rescue/Cargo.
Collapse
Affiliation(s)
- Georg Oberhofer
- California Institute of Technology, Pasadena, California, United States of America
| | - Tobin Ivy
- California Institute of Technology, Pasadena, California, United States of America
| | - Bruce A. Hay
- California Institute of Technology, Pasadena, California, United States of America
| |
Collapse
|
52
|
Shults P, Cohnstaedt LW, Adelman ZN, Brelsfoard C. Next-generation tools to control biting midge populations and reduce pathogen transmission. Parasit Vectors 2021; 14:31. [PMID: 33413518 PMCID: PMC7788963 DOI: 10.1186/s13071-020-04524-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 12/05/2020] [Indexed: 02/06/2023] Open
Abstract
Biting midges of the genus Culicoides transmit disease-causing agents resulting in a significant economic impact on livestock industries in many parts of the world. Localized control efforts, such as removal of larval habitat or pesticide application, can be logistically difficult, expensive and ineffective if not instituted and maintained properly. With these limitations, a population-level approach to the management of Culicoides midges should be investigated as a means to replace or supplement existing control strategies. Next-generation control methods such as Wolbachia- and genetic-based population suppression and replacement are being investigated in several vector species. Here we assess the feasibility and applicability of these approaches for use against biting midges. We also discuss the technical and logistical hurdles needing to be addressed for each method to be successful, as well as emphasize the importance of addressing community engagement and involving stakeholders in the investigation and development of these approaches.
Collapse
Affiliation(s)
- Phillip Shults
- Texas A&M University, 370 Olsen Blvd, College Station, TX, 77843, USA.
| | - Lee W Cohnstaedt
- USDA-ARS Arthropod Borne Animal Disease Research Unit, 1515 College Ave, Manhattan, KS, 66502, USA
| | - Zach N Adelman
- Texas A&M University, 370 Olsen Blvd, College Station, TX, 77843, USA
| | | |
Collapse
|
53
|
Hay BA, Oberhofer G, Guo M. Engineering the Composition and Fate of Wild Populations with Gene Drive. ANNUAL REVIEW OF ENTOMOLOGY 2021; 66:407-434. [PMID: 33035437 DOI: 10.1146/annurev-ento-020117-043154] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Insects play important roles as predators, prey, pollinators, recyclers, hosts, parasitoids, and sources of economically important products. They can also destroy crops; wound animals; and serve as vectors for plant, animal, and human diseases. Gene drive-a process by which genes, gene complexes, or chromosomes encoding specific traits are made to spread through wild populations, even if these traits result in a fitness cost to carriers-provides new opportunities for altering populations to benefit humanity and the environment in ways that are species specific and sustainable. Gene drive can be used to alter the genetic composition of an existing population, referred to as population modification or replacement, or to bring about population suppression or elimination. We describe technologies under consideration, progress that has been made, and remaining technological hurdles, particularly with respect to evolutionary stability and our ability to control the spread and ultimate fate of genes introduced into populations.
Collapse
Affiliation(s)
- Bruce A Hay
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA; ,
- St. John's College, University of Cambridge, Cambridge CB2 1TP, United Kingdom
| | - Georg Oberhofer
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA; ,
| | - Ming Guo
- Departments of Neurology and Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California 90095, USA;
| |
Collapse
|
54
|
femaleless Controls Sex Determination and Dosage Compensation Pathways in Females of Anopheles Mosquitoes. Curr Biol 2021; 31:1084-1091.e4. [PMID: 33417880 PMCID: PMC7955153 DOI: 10.1016/j.cub.2020.12.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 11/09/2020] [Accepted: 12/11/2020] [Indexed: 01/04/2023]
Abstract
The insect sex determination and the intimately linked dosage compensation pathways represent a challenging evolutionary puzzle that has been solved only in Drosophila melanogaster. Analyses of orthologs of the Drosophila genes identified in non-drosophilid taxa1,2 revealed that evolution of sex determination pathways is consistent with a bottom-up mode,3 where only the terminal genes within the pathway are well conserved. doublesex (dsx), occupying a bottom-most position and encoding sex-specific proteins orchestrating downstream sexual differentiation processes, is an ancient sex-determining gene present in all studied species.2,4,5 With the exception of lepidopterans, its female-specific splicing is known to be regulated by transformer (tra) and its co-factor transformer-2 (tra2).6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 Here we show that in the African malaria mosquito Anopheles gambiae, a gene, which likely arose in the Anopheles lineage and which we call femaleless (fle), controls sex determination in females by regulating splicing of dsx and fruitless (fru; another terminal gene within a branch of the sex determination pathway). Moreover, fle represents a novel molecular link between the sex determination and dosage compensation pathways. It is necessary to suppress activation of dosage compensation in females, as demonstrated by the significant upregulation of the female X chromosome genes and a correlated female-specific lethality, but no negative effect on males, in response to fle knockdown. This unexpected property, combined with a high level of conservation in sequence and function in anopheline mosquitoes, makes fle an excellent target for genetic control of all major vectors of human malaria. fle is a new sex determination pathway element conserved in Anopheles mosquitoes fle may have originated in the Anopheles lineage and is highly conserved in Anopheles fle suppresses activation of dosage compensation in females Depletion of fle transcripts is lethal or otherwise deleterious to females
Collapse
|
55
|
Iijima N, Sato K, Kuranaga E, Umetsu D. Differential cell adhesion implemented by Drosophila Toll corrects local distortions of the anterior-posterior compartment boundary. Nat Commun 2020; 11:6320. [PMID: 33303753 PMCID: PMC7729853 DOI: 10.1038/s41467-020-20118-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 11/16/2020] [Indexed: 11/25/2022] Open
Abstract
Maintaining lineage restriction boundaries in proliferating tissues is vital to animal development. A long-standing thermodynamics theory, the differential adhesion hypothesis, attributes cell sorting phenomena to differentially expressed adhesion molecules. However, the contribution of the differential adhesion system during tissue morphogenesis has been unsubstantiated despite substantial theoretical support. Here, we report that Toll-1, a transmembrane receptor protein, acts as a differentially expressed adhesion molecule that straightens the fluctuating anteroposterior compartment boundary in the abdominal epidermal epithelium of the Drosophila pupa. Toll-1 is expressed across the entire posterior compartment under the control of the selector gene engrailed and displays a sharp expression boundary that coincides with the compartment boundary. Toll-1 corrects local distortions of the boundary in the absence of cable-like Myosin II enrichment along the boundary. The reinforced adhesion of homotypic cell contacts, together with pulsed cell contraction, achieves a biased vertex sliding action by resisting the separation of homotypic cell contacts in boundary cells. This work reveals a self-organizing system that integrates a differential adhesion system with pulsed contraction of cells to maintain lineage restriction boundaries.
Collapse
Affiliation(s)
- Norihiro Iijima
- Laboratory for Histogenetic Dynamics, Graduate School of Life Sciences, Tohoku University, Sendai, 980-8578, Japan
| | - Katsuhiko Sato
- Research Institute for Electronic Science, Hokkaido University, Sapporo, 001-0020, Japan
- Global Station for Soft Matter, Global Institution for Collaborative Research and Education, Hokkaido University, Sapporo, 001-0020, Japan
| | - Erina Kuranaga
- Laboratory for Histogenetic Dynamics, Graduate School of Life Sciences, Tohoku University, Sendai, 980-8578, Japan.
| | - Daiki Umetsu
- Laboratory for Histogenetic Dynamics, Graduate School of Life Sciences, Tohoku University, Sendai, 980-8578, Japan.
| |
Collapse
|
56
|
Öztürk-Çolak A, Inami S, Buchler JR, McClanahan PD, Cruz A, Fang-Yen C, Koh K. Sleep Induction by Mechanosensory Stimulation in Drosophila. Cell Rep 2020; 33:108462. [PMID: 33264620 PMCID: PMC7735403 DOI: 10.1016/j.celrep.2020.108462] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 09/11/2020] [Accepted: 11/10/2020] [Indexed: 12/11/2022] Open
Abstract
People tend to fall asleep when gently rocked or vibrated. Experimental studies have shown that rocking promotes sleep in humans and mice. However, the mechanisms underlying the phenomenon are not well understood. A habituation model proposes that habituation, a form of non-associative learning, mediates sleep induction by monotonous stimulation. Here, we show that gentle vibration promotes sleep in Drosophila in part through habituation. Vibration-induced sleep (VIS) leads to increased homeostatic sleep credit and reduced arousability, and can be suppressed by heightened arousal or reduced GABA signaling. Multiple mechanosensory organs mediate VIS, and the magnitude of VIS depends on vibration frequency and genetic background. Sleep induction improves over successive blocks of vibration. Furthermore, training with continuous vibration does not generalize to intermittent vibration, demonstrating stimulus specificity, a characteristic of habituation. Our findings suggest that habituation plays a significant role in sleep induction by vibration. Öztürk-Çolak et al. demonstrate that gentle vibration induces sleep in Drosophila. The authors show that sleep induction improves over multiple vibration sessions, which suggests that habituation, a form of simple learning, plays a significant role in vibration-induced sleep.
Collapse
Affiliation(s)
- Arzu Öztürk-Çolak
- Department of Neuroscience, Jefferson Center for Synaptic Biology, and the Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19106, USA
| | - Sho Inami
- Department of Neuroscience, Jefferson Center for Synaptic Biology, and the Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19106, USA
| | - Joseph R Buchler
- Department of Neuroscience, Jefferson Center for Synaptic Biology, and the Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19106, USA
| | - Patrick D McClanahan
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andri Cruz
- Department of Neuroscience, Jefferson Center for Synaptic Biology, and the Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19106, USA
| | - Christopher Fang-Yen
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kyunghee Koh
- Department of Neuroscience, Jefferson Center for Synaptic Biology, and the Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19106, USA.
| |
Collapse
|
57
|
Morris O, Deng H, Tam C, Jasper H. Warburg-like Metabolic Reprogramming in Aging Intestinal Stem Cells Contributes to Tissue Hyperplasia. Cell Rep 2020; 33:108423. [PMID: 33238124 PMCID: PMC8011352 DOI: 10.1016/j.celrep.2020.108423] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 09/28/2020] [Accepted: 11/02/2020] [Indexed: 12/28/2022] Open
Abstract
In many tissues, stem cell (SC) proliferation is dynamically adjusted to regenerative needs. How SCs adapt their metabolism to meet the demands of proliferation and how changes in such adaptive mechanisms contribute to age-related dysfunction remain poorly understood. Here, we identify mitochondrial Ca2+ uptake as a central coordinator of SC metabolism. Live imaging of genetically encoded metabolite sensors in intestinal SCs (ISCs) of Drosophila reveals that mitochondrial Ca2+ uptake transiently adapts electron transport chain flux to match energetic demand upon proliferative activation. This tight metabolic adaptation is lost in ISCs of old flies, as declines in mitochondrial Ca2+ uptake promote a "Warburg-like" metabolic reprogramming toward aerobic glycolysis. This switch mimics metabolic reprogramming by the oncogene RasV12 and enhances ISC hyperplasia. Our data identify a critical mechanism for metabolic adaptation of tissue SCs and reveal how its decline sets aging SCs on a metabolic trajectory reminiscent of that seen upon oncogenic transformation.
Collapse
Affiliation(s)
- Otto Morris
- Immunology Discovery, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Hansong Deng
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 20092, China; Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945-1400, USA
| | - Christine Tam
- Department of Biomolecular Resources, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Heinrich Jasper
- Immunology Discovery, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA; Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945-1400, USA.
| |
Collapse
|
58
|
Abstract
In life's constant battle for survival, it takes one to kill but two to conquer. Toxin-antitoxin or toxin-antidote (TA) elements are genetic dyads that cheat the laws of inheritance to guarantee their transmission to the next generation. This seemingly simple genetic arrangement—a toxin linked to its antidote—is capable of quickly spreading and persisting in natural populations. TA elements were first discovered in bacterial plasmids in the 1980s and have recently been characterized in fungi, plants, and animals, where they underlie genetic incompatibilities and sterility in crosses between wild isolates. In this review, we provide a unified view of TA elements in both prokaryotic and eukaryotic organisms and highlight their similarities and differences at the evolutionary, genetic, and molecular levels. Finally, we propose several scenarios that could explain the paradox of the evolutionary origin of TA elements and argue that these elements may be key evolutionary players and that the full scope of their roles is only beginning to be uncovered.
Collapse
Affiliation(s)
- Alejandro Burga
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), 1030 Vienna, Austria
| | - Eyal Ben-David
- Department of Human Genetics, Department of Biological Chemistry, and Howard Hughes Medical Institute, University of California, Los Angeles, California 90095, USA
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada, The Hebrew University School of Medicine, Jerusalem 91120, Israel
| | - Leonid Kruglyak
- Department of Human Genetics, Department of Biological Chemistry, and Howard Hughes Medical Institute, University of California, Los Angeles, California 90095, USA
| |
Collapse
|
59
|
EFSA Panel on Genetically Modified Organisms (GMO), Naegeli H, Bresson J, Dalmay T, Dewhurst IC, Epstein MM, Guerche P, Hejatko J, Moreno FJ, Mullins E, Nogué F, Rostoks N, Sánchez Serrano JJ, Savoini G, Veromann E, Veronesi F, Bonsall MB, Mumford J, Wimmer EA, Devos Y, Paraskevopoulos K, Firbank LG. Adequacy and sufficiency evaluation of existing EFSA guidelines for the molecular characterisation, environmental risk assessment and post-market environmental monitoring of genetically modified insects containing engineered gene drives. EFSA J 2020; 18:e06297. [PMID: 33209154 PMCID: PMC7658669 DOI: 10.2903/j.efsa.2020.6297] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Advances in molecular and synthetic biology are enabling the engineering of gene drives in insects for disease vector/pest control. Engineered gene drives (that bias their own inheritance) can be designed either to suppress interbreeding target populations or modify them with a new genotype. Depending on the engineered gene drive system, theoretically, a genetic modification of interest could spread through target populations and persist indefinitely, or be restricted in its spread or persistence. While research on engineered gene drives and their applications in insects is advancing at a fast pace, it will take several years for technological developments to move to practical applications for deliberate release into the environment. Some gene drive modified insects (GDMIs) have been tested experimentally in the laboratory, but none has been assessed in small-scale confined field trials or in open release trials as yet. There is concern that the deliberate release of GDMIs in the environment may have possible irreversible and unintended consequences. As a proactive measure, the European Food Safety Authority (EFSA) has been requested by the European Commission to review whether its previously published guidelines for the risk assessment of genetically modified animals (EFSA, 2012 and 2013), including insects (GMIs), are adequate and sufficient for GDMIs, primarily disease vectors, agricultural pests and invasive species, for deliberate release into the environment. Under this mandate, EFSA was not requested to develop risk assessment guidelines for GDMIs. In this Scientific Opinion, the Panel on Genetically Modified Organisms (GMO) concludes that EFSA's guidelines are adequate, but insufficient for the molecular characterisation (MC), environmental risk assessment (ERA) and post-market environmental monitoring (PMEM) of GDMIs. While the MC,ERA and PMEM of GDMIs can build on the existing risk assessment framework for GMIs that do not contain engineered gene drives, there are specific areas where further guidance is needed for GDMIs.
Collapse
|
60
|
Dhole S, Lloyd AL, Gould F. Gene Drive Dynamics in Natural Populations: The Importance of Density Dependence, Space, and Sex. ANNUAL REVIEW OF ECOLOGY, EVOLUTION, AND SYSTEMATICS 2020; 51:505-531. [PMID: 34366722 PMCID: PMC8340601 DOI: 10.1146/annurev-ecolsys-031120-101013] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The spread of synthetic gene drives is often discussed in the context of panmictic populations connected by gene flow and described with simple deterministic models. Under such assumptions, an entire species could be altered by releasing a single individual carrying an invasive gene drive, such as a standard homing drive. While this remains a theoretical possibility, gene drive spread in natural populations is more complex and merits a more realistic assessment. The fate of any gene drive released in a population would be inextricably linked to the population's ecology. Given the uncertainty often involved in ecological assessment of natural populations, understanding the sensitivity of gene drive spread to important ecological factors is critical. Here we review how different forms of density dependence, spatial heterogeneity, and mating behaviors can impact the spread of self-sustaining gene drives. We highlight specific aspects of gene drive dynamics and the target populations that need further research.
Collapse
Affiliation(s)
- Sumit Dhole
- Department of Entomology and Plant Pathology, North Carolina State University, Raleigh, North Carolina 27695, USA
| | - Alun L Lloyd
- Biomathematics Graduate Program and Department of Mathematics, North Carolina State University, Raleigh, North Carolina 27695-8213, USA
- Genetic Engineering and Society Center, North Carolina State University, Raleigh, North Carolina 27695-7565, USA
| | - Fred Gould
- Department of Entomology and Plant Pathology, North Carolina State University, Raleigh, North Carolina 27695, USA
- Genetic Engineering and Society Center, North Carolina State University, Raleigh, North Carolina 27695-7565, USA
| |
Collapse
|
61
|
The Antiviral Small-Interfering RNA Pathway Induces Zika Virus Resistance in Transgenic Aedes aegypti. Viruses 2020; 12:v12111231. [PMID: 33142991 PMCID: PMC7692394 DOI: 10.3390/v12111231] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/27/2020] [Accepted: 10/28/2020] [Indexed: 01/21/2023] Open
Abstract
The resurgence of arbovirus outbreaks across the globe, including the recent Zika virus (ZIKV) epidemic in 2015–2016, emphasizes the need for innovative vector control methods. In this study, we investigated ZIKV susceptibility to transgenic Aedes aegypti engineered to target the virus by means of the antiviral small-interfering RNA (siRNA) pathway. The robustness of antiviral effector expression in transgenic mosquitoes is strongly influenced by the genomic insertion locus and transgene copy number; we therefore used CRISPR/Cas9 to re-target a previously characterized locus (Chr2:321382225) and engineered mosquitoes expressing an inverted repeat (IR) dsRNA against the NS3/4A region of the ZIKV genome. Small RNA analysis revealed that the IR effector triggered the mosquito’s siRNA antiviral pathway in bloodfed females. Nearly complete (90%) inhibition of ZIKV replication was found in vivo in both midguts and carcasses at 7 or 14 days post-infection (dpi). Furthermore, significantly fewer transgenic mosquitoes contained ZIKV in their salivary glands (p = 0.001), which led to a reduction in the number of ZIKV-containing saliva samples as measured by transmission assay. Our work shows that Ae. aegypti innate immunity can be co-opted to engineer mosquitoes resistant to ZIKV.
Collapse
|
62
|
Chang LW, Tseng IC, Wang LH, Sun YH. Isoform-specific functions of an evolutionarily conserved 3 bp micro-exon alternatively spliced from another exon in Drosophila homothorax gene. Sci Rep 2020; 10:12783. [PMID: 32732884 PMCID: PMC7392893 DOI: 10.1038/s41598-020-69644-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 07/14/2020] [Indexed: 12/03/2022] Open
Abstract
Micro-exons are exons of very small size (usually 3–30 nts). Some micro-exons are alternatively spliced. Their functions, regulation and evolution are largely unknown. Here, we present an example of an alternatively spliced 3 bp micro-exon (micro-Ex8) in the homothorax (hth) gene in Drosophila. Hth is involved in many developmental processes. It contains a MH domain and a TALE-class homeodomain (HD). It binds to another homeodomain Exd via its MH domain to promote the nuclear import of the Hth-Exd complex and serve as a cofactor for Hox proteins. The MH and HD domains in Hth as well as the HTh-Exd interaction are highly conserved in evolution. The alternatively spliced micro-exon lies between the exons encoding the MH and HD domains. We provide clear proof that the micro-Ex8 is produced by alternative splicing from a 48 bp full-length exon 8 (FL-Ex8) and the micro-Ex8 is the first three nt is FL-Ex8. We found that the micro-Ex8 is the ancient form and the 3 + 48 organization of alternatively spliced overlapping exons only emerged in the Schizophora group of Diptera and is absolutely conserved in this group. We then used several strategies to test the in vivo function of the two types of isoforms and found that the micro-Ex8 and FL-Ex8 isoforms have largely overlapping functions but also have non-redundant functions that are tissue-specific, which supports their strong evolutionary conservation. Since the different combinations of protein interaction of Hth with Exd and/or Hox can have different DNA target specificity, our finding of alternatively spliced isoforms adds to the spectrum of structural and functional diversity under developmental regulation.
Collapse
Affiliation(s)
- Ling-Wen Chang
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan, ROC
| | - I-Chieh Tseng
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan, ROC.,Institute of Genomic Sciences, National Yang-Ming University, Taipei, Taiwan, ROC.,Department of Life Science, Chinese Culture University, Taipei, Taiwan, ROC
| | - Lan-Hsin Wang
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.
| | - Y Henry Sun
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan, ROC. .,Institute of Genomic Sciences, National Yang-Ming University, Taipei, Taiwan, ROC.
| |
Collapse
|
63
|
Kojin BB, Biedler JK, Tu Z, Adelman ZN. Characterization of a female germline and early zygote promoter from the transcription factor bZip1 in the dengue mosquito Aedes aegypti. Parasit Vectors 2020; 13:353. [PMID: 32680549 PMCID: PMC7367395 DOI: 10.1186/s13071-020-04216-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 07/06/2020] [Indexed: 11/10/2022] Open
Abstract
Background The wide distribution of Aedes aegypti, the main vector of dengue and yellow fever viruses, currently puts three billion people in the world at risk of infection with these viruses. Continuous transmission of these and other viruses despite aggressive efforts to prevent this emphasizes the need to develop new control strategies. Proposals to control disease transmission based on vector engineering, including both population suppression and population replacement, rely on the development of transgenes under the control of regulatory elements able to drive molecules in a specific tissue, time and strength. Methods Here we report the characterization of a promoter active in both the female germline and early zygote, derived from the transcription factor bZip1 in the mosquito Ae. aegypti, using transposon-based methods and RT-qPCR. Results We generated seven transgenic lines carrying AabZip1-reporter constructs and observed expression in both the ovary and early embryo. RT-qPCR analysis was performed to evaluate transcript expression patterns for each line, confirming that transgenic expression from the AabZip1 promoter largely recapitulated the endogenous expression pattern, albeit the strength of maternal expression appeared to be strongly influenced by chromosomal position. Conclusions This study provides a new regulatory sequence that can be useful for generating transgenic lines that can become a tool in vector control strategies.![]()
Collapse
Affiliation(s)
- Bianca B Kojin
- Department of Entomology and Agrilife Research, Texas A&M University, College Station, TX, USA
| | - James K Biedler
- Department of Biochemistry and the Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, USA
| | - Zhijian Tu
- Department of Biochemistry and the Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, USA
| | - Zach N Adelman
- Department of Entomology and Agrilife Research, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
64
|
Analysis of a Strong Suppressor of Segregation Distorter in Drosophila melanogaster. Genetics 2020; 215:1085-1105. [PMID: 32561521 DOI: 10.1534/genetics.120.303150] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 03/19/2020] [Indexed: 12/30/2022] Open
Abstract
Segregation Di st orter (SD) is a naturally occurring male meiotic drive system in Drosophila melanogaster, characterized by almost exclusive transmission of the SD chromosome owing to dysfunction of sperm receiving the SD+ homolog. Previous studies identified at least three closely linked loci on chromosome 2 required for distortion: Sd, the primary distorting gene; E(SD) (Enhancer of SD), which increases the strength of distortion; and Rsp (Responder), the apparent target of Sd Strength of distortion is also influenced by linked upward modifiers including M(SD) (Modifier of SD) and St(SD) (Stabilizer of SD), and by various unlinked suppressors. Although Sd is known to encode a mutant RanGAP protein, none of the modifiers have been molecularly identified. This work focuses on the genetic and cytological characterization of a strong X-linked suppressor, Su(SD), capable of restoring Mendelian transmission in SD/SD+ males. Sd and its cohort of positive modifiers appear to act semiquantitatively in opposition to Su(SD) with distortion strength depending primarily on the total number of distorting elements rather than which particular elements are present. Su(SD) can also suppress male sterility observed in certain SD genotypes. To facilitate its eventual molecular identification, Su(SD) was localized by deletion mapping to polytene region 13C7-13E4 These studies highlight the polygenic nature of distortion and its dependence on a constellation of positive and negative modifiers, provide insight into the stability of Mendelian transmission in natural populations even when a drive system arises, and pave the way for molecular characterization of Su(SD) whose identity should reveal new information about the mechanism of distortion.
Collapse
|
65
|
Quinn CM, Nolan T. Nuclease-based gene drives, an innovative tool for insect vector control: advantages and challenges of the technology. CURRENT OPINION IN INSECT SCIENCE 2020; 39:77-83. [PMID: 32339930 DOI: 10.1016/j.cois.2020.03.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/09/2020] [Accepted: 03/19/2020] [Indexed: 06/11/2023]
Abstract
Genetic control of insects involves the release of modified insects that contain altered genetic traits and are competent to mate with target populations to introduce the traits therein. Since it relies on mating, this type of control is species-specific, non-toxic, and has the advantage that the released insects can do the difficult task of reaching remote and otherwise inaccessible insect niches. Gene drives are capable of drastically biasing their own transmission and are being developed as a new type of genetic control, one that would be self-sustaining, requiring low numbers in the initial release in order to spread and persist within a population. In this review, the advantages and challenges of building and deploying this technology will be discussed, using mosquito control as an example.
Collapse
Affiliation(s)
| | - Tony Nolan
- Liverpool School of Tropical Medicine, United Kingdom.
| |
Collapse
|
66
|
Peng W, Yu S, Handler AM, Zhang H. Transcriptome Analysis of the Oriental Fruit Fly Bactrocera dorsalis Early Embryos. INSECTS 2020; 11:insects11050323. [PMID: 32456171 PMCID: PMC7290859 DOI: 10.3390/insects11050323] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/28/2020] [Accepted: 04/28/2020] [Indexed: 12/01/2022]
Abstract
The oriental fruit fly, Bactrocera dorsalis (Hendel), is one of the most devastating and highly invasive agricultural pests world-wide, resulting in severe economic loss. Thus, it is of great interest to understand the transcriptional changes that occur during the activation of its zygotic genome at the early stages of embryonic development, especially the expression of genes involved in sex determination and the cellularization processes. In this study, we applied Illumina sequencing to identify B. dorsalis sex determination genes and early zygotic genes by analyzing transcripts from three early embryonic stages at 0–1, 2–4, and 5–8 h post-oviposition, which include the initiation of sex determination and cellularization. These tests generated 13,489 unigenes with an average length of 2185 bp. In total, 1683, 3201 and 3134 unigenes had significant changes in expression levels at times after oviposition including at 2–4 h versus 0–1 h, 5–8 h versus 0–1 h, and 5–8 h versus 2–4 h, respectively. Clusters of gene orthology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) annotations were performed throughout embryonic development to better understand the functions of differentially expressed unigenes. We observed that the RNA binding and spliceosome pathways were highly enriched and overrepresented during the early stage of embryogenesis. Additionally, transcripts for 21 sex-determination and three cellularization genes were identified, and expression pattern analysis revealed that the majority of these genes were highly expressed during embryogenesis. This study is the first assembly performed for B. dorsalis based on Illumina next-generation sequencing technology during embryogenesis. Our data should contribute significantly to the fundamental understanding of sex determination and early embryogenesis in tephritid fruit flies, and provide gene promoter and effector gene candidates for transgenic pest-management strategies for these economically important species.
Collapse
Affiliation(s)
- Wei Peng
- Key Laboratory of Horticultural Plant Biology (MOE), State Key Laboratory of Agricultural Microbiology, China-Australia Joint Research Centre for Horticultural and Urban Pests, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (W.P.); (S.Y.)
- College of Life Sciences, China Jiliang University, Hangzhou 310018, China
| | - Shuning Yu
- Key Laboratory of Horticultural Plant Biology (MOE), State Key Laboratory of Agricultural Microbiology, China-Australia Joint Research Centre for Horticultural and Urban Pests, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (W.P.); (S.Y.)
| | - Alfred M. Handler
- USDA/ARS, Center for Medical, Agricultural and Veterinary Entomology, 1700 SW 23rd Drive, Gainesville, FL 32608, USA;
| | - Hongyu Zhang
- Key Laboratory of Horticultural Plant Biology (MOE), State Key Laboratory of Agricultural Microbiology, China-Australia Joint Research Centre for Horticultural and Urban Pests, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (W.P.); (S.Y.)
- Correspondence:
| |
Collapse
|
67
|
Vesicular transport mediates the uptake of cytoplasmic proteins into mitochondria in Drosophila melanogaster. Nat Commun 2020; 11:2592. [PMID: 32444642 PMCID: PMC7244744 DOI: 10.1038/s41467-020-16335-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 04/27/2020] [Indexed: 12/15/2022] Open
Abstract
Mitochondrial aging, which results in mitochondrial dysfunction, is strongly linked to many age-related diseases. Aging is associated with mitochondrial enlargement and transport of cytosolic proteins into mitochondria. The underlying homeostatic mechanisms that regulate mitochondrial morphology and function, and their breakdown during aging, remain unclear. Here, we identify a mitochondrial protein trafficking pathway in Drosophila melanogaster involving the mitochondria-associated protein Dosmit. Dosmit induces mitochondrial enlargement and the formation of double-membraned vesicles containing cytosolic protein within mitochondria. The rate of vesicle formation increases with age. Vesicles originate from the outer mitochondrial membrane as observed by tracking Tom20 localization, and the process is mediated by the mitochondria-associated Rab32 protein. Dosmit expression level is closely linked to the rate of ubiquitinated protein aggregation, which are themselves associated with age-related diseases. The mitochondrial protein trafficking route mediated by Dosmit offers a promising target for future age-related mitochondrial disease therapies. Mitochondrial dynamics change during ageing, with larger mitochondria and altered protein import in older animals. Here the authors show that Dosmit protein mediates mitochondrial morphology with Rab32 by inducing double-membraned vesicles that regulate protein trafficking into mitochondria.
Collapse
|
68
|
Oberhofer G, Ivy T, Hay BA. Gene drive and resilience through renewal with next generation Cleave and Rescue selfish genetic elements. Proc Natl Acad Sci U S A 2020; 117:9013-9021. [PMID: 32245808 PMCID: PMC7183144 DOI: 10.1073/pnas.1921698117] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Gene drive-based strategies for modifying populations face the problem that genes encoding cargo and the drive mechanism are subject to separation, mutational inactivation, and loss of efficacy. Resilience, an ability to respond to these eventualities in ways that restore population modification with functional genes, is needed for long-term success. Here, we show that resilience can be achieved through cycles of population modification with "Cleave and Rescue" (ClvR) selfish genetic elements. ClvR comprises 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. Cycles of modification can, in principle, be carried out if two ClvR elements targeting different essential genes are located at the same genomic position, and one of them, ClvRn+1, carries a Rescue transgene from an earlier element, ClvRnClvRn+1 should spread within a population of ClvRn, while also bringing about a decrease in its frequency. To test this hypothesis, we first show that multiple ClvRs, each targeting a different essential gene, function when located at a common chromosomal position in Drosophila We then show that when several of these also carry the Rescue from a different ClvR, they spread to transgene fixation in populations fixed for the latter and at its expense. Therefore, genetic modifications of populations can be overwritten with new content, providing an ongoing point of control.
Collapse
Affiliation(s)
- Georg Oberhofer
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Tobin Ivy
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Bruce A Hay
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125;
- St John's College, University of Cambridge, CB2 1TP Cambridge, United Kingdom
| |
Collapse
|
69
|
Lee G, Sehgal R, Wang Z, Park JH. Ultraspiracle-independent anti-apoptotic function of ecdysone receptors is required for the survival of larval peptidergic neurons via suppression of grim expression in Drosophila melanogaster. Apoptosis 2020; 24:256-268. [PMID: 30637539 DOI: 10.1007/s10495-019-01514-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In Drosophila melanogaster a significant number of heterogenous larval neurons in the central nervous system undergo metamorphosis-associated programmed cell death, termed metamorphoptosis. Interestingly distinct groups of doomed larval neurons are eliminated at different metamorphic phases. Although ecdysone hormonal signaling via nuclear ecdysone receptors (EcRs) is known to orchestrate the neuronal metamorphoptosis, little is known about how this signaling controls such diverse neuronal responses. Crustacean cardioactive peptide (CCAP)-producing neurons in the ventral nerve cord are developmentally programmed to die shortly after adult emergence. In this study, we show that disruption of endogenous EcR function by ectopic expression of dominant negative forms of EcRs (EcRDN) causes premature death of larval CCAP neurons in a caspase-dependent manner. This event is rescued by co-expression of individual EcR isoforms. Furthermore, larval CCAP neurons are largely normal in ecr mutants lacking either EcR-A or EcR-B isoforms, suggesting that EcR isoforms redundantly function to protect larval CCAP neurons. Of surprise, a role of Ultraspiracle (Usp), a canonical partner of EcR, is dispensable in the protection of CCAP neurons, whereas both EcR and Usp are required for inducing metamorphoptosis of vCrz neurons shortly after prepupal formation. As a downstream, grim is an essential cell death gene for the EcRDN-mediated CCAP neuronal death, while either hid or rpr function is dispensable. Together, our results suggest that Usp-independent EcR actions protect CCAP neurons from their premature death by repressing grim expression until their normally scheduled apoptosis at post-emergence. Our studies highlight two opposite roles played by EcR function for metamorphoptosis of two different peptidergic neuronal groups, proapoptotic (vCrz) versus antiapoptotic (CCAP), and propose that distinct death timings of doomed larval neurons are determined by differential signaling mechanisms involving EcR.
Collapse
Affiliation(s)
- Gyunghee Lee
- Department of Biochemistry and Cellular and Molecular Biology and NeuroNet Research Center, University of Tennessee, Knoxville, TN, 37996, USA
| | - Ritika Sehgal
- Department of Biochemistry and Cellular and Molecular Biology and NeuroNet Research Center, University of Tennessee, Knoxville, TN, 37996, USA
| | - Zixing Wang
- UT-ORNL Graduate School of Genome Science and Technology Program, University of Tennessee, Knoxville, TN, 37996, USA
| | - Jae H Park
- Department of Biochemistry and Cellular and Molecular Biology and NeuroNet Research Center, University of Tennessee, Knoxville, TN, 37996, USA. .,UT-ORNL Graduate School of Genome Science and Technology Program, University of Tennessee, Knoxville, TN, 37996, USA.
| |
Collapse
|
70
|
Champer J, Kim IK, Champer SE, Clark AG, Messer PW. Performance analysis of novel toxin-antidote CRISPR gene drive systems. BMC Biol 2020; 18:27. [PMID: 32164660 PMCID: PMC7068947 DOI: 10.1186/s12915-020-0761-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 02/28/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND CRISPR gene drive systems allow the rapid spread of a genetic construct throughout a population. Such systems promise novel strategies for the management of vector-borne diseases and invasive species by suppressing a target population or modifying it with a desired trait. However, current homing-type drives have two potential shortcomings. First, they can be thwarted by the rapid evolution of resistance. Second, they lack any mechanism for confinement to a specific target population. In this study, we conduct a comprehensive performance assessment of several new types of CRISPR-based gene drive systems employing toxin-antidote (TA) principles, which should be less prone to resistance and allow for the confinement of drives to a target population due to invasion frequency thresholds. RESULTS The underlying principle of the proposed CRISPR toxin-antidote gene drives is to disrupt an essential target gene while also providing rescue by a recoded version of the target as part of the drive allele. Thus, drive alleles tend to remain viable, while wild-type targets are disrupted and often rendered nonviable, thereby increasing the relative frequency of the drive allele. Using individual-based simulations, we show that Toxin-Antidote Recessive Embryo (TARE) drives targeting an haplosufficient but essential gene (lethal when both copies are disrupted) can enable the design of robust, regionally confined population modification strategies with high flexibility in choosing promoters and targets. Toxin-Antidote Dominant Embryo (TADE) drives require a haplolethal target gene and a germline-restricted promoter, but they could permit faster regional population modification and even regionally confined population suppression. Toxin-Antidote Dominant Sperm (TADS) drives can be used for population modification or suppression. These drives are expected to spread rapidly and could employ a variety of promoters, but unlike TARE and TADE, they would not be regionally confined and also require highly specific target genes. CONCLUSIONS Overall, our results suggest that CRISPR-based TA gene drives provide promising candidates for flexible ecological engineering strategies in a variety of organisms.
Collapse
Affiliation(s)
- Jackson Champer
- Department of Computational Biology, Cornell University, Ithaca, NY, 14853, USA.
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, 14853, USA.
| | - Isabel K Kim
- Department of Computational Biology, Cornell University, Ithaca, NY, 14853, USA
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, 14853, USA
| | - Samuel E Champer
- Department of Computational Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Andrew G Clark
- Department of Computational Biology, Cornell University, Ithaca, NY, 14853, USA
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, 14853, USA
| | - Philipp W Messer
- Department of Computational Biology, Cornell University, Ithaca, NY, 14853, USA
| |
Collapse
|
71
|
Champer J, Lee E, Yang E, Liu C, Clark AG, Messer PW. A toxin-antidote CRISPR gene drive system for regional population modification. Nat Commun 2020; 11:1082. [PMID: 32109227 PMCID: PMC7046741 DOI: 10.1038/s41467-020-14960-3] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 02/06/2020] [Indexed: 01/04/2023] Open
Abstract
Engineered gene drives based on a homing mechanism could rapidly spread genetic alterations through a population. However, such drives face a major obstacle in the form of resistance against the drive. In addition, they are expected to be highly invasive. Here, we introduce the Toxin-Antidote Recessive Embryo (TARE) drive. It functions by disrupting a target gene, forming recessive lethal alleles, while rescuing drive-carrying individuals with a recoded version of the target. Modeling shows that such drives will have threshold-dependent invasion dynamics, spreading only when introduced above a fitness-dependent frequency. We demonstrate a TARE drive in Drosophila with 88-95% transmission by female heterozygotes. This drive was able to spread through a large cage population in just six generations following introduction at 24% frequency without any apparent evolution of resistance. Our results suggest that TARE drives constitute promising candidates for the development of effective, flexible, and regionally confinable drives for population modification.
Collapse
Affiliation(s)
- Jackson Champer
- Department of Computational Biology, Cornell University, Ithaca, NY, 14853, USA.
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, 14853, USA.
| | - Esther Lee
- Department of Computational Biology, Cornell University, Ithaca, NY, 14853, USA
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, 14853, USA
| | - Emily Yang
- Department of Computational Biology, Cornell University, Ithaca, NY, 14853, USA
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, 14853, USA
| | - Chen Liu
- Department of Computational Biology, Cornell University, Ithaca, NY, 14853, USA
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, 14853, USA
| | - Andrew G Clark
- Department of Computational Biology, Cornell University, Ithaca, NY, 14853, USA
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, 14853, USA
| | - Philipp W Messer
- Department of Computational Biology, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
72
|
Abstract
Vector-borne diseases, such as dengue, Zika and malaria, are a major cause of morbidity and mortality worldwide. These diseases have proven difficult to control and currently available management tools are insufficient to eliminate them in many regions. Gene drives have the potential to revolutionize vector-borne disease control. This suite of technologies has advanced rapidly in recent years as a result of the availability of new, more efficient gene editing technologies. Gene drives can favorably bias the inheritance of a linked disease-refractory gene, which could possibly be exploited (i) to generate a vector population incapable of transmitting disease or (ii) to disrupt an essential gene for viability or fertility, which could eventually eliminate a population. Importantly, gene drives vary in characteristics such as their transmission efficiency, confinability and reversibility, and their potential to develop resistance to the drive mechanism. Here, we discuss recent advancements in the gene drive field, and contrast the benefits and limitations of a variety of technologies, as well as approaches to overcome these limitations. We also discuss the current state of each gene drive technology and the technical considerations that need to be addressed on the pathway to field implementation. While there are still many obstacles to overcome, recent progress has brought us closer than ever before to genetic-based vector modification as a tool to support vector-borne disease elimination efforts worldwide.
Collapse
Affiliation(s)
- Robyn R Raban
- Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | - John M Marshall
- Innovative Genomics Institute, Berkeley, CA 94720, USA
- Tata Institute for Genetics and Society, University of California, San Diego, La Jolla, CA 92093, USA
| | - Omar S Akbari
- Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA 92093, USA
- Division of Epidemiology & Biostatistics, School of Public Health, University of California, Berkeley, CA 94720, USA
| |
Collapse
|
73
|
Kandul NP, Liu J, Buchman A, Gantz VM, Bier E, Akbari OS. Assessment of a Split Homing Based Gene Drive for Efficient Knockout of Multiple Genes. G3 (BETHESDA, MD.) 2020; 10:827-837. [PMID: 31882406 PMCID: PMC7003086 DOI: 10.1534/g3.119.400985] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 12/19/2019] [Indexed: 01/08/2023]
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.
Collapse
Affiliation(s)
| | - Junru Liu
- Section of Cell and Developmental Biology and
| | | | | | - Ethan Bier
- Section of Cell and Developmental Biology and
- Tata Institute for Genetics and Society, University of California, San Diego, La Jolla, CA 92093
| | - Omar S Akbari
- Section of Cell and Developmental Biology and
- Tata Institute for Genetics and Society, University of California, San Diego, La Jolla, CA 92093
| |
Collapse
|
74
|
Antiviral Effectors and Gene Drive Strategies for Mosquito Population Suppression or Replacement to Mitigate Arbovirus Transmission by Aedes aegypti. INSECTS 2020; 11:insects11010052. [PMID: 31940960 PMCID: PMC7023000 DOI: 10.3390/insects11010052] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/07/2020] [Accepted: 01/09/2020] [Indexed: 12/11/2022]
Abstract
The mosquito vector Aedes aegypti transmits arthropod-borne viruses (arboviruses) of medical importance, including Zika, dengue, and yellow fever viruses. Controlling mosquito populations remains the method of choice to prevent disease transmission. Novel mosquito control strategies based on genetically manipulating mosquitoes are being developed as additional tools to combat arbovirus transmission. Genetic control of mosquitoes includes two basic strategies: population suppression and population replacement. The former aims to eliminate mosquito populations while the latter aims to replace wild populations with engineered, pathogen-resistant mosquitoes. In this review, we outline suppression strategies being applied in the field, as well as current antiviral effector genes that have been characterized and expressed in transgenic Ae. aegypti for population replacement. We discuss cutting-edge gene drive technologies that can be used to enhance the inheritance of effector genes, while highlighting the challenges and opportunities associated with gene drives. Finally, we present currently available models that can estimate mosquito release numbers and time to transgene fixation for several gene drive systems. Based on the recent advances in genetic engineering, we anticipate that antiviral transgenic Ae. aegypti exhibiting gene drive will soon emerge; however, close monitoring in simulated field conditions will be required to demonstrate the efficacy and utility of such transgenic mosquitoes.
Collapse
|
75
|
Cash SA, Robert MA, Lorenzen MD, Gould F. The impact of local population genetic background on the spread of the selfish element Medea-1 in red flour beetles. Ecol Evol 2020; 10:863-874. [PMID: 32015850 PMCID: PMC6988536 DOI: 10.1002/ece3.5946] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 11/24/2019] [Accepted: 11/27/2019] [Indexed: 12/18/2022] Open
Abstract
Selfish genetic elements have been found in the genomes of many species, yet our understanding of their evolutionary dynamics is only partially understood. A number of distinct selfish Medea elements are naturally present in many populations of the red flour beetle (Tribolium castaneum). Although these Medea elements are predicted by models to increase in frequency within populations because any offspring of a Medea-bearing mother that do not inherit at least one Medea allele will die, experiments demonstrating an increase in a naturally occurring Medea element are lacking. Our survey of the specific Medea element, M1, in the United States showed that it had a patchy geographic distribution. From the survey, it could not be determined if this distribution was caused by a slow process of M1 colonization of discrete populations or if some populations lacked M1 because they had genetic factors conferring resistance to the Medea mechanism. We show that populations with naturally low to intermediate M1 frequencies likely represent transient states during the process of Medea spread. Furthermore, we find no evidence that genetic factors are excluding M1 from US populations where the element is not presently found. We also show how a known suppressor of Medea can impair the increase of M1 in populations and discuss the implications of our findings for pest-management applications of Medea elements.
Collapse
Affiliation(s)
- Sarah A. Cash
- Graduate Program in GeneticsDepartment of Biological SciencesNorth Carolina State UniversityRaleighNorth Carolina
- W. M. Keck Center for Behavioral BiologyNorth Carolina State UniversityRaleighNorth Carolina
| | - Michael A. Robert
- Department of Mathematics, Physics, and StatisticsUniversity of the SciencesPhiladelphiaPennsylvania
| | - Marcé D. Lorenzen
- Department of Entomology and Plant PathologyNorth Carolina State UniversityRaleighNorth Carolina
| | - Fred Gould
- W. M. Keck Center for Behavioral BiologyNorth Carolina State UniversityRaleighNorth Carolina
- Department of Entomology and Plant PathologyNorth Carolina State UniversityRaleighNorth Carolina
- Genetic Engineering and Society CenterNorth Carolina State UniversityRaleighNorth Carolina
| |
Collapse
|
76
|
Maier T, Wheeler NJ, Namigai EKO, Tycko J, Grewelle RE, Woldeamanuel Y, Klohe K, Perez-Saez J, Sokolow SH, De Leo GA, Yoshino TP, Zamanian M, Reinhard-Rupp J. Gene drives for schistosomiasis transmission control. PLoS Negl Trop Dis 2019; 13:e0007833. [PMID: 31856157 PMCID: PMC6922350 DOI: 10.1371/journal.pntd.0007833] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Schistosomiasis is one of the most important and widespread neglected tropical diseases (NTD), with over 200 million people infected in more than 70 countries; the disease has nearly 800 million people at risk in endemic areas. Although mass drug administration is a cost-effective approach to reduce occurrence, extent, and severity of the disease, it does not provide protection to subsequent reinfection. Interventions that target the parasites’ intermediate snail hosts are a crucial part of the integrated strategy required to move toward disease elimination. The recent revolution in gene drive technology naturally leads to questions about whether gene drives could be used to efficiently spread schistosome resistance traits in a population of snails and whether gene drives have the potential to contribute to reduced disease transmission in the long run. Responsible implementation of gene drives will require solutions to complex challenges spanning multiple disciplines, from biology to policy. This Review Article presents collected perspectives from practitioners of global health, genome engineering, epidemiology, and snail/schistosome biology and outlines strategies for responsible gene drive technology development, impact measurements of gene drives for schistosomiasis control, and gene drive governance. Success in this arena is a function of many factors, including gene-editing specificity and efficiency, the level of resistance conferred by the gene drive, how fast gene drives may spread in a metapopulation over a complex landscape, ecological sustainability, social equity, and, ultimately, the reduction of infection prevalence in humans. With combined efforts from across the broad global health community, gene drives for schistosomiasis control could fortify our defenses against this devastating disease in the future.
Collapse
Affiliation(s)
- Theresa Maier
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, United Kingdom
| | - Nicolas James Wheeler
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Global Health Institute of Merck (KGaA), Eysins, Switzerland
| | | | - Josh Tycko
- Department of Genetics, Stanford University School of Medicine, Stanford, California, United States of America
| | - Richard Ernest Grewelle
- Hopkins Marine Station, School of Humanities and Sciences, Stanford University, Pacific Grove, California, United States of America
| | - Yimtubezinash Woldeamanuel
- Department of Microbiology, Immunology & Parasitology, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | | | - Javier Perez-Saez
- Laboratory of Ecohydrology, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Susanne H. Sokolow
- Woods Institute for the Environment, Stanford University, Stanford, California, United States of America
- Marine Science Institute, University of California, Santa Barbara, California, United States of America
| | - Giulio A. De Leo
- Hopkins Marine Station, School of Humanities and Sciences, Stanford University, Pacific Grove, California, United States of America
| | - Timothy P. Yoshino
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Mostafa Zamanian
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | | |
Collapse
|
77
|
Cash SA, Lorenzen MD, Gould F. The distribution and spread of naturally occurring Medea selfish genetic elements in the United States. Ecol Evol 2019; 9:14407-14416. [PMID: 31938528 PMCID: PMC6953677 DOI: 10.1002/ece3.5876] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 10/31/2019] [Accepted: 11/05/2019] [Indexed: 01/01/2023] Open
Abstract
Selfish genetic elements (SGEs) are DNA sequences that are transmitted to viable offspring in greater than Mendelian frequencies. Medea SGEs occur naturally in some populations of red flour beetle (Tribolium castaneum) and are expected to increase in frequency within populations and spread among populations. The large-scale U.S. distributions of Medea-4 (M4) had been mapped based on samples from 1993 to 1995. We sampled beetles in 2011-2014 and show that the distribution of M4 in the United States is dynamic and has shifted southward. By using a genetic marker of Medea-1 (M1), we found five unique geographic clusters with high and low M1 frequencies in a pattern not predicted by microsatellite-based analysis of population structure. Our results indicate the absence of rigid barriers to Medea spread in the United States, so assessment of what factors have limited its current distribution requires further investigation. There is great interest in using synthetic SGEs, including synthetic Medea, to alter or suppress pest populations, but there is concern about unpredicted spread of these SGEs and potential for populations to become resistant to them. The finding of patchy distributions of Medea elements suggests that released synthetic SGEs cannot always be expected to spread uniformly, especially in target species with limited dispersal.
Collapse
Affiliation(s)
- Sarah A. Cash
- Program in GeneticsDepartment of Biological SciencesNorth Carolina State UniversityRaleighNCUSA
| | - Marce D. Lorenzen
- Department of Entomology and Plant PathologyNorth Carolina State UniversityRaleighNCUSA
| | - Fred Gould
- Department of Entomology and Plant PathologyNorth Carolina State UniversityRaleighNCUSA
| |
Collapse
|
78
|
Sánchez C. HM, Wu SL, Bennett JB, Marshall JM. MGD
riv
E: A modular simulation framework for the spread of gene drives through spatially explicit mosquito populations. Methods Ecol Evol 2019. [DOI: 10.1111/2041-210x.13318] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Héctor M. Sánchez C.
- Division of Epidemiology and Biostatistics School of Public Health University of California Berkeley CA USA
| | - Sean L. Wu
- Division of Epidemiology and Biostatistics School of Public Health University of California Berkeley CA USA
| | - Jared B. Bennett
- Biophysics Graduate Group Division of Biological Sciences College of Letters and Science University of California Berkeley CA USA
| | - John M. Marshall
- Division of Epidemiology and Biostatistics School of Public Health University of California Berkeley CA USA
- Innovative Genomics Institute Berkeley CA USA
| |
Collapse
|
79
|
Bull JJ, Remien CH, Gomulkiewicz R, Krone SM. Spatial structure undermines parasite suppression by gene drive cargo. PeerJ 2019; 7:e7921. [PMID: 31681512 PMCID: PMC6824332 DOI: 10.7717/peerj.7921] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 09/18/2019] [Indexed: 12/17/2022] Open
Abstract
Gene drives may be used in two ways to curtail vectored diseases. Both involve engineering the drive to spread in the vector population. One approach uses the drive to directly depress vector numbers, possibly to extinction. The other approach leaves intact the vector population but suppresses the disease agent during its interaction with the vector. This second application may use a drive engineered to carry a genetic cargo that blocks the disease agent. An advantage of the second application is that it is far less likely to select vector resistance to block the drive, but the disease agent may instead evolve resistance to the inhibitory cargo. However, some gene drives are expected to spread so fast and attain such high coverage in the vector population that, if the disease agent can evolve resistance only gradually, disease eradication may be feasible. Here we use simple models to show that spatial structure in the vector population can greatly facilitate persistence and evolution of resistance by the disease agent. We suggest simple approaches to avoid some types of spatial structure, but others may be intrinsic to the populations being challenged and difficult to overcome.
Collapse
Affiliation(s)
- James J. Bull
- Department of Biological Sciences, University of Idaho, Moscow, ID, United States of America
| | - Christopher H. Remien
- Department of Mathematics, University of Idaho, Moscow, ID, United States of America
| | - Richard Gomulkiewicz
- School of Biological Sciences, Washington State University, Pullman, WA, United States of America
| | - Stephen M. Krone
- Department of Mathematics, University of Idaho, Moscow, ID, United States of America
| |
Collapse
|
80
|
Teem JL, Ambali A, Glover B, Ouedraogo J, Makinde D, Roberts A. Problem formulation for gene drive mosquitoes designed to reduce malaria transmission in Africa: results from four regional consultations 2016-2018. Malar J 2019; 18:347. [PMID: 31615576 PMCID: PMC6794889 DOI: 10.1186/s12936-019-2978-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 09/26/2019] [Indexed: 01/10/2023] Open
Abstract
Background Gene drive mosquitoes have been proposed as a possible means to reduce the transmission of malaria in Africa. Because this technology has no prior use-history at this time, environmental risk assessments for gene drive mosquitoes will benefit from problem formulation—an organized and ordered process to identify protection goals and potential pathways to harm to the environment, or animal or human health. Recognizing this need, the New Partnership for Africa’s Development (NEPAD), with support from African and international partners, organized four regional consultative workshops in Africa to initiate this process. Methods The workshops were attended by a diverse set of participants and stakeholders, including scientists, ethicists, health professionals, government regulators in the fields of environment health and biosafety as well government policymakers, who met for 4 days to deliberate on protection goals and pathways relevant to the use of gene drive mosquitoes for malaria control. The goal of the workshops was not to produce a comprehensive and detailed environmental risk assessment of gene drive mosquitoes, but rather to introduce problem formulation as a tool to the stakeholder community, and to serve as a starting point for conducting systematic environmental risk assessments in the future, identifying protection goals related to gene drive mosquitoes that are particular to African stakeholders. Results Participants in the workshops frequently identified human health and biodiversity as being relevant broad protection goals. Results of the deliberations provide insight into the concerns of African participants at an early stage in the development of gene drive organism/products that should be instructive to developers using this technology. Conclusions In general, the African participants of the consultations had a precautionary perspective with regard to environmental risk assessment of gene drive technology. As gene drive technology develops, protection goals will become further refined and candidate products will be further defined. These workshops represent only the beginning of a continuing process that will ultimately inform environmental risk assessment for gene drive mosquitoes to control malaria in Africa.
Collapse
Affiliation(s)
- John L Teem
- ILSI Research Foundation, 740 Fifteenth Street NW, Suite 600, Washington, DC, 20005, USA
| | - Aggrey Ambali
- NEPAD Agency, Industrialization, Science, Technology and Innovation Hub, 230 15th Road, Midrand, South Africa
| | - Barbara Glover
- NEPAD Agency, Industrialization, Science, Technology and Innovation Hub, 230 15th Road, Midrand, South Africa
| | - Jeremy Ouedraogo
- ABNE, NEPAD Regional Office West Africa, Hann Maristes 2, Rue HB 350, BP 17204, Dakar, Senegal
| | - Diran Makinde
- NEPAD Agency, Industrialization, Science, Technology and Innovation Hub, 230 15th Road, Midrand, South Africa
| | - Andrew Roberts
- ILSI Research Foundation, 740 Fifteenth Street NW, Suite 600, Washington, DC, 20005, USA.
| |
Collapse
|
81
|
Barrett LG, Legros M, Kumaran N, Glassop D, Raghu S, Gardiner DM. Gene drives in plants: opportunities and challenges for weed control and engineered resilience. Proc Biol Sci 2019; 286:20191515. [PMID: 31551052 PMCID: PMC6784734 DOI: 10.1098/rspb.2019.1515] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 09/06/2019] [Indexed: 02/07/2023] Open
Abstract
Plant species, populations and communities are under threat from climate change, invasive pathogens, weeds and habitat fragmentation. Despite considerable research effort invested in genome engineering for crop improvement, the development of genetic tools for the management of wild plant populations has rarely been given detailed consideration. Gene drive systems that allow direct genetic management of plant populations via the spread of fitness-altering genetic modifications could be of great utility. However, despite the rapid development of synthetic tools and their enormous promise, little explicit consideration has been given to their application in plants and, to date, they remain untested. This article considers the potential utility of gene drives for the management of wild plant populations, and examines the factors that might influence the design, spread and efficacy of synthetic drives. To gain insight into optimal ways to design and deploy synthetic drive systems, we investigate the diversity of mechanisms underlying natural gene drives and their dynamics within plant populations and species. We also review potential approaches for engineering gene drives and discuss their potential application to plant genomes. We highlight the importance of considering the impact of plant life-history and genetic architecture on the dynamics of drive, investigate the potential for different types of resistance evolution, and touch on the ethical, regulatory and social challenges ahead.
Collapse
Affiliation(s)
- Luke G. Barrett
- CSIRO Agriculture and Food, Canberra, Australian Capital Territory, Australia
- CSIRO Synthetic Biology Future Science Platform, Canberra, Australian Capital Territory, Australia
| | - Mathieu Legros
- CSIRO Agriculture and Food, Canberra, Australian Capital Territory, Australia
- CSIRO Synthetic Biology Future Science Platform, Canberra, Australian Capital Territory, Australia
| | | | - Donna Glassop
- CSIRO Agriculture and Food, Canberra, Australian Capital Territory, Australia
| | - S. Raghu
- CSIRO Health and Biosecurity, Brisbane, Queensland, Australia
| | - Donald M. Gardiner
- CSIRO Agriculture and Food, Canberra, Australian Capital Territory, Australia
| |
Collapse
|
82
|
Backus GA, Delborne JA. Threshold-Dependent Gene Drives in the Wild: Spread, Controllability, and Ecological Uncertainty. Bioscience 2019. [DOI: 10.1093/biosci/biz098] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AbstractGene drive technology could allow the intentional spread of a desired gene throughout an entire wild population in relatively few generations. However, there are major concerns that gene drives could either fail to spread or spread without restraint beyond the targeted population. One potential solution is to use more localized threshold-dependent drives, which only spread when they are released in a population above a critical frequency. However, under certain conditions, small changes in gene drive fitness could lead to divergent outcomes in spreading behavior. In the face of ecological uncertainty, the inability to estimate gene drive fitness in a real-world context could prove problematic because gene drives designed to be localized could spread to fixation in neighboring populations if ecological conditions unexpectedly favor the gene drive. This perspective offers guidance to developers and managers because navigating gene drive spread and controllability could be risky without detailed knowledge of ecological contexts.
Collapse
|
83
|
Xie T, Ho MCW, Liu Q, Horiuchi W, Lin CC, Task D, Luan H, White BH, Potter CJ, Wu MN. A Genetic Toolkit for Dissecting Dopamine Circuit Function in Drosophila. Cell Rep 2019; 23:652-665. [PMID: 29642019 PMCID: PMC5962273 DOI: 10.1016/j.celrep.2018.03.068] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 02/19/2018] [Accepted: 03/15/2018] [Indexed: 01/23/2023] Open
Abstract
The neuromodulator dopamine (DA) plays a key role in motor control, motivated behaviors, and higher-order cognitive processes. Dissecting how these DA neural networks tune the activity of local neural circuits to regulate behavior requires tools for manipulating small groups of DA neurons. To address this need, we assembled a genetic toolkit that allows for an exquisite level of control over the DA neural network in Drosophila. To further refine targeting of specific DA neurons, we also created reagents that allow for the conversion of any existing GAL4 line into Split GAL4 or GAL80 lines. We demonstrated how this toolkit can be used with recently developed computational methods to rapidly generate additional reagents for manipulating small subsets or individual DA neurons. Finally, we used the toolkit to reveal a dynamic interaction between a small subset of DA neurons and rearing conditions in a social space behavioral assay.
Collapse
Affiliation(s)
- Tingting Xie
- School of Life Sciences, Peking University, Beijing 100871, China; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Margaret C W Ho
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Qili Liu
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Wakako Horiuchi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Chun-Chieh Lin
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Darya Task
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Haojiang Luan
- Laboratory of Molecular Biology, National Institute of Mental Health, NIH, Bethesda, MD 20892, USA
| | - Benjamin H White
- Laboratory of Molecular Biology, National Institute of Mental Health, NIH, Bethesda, MD 20892, USA
| | - Christopher J Potter
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Mark N Wu
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
84
|
Dhole S, Lloyd AL, Gould F. Tethered homing gene drives: A new design for spatially restricted population replacement and suppression. Evol Appl 2019; 12:1688-1702. [PMID: 31462923 PMCID: PMC6708424 DOI: 10.1111/eva.12827] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 05/06/2019] [Accepted: 05/10/2019] [Indexed: 12/18/2022] Open
Abstract
Optimism regarding potential epidemiological and conservation applications of modern gene drives is tempered by concern about the possibility of unintended spread of engineered organisms beyond the target population. In response, several novel gene drive approaches have been proposed that can, under certain conditions, locally alter characteristics of a population. One challenge for these gene drives is the difficulty of achieving high levels of localized population suppression without very large releases in the face of gene flow. We present a new gene drive system, tethered homing (TH), with improved capacity for both localization and population suppression. The TH drive is based on driving a payload gene using a homing construct that is anchored to a spatially restricted gene drive. We use a proof-of-concept mathematical model to show the dynamics of a TH drive that uses engineered underdominance as an anchor. This system is composed of a split homing drive and a two-locus engineered underdominance drive linked to one part of the split drive (the Cas endonuclease). We use simple population genetic simulations to show that the tethered homing technique can offer improved localized spread of costly transgenic payload genes. Additionally, the TH system offers the ability to gradually adjust the genetic load in a population after the initial alteration, with minimal additional release effort. We discuss potential solutions for improving localization and the feasibility of creating TH drive systems. Further research with models that include additional biological details will be needed to better understand how TH drives would behave in natural populations, but the preliminary results shown here suggest that tethered homing drives can be a useful addition to the repertoire of localized gene drives.
Collapse
Affiliation(s)
- Sumit Dhole
- Department of Entomology and Plant PathologyNorth Carolina State UniversityRaleighNorth Carolina
| | - Alun L. Lloyd
- Biomathematics Graduate Program and Department of MathematicsNorth Carolina State UniversityRaleighNorth Carolina
- Genetic Engineering and Society CenterNorth Carolina State UniversityRaleighNorth Carolina
| | - Fred Gould
- Department of Entomology and Plant PathologyNorth Carolina State UniversityRaleighNorth Carolina
- Genetic Engineering and Society CenterNorth Carolina State UniversityRaleighNorth Carolina
| |
Collapse
|
85
|
Climatic Conditions: Conventional and Nanotechnology-Based Methods for the Control of Mosquito Vectors Causing Human Health Issues. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16173165. [PMID: 31480254 PMCID: PMC6747303 DOI: 10.3390/ijerph16173165] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/25/2019] [Accepted: 08/27/2019] [Indexed: 12/12/2022]
Abstract
Climate variability is highly impacting on mosquito-borne diseases causing malaria and dengue fever across the globe. Seasonal variability change in temperature and rainfall patterns are impacting on human health. Mosquitoes cause diseases like dengue fever, yellow fever, malaria, Chikungunya, West Nile and Japanese encephalitis. According to estimations by health organizations, annually one million human deaths are caused by vector-borne diseases, and dengue fever has increased about 30-fold over the past 50 years. Similarly, over 200 million cases of malaria are being reported annually. Mosquito-borne diseases are sensitive to temperature, humidity and seasonal variability. Both conventional (environmental, chemical, mechanical, biological etc.) and nanotechnology-based (Liposomes, nano-suspensions and polymer-based nanoparticles) approaches are used for the eradication of Malaria and dengue fever. Now green approaches are used to eradicate mosquitoes to save human health without harming the environment. In this review, the impact of climatic conditions on mosquito-borne diseases along with conventional and nanotechnology-based approaches used for controlling malaria and dengue fever have been discussed. Important recommendations have been made for people to stay healthy.
Collapse
|
86
|
Two-By-One model of cytoplasmic incompatibility: Synthetic recapitulation by transgenic expression of cifA and cifB in Drosophila. PLoS Genet 2019; 15:e1008221. [PMID: 31242186 PMCID: PMC6594578 DOI: 10.1371/journal.pgen.1008221] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 05/30/2019] [Indexed: 01/22/2023] Open
Abstract
Wolbachia are maternally inherited bacteria that infect arthropod species worldwide and are deployed in vector control to curb arboviral spread using cytoplasmic incompatibility (CI). CI kills embryos when an infected male mates with an uninfected female, but the lethality is rescued if the female and her embryos are likewise infected. Two phage WO genes, cifAwMel and cifBwMel from the wMel Wolbachia deployed in vector control, transgenically recapitulate variably penetrant CI, and one of the same genes, cifAwMel, rescues wild type CI. The proposed Two-by-One genetic model predicts that CI and rescue can be recapitulated by transgenic expression alone and that dual cifAwMeland cifBwMel expression can recapitulate strong CI. Here, we use hatch rate and gene expression analyses in transgenic Drosophila melanogaster to demonstrate that CI and rescue can be synthetically recapitulated in full, and strong, transgenic CI comparable to wild type CI is achievable. These data explicitly validate the Two-by-One model in wMel-infected D. melanogaster, establish a robust system for transgenic studies of CI in a model system, and represent the first case of completely engineering male and female animal reproduction to depend upon bacteriophage gene products. Releases of Wolbachia-infected mosquitos are underway worldwide because Wolbachia block replication of Zika and Dengue viruses and spread themselves maternally through arthropod populations via cytoplasmic incompatibility (CI). The CI drive system depends on a Wolbachia-induced sperm modification that results in embryonic lethality when an infected male mates with an uninfected female, but this lethality is rescued when the female and her embryos are likewise infected. We recently reported that the phage WO genes, cifA and cifB, cause the sperm modification and cifA rescues the embryonic lethality caused by the wMel Wolbachia strain deployed in vector control. These reports motivated proposal of the Two-by-One model of CI whereby two genes cause lethality and one gene rescues it. Here we provide unequivocal support for the model in the Wolbachia strain used in vector control via synthetic methods that recapitulate CI and rescue in the absence of a Wolbachia infections. Our results reveal the set of phage WO genes responsible for this powerful genetic drive system, act as a proof-of-concept that these genes alone can induce gene drive like crossing patterns, and establish methodologies and hypotheses for future studies of CI in Drosophila. We discuss the implications of the Two-by-One model towards functional mechanisms of CI, the emergence of incompatibility between Wolbachia strains, vector control applications, and CI gene nomenclature.
Collapse
|
87
|
Oberhofer G, Ivy T, Hay BA. Cleave and Rescue, a novel selfish genetic element and general strategy for gene drive. Proc Natl Acad Sci U S A 2019; 116:6250-6259. [PMID: 30760597 PMCID: PMC6442612 DOI: 10.1073/pnas.1816928116] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
There is great interest in being able to spread beneficial traits throughout wild populations in ways that are self-sustaining. Here, we describe a chromosomal selfish genetic element, CleaveR [Cleave and Rescue (ClvR)], able to achieve this goal. ClvR comprises two linked chromosomal components. One, germline-expressed Cas9 and guide RNAs (gRNAs)-the Cleaver-cleaves and thereby disrupts endogenous copies of a gene whose product is essential. The other, a recoded version of the essential gene resistant to cleavage and gene conversion with cleaved copies-the Rescue-provides essential gene function. ClvR enhances its transmission, and that of linked genes, by creating conditions in which progeny lacking ClvR die because they have no functional copies of the essential gene. In contrast, those who inherit ClvR survive, resulting in an increase in ClvR frequency. ClvR is predicted to spread to fixation under diverse conditions. To test these predictions, we generated a ClvR element in Drosophila melanogasterClvRtko is located on chromosome 3 and uses Cas9 and four gRNAs to disrupt melanogaster technical knockout (tko), an X-linked essential gene. Rescue activity is provided by tko from Drosophila virilisClvRtko results in germline and maternal carryover-dependent inactivation of melanogaster tko (>99% per generation); lethality caused by this loss is rescued by the virilis transgene; ClvRtko activities are robust to genetic diversity in strains from five continents; and uncleavable but functional melanogaster tko alleles were not observed. Finally, ClvRtko spreads to transgene fixation. The simplicity of ClvR suggests it may be useful for altering populations in diverse species.
Collapse
Affiliation(s)
- Georg Oberhofer
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Tobin Ivy
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Bruce A Hay
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| |
Collapse
|
88
|
|
89
|
Engineered resistance to Zika virus in transgenic Aedes aegypti expressing a polycistronic cluster of synthetic small RNAs. Proc Natl Acad Sci U S A 2019; 116:3656-3661. [PMID: 30723148 PMCID: PMC6397566 DOI: 10.1073/pnas.1810771116] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Here, we describe the generation of Aedes aegypti mosquitoes that are engineered to be resistant to Zika virus (ZIKV) transmission. Our results demonstrate that engineered mosquitoes express a polycistronic cluster of synthetic small RNAs designed to target the ZIKV genome. As a result, homozygous mosquitoes were refractory to ZIKV infection, and therefore could not transmit the virus. Additionally, mosquitoes heterozygous for the transgene showed significantly lower levels of viral infection, dissemination, and transmission compared with wild-type mosquitoes; importantly, these levels were low enough to make such mosquitoes unlikely to transmit ZIKV to a susceptible host. Finally, we discuss how such an engineering approach can be used to combat the major health burden of ZIKV, and potentially other arboviruses, in the future. Recent Zika virus (ZIKV) outbreaks have highlighted the necessity for development of novel vector control strategies to combat arboviral transmission, including genetic versions of the sterile insect technique, artificial infection with Wolbachia to reduce population size and/or vectoring competency, and gene drive-based methods. Here, we describe the development of mosquitoes synthetically engineered to impede vector competence to ZIKV. We demonstrate that a polycistronic cluster of engineered synthetic small RNAs targeting ZIKV is expressed and fully processed in Aedes aegypti, ensuring the formation of mature synthetic small RNAs in the midgut where ZIKV resides in the early stages of infection. Critically, we demonstrate that engineered Ae. aegypti mosquitoes harboring the anti-ZIKV transgene have significantly reduced viral infection, dissemination, and transmission rates of ZIKV. Taken together, these compelling results provide a promising path forward for development of effective genetic-based ZIKV control strategies, which could potentially be extended to curtail other arboviruses.
Collapse
|
90
|
Wu Y, Hu W, Biedler JK, Chen XG, Tu ZJ. Pure early zygotic genes in the Asian malaria mosquito Anopheles stephensi. Parasit Vectors 2018; 11:652. [PMID: 30583723 PMCID: PMC6304767 DOI: 10.1186/s13071-018-3220-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The Asian malaria mosquito, Anopheles stephensi, is a major urban malaria vector in the Middle East and on the Indian subcontinent. Early zygotic transcription, which marks the maternal-to-zygotic transition, has not been systematically studied in An. stephensi or any other Anopheles mosquitoes. Improved understanding of early embryonic gene expression in An. stephensi will facilitate genetic and evolutionary studies and help with the development of novel control strategies for this important disease vector. RESULTS We obtained RNA-seq data in biological triplicates from four early An. stephensi embryonic time points. Using these data, we identified 70 and 153 pure early zygotic genes (pEZGs) under stringent and relaxed conditions, respectively. We show that these pEZGs are enriched in functional groups related to DNA-binding transcription regulators, cell cycle modulators, proteases, transport, and cellular metabolism. On average these pEZGs are shorter and have less introns than other An. stephensi genes. Some of the pEZGs may arise de novo while others have clear non-pEZG paralogs. There is no or very limited overlap between An. stephensi pEZGs and Drosophila melanogaster or Aedes aegypti pEZGs. Interestingly, the upstream region of An. stephensi pEZGs lack significant enrichment of a previously reported TAGteam/VBRGGTA motif found in the regulatory region of pEZGs in D. melanogaster and Ae. aegypti. However, a GT-rich motif was found in An. stephensi pEZGs instead. CONCLUSIONS We have identified a number of pEZGs whose predicted functions and structures are consistent with their collective roles in the degradation of maternally deposited components, activation of the zygotic genome, cell division, and metabolism. The pEZGs appear to rapidly turn over within the Dipteran order and even within the Culicidae family. These pEZGs, and the shared regulatory motif, could provide the promoter or regulatory sequences to drive gene expression in the syncytial or early cellular blastoderm, a period when the developing embryo is accessible to genetic manipulation. In addition, these molecular resources may be used to achieve sex separation of mosquitoes for sterile insect technique.
Collapse
Affiliation(s)
- Yang Wu
- Department of Pathogen Biology, School of Public Health, Southern Medical, University, Guangzhou, Guangdong, 510515, People's Republic of China.,Department of Biochemistry, Engel Hall, Blacksburg, VA, 24061, USA.,Fralin Life Science Institute, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Wanqi Hu
- Department of Biochemistry, Engel Hall, Blacksburg, VA, 24061, USA.,Fralin Life Science Institute, Virginia Tech, Blacksburg, VA, 24061, USA
| | - James K Biedler
- Department of Biochemistry, Engel Hall, Blacksburg, VA, 24061, USA.,Fralin Life Science Institute, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Xiao-Guang Chen
- Department of Pathogen Biology, School of Public Health, Southern Medical, University, Guangzhou, Guangdong, 510515, People's Republic of China.
| | - Zhijian Jake Tu
- Department of Biochemistry, Engel Hall, Blacksburg, VA, 24061, USA. .,Fralin Life Science Institute, Virginia Tech, Blacksburg, VA, 24061, USA.
| |
Collapse
|
91
|
Abstract
Background Aedes aegypti is an important mosquito vector that transmits arboviruses that cause devastating diseases including Zika, dengue fever, yellow fever and chikungunya. Improved understanding of gene regulation in the early development of Ae. aegypti will facilitate genetic studies and help the development of novel control strategies of this important disease vector. Results In this study, we demonstrated through transgenic assays that the promoter of an endogenous early zygotic gene KLC2 could drive gene expression in the syncytial blastoderm and early cellular blastoderm, which is a stage that the developing germline and the rest of embryo are accessible to genetic manipulation. An unexpected expression of the reporter gene in transgenic male testes was also observed. Further analysis confirmed the expression of the endogenous KLC2 in the testes, which was not detected in the previous RNA sequencing data. Conclusions Our finding provided a new promoter element that can be used in future genetic studies and applications in Ae. aegypti. Moreover, our transgenic reporter assays showed that cautions are needed when interpreting RNA sequencing data as transient or tissue-specific transcription may go undetected by RNAseq. Electronic supplementary material The online version of this article (10.1186/s13071-018-3210-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wanqi Hu
- Department of Biochemistry, Virginia Tech, 303 Fralin, Blacksburg, VA, 24061, USA.,Fralin Life Science Institute, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Zhijian Jake Tu
- Department of Biochemistry, Virginia Tech, 303 Fralin, Blacksburg, VA, 24061, USA. .,Fralin Life Science Institute, Virginia Tech, Blacksburg, VA, 24061, USA.
| |
Collapse
|
92
|
Chai A, Mateus AM, Oozeer F, Sousa-Nunes R. Spatiotemporally controlled genetic perturbation for efficient large-scale studies of cell non-autonomous effects. eLife 2018; 7:e38393. [PMID: 30479273 PMCID: PMC6320068 DOI: 10.7554/elife.38393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 11/26/2018] [Indexed: 11/13/2022] Open
Abstract
Studies in genetic model organisms have revealed much about the development and pathology of complex tissues. Most have focused on cell-intrinsic gene functions and mechanisms. Much less is known about how transformed, or otherwise functionally disrupted, cells interact with healthy ones toward a favorable or pathological outcome. This is largely due to technical limitations. We developed new genetic tools in Drosophila melanogaster that permit efficient multiplexed gain- and loss-of-function genetic perturbations with separable spatial and temporal control. Importantly, our novel tool-set is independent of the commonly used GAL4/UAS system, freeing the latter for additional, non-autonomous, genetic manipulations; and is built into a single strain, allowing one-generation interrogation of non-autonomous effects. Altogether, our design opens up efficient genome-wide screens on any deleterious phenotype, once plasmid or genome engineering is used to place the desired miRNA(s) or ORF(s) into our genotype. Specifically, we developed tools to study extrinsic effects on neural tumor growth but the strategy presented has endless applications within and beyond neurobiology, and in other model organisms.
Collapse
Affiliation(s)
- Andrea Chai
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and NeuroscienceKing’s College LondonLondonUnited Kingdom
| | - Ana M Mateus
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and NeuroscienceKing’s College LondonLondonUnited Kingdom
| | - Fazal Oozeer
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and NeuroscienceKing’s College LondonLondonUnited Kingdom
| | - Rita Sousa-Nunes
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and NeuroscienceKing’s College LondonLondonUnited Kingdom
| |
Collapse
|
93
|
Shaw WR, Catteruccia F. Vector biology meets disease control: using basic research to fight vector-borne diseases. Nat Microbiol 2018; 4:20-34. [PMID: 30150735 DOI: 10.1038/s41564-018-0214-7] [Citation(s) in RCA: 175] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 06/29/2018] [Indexed: 12/11/2022]
Abstract
Human pathogens that are transmitted by insects are a global problem, particularly those vectored by mosquitoes; for example, malaria parasites transmitted by Anopheles species, and viruses such as dengue, Zika and chikungunya that are carried by Aedes mosquitoes. Over the past 15 years, the prevalence of malaria has been substantially reduced and virus outbreaks have been contained by controlling mosquito vectors using insecticide-based approaches. However, disease control is now threatened by alarming rates of insecticide resistance in insect populations, prompting the need to develop a new generation of specific strategies that can reduce vector-mediated transmission. Here, we review how increased knowledge in insect biology and insect-pathogen interactions is stimulating new concepts and tools for vector control. We focus on strategies that either interfere with the development of pathogens within their vectors or directly impact insect survival, including enhancement of vector-mediated immune control, manipulation of the insect microbiome, or use of powerful new genetic tools such as CRISPR-Cas systems to edit vector genomes. Finally, we offer a perspective on the implementation hurdles as well as the knowledge gaps that must be filled in the coming years to safely realize the potential of these novel strategies to eliminate the scourge of vector-borne disease.
Collapse
Affiliation(s)
- W Robert Shaw
- Harvard T. H. Chan School of Public Health, Department of Immunology and Infectious Diseases, Boston, MA, USA.
| | - Flaminia Catteruccia
- Harvard T. H. Chan School of Public Health, Department of Immunology and Infectious Diseases, Boston, MA, USA.
| |
Collapse
|
94
|
Gantz VM, Akbari OS. Gene editing technologies and applications for insects. CURRENT OPINION IN INSECT SCIENCE 2018; 28:66-72. [PMID: 30551769 PMCID: PMC6296244 DOI: 10.1016/j.cois.2018.05.006] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 05/15/2018] [Accepted: 05/16/2018] [Indexed: 05/09/2023]
Abstract
Initially discovered in bacteria, CRISPR-based genome editing endonucleases have proven remarkably amenable for adaptation to insects. To date, these endonucleases have been utilized in a plethora of both model and non-model insects including diverse flies, bees, beetles, butterflies, moths, and grasshoppers, to name a few, thereby revolutionizing functional genomics of insects. In addition to basic genome editing, they have also been invaluable for advanced genome engineering and synthetic biology applications. Here we explore the recent genome editing advancements in insects for generating site-specific genomic mutations, insertions, deletions, as well as more advanced applications such as Homology Assisted Genome Knock-in (HACK), potential to utilize DNA base editing, generating predictable reciprocal chromosomal translocations, and development gene drives to control the fate of wild populations.
Collapse
Affiliation(s)
- Valentino M Gantz
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA 92092, USA; Tata Institute for Genetics and Society, University of California, San Diego, La Jolla, CA 92093, USA
| | - Omar S Akbari
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA 92092, USA; Tata Institute for Genetics and Society, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
95
|
Burt A, Deredec A. Self-limiting population genetic control with sex-linked genome editors. Proc Biol Sci 2018; 285:rspb.2018.0776. [PMID: 30051868 PMCID: PMC6083257 DOI: 10.1098/rspb.2018.0776] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 06/28/2018] [Indexed: 12/30/2022] Open
Abstract
In male heterogametic species the Y chromosome is transmitted solely from fathers to sons, and is selected for based only on its impacts on male fitness. This fact can be exploited to develop efficient pest control strategies that use Y-linked editors to disrupt the fitness of female descendants. With simple population genetic and dynamic models we show that Y-linked editors can be substantially more efficient than other self-limiting strategies and, while not as efficient as gene drive approaches, are expected to have less impact on non-target populations with which there is some gene flow. Efficiency can be further augmented by simultaneously releasing an autosomal X-shredder construct, in either the same or different males. Y-linked editors may be an attractive option to consider when efficient control of a species is desired in some locales but not others.
Collapse
Affiliation(s)
- Austin Burt
- Life Sciences, Imperial College, Silwood Park, Ascot SL5 7PY, UK
| | - Anne Deredec
- UMR BIOGER, INRA AgroParisTech, Université Paris-Saclay, Avenue Lucien Bretignières, 78850 Thiverval-Grignon, France
| |
Collapse
|
96
|
Mitochondrial dynamics regulates Drosophila intestinal stem cell differentiation. Cell Death Discov 2018; 4:17. [PMID: 30062062 PMCID: PMC6056485 DOI: 10.1038/s41420-018-0083-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 05/21/2018] [Accepted: 06/22/2018] [Indexed: 01/12/2023] Open
Abstract
Differentiation of stem/progenitor cells is associated with a substantial increase in mitochondrial mass and complexity. Mitochondrial dynamics, including the processes of fusion and fission, plays an important role for somatic cell reprogramming and pluripotency maintenance in induced pluripotent cells (iPSCs). However, the role of mitochondrial dynamics during stem/progenitor cell differentiation in vivo remains elusive. Here we found differentiation of Drosophila intestinal stem cell is accompanied with continuous mitochondrial fusion. Mitochondrial fusion defective(opa1RNAi) ISCs contain less mitochondrial membrane potential, reduced ATP, and increased ROS level. Surprisingly, suppressing fusion also resulted in the failure of progenitor cells to differentiate. Cells did not switch on the expression of differentiation markers, and instead continued to show characteristics of progenitor cells. Meanwhile, proliferation or apoptosis was unaffected. The differentiation defect could be rescued by concomitant inhibition of Drp1, a mitochondrial fission molecule. Moreover, ROS scavenger also partially rescues opa1RNAi-associated differentiation defects via down-regulating JNK activity. We propose that mitochondrial fusion plays a pivotal role in controlling the developmental switch of stem cell fate.
Collapse
|
97
|
Noble C, Adlam B, Church GM, Esvelt KM, Nowak MA. Current CRISPR gene drive systems are likely to be highly invasive in wild populations. eLife 2018; 7:33423. [PMID: 29916367 PMCID: PMC6014726 DOI: 10.7554/elife.33423] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 05/15/2018] [Indexed: 12/25/2022] Open
Abstract
Recent reports have suggested that self-propagating CRISPR-based gene drive systems are unlikely to efficiently invade wild populations due to drive-resistant alleles that prevent cutting. Here we develop mathematical models based on existing empirical data to explicitly test this assumption for population alteration drives. Our models show that although resistance prevents spread to fixation in large populations, even the least effective drive systems reported to date are likely to be highly invasive. Releasing a small number of organisms will often cause invasion of the local population, followed by invasion of additional populations connected by very low rates of gene flow. Hence, initiating contained field trials as tentatively endorsed by the National Academies report on gene drive could potentially result in unintended spread to additional populations. Our mathematical results suggest that self-propagating gene drive is best suited to applications such as malaria prevention that seek to affect all wild populations of the target species.
Collapse
Affiliation(s)
- Charleston Noble
- Program for Evolutionary Dynamics, Harvard University, Cambridge, United States.,Department of Genetics, Harvard Medical School, Harvard University, Boston, United States.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston MA, United States
| | - Ben Adlam
- Program for Evolutionary Dynamics, Harvard University, Cambridge, United States.,School of Engineering and Applied Science, Harvard University, Cambridge, United States
| | - George M Church
- Department of Genetics, Harvard Medical School, Harvard University, Boston, United States.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston MA, United States
| | - Kevin M Esvelt
- Massachusetts Institute of Technology Media Lab, Cambridge, United States
| | - Martin A Nowak
- Program for Evolutionary Dynamics, Harvard University, Cambridge, United States.,Department of Mathematics, Harvard University, Cambridge, United States.,Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, United States
| |
Collapse
|
98
|
Ren Q, Awasaki T, Wang YC, Huang YF, Lee T. Lineage-guided Notch-dependent gliogenesis by Drosophila multi-potent progenitors. Development 2018; 145:dev.160127. [PMID: 29764857 DOI: 10.1242/dev.160127] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 05/08/2018] [Indexed: 12/27/2022]
Abstract
Macroglial cells in the central nervous system exhibit regional specialization and carry out region-specific functions. Diverse glial cells arise from specific progenitors in specific spatiotemporal patterns. This raises an interesting possibility that glial precursors with distinct developmental fates exist that govern region-specific gliogenesis. Here, we have mapped the glial progeny produced by the Drosophila type II neuroblasts, which, like vertebrate radial glia cells, yield both neurons and glia via intermediate neural progenitors (INPs). Distinct type II neuroblasts produce different characteristic sets of glia. A single INP can make both astrocyte-like and ensheathing glia, which co-occupy a relatively restrictive subdomain. Blocking apoptosis uncovers further lineage distinctions in the specification, proliferation and survival of glial precursors. Both the switch from neurogenesis to gliogenesis and the subsequent glial expansion depend on Notch signaling. Taken together, lineage origins preconfigure the development of individual glial precursors with involvement of serial Notch actions in promoting gliogenesis.
Collapse
Affiliation(s)
- Qingzhong Ren
- Howard Hughes Medical Institute, Janelia Research Campus, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Takeshi Awasaki
- Howard Hughes Medical Institute, Janelia Research Campus, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Yu-Chun Wang
- Howard Hughes Medical Institute, Janelia Research Campus, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Yu-Fen Huang
- Howard Hughes Medical Institute, Janelia Research Campus, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Tzumin Lee
- Howard Hughes Medical Institute, Janelia Research Campus, 19700 Helix Drive, Ashburn, VA 20147, USA
| |
Collapse
|
99
|
Buchman AB, Ivy T, Marshall JM, Akbari OS, Hay BA. Engineered Reciprocal Chromosome Translocations Drive High Threshold, Reversible Population Replacement in Drosophila. ACS Synth Biol 2018. [PMID: 29608276 DOI: 10.1101/088393] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Replacement of wild insect populations with transgene-bearing individuals unable to transmit disease or survive under specific environmental conditions using gene drive provides a self-perpetuating method of disease prevention. Mechanisms that require the gene drive element and linked cargo to exceed a high threshold frequency in order for spread to occur are attractive because they offer several points of control: they bring about local, but not global population replacement; and transgenes can be eliminated by reintroducing wildtypes into the population so as to drive the frequency of transgenes below the threshold frequency required for drive. Reciprocal chromosome translocations were proposed as a tool for bringing about high threshold population replacement in 1940 and 1968. However, translocations able to achieve this goal have only been reported once, in the spider mite Tetranychus urticae, a haplo-diploid species in which there is strong selection in haploid males for fit homozygotes. We report the creation of engineered translocation-bearing strains of Drosophila melanogaster, generated through targeted chromosomal breakage and homologous recombination. These strains drive high threshold population replacement in laboratory populations. While it remains to be shown that engineered translocations can bring about population replacement in wild populations, these observations suggest that further exploration of engineered translocations as a tool for controlled population replacement is warranted.
Collapse
Affiliation(s)
- Anna B Buchman
- Division of Biology and Biological Engineering , California Institute of Technology , Pasadena , California 91125 , United States
- Division of Biological Sciences , University of California , San Diego , California 92161 , United States
| | - Tobin Ivy
- Division of Biology and Biological Engineering , California Institute of Technology , Pasadena , California 91125 , United States
| | - John M Marshall
- School of Public Health , University of California , Berkeley , California 94720 , United States
| | - Omar S Akbari
- Division of Biology and Biological Engineering , California Institute of Technology , Pasadena , California 91125 , United States
- Division of Biological Sciences , University of California , San Diego , California 92161 , United States
| | - Bruce A Hay
- Division of Biology and Biological Engineering , California Institute of Technology , Pasadena , California 91125 , United States
- Division of Biological Sciences , University of California , San Diego , California 92161 , United States
| |
Collapse
|
100
|
Buchman AB, Ivy T, Marshall JM, Akbari OS, Hay BA. Engineered Reciprocal Chromosome Translocations Drive High Threshold, Reversible Population Replacement in Drosophila. ACS Synth Biol 2018; 7:1359-1370. [PMID: 29608276 DOI: 10.1021/acssynbio.7b00451] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Replacement of wild insect populations with transgene-bearing individuals unable to transmit disease or survive under specific environmental conditions using gene drive provides a self-perpetuating method of disease prevention. Mechanisms that require the gene drive element and linked cargo to exceed a high threshold frequency in order for spread to occur are attractive because they offer several points of control: they bring about local, but not global population replacement; and transgenes can be eliminated by reintroducing wildtypes into the population so as to drive the frequency of transgenes below the threshold frequency required for drive. Reciprocal chromosome translocations were proposed as a tool for bringing about high threshold population replacement in 1940 and 1968. However, translocations able to achieve this goal have only been reported once, in the spider mite Tetranychus urticae, a haplo-diploid species in which there is strong selection in haploid males for fit homozygotes. We report the creation of engineered translocation-bearing strains of Drosophila melanogaster, generated through targeted chromosomal breakage and homologous recombination. These strains drive high threshold population replacement in laboratory populations. While it remains to be shown that engineered translocations can bring about population replacement in wild populations, these observations suggest that further exploration of engineered translocations as a tool for controlled population replacement is warranted.
Collapse
Affiliation(s)
- Anna B Buchman
- Division of Biology and Biological Engineering , California Institute of Technology , Pasadena , California 91125 , United States
- Division of Biological Sciences , University of California , San Diego , California 92161 , United States
| | - Tobin Ivy
- Division of Biology and Biological Engineering , California Institute of Technology , Pasadena , California 91125 , United States
| | - John M Marshall
- School of Public Health , University of California , Berkeley , California 94720 , United States
| | - Omar S Akbari
- Division of Biology and Biological Engineering , California Institute of Technology , Pasadena , California 91125 , United States
- Division of Biological Sciences , University of California , San Diego , California 92161 , United States
| | - Bruce A Hay
- Division of Biology and Biological Engineering , California Institute of Technology , Pasadena , California 91125 , United States
- Division of Biological Sciences , University of California , San Diego , California 92161 , United States
| |
Collapse
|