1
|
Ukegbu CV, Mohamed M, Hoermann A, Qin Y, Kweyamba PA, Lwetoijera DW, Windbichler N, Moore S, Christophides GK, Vlachou D. Nanobody-mediated targeting of Plasmodium falciparum PfPIMMS43 can block malaria transmission in mosquitoes. Commun Biol 2025; 8:683. [PMID: 40301628 PMCID: PMC12041390 DOI: 10.1038/s42003-025-08033-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 04/02/2025] [Indexed: 05/01/2025] Open
Abstract
The transition from ookinete to oocyst is a critical step in the Plasmodium falciparum lifecycle and an important target for malaria transmission-blocking strategies. PfPIMMS43, a surface protein of P. falciparum ookinetes and sporozoites, is critical for this transition and aids the parasite in evading mosquito immune responses. Previous studies demonstrated that polyclonal PfPIMMS43 antibodies reduced P. falciparum infection in Anopheles mosquitoes. Here, building on these findings, we have developed high-affinity single-domain VHH antibodies (nanobodies) derived from llama heavy-chain-only antibodies. We have shown that these nanobodies bind both recombinant and endogenous PfPIMMS43 produced by P. falciparum ookinetes in the mosquito midgut. Importantly, they significantly reduce infection intensity and prevalence of laboratory and field strains of P. falciparum in An. coluzzii and An. gambiae, respectively. Epitope mapping has revealed that the nanobodies target conserved regions in the second half of PfPIMMS43, with homology modelling confirming epitope accessibility. These findings establish PfPIMMS43 as a promising transmission-blocking target. To enhance malaria control and elimination efforts, we propose an innovative strategy in which genetically modified mosquitoes express PfPIMMS43-specific nanobodies in their midguts and spread this trait in wild mosquito populations via gene drive technology.
Collapse
Affiliation(s)
| | - Mgeni Mohamed
- Environmental Health and Ecological Sciences, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Astrid Hoermann
- Department of Life Sciences, Imperial College London, London, UK
| | - Yuyan Qin
- Department of Life Sciences, Imperial College London, London, UK
| | - Prisca A Kweyamba
- Environmental Health and Ecological Sciences, Ifakara Health Institute, Bagamoyo, Tanzania
| | | | | | - Sarah Moore
- Environmental Health and Ecological Sciences, Ifakara Health Institute, Bagamoyo, Tanzania
- Swiss Tropical and Public Health Institute, Kreuzstrasse 2, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | | | - Dina Vlachou
- Department of Life Sciences, Imperial College London, London, UK.
| |
Collapse
|
2
|
He S, Zheng S, Hu Y, Yang Z, Zhu H, Yu B, Wei J, Pan G, Zhou Z, Li C. The unique transmembrane protein NbTMP2 (NBO_555g0004) of Nosema bombycis contributes to host infection. J Invertebr Pathol 2025; 211:108320. [PMID: 40120668 DOI: 10.1016/j.jip.2025.108320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/18/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025]
Abstract
Nosema bombycis is one of the earliest discovered microsporidia and can infect silkworms through both horizontal and transovarial transmission, posing a significant threat to the sericulture industry. Microsporidia can form mature dormant spores with a thick proteinaceous and chitin-rich wall that shields them from environmental stress, leading to difficulties in prevention and control. However, the intracellular proliferative phase is a particularly active life stage for the pathogen. At this stage, the plasma membrane of pathogen is exposed to the cytoplasm of the host cells without the protection of the spore walls, which make it an ideal phase for intervention. In this study, based on transcriptomic data, we identified that a transmembrane protein of N. bombycis, NbTMP2 (NBO_555g0004), was highly expressed after infection with microsporidia. Sequence analysis of the cloned NbTMP2 gene revealed that the protein consists of 253 amino acids, including a signal peptide and a transmembrane domain. Indirect immunofluorescence analysis (IFA) showed that NbTMP2 is localized to the plasma membrane. Furthermore, IFA, RT-qPCR, and western blotting indicated that NbTMP2 is expressed at all developmental stages, with a significant upregulation during the early proliferative phase of N. bombycis. The interference of NbTMP2 confirmed that downregulation of NbTMP2 expression significantly inhibit the proliferation of N. bombycis. In conclusion, NbTMP2 is a membrane protein that plays a role during the development of N. bombycis. This study provides a potential target for inhibiting the proliferation of N. bombycis and lays a foundation for future research on breeding N. bombycis-resistant silkworms.
Collapse
Affiliation(s)
- Shaogang He
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China
| | - Shiyi Zheng
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China; Affiliated Jinhua Hospital, Zhejiang University of Medicine, Jinhua Municipal Central Hospital, Jinhua, Zhejiang 321000, China
| | - Yuanke Hu
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China
| | - Zhonghua Yang
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China
| | - Honglin Zhu
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China
| | - Bin Yu
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China; Laboratory of Tissue and Cell Biology, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing 400016, China
| | - Junhong Wei
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China
| | - Guoqing Pan
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China
| | - Zeyang Zhou
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China; College of Life Sciences, Chongqing Normal University, Chongqing 401331, China
| | - Chunfeng Li
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China.
| |
Collapse
|
3
|
Kefi M, Cardoso-Jaime V, Saab SA, Dimopoulos G. Curing mosquitoes with genetic approaches for malaria control. Trends Parasitol 2024; 40:487-499. [PMID: 38760256 DOI: 10.1016/j.pt.2024.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/15/2024] [Accepted: 04/15/2024] [Indexed: 05/19/2024]
Abstract
Malaria remains a persistent global public health challenge because of the limitations of current prevention tools. The use of transgenic mosquitoes incapable of transmitting malaria, in conjunction with existing methods, holds promise for achieving elimination of malaria and preventing its reintroduction. In this context, population modification involves the spread of engineered genetic elements through mosquito populations that render them incapable of malaria transmission. Significant progress has been made in this field over the past decade in revealing promising targets, optimizing genetic tools, and facilitating the transition from the laboratory to successful field deployments, which are subject to regulatory scrutiny. This review summarizes recent advances and ongoing challenges in 'curing' Anopheles vectors of the malaria parasite.
Collapse
Affiliation(s)
- Mary Kefi
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Victor Cardoso-Jaime
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Sally A Saab
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - George Dimopoulos
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
4
|
Cai JA, Christophides GK. Immune interactions between mosquitoes and microbes during midgut colonization. CURRENT OPINION IN INSECT SCIENCE 2024; 63:101195. [PMID: 38552792 DOI: 10.1016/j.cois.2024.101195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 03/14/2024] [Accepted: 03/21/2024] [Indexed: 04/07/2024]
Abstract
Mosquitoes encounter diverse microbes during their lifetime, including symbiotic bacteria, shaping their midgut ecosystem. The organization of the midgut supports microbiota persistence while defending against potential pathogens. The influx of nutrients during blood feeding triggers bacterial proliferation, challenging host homeostasis. Immune responses, aimed at controlling bacterial overgrowth, impact blood-borne pathogens such as malaria parasites. However, parasites deploy evasion strategies against mosquito immunity. Leveraging these mechanisms could help engineer malaria-resistant mosquitoes, offering a transformative tool for malaria elimination.
Collapse
Affiliation(s)
- Julia A Cai
- Department of Life Sciences, Imperial College London, Exhibition Road, SW7 2AZ London, United Kingdom
| | - George K Christophides
- Department of Life Sciences, Imperial College London, Exhibition Road, SW7 2AZ London, United Kingdom.
| |
Collapse
|
5
|
Weng SC, Masri RA, Akbari OS. Advances and challenges in synthetic biology for mosquito control. Trends Parasitol 2024; 40:75-88. [PMID: 38000957 PMCID: PMC11064511 DOI: 10.1016/j.pt.2023.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/26/2023] [Accepted: 11/01/2023] [Indexed: 11/26/2023]
Abstract
Mosquito-borne illnesses represent a significant global health peril, resulting in approximately one million fatalities annually. West Nile, dengue, Zika, and malaria are continuously expanding their global reach, driven by factors that escalate mosquito populations and pathogen transmission. Innovative control measures are imperative to combat these catastrophic ailments. Conventional approaches, such as eliminating breeding sites and using insecticides, have been helpful, but they face challenges such as insecticide resistance and environmental harm. Given the mounting severity of mosquito-borne diseases, there is promise in exploring innovative approaches using synthetic biology to bolster mosquitoes' resistance to pathogens, or even eliminate the mosquito vectors, as a means of control. This review outlines current strategies, future goals, and the importance of gene editing for global health defenses against mosquito-borne diseases.
Collapse
Affiliation(s)
- Shih-Che Weng
- School of Biological Sciences, Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Reem A Masri
- School of Biological Sciences, Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Omar S Akbari
- School of Biological Sciences, Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
6
|
Yu B, Zheng R, Bian M, Liu T, Lu K, Bao J, Pan G, Zhou Z, Li C. A monoclonal antibody targeting spore wall protein 1 inhibits the proliferation of Nosema bombycis in Bombyx mori. Microbiol Spectr 2023; 11:e0068123. [PMID: 37811955 PMCID: PMC10714992 DOI: 10.1128/spectrum.00681-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 08/24/2023] [Indexed: 10/10/2023] Open
Abstract
IMPORTANCE There are a few reports on the resistance of microsporidia, including Nosema bombycis. Here, the alkali-soluble germination proteins of N. bombycis were used as immunogens to prepare a monoclonal antibody, and its single-chain variable fragments effectively blocked microsporidia infection. Our study has provided novel strategies for microsporidiosis control and demonstrated a useful method for the potential treatment of other microsporidia diseases.
Collapse
Affiliation(s)
- Bin Yu
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
- Chongqing Key Laboratory of Microsporidia Infection and Prevention, Southwest University, Chongqing, China
| | - Rong Zheng
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Maofei Bian
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
- Chongqing Three Gorges Medical College, Chongqing, China
| | - Ting Liu
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Kun Lu
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Jialing Bao
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
- Chongqing Key Laboratory of Microsporidia Infection and Prevention, Southwest University, Chongqing, China
| | - Guoqing Pan
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
- Chongqing Key Laboratory of Microsporidia Infection and Prevention, Southwest University, Chongqing, China
| | - Zeyang Zhou
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
- Chongqing Key Laboratory of Microsporidia Infection and Prevention, Southwest University, Chongqing, China
- College of Life Sciences, Chongqing Normal University, Chongqing, China
| | - Chunfeng Li
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
- Chongqing Key Laboratory of Microsporidia Infection and Prevention, Southwest University, Chongqing, China
| |
Collapse
|
7
|
Green EI, Jaouen E, Klug D, Proveti Olmo R, Gautier A, Blandin S, Marois E. A population modification gene drive targeting both Saglin and Lipophorin impairs Plasmodium transmission in Anopheles mosquitoes. eLife 2023; 12:e93142. [PMID: 38051195 PMCID: PMC10786457 DOI: 10.7554/elife.93142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 11/14/2023] [Indexed: 12/07/2023] Open
Abstract
Lipophorin is an essential, highly expressed lipid transport protein that is secreted and circulates in insect hemolymph. We hijacked the Anopheles coluzzii Lipophorin gene to make it co-express a single-chain version of antibody 2A10, which binds sporozoites of the malaria parasite Plasmodium falciparum. The resulting transgenic mosquitoes show a markedly decreased ability to transmit Plasmodium berghei expressing the P. falciparum circumsporozoite protein to mice. To force the spread of this antimalarial transgene in a mosquito population, we designed and tested several CRISPR/Cas9-based gene drives. One of these is installed in, and disrupts, the pro-parasitic gene Saglin and also cleaves wild-type Lipophorin, causing the anti-malarial modified Lipophorin version to replace the wild type and hitch-hike together with the Saglin drive. Although generating drive-resistant alleles and showing instability in its gRNA-encoding multiplex array, the Saglin-based gene drive reached high levels in caged mosquito populations and efficiently promoted the simultaneous spread of the antimalarial Lipophorin::Sc2A10 allele. This combination is expected to decrease parasite transmission via two different mechanisms. This work contributes to the design of novel strategies to spread antimalarial transgenes in mosquitoes, and illustrates some expected and unexpected outcomes encountered when establishing a population modification gene drive.
Collapse
Affiliation(s)
- Emily I Green
- Inserm U1257, CNRS UPR9022, University of StrasbourgStrasbourgFrance
| | - Etienne Jaouen
- Inserm U1257, CNRS UPR9022, University of StrasbourgStrasbourgFrance
| | - Dennis Klug
- Inserm U1257, CNRS UPR9022, University of StrasbourgStrasbourgFrance
| | | | - Amandine Gautier
- Inserm U1257, CNRS UPR9022, University of StrasbourgStrasbourgFrance
| | - Stéphanie Blandin
- Inserm U1257, CNRS UPR9022, University of StrasbourgStrasbourgFrance
| | - Eric Marois
- Inserm U1257, CNRS UPR9022, University of StrasbourgStrasbourgFrance
| |
Collapse
|
8
|
Kormos A, Dimopoulos G, Bier E, Lanzaro GC, Marshall JM, James AA. Conceptual risk assessment of mosquito population modification gene-drive systems to control malaria transmission: preliminary hazards list workshops. Front Bioeng Biotechnol 2023; 11:1261123. [PMID: 37965050 PMCID: PMC10641379 DOI: 10.3389/fbioe.2023.1261123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 10/09/2023] [Indexed: 11/16/2023] Open
Abstract
The field-testing and eventual adoption of genetically-engineered mosquitoes (GEMs) to control vector-borne pathogen transmission will require them meeting safety criteria specified by regulatory authorities in regions where the technology is being considered for use and other locales that might be impacted. Preliminary risk considerations by researchers and developers may be useful for planning the baseline data collection and field research used to address the anticipated safety concerns. Part of this process is to identify potential hazards (defined as the inherent ability of an entity to cause harm) and their harms, and then chart the pathways to harm and evaluate their probability as part of a risk assessment. The University of California Malaria Initiative (UCMI) participated in a series of workshops held to identify potential hazards specific to mosquito population modification strains carrying gene-drive systems coupled to anti-parasite effector genes and their use in a hypothetical island field trial. The hazards identified were placed within the broader context of previous efforts discussed in the scientific literature. Five risk areas were considered i) pathogens, infections and diseases, and the impacts of GEMs on human and animal health, ii) invasiveness and persistence of GEMs, and interactions of GEMs with target organisms, iii) interactions of GEMs with non-target organisms including horizontal gene transfer, iv) impacts of techniques used for the management of GEMs and v) evolutionary and stability considerations. A preliminary hazards list (PHL) was developed and is made available here. This PHL is useful for internal project risk evaluation and is available to regulators at prospective field sites. UCMI project scientists affirm that the subsequent processes associated with the comprehensive risk assessment for the application of this technology should be driven by the stakeholders at the proposed field site and areas that could be affected by this intervention strategy.
Collapse
Affiliation(s)
- Ana Kormos
- Vector Genetics Laboratory, University of California, Davis, Davis, CA, United States
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Malaria Research Institute, Johns Hopkins University, Baltimore, MD, United States
| | - Ethan Bier
- Department of Cell and Developmental Biology, University of California, San Diego, San Diego, CA, United States
| | - Gregory C. Lanzaro
- Vector Genetics Laboratory, University of California, Davis, Davis, CA, United States
| | - John M. Marshall
- Divisions of Epidemiology and Biostatistics, School of Public Health, University of California, Berkeley, Berkeley, CA, United States
| | - Anthony A. James
- Departments of Microbiology and Molecular Genetics and Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
9
|
Cardoso-Jaime V, Maya-Maldonado K, Tsutsumi V, Hernández-Martínez S. Mosquito pericardial cells upregulate Cecropin expression after an immune challenge. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 147:104745. [PMID: 37268262 DOI: 10.1016/j.dci.2023.104745] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/04/2023]
Abstract
Most mosquito-transmitted pathogens grow or replicate in the midgut before invading the salivary glands. Pathogens are exposed to several immunological factors along the way. Recently, it was shown that hemocytes gather near the periostial region of the heart to efficiently phagocytose pathogens circulating in the hemolymph. Nerveless, not all pathogens can be phagocyted by hemocytes and eliminated by lysis. Interestingly, some studies have shown that pericardial cells (PCs) surrounding periostial regions, may produce humoral factors, such as lysozymes. Our current work provides evidence that Anopheles albimanus PCs are a major producer of Cecropin 1 (Cec1). Furthermore, our findings reveal that after an immunological challenge, PCs upregulate Cec1 expression. We conclude that PCs are positioned in a strategic location that could allow releasing humoral components, such as cecropin, to lyse pathogens on the heart or circulating in the hemolymph, implying that PCs could play a significant role in the systemic immune response.
Collapse
Affiliation(s)
- Victor Cardoso-Jaime
- Centro de Investigaciones Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública. Av. Universidad 655, Santa María Ahuacatitlan, Cuernavaca, Morelos, C.P. 62100, Mexico; Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados, IPN. Av. Instituto Politécnico Nacional 2508, Gustavo A. Madero, Ciudad de México, C.P. 07360, Mexico
| | - Krystal Maya-Maldonado
- Centro de Investigaciones Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública. Av. Universidad 655, Santa María Ahuacatitlan, Cuernavaca, Morelos, C.P. 62100, Mexico; Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados, IPN. Av. Instituto Politécnico Nacional 2508, Gustavo A. Madero, Ciudad de México, C.P. 07360, Mexico
| | - Víctor Tsutsumi
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados, IPN. Av. Instituto Politécnico Nacional 2508, Gustavo A. Madero, Ciudad de México, C.P. 07360, Mexico.
| | - Salvador Hernández-Martínez
- Centro de Investigaciones Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública. Av. Universidad 655, Santa María Ahuacatitlan, Cuernavaca, Morelos, C.P. 62100, Mexico.
| |
Collapse
|
10
|
Carballar-Lejarazú R, Dong Y, Pham TB, Tushar T, Corder RM, Mondal A, Sánchez C. HM, Lee HF, Marshall JM, Dimopoulos G, James AA. Dual effector population modification gene-drive strains of the African malaria mosquitoes, Anopheles gambiae and Anopheles coluzzii. Proc Natl Acad Sci U S A 2023; 120:e2221118120. [PMID: 37428915 PMCID: PMC10629562 DOI: 10.1073/pnas.2221118120] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 06/05/2023] [Indexed: 07/12/2023] Open
Abstract
Proposed genetic approaches for reducing human malaria include population modification, which introduces genes into vector mosquitoes to reduce or prevent parasite transmission. We demonstrate the potential of Cas9/guide RNA (gRNA)-based gene-drive systems linked to dual antiparasite effector genes to spread rapidly through mosquito populations. Two strains have an autonomous gene-drive system coupled to dual anti-Plasmodium falciparum effector genes comprising single-chain variable fragment monoclonal antibodies targeting parasite ookinetes and sporozoites in the African malaria mosquitoes Anopheles gambiae (AgTP13) and Anopheles coluzzii (AcTP13). The gene-drive systems achieved full introduction within 3 to 6 mo after release in small cage trials. Life-table analyses revealed no fitness loads affecting AcTP13 gene-drive dynamics but AgTP13 males were less competitive than wild types. The effector molecules reduced significantly both parasite prevalence and infection intensities. These data supported transmission modeling of conceptual field releases in an island setting that shows meaningful epidemiological impacts at different sporozoite threshold levels (2.5 to 10 k) for human infection by reducing malaria incidence in optimal simulations by 50 to 90% within as few as 1 to 2 mo after a series of releases, and by ≥90% within 3 mo. Modeling outcomes for low sporozoite thresholds are sensitive to gene-drive system fitness loads, gametocytemia infection intensities during parasite challenges, and the formation of potentially drive-resistant genome target sites, extending the predicted times to achieve reduced incidence. TP13-based strains could be effective for malaria control strategies following validation of sporozoite transmission threshold numbers and testing field-derived parasite strains. These or similar strains are viable candidates for future field trials in a malaria-endemic region.
Collapse
Affiliation(s)
| | - Yuemei Dong
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Malaria Research Institute, Johns Hopkins University, Baltimore, MD21205
| | - Thai Binh Pham
- Department of Microbiology & Molecular Genetics, University of California, Irvine, CA92697-4025
| | - Taylor Tushar
- Department of Microbiology & Molecular Genetics, University of California, Irvine, CA92697-4025
| | - Rodrigo M. Corder
- Divisions of Epidemiology and Biostatistics, School of Public Health, University of California, Berkeley, CA94720
| | - Agastya Mondal
- Divisions of Epidemiology and Biostatistics, School of Public Health, University of California, Berkeley, CA94720
| | - Héctor M. Sánchez C.
- Divisions of Epidemiology and Biostatistics, School of Public Health, University of California, Berkeley, CA94720
| | - Hsu-Feng Lee
- Department of Microbiology & Molecular Genetics, University of California, Irvine, CA92697-4025
| | - John M. Marshall
- Divisions of Epidemiology and Biostatistics, School of Public Health, University of California, Berkeley, CA94720
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Malaria Research Institute, Johns Hopkins University, Baltimore, MD21205
| | - Anthony A. James
- Department of Microbiology & Molecular Genetics, University of California, Irvine, CA92697-4025
- Department of Molecular Biology & Biochemistry, University of California, Irvine, CA92697-3900
| |
Collapse
|
11
|
|
12
|
Bottino-Rojas V, James AA. Use of Insect Promoters in Genetic Engineering to Control Mosquito-Borne Diseases. Biomolecules 2022; 13:16. [PMID: 36671401 PMCID: PMC9855440 DOI: 10.3390/biom13010016] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 12/16/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022] Open
Abstract
Mosquito transgenesis and gene-drive technologies provide the basis for developing promising new tools for vector-borne disease prevention by either suppressing wild mosquito populations or reducing their capacity from transmitting pathogens. Many studies of the regulatory DNA and promoters of genes with robust sex-, tissue- and stage-specific expression profiles have supported the development of new tools and strategies that could bring mosquito-borne diseases under control. Although the list of regulatory elements available is significant, only a limited set of those can reliably drive spatial-temporal expression. Here, we review the advances in our ability to express beneficial and other genes in mosquitoes, and highlight the information needed for the development of new mosquito-control and anti-disease strategies.
Collapse
Affiliation(s)
- Vanessa Bottino-Rojas
- Department of Microbiology and Molecular Genetics, University of California, Irvine, CA 92697, USA
| | - Anthony A. James
- Department of Microbiology and Molecular Genetics, University of California, Irvine, CA 92697, USA
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697, USA
| |
Collapse
|
13
|
Hoermann A, Habtewold T, Selvaraj P, Del Corsano G, Capriotti P, Inghilterra MG, Kebede TM, Christophides GK, Windbichler N. Gene drive mosquitoes can aid malaria elimination by retarding Plasmodium sporogonic development. SCIENCE ADVANCES 2022; 8:eabo1733. [PMID: 36129981 PMCID: PMC9491717 DOI: 10.1126/sciadv.abo1733] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 08/04/2022] [Indexed: 05/12/2023]
Abstract
Gene drives hold promise for the genetic control of malaria vectors. The development of vector population modification strategies hinges on the availability of effector mechanisms impeding parasite development in transgenic mosquitoes. We augmented a midgut gene of the malaria mosquito Anopheles gambiae to secrete two exogenous antimicrobial peptides, magainin 2 and melittin. This small genetic modification, capable of efficient nonautonomous gene drive, hampers oocyst development in both Plasmodium falciparum and Plasmodium berghei. It delays the release of infectious sporozoites, while it simultaneously reduces the life span of homozygous female transgenic mosquitoes. Modeling the spread of this modification using a large-scale agent-based model of malaria epidemiology reveals that it can break the cycle of disease transmission across a range of transmission intensities.
Collapse
Affiliation(s)
- Astrid Hoermann
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Tibebu Habtewold
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Prashanth Selvaraj
- Institute for Disease Modeling, Bill and Melinda Gates Foundation, Seattle, WA 98109, USA
| | | | - Paolo Capriotti
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | | | - Temesgen M. Kebede
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | | | | |
Collapse
|
14
|
Zhang R, Zheng S, Huang H, Sun X, Huang Y, Wei J, Pan G, Li C, Zhou Z. Expression of anti-NbHK single-chain antibody in fusion with NSlmb enhances the resistance to Nosema bombycis in Sf9-III cells. BULLETIN OF ENTOMOLOGICAL RESEARCH 2022; 112:502-508. [PMID: 35382911 DOI: 10.1017/s0007485321001036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Nosema bombycis is a destructive and specific intracellular parasite of silkworm, which is extremely harmful to the silkworm industry. N. bombycis is considered as a quarantine pathogen of sericulture because of its long incubation period and horizontal and vertical transmission. Herein, two single-chain antibodies targeting N. bombycis hexokinase (NbHK) were cloned and expressed in fusion with the N-terminal of Slmb (a Drosophila melanogaster FBP), which contains the F-box domain. Western blotting demonstrated that Sf9-III cells expressed NSlmb-scFv-7A and NSlmb-scFv-6H, which recognized native NbHK. Subsequently, the NbHK was degraded by host ubiquitination system. When challenged with N. bombycis, the transfected Sf9-III cells exhibited better resistance relative to the controls, demonstrating that NbHK is a prospective target for parasite controls and this approach represents a potential solution for constructing N. bombycis-resistant Bombyx mori.
Collapse
Affiliation(s)
- Renze Zhang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China
| | - Shiyi Zheng
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China
- Affiliated Jinhua Hospital, Zhejiang University of Medicine, Jinhua Municipal Central Hospital, Jinhua, Zhejiang 321000, China
| | - Hongyun Huang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China
| | - Xi Sun
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China
| | - Yukang Huang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China
| | - Junhong Wei
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China
| | - Guoqing Pan
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China
| | - Chunfeng Li
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China
| | - Zeyang Zhou
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, China
| |
Collapse
|
15
|
Leung S, Windbichler N, Wenger EA, Bever CA, Selvaraj P. Population replacement gene drive characteristics for malaria elimination in a range of seasonal transmission settings: a modelling study. Malar J 2022; 21:226. [PMID: 35883100 PMCID: PMC9327287 DOI: 10.1186/s12936-022-04242-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 07/11/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Gene drives are a genetic engineering method where a suite of genes is inherited at higher than Mendelian rates and has been proposed as a promising new vector control strategy to reinvigorate the fight against malaria in sub-Saharan Africa. METHODS Using an agent-based model of malaria transmission with vector genetics, the impacts of releasing population-replacement gene drive mosquitoes on malaria transmission are examined and the population replacement gene drive system parameters required to achieve local elimination within a spatially-resolved, seasonal Sahelian setting are quantified. The performance of two different gene drive systems-"classic" and "integral"-are evaluated. Various transmission regimes (low, moderate, and high-corresponding to annual entomological inoculation rates of 10, 30, and 80 infectious bites per person) and other simultaneous interventions, including deployment of insecticide-treated nets (ITNs) and passive healthcare-seeking, are also simulated. RESULTS Local elimination probabilities decreased with pre-existing population target site resistance frequency, increased with transmission-blocking effectiveness of the introduced antiparasitic gene and drive efficiency, and were context dependent with respect to fitness costs associated with the introduced gene. Of the four parameters, transmission-blocking effectiveness may be the most important to focus on for improvements to future gene drive strains because a single release of classic gene drive mosquitoes is likely to locally eliminate malaria in low to moderate transmission settings only when transmission-blocking effectiveness is very high (above ~ 80-90%). However, simultaneously deploying ITNs and releasing integral rather than classic gene drive mosquitoes significantly boosts elimination probabilities, such that elimination remains highly likely in low to moderate transmission regimes down to transmission-blocking effectiveness values as low as ~ 50% and in high transmission regimes with transmission-blocking effectiveness values above ~ 80-90%. CONCLUSION A single release of currently achievable population replacement gene drive mosquitoes, in combination with traditional forms of vector control, can likely locally eliminate malaria in low to moderate transmission regimes within the Sahel. In a high transmission regime, higher levels of transmission-blocking effectiveness than are currently available may be required.
Collapse
Affiliation(s)
- Shirley Leung
- Institute for Disease Modeling, Bill & Melinda Gates Foundation, Seattle, WA, USA
| | - Nikolai Windbichler
- Department of Life Sciences, Imperial College London, South Kensington, London, UK
| | - Edward A Wenger
- Institute for Disease Modeling, Bill & Melinda Gates Foundation, Seattle, WA, USA
| | - Caitlin A Bever
- Institute for Disease Modeling, Bill & Melinda Gates Foundation, Seattle, WA, USA
| | - Prashanth Selvaraj
- Institute for Disease Modeling, Bill & Melinda Gates Foundation, Seattle, WA, USA.
| |
Collapse
|
16
|
Fofana A, Yerbanga RS, Bilgo E, Ouedraogo GA, Gendrin M, Ouedraogo JB. The Strategy of Paratransgenesis for the Control of Malaria Transmission. FRONTIERS IN TROPICAL DISEASES 2022. [DOI: 10.3389/fitd.2022.867104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Insect-borne diseases are responsible for important burdens on health worldwide particularly in Africa. Malaria alone causes close to half a million deaths every year, mostly in developing, tropical and subtropical countries, with 94% of the global deaths in 2019 occurring in the WHO African region. With several decades, vector control measures have been fundamental to fight against malaria. Considering the spread of resistance to insecticides in mosquitoes and to drugs in parasites, the need for novel strategies to inhibit the transmission of the disease is pressing. In recent years, several studies have focused on the interaction of malaria parasites, bacteria and their insect vectors. Their findings suggested that the microbiota of mosquitoes could be used to block Plasmodium transmission. A strategy, termed paratransgenesis, aims to interfere with the development of malaria parasites within their vectors through genetically-modified microbes, which produce antimalarial effectors inside the insect host. Here we review the progress of the paratransgenesis approach. We provide a historical perspective and then focus on the choice of microbial strains and on genetic engineering strategies. We finally describe the different steps from laboratory design to field implementation to fight against malaria.
Collapse
|
17
|
Carballar-Lejarazú R, Tushar T, Pham T, James AA. Cas9-mediated maternal effect and derived resistance alleles in a gene-drive strain of the African malaria vector mosquito, Anopheles gambiae. Genetics 2022; 221:iyac055. [PMID: 35389492 PMCID: PMC9157122 DOI: 10.1093/genetics/iyac055] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/30/2022] [Indexed: 11/24/2022] Open
Abstract
CRISPR/Cas9 technologies are important tools for the development of gene-drive systems to modify mosquito vector populations to control the transmission of pathogens that cause diseases such as malaria. However, one of the challenges for current Cas9-based drive systems is their ability to produce drive-resistant alleles resulting from insertions and deletions (indels) caused principally by nonhomologous end-joining following chromosome cleavage. Rapid increases in the frequency of such alleles may impair gene-drive dynamics. We explored the generation of indels in the germline and somatic cells in female gene-drive lineages using a series of selective crosses between a gene-drive line, AgNosCd-1, and wild-type mosquitoes. We find that potential drive-resistant mutant alleles are generated largely during embryonic development, most likely caused by deposition of the Cas9 endonuclease and guide RNAs in oocytes and resulting embryos by homozygous and hemizygous gene-drive mothers.
Collapse
Affiliation(s)
- Rebeca Carballar-Lejarazú
- Department of Microbiology & Molecular Genetics, University of California, Irvine, Irvine, CA 92697-4025, USA
| | - Taylor Tushar
- Department of Microbiology & Molecular Genetics, University of California, Irvine, Irvine, CA 92697-4025, USA
| | - Thai Binh Pham
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA 92697-3900, USA
| | - Anthony A James
- Department of Microbiology & Molecular Genetics, University of California, Irvine, Irvine, CA 92697-4025, USA
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA 92697-3900, USA
| |
Collapse
|
18
|
Pascini TV, Jeong YJ, Huang W, Pala ZR, Sá JM, Wells MB, Kizito C, Sweeney B, Alves E Silva TL, Andrew DJ, Jacobs-Lorena M, Vega-Rodríguez J. Transgenic Anopheles mosquitoes expressing human PAI-1 impair malaria transmission. Nat Commun 2022; 13:2949. [PMID: 35618711 PMCID: PMC9135733 DOI: 10.1038/s41467-022-30606-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 04/22/2022] [Indexed: 11/08/2022] Open
Abstract
In mammals, the serine protease plasmin degrades extracellular proteins during blood clot removal, tissue remodeling, and cell migration. The zymogen plasminogen is activated into plasmin by two serine proteases: tissue-type plasminogen activator (tPA) and urokinase-type plasminogen activator (uPA), a process regulated by plasminogen activator inhibitor 1 (PAI-1), a serine protease inhibitor that specifically inhibits tPA and uPA. Plasmodium gametes and sporozoites use tPA and uPA to activate plasminogen and parasite-bound plasmin degrades extracellular matrices, facilitating parasite motility in the mosquito and the mammalian host. Furthermore, inhibition of plasminogen activation by PAI-1 strongly blocks infection in both hosts. To block parasite utilization of plasmin, we engineered Anopheles stephensi transgenic mosquitoes constitutively secreting human PAI-1 (huPAI-1) in the midgut lumen, in the saliva, or both. Mosquitoes expressing huPAI-1 strongly reduced rodent and human Plasmodium parasite transmission to mosquitoes, showing that co-opting plasmin for mosquito infection is a conserved mechanism among Plasmodium species. huPAI-1 expression in saliva induced salivary gland deformation which affects sporozoite invasion and P. berghei transmission to mice, resulting in significant levels of protection from malaria. Targeting the interaction of malaria parasites with the fibrinolytic system using genetically engineered mosquitoes could be developed as an intervention to control malaria transmission.
Collapse
Affiliation(s)
- Tales V Pascini
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rm 2E20A, Rockville, MD, 20852, USA
| | - Yeong Je Jeong
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rm 2E20A, Rockville, MD, 20852, USA
| | - Wei Huang
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Zarna R Pala
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rm 2E20A, Rockville, MD, 20852, USA
| | - Juliana M Sá
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rm 2E20A, Rockville, MD, 20852, USA
| | - Michael B Wells
- Department of Cell Biology, Johns Hopkins University School of Medicine, 725 N. Wolfe Street, G10 Hunterian, Baltimore, MD, 21205, USA
- Department of Biomedical Sciences, Idaho College of Osteopathic Medicine, Meridian, ID, 83642, USA
| | - Christopher Kizito
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Brendan Sweeney
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rm 2E20A, Rockville, MD, 20852, USA
| | - Thiago L Alves E Silva
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rm 2E20A, Rockville, MD, 20852, USA
| | - Deborah J Andrew
- Department of Cell Biology, Johns Hopkins University School of Medicine, 725 N. Wolfe Street, G10 Hunterian, Baltimore, MD, 21205, USA
| | - Marcelo Jacobs-Lorena
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Joel Vega-Rodríguez
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rm 2E20A, Rockville, MD, 20852, USA.
| |
Collapse
|
19
|
Reid WR, Olson KE, Franz AWE. Current Effector and Gene-Drive Developments to Engineer Arbovirus-Resistant Aedes aegypti (Diptera: Culicidae) for a Sustainable Population Replacement Strategy in the Field. JOURNAL OF MEDICAL ENTOMOLOGY 2021; 58:1987-1996. [PMID: 33704462 PMCID: PMC8421695 DOI: 10.1093/jme/tjab030] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Indexed: 05/13/2023]
Abstract
Arthropod-borne viruses (arboviruses) such as dengue, Zika, and chikungunya viruses cause morbidity and mortality among human populations living in the tropical regions of the world. Conventional mosquito control efforts based on insecticide treatments and/or the use of bednets and window curtains are currently insufficient to reduce arbovirus prevalence in affected regions. Novel, genetic strategies that are being developed involve the genetic manipulation of mosquitoes for population reduction and population replacement purposes. Population replacement aims at replacing arbovirus-susceptible wild-type mosquitoes in a target region with those that carry a laboratory-engineered antiviral effector to interrupt arboviral transmission in the field. The strategy has been primarily developed for Aedes aegypti (L.), the most important urban arbovirus vector. Antiviral effectors based on long dsRNAs, miRNAs, or ribozymes destroy viral RNA genomes and need to be linked to a robust gene drive to ensure their fixation in the target population. Synthetic gene-drive concepts are based on toxin/antidote, genetic incompatibility, and selfish genetic element principles. The CRISPR/Cas9 gene editing system can be configurated as a homing endonuclease gene (HEG) and HEG-based drives became the preferred choice for mosquitoes. HEGs are highly allele and nucleotide sequence-specific and therefore sensitive to single-nucleotide polymorphisms/resistant allele formation. Current research efforts test new HEG-based gene-drive designs that promise to be less sensitive to resistant allele formation. Safety aspects in conjunction with gene drives are being addressed by developing procedures that would allow a recall or overwriting of gene-drive transgenes once they have been released.
Collapse
Affiliation(s)
- William R Reid
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
| | - Ken E Olson
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Alexander W E Franz
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
- Corresponding author, e-mail:
| |
Collapse
|
20
|
Adelman ZN, Kojin BB. Malaria-Resistant Mosquitoes (Diptera: Culicidae); The Principle is Proven, But Will the Effectors Be Effective? JOURNAL OF MEDICAL ENTOMOLOGY 2021; 58:1997-2005. [PMID: 34018548 DOI: 10.1093/jme/tjab090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Indexed: 06/12/2023]
Abstract
Over the last few decades, a substantial number of anti-malarial effector genes have been evaluated for their ability to block parasite infection in the mosquito vector. While many of these approaches have yielded significant effects on either parasite intensity or prevalence of infection, just a few have been able to completely block transmission. Additionally, many approaches, while effective against the parasite, also disrupt or alter important aspects of mosquito physiology, leading to corresponding changes in lifespan, reproduction, and immunity. As the most promising approaches move towards field-based evaluation, questions of effector gene robustness and durability move to the forefront. In this forum piece, we critically evaluate past effector gene approaches with an eye towards developing a deeper pipeline to augment the current best candidates.
Collapse
Affiliation(s)
- Zach N Adelman
- Department of Entomology and AgriLife Research, Texas A&M University, College Station, TX, USA
| | - Bianca B Kojin
- Department of Entomology and AgriLife Research, Texas A&M University, College Station, TX, USA
| |
Collapse
|
21
|
Dong S, Dong Y, Simões ML, Dimopoulos G. Mosquito transgenesis for malaria control. Trends Parasitol 2021; 38:54-66. [PMID: 34483052 DOI: 10.1016/j.pt.2021.08.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/01/2021] [Accepted: 08/03/2021] [Indexed: 12/14/2022]
Abstract
Malaria is one of the deadliest diseases. Because of the ineffectiveness of current malaria-control methods, several novel mosquito vector-based control strategies have been proposed to supplement existing control strategies. Mosquito transgenesis and gene drive have emerged as promising tools for preventing the spread of malaria by either suppressing mosquito populations by self-destructing mosquitoes or replacing mosquito populations with disease-refractory populations. Here we review the development of mosquito transgenesis and its application for malaria control, highlighting the transgenic expression of antiparasitic effector genes, inactivation of host factor genes, and manipulation of miRNAs and lncRNAs. Overall, from a malaria-control perspective, mosquito transgenesis is not envisioned as a stand-alone approach; rather, its use is proposed as a complement to existing vector-control strategies.
Collapse
Affiliation(s)
- Shengzhang Dong
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Yuemei Dong
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Maria L Simões
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
22
|
Verma P, Reeves RG, Gokhale CS. A common gene drive language eases regulatory process and eco-evolutionary extensions. BMC Ecol Evol 2021; 21:156. [PMID: 34372763 PMCID: PMC8351217 DOI: 10.1186/s12862-021-01881-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 07/12/2021] [Indexed: 02/08/2023] Open
Abstract
Background Synthetic gene drive technologies aim to spread transgenic constructs into wild populations even when they impose organismal fitness disadvantages. The extraordinary diversity of plausible drive mechanisms and the range of selective parameters they may encounter makes it very difficult to convey their relative predicted properties, particularly where multiple approaches are combined. The sheer number of published manuscripts in this field, experimental and theoretical, the numerous techniques resulting in an explosion in the gene drive vocabulary hinder the regulators’ point of view. We address this concern by defining a simplified parameter based language of synthetic drives. Results Employing the classical population dynamics approach, we show that different drive construct (replacement) mechanisms can be condensed and evaluated on an equal footing even where they incorporate multiple replacement drives approaches. Using a common language, it is then possible to compare various model properties, a task desired by regulators and policymakers. The generalization allows us to extend the study of the invasion dynamics of replacement drives analytically and, in a spatial setting, the resilience of the released drive constructs. The derived framework is available as a standalone tool. Conclusion Besides comparing available drive constructs, our tool is also useful for educational purpose. Users can also explore the evolutionary dynamics of future hypothetical combination drive scenarios. Thus, our results appraise the properties and robustness of drives and provide an intuitive and objective way for risk assessment, informing policies, and enhancing public engagement with proposed and future gene drive approaches.
Collapse
Affiliation(s)
- Prateek Verma
- Research Group for Theoretical Models of Eco-evolutionary Dynamics, Department of Evolutionary Theory, Max Planck Institute for Evolutionary Biology, Plön, Germany.
| | - R Guy Reeves
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Biology, Plön, Germany
| | - Chaitanya S Gokhale
- Research Group for Theoretical Models of Eco-evolutionary Dynamics, Department of Evolutionary Theory, Max Planck Institute for Evolutionary Biology, Plön, Germany
| |
Collapse
|
23
|
Schember I, Halfon MS. Identification of new Anopheles gambiae transcriptional enhancers using a cross-species prediction approach. INSECT MOLECULAR BIOLOGY 2021; 30:410-419. [PMID: 33866636 PMCID: PMC8266755 DOI: 10.1111/imb.12705] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 02/09/2021] [Accepted: 03/31/2021] [Indexed: 06/12/2023]
Abstract
The success of transgenic mosquito vector control approaches relies on well-targeted gene expression, requiring the identification and characterization of a diverse set of mosquito promoters and transcriptional enhancers. However, few enhancers have been characterized in Anopheles gambiae to date. Here, we employ the SCRMshaw method we previously developed to predict enhancers in the A. gambiae genome, preferentially targeting vector-relevant tissues such as the salivary glands, midgut and nervous system. We demonstrate a high overall success rate, with at least 8 of 11 (73%) tested sequences validating as enhancers in an in vivo xenotransgenic assay. Four tested sequences drive expression in either the salivary gland or the midgut, making them directly useful for probing the biology of these infection-relevant tissues. The success of our study suggests that computational enhancer prediction should serve as an effective means for identifying A. gambiae enhancers with activity in tissues involved in malaria propagation and transmission.
Collapse
Affiliation(s)
- Isabella Schember
- Department of Biochemistry, University at Buffalo-State University of New York, Buffalo, NY 14203
| | - Marc S. Halfon
- Department of Biochemistry, University at Buffalo-State University of New York, Buffalo, NY 14203
- Department of Biomedical Informatics, University at Buffalo-State University of New York, Buffalo, NY 14203
- Department of Biological Sciences, University at Buffalo-State University of New York, Buffalo, NY 14203
- NY State Center of Excellence in Bioinformatics & Life Sciences, Buffalo, NY 14203
- Department of Molecular and Cellular Biology and Program in Cancer Genetics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263
| |
Collapse
|
24
|
Caragata EP, Dong S, Dong Y, Simões ML, Tikhe CV, Dimopoulos G. Prospects and Pitfalls: Next-Generation Tools to Control Mosquito-Transmitted Disease. Annu Rev Microbiol 2021; 74:455-475. [PMID: 32905752 DOI: 10.1146/annurev-micro-011320-025557] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Mosquito-transmitted diseases, including malaria and dengue, are a major threat to human health around the globe, affecting millions each year. A diverse array of next-generation tools has been designed to eliminate mosquito populations or to replace them with mosquitoes that are less capable of transmitting key pathogens. Many of these new approaches have been built on recent advances in CRISPR/Cas9-based genome editing. These initiatives have driven the development of pathogen-resistant lines, new genetics-based sexing methods, and new methods of driving desirable genetic traits into mosquito populations. Many other emerging tools involve microorganisms, including two strategies involving Wolbachia that are achieving great success in the field. At the same time, other mosquito-associated bacteria, fungi, and even viruses represent untapped sources of new mosquitocidal or antipathogen compounds. Although there are still hurdles to be overcome, the prospect that such approaches will reduce the impact of these diseases is highly encouraging.
Collapse
Affiliation(s)
- E P Caragata
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205, USA; , , , , ,
| | - S Dong
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205, USA; , , , , ,
| | - Y Dong
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205, USA; , , , , ,
| | - M L Simões
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205, USA; , , , , ,
| | - C V Tikhe
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205, USA; , , , , ,
| | - G Dimopoulos
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205, USA; , , , , ,
| |
Collapse
|
25
|
Piergentili R, Del Rio A, Signore F, Umani Ronchi F, Marinelli E, Zaami S. CRISPR-Cas and Its Wide-Ranging Applications: From Human Genome Editing to Environmental Implications, Technical Limitations, Hazards and Bioethical Issues. Cells 2021; 10:cells10050969. [PMID: 33919194 PMCID: PMC8143109 DOI: 10.3390/cells10050969] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/18/2021] [Accepted: 04/19/2021] [Indexed: 12/14/2022] Open
Abstract
The CRISPR-Cas system is a powerful tool for in vivo editing the genome of most organisms, including man. During the years this technique has been applied in several fields, such as agriculture for crop upgrade and breeding including the creation of allergy-free foods, for eradicating pests, for the improvement of animal breeds, in the industry of bio-fuels and it can even be used as a basis for a cell-based recording apparatus. Possible applications in human health include the making of new medicines through the creation of genetically modified organisms, the treatment of viral infections, the control of pathogens, applications in clinical diagnostics and the cure of human genetic diseases, either caused by somatic (e.g., cancer) or inherited (mendelian disorders) mutations. One of the most divisive, possible uses of this system is the modification of human embryos, for the purpose of preventing or curing a human being before birth. However, the technology in this field is evolving faster than regulations and several concerns are raised by its enormous yet controversial potential. In this scenario, appropriate laws need to be issued and ethical guidelines must be developed, in order to properly assess advantages as well as risks of this approach. In this review, we summarize the potential of these genome editing techniques and their applications in human embryo treatment. We will analyze CRISPR-Cas limitations and the possible genome damage caused in the treated embryo. Finally, we will discuss how all this impacts the law, ethics and common sense.
Collapse
Affiliation(s)
- Roberto Piergentili
- Institute of Molecular Biology and Pathology, Italian National Research Council (CNR-IBPM), 00185 Rome, Italy;
| | - Alessandro Del Rio
- Department of Anatomical, Histological, Forensic, and Orthopedic Sciences, Sapienza University of Rome, 00161 Rome, Italy; (F.U.R.); (E.M.); (S.Z.)
- Correspondence: or
| | - Fabrizio Signore
- Obstetrics and Gynecology Department, USL Roma2, Sant’Eugenio Hospital, 00144 Rome, Italy;
| | - Federica Umani Ronchi
- Department of Anatomical, Histological, Forensic, and Orthopedic Sciences, Sapienza University of Rome, 00161 Rome, Italy; (F.U.R.); (E.M.); (S.Z.)
| | - Enrico Marinelli
- Department of Anatomical, Histological, Forensic, and Orthopedic Sciences, Sapienza University of Rome, 00161 Rome, Italy; (F.U.R.); (E.M.); (S.Z.)
| | - Simona Zaami
- Department of Anatomical, Histological, Forensic, and Orthopedic Sciences, Sapienza University of Rome, 00161 Rome, Italy; (F.U.R.); (E.M.); (S.Z.)
| |
Collapse
|
26
|
Hoermann A, Tapanelli S, Capriotti P, Del Corsano G, Masters EK, Habtewold T, Christophides GK, Windbichler N. Converting endogenous genes of the malaria mosquito into simple non-autonomous gene drives for population replacement. eLife 2021; 10:58791. [PMID: 33845943 PMCID: PMC8043746 DOI: 10.7554/elife.58791] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 03/21/2021] [Indexed: 12/15/2022] Open
Abstract
Gene drives for mosquito population replacement are promising tools for malaria control. However, there is currently no clear pathway for safely testing such tools in endemic countries. The lack of well-characterized promoters for infection-relevant tissues and regulatory hurdles are further obstacles for their design and use. Here we explore how minimal genetic modifications of endogenous mosquito genes can convert them directly into non-autonomous gene drives without disrupting their expression. We co-opted the native regulatory sequences of three midgut-specific loci of the malaria vector Anopheles gambiae to host a prototypical antimalarial molecule and guide-RNAs encoded within artificial introns that support efficient gene drive. We assess the propensity of these modifications to interfere with the development of Plasmodium falciparum and their effect on fitness. Because of their inherent simplicity and passive mode of drive such traits could form part of an acceptable testing pathway of gene drives for malaria eradication.
Collapse
Affiliation(s)
- Astrid Hoermann
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Sofia Tapanelli
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Paolo Capriotti
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | | | - Ellen Kg Masters
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Tibebu Habtewold
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | | | - Nikolai Windbichler
- Department of Life Sciences, Imperial College London, London, United Kingdom
| |
Collapse
|
27
|
Ukegbu CV, Christophides GK, Vlachou D. Identification of Three Novel Plasmodium Factors Involved in Ookinete to Oocyst Developmental Transition. Front Cell Infect Microbiol 2021; 11:634273. [PMID: 33791240 PMCID: PMC8005625 DOI: 10.3389/fcimb.2021.634273] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 02/10/2021] [Indexed: 12/11/2022] Open
Abstract
Plasmodium falciparum malaria remains a major cause of global morbidity and mortality, mainly in sub-Saharan Africa. The numbers of new malaria cases and deaths have been stable in the last years despite intense efforts for disease elimination, highlighting the need for new approaches to stop disease transmission. Further understanding of the parasite transmission biology could provide a framework for the development of such approaches. We phenotypically and functionally characterized three novel genes, PIMMS01, PIMMS57, and PIMMS22, using targeted disruption of their orthologs in the rodent parasite Plasmodium berghei. PIMMS01 and PIMMS57 are specifically and highly expressed in ookinetes, while PIMMS22 transcription starts already in gametocytes and peaks in sporozoites. All three genes show strong phenotypes associated with the ookinete to oocyst transition, as their disruption leads to very low numbers of oocysts and complete abolishment of transmission. PIMMS22 has a secondary essential function in the oocyst. Our results enrich the molecular understanding of the parasite-vector interactions and identify PIMMS01, PIMMS57, and PIMMS22 as new targets of transmission blocking interventions.
Collapse
Affiliation(s)
- Chiamaka V Ukegbu
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - George K Christophides
- Department of Life Sciences, Imperial College London, London, United Kingdom.,The Cyprus Institute, Nicosia, Cyprus
| | - Dina Vlachou
- Department of Life Sciences, Imperial College London, London, United Kingdom.,The Cyprus Institute, Nicosia, Cyprus
| |
Collapse
|
28
|
Terradas G, Buchman AB, Bennett JB, Shriner I, Marshall JM, Akbari OS, Bier E. Inherently confinable split-drive systems in Drosophila. Nat Commun 2021; 12:1480. [PMID: 33674604 PMCID: PMC7935863 DOI: 10.1038/s41467-021-21771-7] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 02/05/2021] [Indexed: 02/06/2023] Open
Abstract
CRISPR-based gene-drive systems, which copy themselves via gene conversion mediated by the homology-directed repair (HDR) pathway, have the potential to revolutionize vector control. However, mutant alleles generated by the competing non-homologous end-joining (NHEJ) pathway, resistant to Cas9 cleavage, can interrupt the spread of gene-drive elements. We hypothesized that drives targeting genes essential for viability or reproduction also carrying recoded sequences that restore endogenous gene functionality should benefit from dominantly-acting maternal clearance of NHEJ alleles combined with recessive Mendelian culling processes. Here, we test split gene-drive (sGD) systems in Drosophila melanogaster that are inserted into essential genes required for viability (rab5, rab11, prosalpha2) or fertility (spo11). In single generation crosses, sGDs copy with variable efficiencies and display sex-biased transmission. In multigenerational cage trials, sGDs follow distinct drive trajectories reflecting their differential tendencies to induce target chromosome damage and/or lethal/sterile mosaic Cas9-dependent phenotypes, leading to inherently confinable drive outcomes. NHEJ alleles and Cas9 remnants after a gene drive introduction are scientific and public concerns. Here, the authors use split drives with recoded rescue elements to target essential genes and minimize the appearance of NHEJ alleles while also leaving no trace of Cas9.
Collapse
Affiliation(s)
- Gerard Terradas
- Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA, USA.,Tata Institute for Genetics and Society, University of California, San Diego, La Jolla, CA, USA
| | - Anna B Buchman
- Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA, USA
| | - Jared B Bennett
- Biophysics Graduate Group, Division of Biological Sciences, College of Letters and Science, University of California, Berkeley, CA, USA
| | - Isaiah Shriner
- Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA, USA
| | - John M Marshall
- Divisions of Epidemiology and Biostatistics, School of Public Health, University of California, Berkeley, CA, USA.,Innovative Genomics Institute, Berkeley, CA, USA
| | - Omar S Akbari
- Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA, USA
| | - Ethan Bier
- Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA, USA. .,Tata Institute for Genetics and Society, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
29
|
Zheng S, Huang Y, Huang H, Yu B, Zhou N, Wei J, Pan G, Li C, Zhou Z. The role of NbTMP1, a surface protein of sporoplasm, in Nosema bombycis infection. Parasit Vectors 2021; 14:81. [PMID: 33494800 PMCID: PMC7836179 DOI: 10.1186/s13071-021-04595-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 01/11/2021] [Indexed: 12/02/2022] Open
Abstract
Background Nosema bombycis is a unicellular eukaryotic pathogen of the silkworm, Bombyx mori, and is an economic and occupational hazard in the silkworm industry. Because of its long incubation period and horizontal and vertical transmission, it is subject to quarantine measures in sericulture production. The microsporidian life-cycle includes a dormant extracellular phase and intracellular proliferation phase, with the proliferation period being the most active period. This latter period lacks spore wall protection and may be the most susceptible stage for control. Methods In order to find suitable target for the selective breeding of N. bombycis-resistant silkworm strains, we screen highly expressed membrane proteins from the transcriptome data of N. bombycis. The subcellular localization of the candidate protein was verified by Indirect immunofluorescence analysis (IFA) and immunoelectron microscopy (IEM), and its role in N. bombycis proliferation was verified by RNAi. Results The N. bombycis protein (NBO_76g0014) was identified as a transmembrane protein and named NbTMP1. It is homologous with hypothetical proteins NGRA_1734 from Nosema granulosis. NbTMP1 has a transmembrane region of 23 amino acids at the N-terminus. Indirect immunofluorescence analysis (IFA) results suggest that NbTMP1 is secreted on the plasma membrane as the spores develop. Western blot and qRT-PCR analysis showed that NbTMP1 was expressed in all developmental stages of N. bombycis in infected cells and in the silkworm midgut. Downregulation of NbTMP1 expression resulted in significant inhibition of N. bombycis proliferation. Conclusions We confirmed that NbTMP1 is a membrane protein of N. bombycis. Reduction of the transcription level of NbTMP1 significantly inhibited N. bombycis proliferation, and this protein may be a target for the selective breeding of N. bombycis-resistant silkworm strains.
![]()
Collapse
Affiliation(s)
- Shiyi Zheng
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, China.,Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing, 400715, China.,Affiliated Jinhua Hospital, Zhejiang University of Medicine-Jinhua Municipal Central Hospital, Jinhua, 321000, Zhejiang, China
| | - Yukang Huang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, China.,Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing, 400715, China
| | - Hongyun Huang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, China.,Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing, 400715, China
| | - Bin Yu
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, China.,Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing, 400715, China
| | - Ni Zhou
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, China.,Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing, 400715, China
| | - Junhong Wei
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, China.,Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing, 400715, China
| | - Guoqing Pan
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, China.,Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing, 400715, China
| | - Chunfeng Li
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, China. .,Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing, 400715, China.
| | - Zeyang Zhou
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, China.,Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing, 400715, China.,College of Life Sciences, Chongqing Normal University, Chongqing, 401331, China
| |
Collapse
|
30
|
Adolfi A, Gantz VM, Jasinskiene N, Lee HF, Hwang K, Terradas G, Bulger EA, Ramaiah A, Bennett JB, Emerson JJ, Marshall JM, Bier E, James AA. Efficient population modification gene-drive rescue system in the malaria mosquito Anopheles stephensi. Nat Commun 2020; 11:5553. [PMID: 33144570 PMCID: PMC7609566 DOI: 10.1038/s41467-020-19426-0] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 10/13/2020] [Indexed: 12/27/2022] Open
Abstract
Cas9/gRNA-mediated gene-drive systems have advanced development of genetic technologies for controlling vector-borne pathogen transmission. These technologies include population suppression approaches, genetic analogs of insecticidal techniques that reduce the number of insect vectors, and population modification (replacement/alteration) approaches, which interfere with competence to transmit pathogens. Here, we develop a recoded gene-drive rescue system for population modification of the malaria vector, Anopheles stephensi, that relieves the load in females caused by integration of the drive into the kynurenine hydroxylase gene by rescuing its function. Non-functional resistant alleles are eliminated via a dominantly-acting maternal effect combined with slower-acting standard negative selection, and rare functional resistant alleles do not prevent drive invasion. Small cage trials show that single releases of gene-drive males robustly result in efficient population modification with ≥95% of mosquitoes carrying the drive within 5-11 generations over a range of initial release ratios.
Collapse
Affiliation(s)
- Adriana Adolfi
- Department of Microbiology & Molecular Genetics, University of California, Irvine, CA, 92697-3900, USA
- Liverpool School of Tropical Medicine, Vector Biology Department, L3 5QA, Liverpool, UK
| | - Valentino M Gantz
- Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA, 92093-0349, USA
| | - Nijole Jasinskiene
- Department of Microbiology & Molecular Genetics, University of California, Irvine, CA, 92697-3900, USA
| | - Hsu-Feng Lee
- Department of Microbiology & Molecular Genetics, University of California, Irvine, CA, 92697-3900, USA
| | - Kristy Hwang
- Department of Microbiology & Molecular Genetics, University of California, Irvine, CA, 92697-3900, USA
| | - Gerard Terradas
- Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA, 92093-0349, USA
- Tata Institute for Genetics and Society (TIGS)-UCSD, La Jolla, CA, 92093-0335, USA
| | - Emily A Bulger
- Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA, 92093-0349, USA
- Tata Institute for Genetics and Society (TIGS)-UCSD, La Jolla, CA, 92093-0335, USA
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, CA, 94158, USA
- The Gladstone Institutes, San Francisco, CA, 94158, USA
| | - Arunachalam Ramaiah
- Department of Ecology and Evolutionary Biology, University of California, Irvine, CA, 92697-2525, USA
- Tata Institute for Genetics and Society (TIGS)-India, Bangalore, KA, 560065, India
| | - Jared B Bennett
- Biophysics Graduate Group, Division of Biological Sciences, College of Letters and Science, University of California, Berkeley, CA, 94720, USA
| | - J J Emerson
- Department of Ecology and Evolutionary Biology, University of California, Irvine, CA, 92697-2525, USA
| | - John M Marshall
- Division of Epidemiology & Biostatistics, School of Public Health, University of California, Berkeley, CA, 94720, USA
- Innovative Genomics Institute, Berkeley, CA, 94720, USA
| | - Ethan Bier
- Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA, 92093-0349, USA
- Tata Institute for Genetics and Society (TIGS)-UCSD, La Jolla, CA, 92093-0335, USA
| | - Anthony A James
- Department of Microbiology & Molecular Genetics, University of California, Irvine, CA, 92697-3900, USA.
- Department of Molecular Biology & Biochemistry, University of California, Irvine, CA, 92697-4025, USA.
| |
Collapse
|
31
|
Qasim M, Xiao H, He K, Omar MAA, Liu F, Ahmed S, Li F. Genetic engineering and bacterial pathogenesis against the vectorial capacity of mosquitoes. Microb Pathog 2020; 147:104391. [PMID: 32679245 DOI: 10.1016/j.micpath.2020.104391] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/05/2020] [Accepted: 07/09/2020] [Indexed: 12/19/2022]
Abstract
Mosquitoes are the main vector of multiple diseases worldwide and transmit viral (malaria, chikungunya, encephalitis, yellow fever, as well as dengue fever), as well as bacterial diseases (tularemia). To manage the outbreak of mosquito populations, various management programs include the application of chemicals, followed by biological and genetic control. Here we aimed to focus on the role of bacterial pathogenesis and molecular tactics for the management of mosquitoes and their vectorial capacity. Bacterial pathogenesis and molecular manipulations have a substantial impact on the biology of mosquitoes, and both strategies change the gene expression and regulation of disease vectors. The strategy for genetic modification is also proved to be excellent for the management of mosquitoes, which halt the development of population via incompatibility of different sex. Therefore, the purpose of the present discussion is to illustrate the impact of both approaches against the vectorial capacity of mosquitoes. Moreover, it could be helpful to understand the relationship of insect-pathogen and to manage various insect vectors as well as diseases.
Collapse
Affiliation(s)
- Muhammad Qasim
- Ministry of Agricultural and Rural Affairs Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Insect Sciences, College of Agriculture & Biotechnology, Zhejiang University, Hangzhou, 310058, China.
| | - Huamei Xiao
- Ministry of Agricultural and Rural Affairs Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Insect Sciences, College of Agriculture & Biotechnology, Zhejiang University, Hangzhou, 310058, China; College of Life Sciences and Resource Environment, Key Laboratory of Crop Growth and Development Regulation of Jiangxi Province, Yichun University, Yichun, 336000, China
| | - Kang He
- Ministry of Agricultural and Rural Affairs Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Insect Sciences, College of Agriculture & Biotechnology, Zhejiang University, Hangzhou, 310058, China
| | - Mohamed A A Omar
- Ministry of Agricultural and Rural Affairs Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Insect Sciences, College of Agriculture & Biotechnology, Zhejiang University, Hangzhou, 310058, China
| | - Feiling Liu
- Ministry of Agricultural and Rural Affairs Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Insect Sciences, College of Agriculture & Biotechnology, Zhejiang University, Hangzhou, 310058, China
| | - Sohail Ahmed
- Department of Entomology, University of Agriculture, Faisalabad, 38040, Pakistan
| | - Fei Li
- Ministry of Agricultural and Rural Affairs Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Insect Sciences, College of Agriculture & Biotechnology, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
32
|
Dong Y, Simões ML, Dimopoulos G. Versatile transgenic multistage effector-gene combinations for Plasmodium falciparum suppression in Anopheles. SCIENCE ADVANCES 2020; 6:eaay5898. [PMID: 32426491 PMCID: PMC7220273 DOI: 10.1126/sciadv.aay5898] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 02/27/2020] [Indexed: 05/14/2023]
Abstract
The malaria parasite's complex journey through the Anopheles mosquito vector provides multiple opportunities for targeting Plasmodium with recombinant effectors at different developmental stages and different host tissues. We have designed and expressed transgenes that efficiently suppress Plasmodium infection by targeting the parasite with multiple independent endogenous and exogenous effectors at multiple infection stages to potentiate suppression and minimize the probability for development of resistance to develop. We have also addressed the fitness impact of transgene expression on the mosquito. We show that highly potent suppression can be achieved by targeting both pre-oocyst stages by transgenically overexpressing either the endogenous immune deficiency immune pathway transcription factor Rel2 or a polycistronic mRNA encoding multiple antiparasitic effectors and simultaneously targeting the sporozoite stages with an anti-sporozoite single-chain antibody fused to the antiparasitic protein Scorpine. Expression of the selected endogenous effector systems appears to pose a lower fitness cost than does the use of foreign genes.
Collapse
|
33
|
Cas9-Mediated Gene-Editing in the Malaria Mosquito Anopheles stephensi by ReMOT Control. G3-GENES GENOMES GENETICS 2020; 10:1353-1360. [PMID: 32122959 PMCID: PMC7144067 DOI: 10.1534/g3.120.401133] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Innovative tools are essential for advancing malaria control and depend on an understanding of molecular mechanisms governing transmission of malaria parasites by Anopheles mosquitoes. CRISPR/Cas9-based gene disruption is a powerful method to uncover underlying biology of vector-pathogen interactions and can itself form the basis of mosquito control strategies. However, embryo injection methods used to genetically manipulate mosquitoes (especially Anopheles) are difficult and inefficient, particularly for non-specialist laboratories. Here, we adapted the ReMOT Control (Receptor-mediated Ovary Transduction of Cargo) technique to deliver Cas9 ribonucleoprotein complex to adult mosquito ovaries, generating targeted and heritable mutations in the malaria vector Anopheles stephensi without injecting embryos. In Anopheles, ReMOT Control gene editing was as efficient as standard embryo injections. The application of ReMOT Control to Anopheles opens the power of CRISPR/Cas9 methods to malaria laboratories that lack the equipment or expertise to perform embryo injections and establishes the flexibility of ReMOT Control for diverse mosquito species.
Collapse
|
34
|
PIMMS43 is required for malaria parasite immune evasion and sporogonic development in the mosquito vector. Proc Natl Acad Sci U S A 2020; 117:7363-7373. [PMID: 32165544 PMCID: PMC7132314 DOI: 10.1073/pnas.1919709117] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Malaria is transmitted among humans through mosquito bites. Here, we characterize a protein found on the surface of mosquito stages of malaria parasites and reveal that it serves to evade the mosquito immune system and ensure disease transmission. Neutralization of PIMMS43 (Plasmodium Infection of the Mosquito Midgut Screen 43), either by eliminating it from the parasite genome or by preincubating parasites with antibodies that bind to the PIMMS43 protein, inhibits mosquito infection with malaria parasites. Differences in PIMMS43 detected between African malaria parasite populations suggest that these populations have adapted for transmission by different mosquito vectors that are also differentially distributed across the continent. We conclude that targeting PIMMS43 can block malaria parasites inside mosquitoes before they can infect humans. After being ingested by a female Anopheles mosquito during a bloodmeal on an infected host, and before they can reach the mosquito salivary glands to be transmitted to a new host, Plasmodium parasites must establish an infection of the mosquito midgut in the form of oocysts. To achieve this, they must first survive a series of robust innate immune responses that take place prior to, during, and immediately after ookinete traversal of the midgut epithelium. Understanding how parasites may evade these responses could highlight new ways to block malaria transmission. We show that an ookinete and sporozoite surface protein designated as PIMMS43 (Plasmodium Infection of the Mosquito Midgut Screen 43) is required for parasite evasion of the Anopheles coluzzii complement-like response. Disruption of PIMMS43 in the rodent malaria parasite Plasmodium berghei triggers robust complement activation and ookinete elimination upon mosquito midgut traversal. Silencing components of the complement-like system through RNAi largely restores ookinete-to-oocyst transition but oocysts remain small in size and produce a very small number of sporozoites that additionally are not infectious, indicating that PIMMS43 is also essential for sporogonic development in the oocyst. Antibodies that bind PIMMS43 interfere with parasite immune evasion when ingested with the infectious blood meal and significantly reduce the prevalence and intensity of infection. PIMMS43 genetic structure across African Plasmodium falciparum populations indicates allelic adaptation to sympatric vector populations. These data add to our understanding of mosquito–parasite interactions and identify PIMMS43 as a target of malaria transmission blocking.
Collapse
|
35
|
Senderskiy I, Ignatieva A, Dolgikh V. Vairimorpha (Nosema) ceranae (Opisthosporidia: Microsporidia) in vitro Infection of Sf9 Insect Cell Line as an Experimental Model of Parasite – Host Interrelations. BIO WEB OF CONFERENCES 2020. [DOI: 10.1051/bioconf/20201800026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Microsporidia Vairimorpha (Nosema) ceranae is an emergent parasite of honeybee Apis mellifera and has a great concern to apiculture due to high pathogenesity and fumagillin resistance. Recently RNA interference and single-chain antibodies technologies are proposed to create unsusceptible to intracellular parasites breeds of insects. In this study we established cell culture model system to provide a way to investigate the molecular basis of interactions between honeybee and its pathogen. As the first step in this direction V. ceranae spores were isolated from living bees, properly sterilized and in vitro germinated with Sf9 lepidopteran-derived cell line. The entire life circle of this parasite was achieved. This should allow us to develop the novel treatment strategy directed to inhibit propagation of V. ceranae intracellular stages with recombinant antibodies targeted to the vitally important membrane transporters.
Collapse
|
36
|
Dolgikh VV, Timofeev SA, Zhuravlyov VS, Senderskiy IV. Construction and heterologous overexpression of two chimeric proteins carrying outer hydrophilic loops of Vairimorpha ceranae and Nosema bombycis ATP/ADP carriers. J Invertebr Pathol 2020; 171:107337. [PMID: 32035083 DOI: 10.1016/j.jip.2020.107337] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 02/02/2020] [Accepted: 02/04/2020] [Indexed: 01/15/2023]
Abstract
Microsporidia Nosema bombycis and Vairimorpha ceranae cause destructive epizootics of honey bees and silkworms. Insufficient efficiency of the antibiotic fumagillin against V. ceranae, its toxicity and the absence of effective methods of N. bombycis treatment demand the discovery of novel strategies to suppress infections of domesticated insects. RNA interference is one such novel treatment strategy. Another one implies that the intracellular development of microsporidia may be suppressed by single-chain antibodies (scFv fragments) against functionally important parasite proteins. Important components of microsporidian metabolism are non-mitochondrial, plastidic-bacterial ATP/ADP carriers. These membrane transporters import host-derived ATP and provide the capacity to pathogens for energy parasitism. Here, we analyzed membrane topology of four V. ceranae and three N. bombycis ATP/ADP transporters to construct two fusion proteins carrying their outer hydrophilic loops contacting with infected host cell cytoplasm. Interestingly, full-size genes of N. bombycis transporters may be derived from the Asian swallowtail Papilio xuthus genome sequencing project. Synthesis of the artificial genes was followed by overexpression of recombinant proteins in E. coli as insoluble inclusion bodies. The gene fragments encoding the loops of individual transporters were also effectively expressed in bacteria. The chimeric antigens may be used to construct immune libraries or select microsporidia-suppressing scFv fragments from synthetic, semisynthetic, naïve and immune antibody libraries. A further expression of such antibodies in insect cells may increase their resistance to microsporidial infections.
Collapse
Affiliation(s)
- Viacheslav V Dolgikh
- Laboratory of Molecular Plant Protection, All-Russian Institute of Plant Protection, St. Petersburg, Pushkin, Russia.
| | - Sergey A Timofeev
- Laboratory of Molecular Plant Protection, All-Russian Institute of Plant Protection, St. Petersburg, Pushkin, Russia
| | - Vladimir S Zhuravlyov
- Laboratory of Molecular Plant Protection, All-Russian Institute of Plant Protection, St. Petersburg, Pushkin, Russia
| | - Igor V Senderskiy
- Laboratory of Molecular Plant Protection, All-Russian Institute of Plant Protection, St. Petersburg, Pushkin, Russia
| |
Collapse
|
37
|
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
|
38
|
Buchman A, Gamez S, Li M, Antoshechkin I, Li HH, Wang HW, Chen CH, Klein MJ, Duchemin JB, Crowe JE, Paradkar PN, Akbari OS. Broad dengue neutralization in mosquitoes expressing an engineered antibody. PLoS Pathog 2020; 16:e1008103. [PMID: 31945137 PMCID: PMC6964813 DOI: 10.1371/journal.ppat.1008103] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 09/23/2019] [Indexed: 12/11/2022] Open
Abstract
With dengue virus (DENV) becoming endemic in tropical and subtropical regions worldwide, there is a pressing global demand for effective strategies to control the mosquitoes that spread this disease. Recent advances in genetic engineering technologies have made it possible to create mosquitoes with reduced vector competence, limiting their ability to acquire and transmit pathogens. Here we describe the development of Aedes aegypti mosquitoes synthetically engineered to impede vector competence to DENV. These mosquitoes express a gene encoding an engineered single-chain variable fragment derived from a broadly neutralizing DENV human monoclonal antibody and have significantly reduced viral infection, dissemination, and transmission rates for all four major antigenically distinct DENV serotypes. Importantly, this is the first engineered approach that targets all DENV serotypes, which is crucial for effective disease suppression. These results provide a compelling route for developing effective genetic-based DENV control strategies, which could be extended to curtail other arboviruses.
Collapse
Affiliation(s)
- Anna Buchman
- Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California, United States of America
| | - Stephanie Gamez
- Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California, United States of America
| | - Ming Li
- Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California, United States of America
| | - Igor Antoshechkin
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, United States of America
| | - Hsing-Han Li
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Hsin-Wei Wang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Chun-Hong Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Melissa J. Klein
- CSIRO Health and Biosecurity, Australian Animal Health Laboratory, Geelong, VIC, Australia
| | - Jean-Bernard Duchemin
- CSIRO Health and Biosecurity, Australian Animal Health Laboratory, Geelong, VIC, Australia
| | - James E. Crowe
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Departments of Pediatrics, Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Prasad N. Paradkar
- CSIRO Health and Biosecurity, Australian Animal Health Laboratory, Geelong, VIC, Australia
| | - Omar S. Akbari
- Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California, United States of America
- Tata Institute for Genetics and Society-UCSD, La Jolla, California, United States of America
| |
Collapse
|
39
|
Experimental population modification of the malaria vector mosquito, Anopheles stephensi. PLoS Genet 2019; 15:e1008440. [PMID: 31856182 PMCID: PMC6922335 DOI: 10.1371/journal.pgen.1008440] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 09/19/2019] [Indexed: 12/14/2022] Open
Abstract
Small laboratory cage trials of non-drive and gene-drive strains of the Asian malaria vector mosquito, Anopheles stephensi, were used to investigate release ratios and other strain properties for their impact on transgene spread during simulated population modification. We evaluated the effects of transgenes on survival, male contributions to next-generation populations, female reproductive success and the impact of accumulation of gene drive-resistant genomic target sites resulting from nonhomologous end-joining (NHEJ) mutagenesis during Cas9, guide RNA-mediated cleavage. Experiments with a non-drive, autosomally-linked malaria-resistance gene cassette showed ‘full introduction’ (100% of the insects have at least one copy of the transgene) within 8 weeks (≤ 3 generations) following weekly releases of 10:1 transgenic:wild-type males in an overlapping generation trial design. Male release ratios of 1:1 resulted in cages where mosquitoes with at least one copy of the transgene fluctuated around 50%. In comparison, two of three cages in which the malaria-resistance genes were linked to a gene-drive system in an overlapping generation, single 1:1 release reached full introduction in 6–8 generations with a third cage at ~80% within the same time. Release ratios of 0.1:1 failed to establish the transgenes. A non-overlapping generation, single-release trial of the same gene-drive strain resulted in two of three cages reaching 100% introduction within 6–12 generations following a 1:1 transgenic:wild-type male release. Two of three cages with 0.33:1 transgenic:wild-type male single releases achieved full introduction in 13–16 generations. All populations exhibiting full introduction went extinct within three generations due to a significant load on females having disruptions of both copies of the target gene, kynurenine hydroxylase. While repeated releases of high-ratio (10:1) non-drive constructs could achieve full introduction, results from the 1:1 release ratios across all experimental designs favor the use of gene drive, both for efficiency and anticipated cost of the control programs. The experimental introduction of manipulated genes into insect species has a long history in basic genetics. Recent advances in genome editing technologies have spurred considerable effort to exploit these methodologies to provide genetic solutions to some of the worst medical and agricultural problems caused by insects. Insect population suppression and population modification approaches have been proposed to control transmission of vector-borne diseases, including malaria. We used small cage trials to explore the efficacy of non-drive and gene-drive releases to deliver anti-malarial effector genes to a vector mosquito, Anopheles stephensi. We show that both approaches can work to introduce genes to high percentages, but as expected, the gene-drive approaches were more efficient in that they needed only a single release with a much lower number of released insects. The gene-drive females in our studies exhibited a significant load that resulted in some cage populations going to extinction. Furthermore, the accumulation of drive-resistant target genes prevented full introduction of the transgenes in those cages that did not go extinct. While none of the strains evaluated here are proposed for open release, these laboratory cage trials reveal features that can be used to improve next-generation gene-drive strains for population modification.
Collapse
|
40
|
Mumford JD, Long CA, Weaver SC, Miura K, Wang E, Rotenberry R, Dotson EM, Benedict MQ. Plasmodium falciparum (Haemosporodia: Plasmodiidae) and O'nyong-nyong Virus Development in a Transgenic Anopheles gambiae (Diptera: Culicidae) Strain. JOURNAL OF MEDICAL ENTOMOLOGY 2019; 56:936-941. [PMID: 30924861 PMCID: PMC6595505 DOI: 10.1093/jme/tjz032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Indexed: 06/09/2023]
Abstract
Transgenic Anopheles gambiae Giles (Diptera: Culicidae) mosquitoes have been developed that confer sexual sterility on males that carry a transgene encoding a protein which cuts ribosomal DNA. A relevant risk concern with transgenic mosquitoes is that their capacity to transmit known pathogens could be greater than the unmodified form. In this study, the ability to develop two human pathogens in these transgenic mosquitoes carrying a homing endonuclease which is expressed in the testes was compared with its nontransgenic siblings. Infections were performed with Plasmodium falciparum (Welch) and o'nyong-nyong virus (ONNV) and the results between the transgenic and nontransgenic sibling females were compared. There was no difference observed with ONNV isolate SG650 in intrathoracic infections or the 50% oral infectious dose measured at 14 d postinfection or in mean body titers. Some significant differences were observed for leg titers at the medium and highest doses for those individuals in which virus titer could be detected. No consistent difference was observed between the transgenic and nontransgenic comparator females in their ability to develop P. falciparum NF54 strain parasites. This particular transgene caused no significant effect in the ability of mosquitoes to become infected by these two pathogens in this genetic background. These results are discussed in the context of risk to human health if these transgenic individuals were present in the environment.
Collapse
Affiliation(s)
- John D Mumford
- Imperial College London, Centre for Environmental Policy, Silwood Park Campus, Ascot, Berkshire, UK
| | - Carole A Long
- NIH, NIAID, Laboratory of Malaria and Vector Research, Malaria Immunology Section, Twinbrook Pkwy, Rockville, MD
| | - Scott C Weaver
- Institute for Human Infections and Immunity and Department of Microbiology & Immunology, University of Texas Medical Branch (UTMB), Galveston, TX
| | - Katzutoyo Miura
- NIH, NIAID, Laboratory of Malaria and Vector Research, Malaria Immunology Section, Twinbrook Pkwy, Rockville, MD
| | - Eryu Wang
- Institute for Human Infections and Immunity and Department of Microbiology & Immunology, University of Texas Medical Branch (UTMB), Galveston, TX
| | - Rachel Rotenberry
- Centers for Disease Control and Prevention, DPDM/Entomology Branch, Atlanta, GA
| | - Ellen M Dotson
- Centers for Disease Control and Prevention, DPDM/Entomology Branch, Atlanta, GA
| | - Mark Q Benedict
- Centers for Disease Control and Prevention, DPDM/Entomology Branch, Atlanta, GA
| |
Collapse
|
41
|
Nash A, Urdaneta GM, Beaghton AK, Hoermann A, Papathanos PA, Christophides GK, Windbichler N. Integral gene drives for population replacement. Biol Open 2019; 8:bio037762. [PMID: 30498016 PMCID: PMC6361204 DOI: 10.1242/bio.037762] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 11/21/2018] [Indexed: 01/13/2023] Open
Abstract
A first generation of CRISPR-based gene drives has now been tested in the laboratory in a number of organisms, including malaria vector mosquitoes. Challenges for their use in the area-wide genetic control of vector-borne disease have been identified, including the development of target site resistance, their long-term efficacy in the field, their molecular complexity, and practical and legal limitations for field testing of both gene drive and coupled anti-pathogen traits. We have evaluated theoretically the concept of integral gene drive (IGD) as an alternative paradigm for population replacement. IGDs incorporate a minimal set of molecular components, including drive and anti-pathogen effector elements directly embedded within endogenous genes - an arrangement that in theory allows targeting functionally conserved coding sequences without disrupting their function. Autonomous and non-autonomous IGD strains could be generated, optimized, regulated and imported independently. We performed quantitative modeling comparing IGDs with classical replacement drives and show that selection for the function of the hijacked host gene can significantly reduce the establishment of resistant alleles in the population, while drive occurring at multiple genomic loci prolongs the duration of transmission blockage in the face of pre-existing target site variation. IGD thus has potential as a more durable and flexible population replacement strategy.
Collapse
Affiliation(s)
- Alexander Nash
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, South Kensington Campus, London SW7 2AZ, UK
| | - Giulia Mignini Urdaneta
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, South Kensington Campus, London SW7 2AZ, UK
| | - Andrea K Beaghton
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, South Kensington Campus, London SW7 2AZ, UK
| | - Astrid Hoermann
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, South Kensington Campus, London SW7 2AZ, UK
| | - Philippos Aris Papathanos
- Centre of Functional Genomics, Department of Experimental Medicine, University of Perugia, Perugia 06123, Italy
- Department of Entomology, Robert H. Smith Faculty of Agriculture, Food and Environment, Hebrew University of Jerusalem, Rehovot 7610001, Israel
| | - George K Christophides
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, South Kensington Campus, London SW7 2AZ, UK
| | - Nikolai Windbichler
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, South Kensington Campus, London SW7 2AZ, UK
| |
Collapse
|
42
|
Adolfi A, Lycett GJ. Opening the toolkit for genetic analysis and control of Anopheles mosquito vectors. CURRENT OPINION IN INSECT SCIENCE 2018; 30:8-18. [PMID: 30553490 DOI: 10.1016/j.cois.2018.07.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 07/24/2018] [Indexed: 06/09/2023]
Abstract
Anopheles is the only genus of mosquitoes that transmit human malaria and consequently the focus of large scale genome and transcriptome-wide association studies. Genetic tools to define the function of the candidate genes arising from these analyses are vital. Moreover, genome editing offers the potential to modify Anopheles population structure at local and global scale to provide complementary tools towards the ultimate goal of malaria elimination. Major breakthroughs in Anopheles genetic analysis came with the development of germline transformation and RNA interference technology. Yet, the field has been revolutionised again by precise genome editing now possible through site-specific nucleases. Here we review the components of the current genetic toolkit available to study Anopheles, focusing particularly on how these technical advances are used to gain insight into malaria transmission and the design of genetic methods to control Anopheles vectors.
Collapse
Affiliation(s)
- Adriana Adolfi
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, CA 92697-4500, USA
| | - Gareth John Lycett
- Vector Biology Department, Liverpool School of Tropical Medicine, Liverpool, L3 5QA, UK
| |
Collapse
|
43
|
Bilgo E, Vantaux A, Sanon A, Ilboudo S, Dabiré RK, Jacobs-Lorena M, Diabate A. Field assessment of potential sugar feeding stations for disseminating bacteria in a paratransgenic approach to control malaria. Malar J 2018; 17:367. [PMID: 30333029 PMCID: PMC6192189 DOI: 10.1186/s12936-018-2516-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 10/08/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Using bacteria to express and deliver anti-parasite molecules in mosquitoes is among the list of genetic tools to control malaria. The introduction and spread of transgenic bacteria through wild adult mosquitoes is one of the major challenges of this strategy. In prospect of future field experiments, an open field study with blank (without bacteria) attractive sugar bait (ASB) was performed under the assumption that transgenic bacteria would be spread to all sugar fed mosquitoes. METHODS Two types of ASB stations were developed, one with clay pots (CP) placed at mosquito resting sites and one with window entry traps (WET) placed inside inhabited houses. The ASB consisted in either glucose, honey or fruit cocktail solutions. In addition, mark-release-recapture (MRR) experiment of mosquitoes after feeding them with glucose was also conducted to check the proportion of the mosquito population that can be reached by the two ASB stations as well as its suitability to complement the ASB stations for disseminating bacteria. RESULTS Overall, 88% of the mosquitoes were collected in the WET_ASB. The CP_ASB stations were much less attractive with the highest average of 82 ± 11 mosquitoes/day in the CP near the wood piles. The proportions of sugar fed mosquitoes upon ASB were low in both type of ASB stations, ~ 2% and ~ 14% in WET and CP, respectively. Honey solution was the most attractive solution compared to the glucose and the fruit cocktail solutions. The recapture rate in the MRR experiment was low: ~ 4.1% over 7 days. CONCLUSION The WET_ASB looks promising to disseminate transgenic bacteria to endophilic West Africa Anopheles mosquito. However, this feeding station may not be fully effective and could be combined with the CP_ASB to also target outdoor resting mosquitoes. Overall, efforts are needed to improve the mosquito-feeding rates upon ASB.
Collapse
Affiliation(s)
- Etienne Bilgo
- Institut de Recherche en Sciences de la Santé/Centre Muraz, Bobo-Dioulasso, Burkina Faso. .,Laboratoire d'Entomologie Fondamentale et Appliqué/UFR-SVT/Université Ouaga I, Pr. Joseph KI-Zerbo, Ouagadougou, Burkina Faso.
| | | | - Antoine Sanon
- Laboratoire d'Entomologie Fondamentale et Appliqué/UFR-SVT/Université Ouaga I, Pr. Joseph KI-Zerbo, Ouagadougou, Burkina Faso
| | - Seni Ilboudo
- Institut de Recherche en Sciences de la Santé/Centre Muraz, Bobo-Dioulasso, Burkina Faso
| | - Roch K Dabiré
- Institut de Recherche en Sciences de la Santé/Centre Muraz, Bobo-Dioulasso, Burkina Faso
| | - Marcelo Jacobs-Lorena
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Abdoulaye Diabate
- Institut de Recherche en Sciences de la Santé/Centre Muraz, Bobo-Dioulasso, Burkina Faso
| |
Collapse
|
44
|
Rami A, Raz A, Zakeri S, Dinparast Djadid N. Isolation and identification of Asaia sp. in Anopheles spp. mosquitoes collected from Iranian malaria settings: steps toward applying paratransgenic tools against malaria. Parasit Vectors 2018; 11:367. [PMID: 29950179 PMCID: PMC6022440 DOI: 10.1186/s13071-018-2955-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 06/15/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In recent years, the genus Asaia (Rhodospirillales: Acetobacteraceae) has been isolated from different Anopheles species and presented as a promising tool to combat malaria. This bacterium has unique features such as presence in different organs of mosquitoes (midgut, salivary glands and reproductive organs) of female and male mosquitoes and vertical and horizontal transmission. These specifications lead to the possibility of introducing Asaia as a robust candidate for malaria vector control via paratransgenesis technology. Several studies have been performed on the microbiota of Anopheles mosquitoes (Diptera: Culicidae) in Iran and the Middle East to find a suitable candidate for controlling the malaria based on paratransgenesis approaches. The present study is the first report of isolation, biochemical and molecular characterization of the genus Asaia within five different Anopheles species which originated from different zoogeographical zones in the south, east, and north of Iran. METHODS Mosquitoes originated from field-collected and laboratory-reared colonies of five Anopheles spp. Adult mosquitoes were anesthetized; their midguts were isolated by dissection, followed by grinding the midgut contents which were then cultured in enrichment broth media and later in CaCO3 agar plates separately. Morphological, biochemical and physiological characterization were carried out after the appearance of colonies. For molecular confirmation, selected colonies were cultured, their DNAs were extracted and PCR was performed on the 16S ribosomal RNA gene using specific newly designed primers. RESULTS Morphological, biochemical, physiological and molecular results indicated that all isolates are members of the genus Asaia. CONCLUSIONS Contrary to previous opinions, our findings show that Asaia bacteria are present in both insectary-reared colonies and field-collected mosquitoes and can be isolated by simple and specific methods. Furthermore, with respect to the fact that we isolated Asaia within the different Anopheles specimens from distinct climatic and zoogeographical regions, it is promising and may be concluded that species of this genus can tolerate the complicated environmental conditions of the vector-borne diseases endemic regions. Therefore, it can be considered as a promising target in paratransgenesis and vector control programs. However, we suggest that introducing the new technologies such as next generation sequencing and robust in silico approaches may pave the way to find a unique biomarker for rapid and reliable differentiation of the Asaia species.
Collapse
Affiliation(s)
- Abbas Rami
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Abbasali Raz
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Sedigheh Zakeri
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Navid Dinparast Djadid
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
45
|
Huang Y, Chen J, Sun B, Zheng R, Li B, Li Z, Tan Y, Wei J, Pan G, Li C, Zhou Z. Engineered resistance to Nosema bombycis by in vitro expression of a single-chain antibody in Sf9-III cells. PLoS One 2018; 13:e0193065. [PMID: 29447266 PMCID: PMC5814085 DOI: 10.1371/journal.pone.0193065] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 02/02/2018] [Indexed: 11/18/2022] Open
Abstract
Nosema bombycis is a destructive, obligate intracellular parasite of the Bombyx mori. In this study, a single-chain variable fragment (scFv) dependent technology is developed for the purpose of inhibiting parasite proliferation in insect cells. The scFv-G4, which we prepared from a mouse G4 monoclonal antibody, can target the N. bombycis spore wall protein 12 (NbSWP12). Indirect immunofluorescence assays showed that NbSWP12 located mainly on the outside of the N. bombycis cytoskeleton, although some of it co-localized with β-tubulin in the meront-stage of parasites. When meront division began, NbSWP12 became concentrated at both ends of each meront. Western blotting showed that scFv-G4 could express in Sf9-III cells and recognized native NbSWP12. The transgenic Sf9-III cell line showed better resistance than the controls when challenged with N. bombycis, indicating that NbSWP12 is a promising target in this parasite and this scFv dependent strategy could be a solution for construction of N. bombycis-resistant Bombyx mori.
Collapse
Affiliation(s)
- Yukang Huang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, P. R. China
| | - Jie Chen
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, P. R. China
- Key Laboratory for Sericulture Functional Genomics and Biotechnology of Agricultural Ministry, Southwest University, Chongqing, P. R. China
| | - Bin Sun
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, P. R. China
| | - Rong Zheng
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, P. R. China
| | - Boning Li
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, P. R. China
| | - Zeng Li
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, P. R. China
| | - Yaoyao Tan
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, P. R. China
| | - Junhong Wei
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, P. R. China
- Key Laboratory for Sericulture Functional Genomics and Biotechnology of Agricultural Ministry, Southwest University, Chongqing, P. R. China
| | - Guoqing Pan
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, P. R. China
- Key Laboratory for Sericulture Functional Genomics and Biotechnology of Agricultural Ministry, Southwest University, Chongqing, P. R. China
| | - Chunfeng Li
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, P. R. China
- Key Laboratory for Sericulture Functional Genomics and Biotechnology of Agricultural Ministry, Southwest University, Chongqing, P. R. China
- * E-mail: (CL); (ZZ)
| | - Zeyang Zhou
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, P. R. China
- Key Laboratory for Sericulture Functional Genomics and Biotechnology of Agricultural Ministry, Southwest University, Chongqing, P. R. China
- College of Life Sciences, Chongqing Normal University, Chongqing, P. R. China
- * E-mail: (CL); (ZZ)
| |
Collapse
|
46
|
Abstract
The rapid spread of mosquito resistance to currently available insecticides, and the current lack of an efficacious malaria vaccine are among many challenges that affect large-scale efforts for malaria control. As goals of malaria elimination and eradication are put forth, new vector-control paradigms and tools and/or further optimization of current vector-control products are required to meet public health demands. Vector control remains the most effective measure to prevent malaria transmission and present gains against malaria mortality and morbidity may be maintained as long as vector-intervention strategies are sustained and adapted to underlying vector-related transmission dynamics. The following provides a brief overview of vector-control strategies and tools either in use or under development and evaluation that are intended to exploit key entomological parameters toward driving down transmission.
Collapse
Affiliation(s)
- Neil F Lobo
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana 46556
| | - Nicole L Achee
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana 46556
| | - John Greico
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana 46556
| | - Frank H Collins
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana 46556
| |
Collapse
|
47
|
Abstract
Vector control strategies based on population modification of Anopheline mosquitoes may have a significant role in the malaria eradication agenda. They could consolidate elimination gains by providing barriers to the reintroduction of parasites and competent vectors, and allow resources to be allocated to new control sites while maintaining treated areas free of malaria. Synthetic biological approaches are being used to generate transgenic mosquitoes for population modification. Proofs-of-principle exist for mosquito transgenesis, the construction of anti-parasite effector genes and gene-drive systems for rapidly introgressing beneficial genes into wild populations. Key challenges now are to develop field-ready strains of mosquitoes that incorporate features that maximize safety and efficacy, and specify pathways from discovery to development. We propose three pathways and a framework for target product profiles that maximize safety and efficacy while meeting the demands of the complexity of malaria transmission, and the regulatory and social diversity of potential end-users and stakeholders.
Collapse
Affiliation(s)
| | - Anthony A. James
- Department of Microbiology & Molecular Genetics, University of California, Irvine, CA, USA
- Department of Molecular Biology & Biochemistry, University of California, Irvine, CA, USA
| |
Collapse
|
48
|
Wang S, Dos-Santos ALA, Huang W, Liu KC, Oshaghi MA, Wei G, Agre P, Jacobs-Lorena M. Driving mosquito refractoriness to Plasmodium falciparum with engineered symbiotic bacteria. Science 2017; 357:1399-1402. [PMID: 28963255 PMCID: PMC9793889 DOI: 10.1126/science.aan5478] [Citation(s) in RCA: 172] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 08/24/2017] [Indexed: 12/30/2022]
Abstract
The huge burden of malaria in developing countries urgently demands the development of novel approaches to fight this deadly disease. Although engineered symbiotic bacteria have been shown to render mosquitoes resistant to the parasite, the challenge remains to effectively introduce such bacteria into mosquito populations. We describe a Serratia bacterium strain (AS1) isolated from Anopheles ovaries that stably colonizes the mosquito midgut, female ovaries, and male accessory glands and spreads rapidly throughout mosquito populations. Serratia AS1 was genetically engineered for secretion of anti-Plasmodium effector proteins, and the recombinant strains inhibit development of Plasmodium falciparum in mosquitoes.
Collapse
Affiliation(s)
- Sibao Wang
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, China,Corresponding author. (S.W.); (M.J.-L.)
| | - André L. A. Dos-Santos
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Wei Huang
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Kun Connie Liu
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Mohammad Ali Oshaghi
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Ge Wei
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Peter Agre
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Marcelo Jacobs-Lorena
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA,Corresponding author. (S.W.); (M.J.-L.)
| |
Collapse
|
49
|
Xu X, Zhang R, Chen X. Application of a single-chain fragment variable (scFv) antibody for the confirmatory diagnosis of hydatid disease in non-endemic areas. ELECTRON J BIOTECHN 2017. [DOI: 10.1016/j.ejbt.2017.07.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
50
|
Pila EA, Li H, Hambrook JR, Wu X, Hanington PC. Schistosomiasis from a Snail's Perspective: Advances in Snail Immunity. Trends Parasitol 2017; 33:845-857. [PMID: 28803793 DOI: 10.1016/j.pt.2017.07.006] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 07/14/2017] [Accepted: 07/17/2017] [Indexed: 12/17/2022]
Abstract
The snail's immune response is an important determinant of schistosome infection success, acting in concert with host, parasite, and environmental factors. Coordinated by haemocytes and humoral factors, it possesses immunological hallmarks such as pattern recognition receptors, and predicted gastropod-unique factors like the immunoglobulin superfamily domain-containing fibrinogen-related proteins. Investigations into mechanisms that underpin snail-schistosome compatibility have advanced quickly, contributing functional insight to many observational studies. While the snail's immune response is important to continue studying from the perspective of evolutionary immunology, as the foundational determinants of snail-schistosome compatibility continue to be discovered, the possibility of exploiting the snail for schistosomiasis control moves closer into reach. Here, we review the current understanding of immune mechanisms that influence compatibility between Schistosoma mansoni and Biomphalaria glabrata.
Collapse
Affiliation(s)
- Emmanuel A Pila
- School of Public Health, University of Alberta, Edmonton, AB T6G2G7, Canada; These authors contributed equally to this manuscript
| | - Hongyu Li
- School of Public Health, University of Alberta, Edmonton, AB T6G2G7, Canada; Ocean College, Qinzhou University, Qinzhou, Guangxi 535099, China; These authors contributed equally to this manuscript
| | - Jacob R Hambrook
- School of Public Health, University of Alberta, Edmonton, AB T6G2G7, Canada; These authors contributed equally to this manuscript
| | - Xinzhong Wu
- Ocean College, Qinzhou University, Qinzhou, Guangxi 535099, China
| | - Patrick C Hanington
- School of Public Health, University of Alberta, Edmonton, AB T6G2G7, Canada.
| |
Collapse
|