Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Front Immunol ; 12: 720550, 2021.
Article in English | MEDLINE | ID: mdl-34733274

ABSTRACT

Targeted delivery of antigen to antigen presenting cells (APCs) is an efficient way to induce robust antigen-specific immune responses. Here, we present a novel DNA vaccine that targets the Plasmodium falciparum reticulocyte-binding protein homolog 5 (PfRH5), a leading blood-stage antigen of the human malaria pathogen, to APCs. The vaccine is designed as bivalent homodimers where each chain is composed of an amino-terminal single chain fragment variable (scFv) targeting unit specific for major histocompatibility complex class II (MHCII) expressed on APCs, and a carboxyl-terminal antigenic unit genetically linked by the dimerization unit. This vaccine format, named "Vaccibody", has previously been successfully applied for antigens from other infectious diseases including influenza and HIV, as well as for tumor antigens. Recently, the crystal structure and key functional antibody epitopes for the truncated version of PfRH5 (PfRH5ΔNL) were characterized, suggesting PfRH5ΔNL to be a promising candidate for next-generation PfRH5 vaccine design. In this study, we explored the APC-targeting strategy for a PfRH5ΔNL-containing DNA vaccine. BALB/c mice immunized with the targeted vaccine induced higher PfRH5-specific IgG1 antibody responses than those vaccinated with a non-targeted vaccine or antigen alone. The APC-targeted vaccine also efficiently induced rapid IFN-γ and IL-4 T cell responses. Furthermore, the vaccine-induced PfRH5-specific IgG showed inhibition of growth of the P. falciparum 3D7 clone parasite in vitro. Finally, sera obtained after vaccination with this targeted vaccine competed for the same epitopes as PfRH5-specific mAbs from vaccinated humans. Robust humoral responses were also induced by a similar P. vivax Duffy-binding protein (PvDBP)-containing targeted DNA vaccine. Our data highlight a novel targeted vaccine platform for the development of vaccines against blood-stage malaria.


Subject(s)
Antibodies, Protozoan/immunology , Antigen-Presenting Cells/immunology , Carrier Proteins/immunology , Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control , Plasmodium falciparum/immunology , T-Lymphocytes/immunology , Vaccines, DNA/immunology , Animals , Antibodies, Neutralizing/immunology , Antibody Specificity/immunology , Antigen-Presenting Cells/metabolism , Antigens, Protozoan/immunology , Disease Models, Animal , Epitopes/immunology , Female , Gene Order , Genetic Vectors/genetics , Immunization , Malaria, Falciparum/immunology , Malaria, Falciparum/metabolism , Mice , T-Lymphocytes/metabolism
2.
Malar J ; 18(1): 300, 2019 Sep 02.
Article in English | MEDLINE | ID: mdl-31477111

ABSTRACT

BACKGROUND: The ability to report vaccine-induced IgG responses in terms of µg/mL, as opposed arbitrary units (AU), enables a more informed interpretation of the magnitude of the immune response, and better comparison between vaccines targeting different antigens. However, these interpretations rely on the accuracy of the methodology, which is used to generate ELISA data in µg/mL. In a previous clinical trial of a vaccine targeting the apical membrane antigen 1 (AMA1) from Plasmodium falciparum, three laboratories (Oxford, NIH and WRAIR) reported ELISA data in µg/mL that were correlated but not concordant. This current study sought to harmonize the methodology used to generate a conversion factor (CF) for ELISA analysis of human anti-AMA1 IgG responses across the three laboratories. METHODS: Purified IgG was distributed to the three laboratories and, following a set protocol provided by NIH, AMA1-specific human IgG was affinity purified. A new "harmonized CF" was generated by each laboratory using their in-house ELISA, and the original clinical trial ELISA data were re-analysed accordingly. RESULTS: Statistical analysis showed that the data remained highly correlated across all three laboratories, although only Oxford and NIH were able to harmonize their CF for ELISA and generate concordant data. CONCLUSIONS: This study enabled two out of the three laboratories to harmonize their µg/mL readouts for the human anti-AMA1 IgG ELISA, but results reported from WRAIR are ~ twofold higher. Given the need to validate such information for each species and antigen of interest, it is important to bear in mind these likely differences when interpreting µg/mL ELISA data in the future.


Subject(s)
Antibodies, Protozoan/analysis , Clinical Laboratory Techniques/standards , Enzyme-Linked Immunosorbent Assay/standards , Immunoglobulin G/analysis , Malaria Vaccines/immunology , Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , Clinical Trials as Topic , Humans , Immunoglobulin G/immunology , Malaria, Falciparum/prevention & control , Membrane Proteins/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology
3.
Front Immunol ; 10: 1254, 2019.
Article in English | MEDLINE | ID: mdl-31214195

ABSTRACT

The malaria genome encodes over 5,000 proteins and many of these have also been proposed to be potential vaccine candidates, although few of these have been tested clinically. RH5 is one of the leading blood-stage Plasmodium falciparum malaria vaccine antigens and Phase I/II clinical trials of vaccines containing this antigen are currently underway. Its likely mechanism of action is to elicit antibodies that can neutralize merozoites by blocking their invasion of red blood cells (RBC). However, many other antigens could also elicit neutralizing antibodies against the merozoite, and most of these have never been compared directly to RH5. The objective of this study was to compare a range of blood-stage antigens to RH5, to identify any antigens that outperform or synergize with anti-RH5 antibodies. We selected 55 gene products, covering 15 candidate antigens that have been described in the literature and 40 genes selected on the basis of bioinformatics functional prediction. We were able to make 20 protein-in-adjuvant vaccines from the original selection. Of these, S-antigen and CyRPA robustly elicited antibodies with neutralizing properties. Anti-CyRPA IgG generally showed additive GIA with anti-RH5 IgG, although high levels of anti-CyRPA-specific rabbit polyclonal IgG were required to achieve 50% GIA. Our data suggest that further vaccine antigen screening efforts are required to identify a second merozoite target with similar antibody-susceptibility to RH5.


Subject(s)
Erythrocytes/immunology , Erythrocytes/parasitology , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Merozoites/immunology , Plasmodium falciparum/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Protozoan/immunology , Antibody Specificity/immunology , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Female , Humans , Immunoglobulin G/immunology , Malaria, Falciparum/prevention & control , Mice , Plasmodium falciparum/genetics , Plasmodium falciparum/growth & development , Polymorphism, Genetic , Protozoan Proteins/genetics , Protozoan Proteins/immunology
4.
Cell ; 178(1): 216-228.e21, 2019 06 27.
Article in English | MEDLINE | ID: mdl-31204103

ABSTRACT

The Plasmodium falciparum reticulocyte-binding protein homolog 5 (PfRH5) is the leading target for next-generation vaccines against the disease-causing blood-stage of malaria. However, little is known about how human antibodies confer functional immunity against this antigen. We isolated a panel of human monoclonal antibodies (mAbs) against PfRH5 from peripheral blood B cells from vaccinees in the first clinical trial of a PfRH5-based vaccine. We identified a subset of mAbs with neutralizing activity that bind to three distinct sites and another subset of mAbs that are non-functional, or even antagonistic to neutralizing antibodies. We also identify the epitope of a novel group of non-neutralizing antibodies that significantly reduce the speed of red blood cell invasion by the merozoite, thereby potentiating the effect of all neutralizing PfRH5 antibodies as well as synergizing with antibodies targeting other malaria invasion proteins. Our results provide a roadmap for structure-guided vaccine development to maximize antibody efficacy against blood-stage malaria.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Protozoan/immunology , Erythrocytes/parasitology , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Adolescent , Adult , Animals , Binding Sites , Carrier Proteins/immunology , Cross Reactions/immunology , Epitopes/immunology , Female , HEK293 Cells , Healthy Volunteers , Humans , Malaria, Falciparum/parasitology , Male , Merozoites/physiology , Middle Aged , Plasmodium falciparum/metabolism , Protozoan Proteins/immunology , Rabbits , Rats , Rats, Sprague-Dawley , Young Adult
5.
Nat Microbiol ; 4(9): 1497-1507, 2019 09.
Article in English | MEDLINE | ID: mdl-31133755

ABSTRACT

The most widespread form of malaria is caused by Plasmodium vivax. To replicate, this parasite must invade immature red blood cells through a process requiring interaction of the P. vivax Duffy binding protein (PvDBP) with its human receptor, the Duffy antigen receptor for chemokines. Naturally acquired antibodies that inhibit this interaction associate with clinical immunity, suggesting PvDBP as a leading candidate for inclusion in a vaccine to prevent malaria due to P. vivax. Here, we isolated a panel of monoclonal antibodies from human volunteers immunized in a clinical vaccine trial of PvDBP. We screened their ability to prevent PvDBP from binding to the Duffy antigen receptor for chemokines, and their capacity to block red blood cell invasion by a transgenic Plasmodium knowlesi parasite genetically modified to express PvDBP and to prevent reticulocyte invasion by multiple clinical isolates of P. vivax. This identified a broadly neutralizing human monoclonal antibody that inhibited invasion of all tested strains of P. vivax. Finally, we determined the structure of a complex of this antibody bound to PvDBP, indicating the molecular basis for inhibition. These findings will guide future vaccine design strategies and open up possibilities for testing the prophylactic use of such an antibody.


Subject(s)
Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , Malaria Vaccines/immunology , Malaria, Vivax/prevention & control , Plasmodium vivax/immunology , Protozoan Proteins/immunology , Receptors, Cell Surface/immunology , Antibodies, Protozoan/chemistry , Antigens, Protozoan/chemistry , Antigens, Protozoan/genetics , Antigens, Protozoan/metabolism , Crystallography, X-Ray , Duffy Blood-Group System/metabolism , Epitopes, B-Lymphocyte , Erythrocytes/parasitology , Genetic Variation , Humans , Immunoglobulin Fab Fragments , Malaria Vaccines/administration & dosage , Malaria, Vivax/parasitology , Plasmodium knowlesi/genetics , Plasmodium knowlesi/growth & development , Plasmodium knowlesi/immunology , Plasmodium vivax/genetics , Plasmodium vivax/growth & development , Protein Binding , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Reticulocytes/parasitology
6.
Cell Rep ; 27(1): 172-186.e7, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30943399

ABSTRACT

We describe therapeutic monoclonal antibodies isolated from human volunteers vaccinated with recombinant adenovirus expressing Ebola virus glycoprotein (EBOV GP) and boosted with modified vaccinia virus Ankara. Among 82 antibodies isolated from peripheral blood B cells, almost half neutralized GP pseudotyped influenza virus. The antibody response was diverse in gene usage and epitope recognition. Although close to germline in sequence, neutralizing antibodies with binding affinities in the nano- to pico-molar range, similar to "affinity matured" antibodies from convalescent donors, were found. They recognized the mucin-like domain, glycan cap, receptor binding region, and the base of the glycoprotein. A cross-reactive cocktail of four antibodies, targeting the latter three non-overlapping epitopes, given on day 3 of EBOV infection, completely protected guinea pigs. This study highlights the value of experimental vaccine trials as a rich source of therapeutic human monoclonal antibodies.


Subject(s)
Antibodies, Monoclonal/isolation & purification , Ebola Vaccines/isolation & purification , Ebola Vaccines/therapeutic use , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/therapy , Vaccination , Adolescent , Adult , Animals , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/isolation & purification , Antibodies, Neutralizing/therapeutic use , Antibodies, Viral/blood , Antibodies, Viral/isolation & purification , Antibodies, Viral/therapeutic use , Cells, Cultured , Dogs , Female , Guinea Pigs , HEK293 Cells , Hemorrhagic Fever, Ebola/blood , Hemorrhagic Fever, Ebola/immunology , Humans , Madin Darby Canine Kidney Cells , Male , Middle Aged , Vaccination/methods , Young Adult
7.
JCI Insight ; 2(21)2017 11 02.
Article in English | MEDLINE | ID: mdl-29093263

ABSTRACT

The development of a highly effective vaccine remains a key strategic goal to aid the control and eventual eradication of Plasmodium falciparum malaria. In recent years, the reticulocyte-binding protein homolog 5 (RH5) has emerged as the most promising blood-stage P. falciparum candidate antigen to date, capable of conferring protection against stringent challenge in Aotus monkeys. We report on the first clinical trial to our knowledge to assess the RH5 antigen - a dose-escalation phase Ia study in 24 healthy, malaria-naive adult volunteers. We utilized established viral vectors, the replication-deficient chimpanzee adenovirus serotype 63 (ChAd63), and the attenuated orthopoxvirus modified vaccinia virus Ankara (MVA), encoding RH5 from the 3D7 clone of P. falciparum. Vaccines were administered i.m. in a heterologous prime-boost regimen using an 8-week interval and were well tolerated. Vaccine-induced anti-RH5 serum antibodies exhibited cross-strain functional growth inhibition activity (GIA) in vitro, targeted linear and conformational epitopes within RH5, and inhibited key interactions within the RH5 invasion complex. This is the first time to our knowledge that substantial RH5-specific responses have been induced by immunization in humans, with levels greatly exceeding the serum antibody responses observed in African adults following years of natural malaria exposure. These data support the progression of RH5-based vaccines to human efficacy testing.


Subject(s)
Antibodies, Neutralizing , Carrier Proteins/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Protozoan Proteins/immunology , Vaccination , Adaptive Immunity , Adult , Antibodies, Protozoan/blood , Carrier Proteins/genetics , Epitopes/immunology , Female , Genetic Vectors , Humans , Immunization , Male , Middle Aged , Plasmodium falciparum/genetics , Vaccinia virus , Young Adult
8.
JCI Insight ; 2(12)2017 Jun 15.
Article in English | MEDLINE | ID: mdl-28614791

ABSTRACT

BACKGROUND: Plasmodium vivax is the most widespread human malaria geographically; however, no effective vaccine exists. Red blood cell invasion by the P. vivax merozoite depends on an interaction between the Duffy antigen receptor for chemokines (DARC) and region II of the parasite's Duffy-binding protein (PvDBP_RII). Naturally acquired binding-inhibitory antibodies against this interaction associate with clinical immunity, but it is unknown whether these responses can be induced by human vaccination. METHODS: Safety and immunogenicity of replication-deficient chimpanzee adenovirus serotype 63 (ChAd63) and modified vaccinia virus Ankara (MVA) viral vectored vaccines targeting PvDBP_RII (Salvador I strain) were assessed in an open-label dose-escalation phase Ia study in 24 healthy UK adults. Vaccines were delivered by the intramuscular route in a ChAd63-MVA heterologous prime-boost regimen using an 8-week interval. RESULTS: Both vaccines were well tolerated and demonstrated a favorable safety profile in malaria-naive adults. PvDBP_RII-specific ex-vivo IFN-γ T cell, antibody-secreting cell, memory B cell, and serum IgG responses were observed after the MVA boost immunization. Vaccine-induced antibodies inhibited the binding of vaccine homologous and heterologous variants of recombinant PvDBP_RII to the DARC receptor, with median 50% binding-inhibition titers greater than 1:100. CONCLUSION: We have demonstrated for the first time to our knowledge that strain-transcending antibodies can be induced against the PvDBP_RII antigen by vaccination in humans. These vaccine candidates warrant further clinical evaluation of efficacy against the blood-stage P. vivax parasite. TRIAL REGISTRATION: Clinicaltrials.gov NCT01816113. FUNDING: Support was provided by the UK Medical Research Council, UK National Institute of Health Research Oxford Biomedical Research Centre, and the Wellcome Trust.

9.
Int J Parasitol ; 47(7): 435-446, 2017 06.
Article in English | MEDLINE | ID: mdl-28153778

ABSTRACT

Development of bespoke biomanufacturing processes remains a critical bottleneck for translational studies, in particular when modest quantities of a novel product are required for proof-of-concept Phase I/II clinical trials. In these instances the ability to develop a biomanufacturing process quickly and relatively cheaply, without risk to product quality or safety, provides a great advantage by allowing new antigens or concepts in immunogen design to more rapidly enter human testing. These challenges with production and purification are particularly apparent when developing recombinant protein-based vaccines for difficult parasitic diseases, with Plasmodium falciparum malaria being a prime example. To that end, we have previously reported the expression of a novel protein vaccine for malaria using the ExpreS2Drosophila melanogaster Schneider 2 stable cell line system, however, a very low overall process yield (typically <5% recovery of hexa-histidine-tagged protein) meant the initial purification strategy was not suitable for scale-up and clinical biomanufacture of such a vaccine. Here we describe a newly available affinity purification method that was ideally suited to purification of the same protein which encodes the P. falciparum reticulocyte-binding protein homolog 5 - currently the leading antigen for assessment in next generation vaccines aiming to prevent red blood cell invasion by the blood-stage parasite. This purification system makes use of a C-terminal tag known as 'C-tag', composed of the four amino acids, glutamic acid - proline - glutamic acid - alanine (E-P-E-A), which is selectively purified on a CaptureSelect™ affinity resin coupled to a camelid single chain antibody, called NbSyn2. The C-terminal fusion of this short C-tag to P. falciparum reticulocyte-binding protein homolog 5 achieved >85% recovery and >70% purity in a single step purification directly from clarified, concentrated Schneider 2 cell supernatant under mild conditions. Biochemical and immunological analysis showed that the C-tagged and hexa-histidine-tagged P. falciparum reticulocyte-binding protein homolog 5 proteins are comparable. The C-tag technology has the potential to form the basis of a current good manufacturing practice-compliant platform, which could greatly improve the speed and ease with which novel protein-based products progress to clinical testing.


Subject(s)
Carrier Proteins/chemistry , Malaria Vaccines/immunology , Plasmodium falciparum/metabolism , Animals , Carrier Proteins/immunology , Carrier Proteins/metabolism , Cell Line , Cloning, Molecular , Rabbits
10.
Sci Rep ; 6: 30357, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27457156

ABSTRACT

The Plasmodium falciparum reticulocyte-binding protein homolog 5 (PfRH5) has recently emerged as a leading candidate antigen against the blood-stage human malaria parasite. However it has proved challenging to identify a heterologous expression platform that can produce a soluble protein-based vaccine in a manner compliant with current Good Manufacturing Practice (cGMP). Here we report the production of full-length PfRH5 protein using a cGMP-compliant platform called ExpreS(2), based on a Drosophila melanogaster Schneider 2 (S2) stable cell line system. Five sequence variants of PfRH5 were expressed that differed in terms of mutagenesis strategies to remove potential N-linked glycans. All variants bound the PfRH5 receptor basigin and were recognized by a panel of monoclonal antibodies. Analysis following immunization of rabbits identified quantitative and qualitative differences in terms of the functional IgG antibody response against the P. falciparum parasite. The antibodies induced by one protein variant were shown to be qualitatively similar to responses induced by other vaccine platforms. This work identifies Drosophila S2 cells as a clinically-relevant platform suited for the production of 'difficult-to-make' proteins from Plasmodium parasites, and identifies a PfRH5 sequence variant that can be used for clinical production of a non-glycosylated, soluble full-length protein vaccine immunogen.


Subject(s)
Carrier Proteins/immunology , Malaria Vaccines/immunology , Plasmodium falciparum/immunology , Animals , Antibodies, Monoclonal/immunology , Basigin/immunology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Drosophila melanogaster , Immunoglobulin G/immunology , Malaria Vaccines/genetics , Mutation
11.
J Infect Dis ; 213(11): 1743-51, 2016 Jun 01.
Article in English | MEDLINE | ID: mdl-26908756

ABSTRACT

BACKGROUND: Models of controlled human malaria infection (CHMI) initiated by mosquito bite have been widely used to assess efficacy of preerythrocytic vaccine candidates in small proof-of-concept phase 2a clinical trials. Efficacy testing of blood-stage malaria parasite vaccines, however, has generally relied on larger-scale phase 2b field trials in malaria-endemic populations. We report the use of a blood-stage P. falciparum CHMI model to assess blood-stage vaccine candidates, using their impact on the parasite multiplication rate (PMR) as the primary efficacy end point. METHODS: Fifteen healthy United Kingdom adult volunteers were vaccinated with FMP2.1, a protein vaccine that is based on the 3D7 clone sequence of apical membrane antigen 1 (AMA1) and formulated in Adjuvant System 01 (AS01). Twelve vaccinees and 15 infectivity controls subsequently underwent blood-stage CHMI. Parasitemia was monitored by quantitative real-time polymerase chain reaction (PCR) analysis, and PMR was modeled from these data. RESULTS: FMP2.1/AS01 elicited anti-AMA1 T-cell and serum antibody responses. Analysis of purified immunoglobulin G showed functional growth inhibitory activity against P. falciparum in vitro. There were no vaccine- or CHMI-related safety concerns. All volunteers developed blood-stage parasitemia, with no impact of the vaccine on PMR. CONCLUSIONS: FMP2.1/AS01 demonstrated no efficacy after blood-stage CHMI. However, the model induced highly reproducible infection in all volunteers and will accelerate proof-of-concept testing of future blood-stage vaccine candidates. CLINICAL TRIALS REGISTRATION: NCT02044198.


Subject(s)
Antigens, Protozoan/immunology , Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control , Membrane Proteins/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Adult , Enzyme-Linked Immunospot Assay , Erythrocytes/parasitology , Female , Humans , Immunogenicity, Vaccine , Life Cycle Stages , Malaria, Falciparum/parasitology , Male , Middle Aged , Models, Biological , Plasmodium falciparum/physiology , Young Adult
12.
PLoS Negl Trop Dis ; 6(7): e1724, 2012.
Article in English | MEDLINE | ID: mdl-22802980

ABSTRACT

BACKGROUND: Aedes albopictus, the Asian tiger mosquito, is a vector of several arboviruses including dengue and chikungunya, and is also a significant nuisance mosquito. It is one of the most invasive of mosquitoes with a relentlessly increasing geographic distribution. Conventional control methods have so far failed to control Ae. albopictus adequately. Novel genetics-based strategies offer a promising alternative or aid towards efficient control of this mosquito. METHODOLOGY/PRINCIPAL FINDINGS: We describe here the isolation, characterisation and use of the Ae. albopictus Actin-4 gene to drive a dominant lethal gene in the indirect flight muscles of Ae. albopictus, thus inducing a conditional female-specific late-acting flightless phenotype. We also show that in this context, the Actin-4 regulatory regions from both Ae. albopictus and Ae. aegypti can be used to provide conditional female-specific flightlessness in either species. CONCLUSION/SIGNIFICANCE: With the disease-transmitting females incapacitated, the female flightless phenotype encompasses a genetic sexing mechanism and would be suitable for controlling Ae. albopictus using a male-only release approach as part of an integrated pest management strategy.


Subject(s)
Aedes/physiology , Disease Vectors , Flight, Animal , Mosquito Control/methods , Actins/genetics , Aedes/genetics , Animals , Female , Male , Phenotype
13.
PLoS Negl Trop Dis ; 4(8): e788, 2010 Aug 17.
Article in English | MEDLINE | ID: mdl-20808959

ABSTRACT

BACKGROUND: The Asian tiger mosquito, Aedes albopictus (Skuse), is a vector of several arboviruses including dengue and chikungunya. This highly invasive species originating from Southeast Asia has travelled the world in the last 30 years and is now established in Europe, North and South America, Africa, the Middle East and the Caribbean. In the absence of vaccine or antiviral drugs, efficient mosquito control strategies are crucial. Conventional control methods have so far failed to control Ae. albopictus adequately. METHODOLOGY/PRINCIPAL FINDINGS: Germline transformation of Aedes albopictus was achieved by micro-injection of embryos with a piggyBac-based transgene carrying a 3xP3-ECFP marker and an attP site, combined with piggyBac transposase mRNA and piggyBac helper plasmid. Five independent transgenic lines were established, corresponding to an estimated transformation efficiency of 2-3%. Three lines were re-injected with a second-phase plasmid carrying an attB site and a 3xP3-DsRed2 marker, combined with PhiC31 integrase mRNA. Successful site-specific integration was observed in all three lines with an estimated transformation efficiency of 2-6%. CONCLUSIONS/SIGNIFICANCE: Both piggybac- and site-specific PhiC31-mediated germline transformation of Aedes albopictus were successfully achieved. This is the first report of Ae. albopictus germline transformation and engineering, a key step towards studying and controlling this species using novel molecular techniques and genetic control strategies.


Subject(s)
Aedes/genetics , Recombination, Genetic , Transformation, Genetic , Animals , Animals, Genetically Modified , Attachment Sites, Microbiological , Female , Genes, Reporter , Green Fluorescent Proteins/genetics , Integrases/genetics , Luminescent Proteins/genetics , Male , Microinjections/methods , Plasmids , Transposases/genetics , Red Fluorescent Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...