Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Critical Reviews™ in Eukaryotic Gene Expression, 27(4):331–340 (2017)

Genetically Modified Aedes aegypti to Control


Dengue: A Review
Muhammad Qsim,a Usman Ali Ashfaq,a,* Muhammad Zubair Yousaf,b Muhammad Shareef
Masoud,a Ijaz Rasul,a Namrah Noor,a & Azfar Hussainc
Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad-38000,
a

Pakistan; bCentres of Excellence in Science & Applied Technologies, Islamabad, Pakistan; cBuffalo Research Institute
Pattoki, Pakistan

*Address all correspondence to: Usman Ali Ashfaq, Department of Bioinformatics and Biotechnology, Government College University Faisalabad,
Pakistan; Tel./Fax: +92 03314728790, E-mail: usmancemb@gmail.com

ABSTRACT: Dengue is an acute infectious disease of viral etiology characterized by lymphadenopathy, leucopenia,
headache, biphasic fever, pain in various parts of the body, rashes, and extreme physical weakness. It is a vector-borne
disease caused by a positive-stranded RNA virus of the family Flaviviridae, genus Flavivirus. Dengue inflicts a sig-
nificant health, economic, and social burden on populations of endemic areas. Dengue virus is transmitted to humans
by the mosquito vector Aedes aegypti. Vaccines against dengue viruses have been claimed to be developed, but as yet
no effective treatment is available. Alternative therapeutic strategies to overcome this disease and its spread are direly
needed. A traditional sterile insect technique (SIT) harms the health of male insects, leading to their reduced ability to
compete for wild-type female insects for breeding. Oxitec (Abingdon, UK) has developed genetically modified (GM)
strains of A. aegypti via the release of insects carrying a dominant lethal (RIDL) strategy. RIDL male mosquitoes offer
a resolution to many of the limitations of traditional SIT, which has resulted in reduced application of SIT in mosqui-
toes. The technique using RIDL mosquitoes is considered to be ecologically friendly and specific. Homing endonu-
clease genes, also called selfish genes, can also be used in genetic modification methods in such a way that the vector
population and its competency can be reduced. GM mosquitoes carrying a gene that transcribes RNA interference can
also be crucial to control expression of RNA viruses. The RNA virus interference pathway is one of the most critical
components of the innate immune system of insects that can frustrate a variety of RNA viruses such as Flaviviruses.
Here, we summarize and focus on alternative techniques used to control dengue spread.

KEY WORDS: RNA interference, CRISPR-Cas9, Aedes aegypti, dengue, sterile insect technique

I. INTRODUCTION response in approximately 7 d. Secondary infection


is more severe and can cause dengue hemorrhagic
Dengue, probably the most important arthropod- fever or dengue shock syndrome (DSS).
borne viral (arbovirus) disease in terms of human Dengue, a member of the family Flaviviridae, is
morbidity and mortality, is a major public health a positive-sense RNA virus. Dengue virus has four
problem worldwide, with ~ 2.5 billion individuals major serotypes (DENV1–4) that are antigenically
infected, leading to nearly 25,000 deaths per year.1,2 distinct from one another, but all cause disease.6,7
Clinical signs and symptoms can vary from acute Dengue virus is 50-nm-containing lipid mem-
fever with headache, chills, and muscle or joint pain brane, with 180 identical copies of envelop (E) pro-
to severe life-threatening symptoms. It is estimated tein present at the surface of the viral membrane’s
that more than 100 countries with populated of short transmembrane segment. The virus genome
~ 50–100 million individuals are influenced by this (of about 11,000 bases) is translated into a single
infection, with Asia, Central and South America, large polyprotein that is cleaved into three structural
and Africa the main areas affected.3–5 Dengue infec- and seven nonstructural genes. Short noncoding re-
tions may be categorized as primary or secondary. gions are also present on both the 5′ and 3′ ends.
Primary infection causes an acute fever known as Structural proteins include capsid (C), E glycopro-
dengue fever that can be cured by a complex immune tein, and membrane (M). Nonstructural proteins are

1045-4403/17/$35.00 © 2017 Begell House, Inc. www.begellhouse.com 331


332 Qsim et al.

classed as NS1, NS2A, NS2B, NS3, NS4A, NS4B, countries have escaped the morbidity and mortality
and NS5 (Fig. 1).8,9 Structural proteins play a vital that mosquitoes and their arthropod associates inflict
part in the formational organization of virus and on humans via diseases such as dengue and malaria.
viral entrance into host cells, whereas nonstructural A worldwide increase of vectors is one of the major
proteins are key in viral replication and other cel- reasons for the emergence of different diseases in
lular functions.3,4 new areas. Therefore, there is an increasing demand
No specific vaccine or therapeutic drug is avail- to develop strategies for vector control. Since 1950s,
able yet for dengue. The Sanofi Pasture (Lyon, dengue has expanded geographically worldwide at a
France) vaccine chimeric yellow fever virus-DENV high rate. The rise in dengue spread is alarming and
tetravalent dengue vaccine (CYD-TDV) is the most is responsible for many casualties.12 Several factors
recent vaccine in clinical trials now. This vaccine, are involved in the spread of dengue fever, and the
formed by mixing all four serotype recombinant vi- Aedes aegypti mosquito vector is one of them.10 The
ruses, exhibited good results in initial trials, but its Aedes mosquito is thus a good target for overcoming
overall efficacy is low, especially against dengue dengue fever, but comprehensive work is required to
virus serotype 2 (DENV2).10 A study in India demon- control this vector.
strated the development of a vaccine for the preven- Because dengue can be controlled by reduction or
tion of dengue fever via the yeast Pichia pastoris. elimination of the vector population, the prevention
Noninfectious dengue-virus-like particles were cre- of dengue virus transmission relies heavily on vector
ated to activate virus-specific immunity. Animal trials control. Traditional methods of vector control such
using this vaccine yielded good results, but it is not as bed nets and space spraying are largely ineffective
yet commercially available.11 There is thus a strong against this daytime biting mosquito, so new strate-
need to find alternative ways to control this disease. gies are needed. Vector control strategies are classed
Vector-borne diseases are key threats to human into two basic methods: population suppression (re-
health and pose a dreadful and intolerable public duce) or population elimination (replace), both with
health load on society. To a great extent, developed the aid of genetically modified (GM) mosquitoes

FIG. 1: Genome of the dengue virus

Critical Reviews™ in Eukaryotic Gene Expression


GM Aedes aegypti to Control Dengue 333

(GMMs).13 In this review, we focus on several inno- New World screwworm fly in the United States.21
vative techniques for controlling the dengue vector. Although a series of tests were conducted in 1970s
with some success, no SIT programs are currently
in large-scale operation against mosquito species.22
II. SELF-LIMITING POPULATION-
A. aegypti, a mosquito species fit for mass rearing,
SUPPRESSION MECHANISMS
appears to be reasonably uniform across large areas,
A. Sterile Insect Technique without the problems of subspecies and barriers to
mating.23 Recently, to predict effectiveness of SIT,
The sterile insect technique (SIT) is a species-spe- various studies focused on designing mathematical
cific, environmental friendly method of biological models to simulate the application of the technique
insect control that is based on the release of large to A. aegypti.24
numbers of sterile insects into the environment.
With the population-suppression method, the vector 1. SIT Advantages versus Traditional
number is reduced to minimize disease transmission. Strategies
In population replacement, the wild-type vector is
replaced with a GM vector (GMV) that is incapable • SIT is species specific, thus targeted species
of transmitting the disease.14 For this method, a ge- populations only are eliminated.
netic control system is devised based on the release • The mating behavior of male insects is highly
of radiation-sterilized insects. This technique en- efficient.
compasses mating of male GMMs with native fe- • Wild insect density is condensed, making it
males, resulting in decreased female reproductive more effective.25
potential.15 Wild female pairing to a sterile male pro-
duces fewer or no children, so the population tends 2. SIT Limitations
to decrease. If enough sterile insects are released for
an appropriate period, the target population is con- • A loss of competitiveness mating for wild-type
trolled or even eradicated locally.16 Large numbers insects occurs, due to sterilization by irradiation.
of sterile males released into the environment mate • The success of SIT is highly dependent on
with wild-type females, reducing the reproductive release of male mosquitoes only; sterile fe-
capacity of mosquitoes.17 It is noteworthy that re- males bite and consequently transmit disease
duction of mating competitiveness and sterile fe- (male mosquitoes do not bite). Therefore, SIT
males do not reduce the population, and the fertility involves labor-intensive gender-based sep­
of irradiated males is a residual effect that limits the aration of male-only populations.
approaches to mosquito control using SIT. With SIT, • Irradiation of pupae appears to harm insects;
liberated male mosquitoes do not take blood meals, irradiation as adults is less harmful but oper­
resulting in a density-dependent reduction due to le- ation­ally much more difficult. In some trials,
thality performance at very early stages, leading to sterilizing chemicals were used, such as thio­
larval death.18,19 The reduction in fecundity declines tepa, which was effective for sterilization but
in both arthropod-borne pathogen transmission as led to trace contamination with this mutagenic
well as the insect population. chemical.
Numerous sterilizing techniques for mosquito • Unlike agricultural pests against which the
control such as chemosterilization, ionizing radiation, main SIT programs are directed at present,
cytoplasmic incompatibility, and chromosome trans- mosquito populations can be regulated pri-
location have been in use since the 1970s.20 SIT has marily by density-dependent effects in which
been used successfully for more than 50 yr to control a very fruitful population is maintained at a
an entire area and/or eliminate several important ag- level stable for limited resource constraints.
ricultural pests and disease vectors, including the re- • Fitness costs and operational difficulty of ir-
gional elimination of the Mediterranean fruit fly and radiation in SIT field operations.5,6,11

Volume 27, Issue 4, 2017


334 Qsim et al.

To overcome these SIT limitations, GM female 1. Construction of GMMs


mosquitoes may be used. In female transgenic mos-
quitoes, OX3604C genes are present to produce the GM male mosquitoes have been developed that con-
flightless female-tetracycline-repressible pheno- tain the lethal gene to combat the spread of dengue
type. Thus, flightless females become incapable of fever. LA513 is a dependent piggyBac-based trans-
mating (Fig. 2).26 poson containing 8.4 kb of nucleotides. Inserting the
LA513 transposon within A. aegypti mosquitoes by
B. Release of Insects Carrying a Dominant genetic engineering techniques has been performed
Lethal Gene to produce toxins in the mosquitoes during the larval
stage under normal conditions. This then causes the
Population studies using SIT restricted mosquito death of larvae. Transposon LA513 mainly consists of
control due to the somatic damage, producing an as- DsRed2 and tetracycline-repressible transcriptional
sociated performance reduction in the sterile insects activator (tTAV) genes. OX513A individuals can be
that inevitably accompanies radiation-based steril- detected under fluorescent light due to the expression
ization.15 However, modern genetics can overcome of a second, dominantly inherited, introduced gene that
this problem by inserting in insects an engineered encodes for red fluorescent protein (DsRed2). tTAV is
self-limiting gene that eliminates the need for ra- controlled by its binding site tetO, the minimal pro-
diation sterilization in a similar SIT program. The moter from Drosophila heat-shock protein (Hsp70),
release of insects carrying a dominant lethal (RIDL) and the 3′ untranslated region (UTR) sequence from
strategy reduces these limitations by way of the re- the Drosophila gene Dmelfs(1)K10. tTAV binds with
combinant DNA technology that was introduced by its binding site tetO, executing a high-level expression
Oxitec (Abington, UK). RIDL uses GM male and of tTAV that is toxic under normal conditions. In the
female mosquitoes.27,28 OX513A RIDL bisex is a presence of tetracycline, tTAV binds with tetracycline
GM strain from which only males are released to instead of tetO, leading to the reduced expression of
mate with wild females. The offspring of these mat- tTAV. Therefore, this construction gives a very high-
ings die due to late larvae or pupae. Thus, the target level expression of tTAV in the absence of tetracycline
population can easily reduce.16,27 At the same time, but executes minimal to no expression in the presence
some or all of the descendants of GM mosquitoes of tetracycline. The high-level expression of tTAV may
die because of inheriting one or more dominant le- be due to the interaction with the VP16 domain tran-
thal mutations, resulting in the tendency for the pop- scription factor, which provides a key to the building
ulation to decline.28,29 of this deadly system (Fig. 3).29,30

FIG. 2: The GM flightless female lethal gene is inserted into the vector (GM male) that breeds with wild-type females
to produce flightless females

Critical Reviews™ in Eukaryotic Gene Expression


GM Aedes aegypti to Control Dengue 335

Fluorescent Marker

FIG. 3: LA513 transposon structure and function. LA513 is an 8.4-kb piggyBac-based transposon. DsRed is the
fluorescent marker that is used to identify the transgenic gene. The Act5C promoter is used to drive the expression of
DsRed. tTAV is a tetracycline-repressible transcription activator that is controlled by its binding site tetO, the minimal
promoter of Drosophila Hsp70, and a 3’ UTR sequence of Drosophila fs(1)K10 that all control tTAV.

When GMMs are released into the environment HEGs are inserted in their own recognition site, and
and mate with wild-type females, their offspring in- recombination repair machinery then uses the HEG-
herit this trait lethality and die before reaching the age enclosing strand as a template. Thus, another HEG-
of maturity (during the larval stage), resulting in a de- enclosing strand is formed and the number of HEGs
crease of the local mosquito swarms.26 As described increases. In this manner, HEGs serve as the vehicle
above, the RIDL strategy uses the GM A. aegypti for driving genes through a population.
OX513 abisexual strain (Fig. 4).29 Furthermore, by HEGs can also be planned to identify specific
means of GM male A. aegypti, reduction or elimina- sequences of mosquito genes required for vector
tion of the mosquito population of that species can proficiency.32 On the other hand, HEGs can be used
minimize transmission of the dengue virus to humans. for population suppression by pointing genes to en-
courage infertility and decrease survival or sex ratio
C. Homing Endonuclease Genes alterations. So far, HEGs have been positively engi-
neered into A. aegypti.26
Homing endonuclease genes (HEGs) were first
discovered in bacteria.6 For insect control, HEGs III. POPULATION REPLACEMENT FOR
provide a powerful mechanism for driving genes CONTROLLING THE DENGUE VECTOR
through populations, but although they are pri-
marily used as gene drivers, self-limiting forms of Population-replacement techniques involve the in-
HEG control are also possible.31 HEGs encode en- sertion of additional or foreign genes to prevent
donuclease enzymes that recognize the site, cleave a mosquitoes from transmitting disease. These tech-
20–30-bp sequence present on the chromosome, and niques may include inserting an antipathogen gene,
insert HEGs into this site.32 HEGs could be used to immune system up-regulator, or a combination of
dislocate genes that are essential to pathogen spread transgenic approaches that will ultimately result in
or activate homologous recombination and gene al- the reduction or elimination of disease transmission
teration to increase the presence of transgenic an- from mosquito to human. To assess whether a mos-
tipathogen genes.6 quito has reduced disease transmission capability,
Although HEGs are naturally present in mi- the virus titer in the experimental transgenic mos-
crobes, those that are genetically engineered can quito’s midgut and salivary glands can be measured
perform functions in mosquitoes.33 Endonucleases and compared to a wild-type control.35,36
enzymes that recognize the site present only once At least three genetic-transformation systems
in the genome, cleave a 20–30-bp sequence, and in- have been described and used successfully in A.
serted HEGs are then saved from their own activity.34 aegypti to generate GMVs. These transformation

Volume 27, Issue 4, 2017


336

FIG. 4: RIDL mechanism. The LA513 transposon inserts in the produced cultured cell GM larvae, which is kept in the presence of tetracycline to prohibit
gene expression. Eggs are produced after breeding. Lethal genes are expressed and the larvae die due to the absence of tetracycline; wild-type male
progeny are not formed.

Critical Reviews™ in Eukaryotic Gene Expression


Qsim et al.
GM Aedes aegypti to Control Dengue 337

systems are based on the class II transposable el- SINV was converted into dsRNA intermediary, ex-
ements (TEs) Mariner (Mos1), Hermes, and pig- pressing DENV2-prM to activate the RNAi mecha-
gyBac. Mos1 and piggyBac are the most commonly nism. The combination of DENV2 and recombinant
used TEs for generating GMVs.37 SINV with the DENV2-prM sequence reduced
DENV replication in mosquitoes.38
A. Control through Gene Manipulation In a mosquito cell-culture expression of a
via RNA Interference dsRNA, its hairpin structure is a strong catalyst for
the RNAi mechanism. To make the hairpin struc-
RNA interference (RNAi) refers to a gene silencing ture, 567 nucleotides of the prM sequence from
mechanism through which double-stranded (ds) DENV2 represents the sense orientation, and the
RNA is delivered to cells or organisms. Introduction first 290 nucleotides repeat in an antisense orienta-
of dsRNA into invertebrates can be achieved by ex- tion. DENV2-prM is cloned into plasmid, and after
ogenous feeding or injection to create a significant transcription, DENV2-specific dsRNA is formed,
silencing phenotype.31 Use of the RNAi strategy which halts DENV2 replication. Transfection of
(Fig. 5) against the recurrence of dengue virus can C6/36 (Aedes albopictus) with dsRNA of the ar-
operate effectively in mosquitoes and mosquito bovirus genome is performed, showing that Aedes
cells. The first evidence in this regard occurred species have an RNAi mechanism that is similar to
when the recombinant Sindbis virus (SINV, a sin- other organisms.35 Blair et al. and Sánchez-Vargas
gle-stranded RNA virus) that had a DENV2–pre- and colleagues demonstrated that RNAi has an im-
membrane protein (prM) sequence was transformed portant role in the reduction of dengue infection.39,40
into adult female mosquitoes. During replication, These authors found that adeno-associated virus

FIG. 5: RNAi mechanism. dsRNA enters the cell and degrades by dicer, resulting in the production of siRNA.
Afterward, the RISC complex binds with siRNA to target and degrade the messenger RNA.

Volume 27, Issue 4, 2017


338 Qsim et al.

small interfering (si)RNA infected the dendritic B. Genome Engineering with CRISPR/Cas9
cells and decreased dengue infection in humans.
Mosquito cell line culture (C6/36, A. albopictus) is A relatively new approach to the gene-drive system
transformed by way of plasmid that contains the in- widely used nowadays for dengue vector control is
verted sequence of the dengue prM gene and forms CRISPR-associated Cas9 gene editing. Clustered
290-bp dsRNA. These transformed cells enhance re- regularly interspaced palindromic repeats (CRISPR)
sistance to DENV2. Hence, infection of transformed and CRISPR-associated (Cas) genes are compo-
cells with DENV2 shows decreased numbers of viral nents of an adaptive immune system that are found
RNA. These transformed mosquito cells contain all in a wide variety of bacteria and archaea.44
the machinery of RNAi. This finding proved that
RNAi has an important role in reducing DENV2.41 1. Mechanism
Transposons (jumping genes) that contain the in-
verted repeat sequence of the prM gene of dengue The CRISPR-Cas9–based gene-drive system works
virus are transformed in a mosquito’s embryo, along by induction of a dsDNA break (DSB) at a tar­get
with the carboxypeptidase A promoter. With that site. The chief components of the CRISPR-Cas9
promoter in place, mosquitoes express the 578-bp system are (1) a synthetic single-guide RNA (sgRNA),
dsRNA product after they imbibe a bloodmeal. When which is a small RNA containing 17–20 bases of
GMM Carb 77 is attached to DENV2 after a blood complementarity to a specific genomic sequence,
meal, dsRNA is expressed along with viral replica- and (2) the Cas9 nuclease derived from Strep­to­­­
tion, thus preventing viral spread from the vector to coccus pyogenes (SpCas9). The complex of SpCas9
the host via the salivary glands. In this way, the RNAi and sgRNA induces a DSB at sequences of the ge-
mechanism accounts for the resistance of DENV2.36,39 nome that are directly 5′ to a protospacer-adjacent
The siRNA-mediated silencing of receptors motif (PAM) and are complementary to the sgRNA
and facility of clathrin-mediated endocytosis pre- recognition site. The PAM sequence for SpCas9 is
vent entry of the dengue virus. The proliferation of NGG, which occurs approximately once every 17 bp
HepG2 cells reduces the virus and inhibits dengue in the A. aegypti genome, making it possible to target
fever from developing into serious stages. Specified essentially any locus.45 Injection of CRISPR-Cas9
cellular genes are involved in endocytosis opera- reagents for genome editing at the preblastoderm-
tions and dynamics of the cell structure, an important stage embryo results in germ-line mutant pheno-
task of infection DENV. siRNA-directing genes are types. DNA repair in a homology-dependent manner
linked with clathrin-mediated endocytosis.39 using the Cas9 system leads to a net increase in copy
Villegas-Rosales et al.42 projected that three number. Thus, generation of a loss-of-function mu-
siRNAs have ability to silence the four DENV se- tant allele by site-directed mutagenesis in a heritable
rotype genomes by pointing out NS4B and NS5 se- manner is a key feature of gene-drive studies for
quences. siRNA along with RNAi self-processing vector control.46
machinery have a role in the preclusion of unadorned
dengue infection. In the development of new thera- IV. CONCLUSION
peutic drugs, RNAi can play an important part.37,39
RNAi-mediated gene silencing assays in A. ae- Dengue is a mosquito-borne disease caused by any
gypti cells reveal the role of Unk7703 and sterol car- one of four closely related dengue viruses (DENV1–
rier protein 2 (SCP2) in DENV restriction and host 4). Classic dengue fever, also known as break bone
factor, respectively. Unk7703 is conserved in Aedes fever, is characterized by an acute onset of high
culex and Anopheles and is involved in cell signaling fever. Frontal headache, retro-orbital pain, pain in
pathways. Similarly, SCP2 facilitates cholesterol muscles and joints, anorexia, nausea, hemorrhagic man­
uptake in A. aegypti cells. Knockdown or chemical ifestations, rash, and leucopenia are the major symp-
inhibition of SCP2 effectively inhibits DENV repli- toms of the disease. The A. aegypti mosquito is the
cation in engineered A. aegypti Aag2 cells.43 main vector that transmits viruses that cause dengue.

Critical Reviews™ in Eukaryotic Gene Expression


GM Aedes aegypti to Control Dengue 339

A. aegypti belongs to the Flaviviridae family and 10. Screaton G, Mongkolsapaya J, Yacoub S, Roberts C. New
contains different structural and nonstructural pro- insights into the immunopathology and control of dengue
virus infection. Nature Rev Immunol. 2015;15(12):745–59.
teins. The severity of dengue fever leads to hemor- 11. Thisyakorn U, Thisyakorn C. Latest developments and
rhagic fever and eventually DSS. Millions of people future directions in dengue vaccines. Ther Adv Vaccines.
around the globe are affected every year by dengue 2014;2(1):3–9.
fever. Currently, no commercial vaccine is available 12. Ostera GR, Gostin LO. Biosafety concerns involving genet-
ically modified mosquitoes to combat malaria and dengue
against the dengue virus.
in developing countries. JAMA. 2011;305(9):930–1.
A combination of sterile insect technique (SIT) 13. Olson K, Alphey L, Carlson J, James A. Genetic approaches
and genetic engineering approach (RIDL) can be used in Aedes aegypti for control of dengue. In: Knols BGJ, Louis
to reduce the dengue vector population and overcome C, editors. Bridging laboratory and field research for genetic
the limitations of traditional SIT. With the advent control of disease vectors. The Netherlands: Springer; 2006.
p. 77–87.
of modern genetics, approaches including the use of 14. Antonelli T, Wilke ABB, Marrelli MT. Genetic control of
HEGs, RNAi, and CRISPR-Cas9 show promising re- mosquitoes: Population suppression strategies. Rev Inst
sults in vector control. However, large-scale produc- Med Trop Sao Paulo. 2012;54(5):287–92.
tion and field study of these techniques is still required. 15. Carvalho DO, McKemey AR, Garziera L, Lacroix R,
Donnelly CA, Alphey L, Malavasi A, Capurro ML. Sup­
pression of a field population of Aedes aegypti in Brazil
REFERENCES by sustained release of transgenic male mosquitoes. PLoS
Negl Trop Dis. 2015;9(7):e0003864.
1. ul Qamar MT, Kiran S, Ashfaq UA, Javed MR, Anwar F, 16. Atkinson MP, Su Z, Alphey N, Alphey LS, Coleman PG,
Ali MA, Gilani AuH. Discovery of novel dengue NS2B/ Wein LM. Analyzing the control of mosquito-borne dis-
NS3 protease inhibitors using pharmacophore modeling eases by a dominant lethal genetic system. Proc Natl Acad
and molecular docking based virtual screening of the Sci. 2007;104(22):9540–5.
ZINC database. Int J Pharmacol. 2016;12(6):621–32. 17. Coleman PG, Alphey L. Genetic control of vector pop-
2. Idrees S, Ashfaq UA. Discovery and design of cyclic pep- ulations: An imminent prospect. Trop Med Int Health.
tides as dengue virus inhibitors through structure-based mo- 2004;9(4):433–7.
lecular docking. Asian Pac J Trop Med. 2014;7(7):513–6. 18. Lacroix R, McKemey AR, Raduan N, Wee LK, Ming WH,
3. ul Qamar MT, Mumtaz A, Ashfaq UA, Adeel MM, Fatima Ney TG, AA SR, Salman S, Subramaniam S, Nordin O.
T. Potential of plant alkaloids as dengue ns3 protease in- Open field release of genetically engineered sterile male
hibitors: Molecular docking and simulation approach. Aedes aegypti in Malaysia. PLos One. 2012;7(8):e42771.
Bangladesh J Pharmacol. 2014;9(3):262–7. 19. Yakob L, Alphey L, Bonsall MB. Aedes aegypti control:
4. ul Qamar T, Mumtaz A, Ashfaq UA, Azhar S, Fatima T, Has­san The concomitant role of competition, space and transgenic
M, Hussain SS, Akram W, Idrees S. Computer aided screening technologies. J Applied Ecol. 2008;45(4):1258–65.
of phytochemicals from garcinia against the den­gue NS2B/ 20. Araújo HR, Carvalho DO, Ioshino RS, Costa-da-Silva AL,
NS3 protease. Bioinformation. 2014;10(3):115–8. Capurro ML. Aedes aegypti control strategies in Brazil:
5. ul Qamar MT, Mumtaz A, Rabbia Naseem AA, Fatima T, Incorporation of new technologies to overcome the per-
Jabbar T, Ahmad Z, Ashfaq UA. Molecular docking based sistence of dengue epidemics. Insects. 2015;6(2):576–94.
screening of plant flavonoids as dengue NS1 inhibitors. 21. Helinski ME, El-Sayed B, Knols BG. The sterile insect
technique: Can established technology beat malaria?
Bioinformation. 2014;10(7):460.
Entomologische Berichten. 2006;66(1):13–20.
6. Traver BE, Anderson MA, Adelman ZN. Homing endonu-
22. de Valdez MRW, Nimmo D, Betz J, Gong H-F, James AA,
cleases catalyze double-stranded DNA breaks and somatic
Alphey L, Black WC. Genetic elimination of dengue vector
transgene excision in Aedes aegypti. Insect Mol Biol. 2009;
mosquitoes. Proc Natl Acad Sci. 2011;108(12):4772–5.
18(5):623–33. 23. Mwasunda JA. Modelling the effect of sterile insect tech-
7. McCall P, Kittayapong P, editors. Control of dengue vec- nology for control of anopheles mosquito population in
tors: Tools and strategies. Report of the Scientific Working Tanzania [dissertation]. Dar es Salaam (Tanzania): Uni­ver­
Group Meeting on Dengue; Geneva, Switzerland. Oct 1–5, sity of Dar es Salaam; 2012.
2006. 24. Evans TPO, Bishop SR. A spatial model with pulsed re-
8. Alcaraz-Estrada SL, Yocupicio-Monroy M, del Angel RM. leases to compare strategies for the sterile insect tech-
Insights into dengue virus genome replication. Future Virol. nique applied to the mosquito Aedes aegypti. Math Biosci.
2010;5(5):575–92. 2014;254:6–27.
9. Nukui Y, Tajima S, Kotaki A, Ito M, Takasaki T, Koike K, 25. World Health Organization. Progress and prospects for the
Kurane I. Novel dengue virus type 1 from travelers to Yap use of genetically modified mosquitoes to inhibit disease
State, Micronesia. Emerg Infect Dis. 2006;12(2):343–6. transmission. Geneva, Switzerland. May 4–6, 2009.

Volume 27, Issue 4, 2017


340 Qsim et al.

26. Wallace H. Genetically modified mosquitoes: Ongoing 37. Franz AW, Sanchez-Vargas I, Adelman ZN, Blair CD,
concerns. Penang, Malaysia: Third World Network; 2013. Beaty BJ, James AA, Olson KE. Engineering RNA in-
27. Wilke ABB, Nimmo DD, St John O, Kojin BB, Capurro terference-based resistance to dengue virus type 2 in ge-
ML, Marrelli MT. Mini-review: Genetic enhancements to netically modified Aedes aegypti. Proc Natl Acad Sci.
the sterile insect technique to control mosquito popula- 2006;103(11):4198–203.
tions. Asia Pac J Mol Biol Biotechnol. 2009;17(3):65–74. 38. Burt A. Site-specific selfish genes as tools for the control
28. Phuc HK, Andreasen MH, Burton RS, Vass C, Epton MJ, and genetic engineering of natural populations. Proc Royal
Pape G, Fu G, Condon KC, Scaife S, Donnelly CA. Late- Soc London B Biol Sci. 2003;270(1518):921–8.
acting dominant lethal genetic systems and mosquito con- 39. Blair CD, Sanchez-Vargas I, Franz AW, Olson KE. Ren­
trol. BMC Biol. 2007;5(1):11. dering mosquitoes resistant to arboviruses through RNA
29. Alphey L, Benedict M, Bellini R, Clark GG, Dame DA, interference. Microbe. 2006;1(10):466.
Service MW, Dobson SL. Sterile-insect methods for con- 40. Idrees S, Ashfaq UA. RNAi: Antiviral therapy against
trol of mosquito-borne diseases: An analysis. Vect Borne dengue virus. Asian Pac J Trop Biomed. 2013;3(3):232–6.
Zoonotic Dis. 2010;10(3):295–311. 41. Sánchez-Vargas I, Scott JC, Poole-Smith BK, Franz AW,
30. Wilke ABB, Marrelli MT. Genetic control of mosquitoes: Barbosa-Solomieu V, Wilusz J, Olson KE, Blair CD.
Population suppression strategies. Rev Inst Med Trop Sao Dengue virus type 2 infections of Aedes aegypti are mod-
Paulo. 2012;54(5):287–92.
ulated by the mosquito’s RNA interference pathway. PLoS
31. Leftwich PT, Bolton M, Chapman T. Evolutionary bi-
Pathol. 2009;5(2):e1000299.
ology and genetic techniques for insect control. Evol Appl.
42. Villegas-Rosales PM, Méndez-Tenorio A, Ortega-Soto E,
2016;9(1):212–30.
Barrón LB. Bioinformatics prediction of siRNAs as poten-
32. McGraw EA, O’neill SL. Beyond insecticides: New think-
tial. Bioinformation. 2012;8:519–22.
­ing on an ancient problem. Nature Rev Microbiol. 2013;11
43. Jupatanakul N, Sim S, Angleró-Rodríguez YI, Souza-
(3):181–93.
33. Massonnet-Bruneel B, Corre-Catelin N, Lacroix R, Lees Neto J, Das S, Poti KE, Rossi SL, Bergren N, Vasilakis
RS, Hoang KP, Nimmo D, Alphey L, Reiter P. Fitness of N, Dimopoulos G. Engineered Aedes aegypti JAK/STAT
transgenic mosquito Aedes aegypti males carrying a domi- pathway-mediated immunity to dengue virus. PLoS Negl
nant lethal genetic system. PLos One. 2013;8(5):e62711. Trop Dis. 2017;11(1):e0005187.
34. Deredec A, Godfray HCJ, Burt A. Requirements for effec- 44. Gantz VM, Jasinskiene N, Tatarenkova O, Fazekas A, Macias
tive malaria control with homing endonuclease genes. Proc VM, Bier E, James AA. Highly efficient Cas9-mediated
Natl Acad Sci. 2011;108(43):E874–80. gene drive for population modification of the malaria
35. Windbichler N, Menichelli M, Papathanos PA, Thyme SB, Li vec­tor mosquito Anopheles stephensi. Proc Natl Acad Sci.
H, Ulge UY, Hovde BT, Baker D, Monnat RJ, Burt A. A syn- 2015;112(49):E6736–43.
thetic homing endonuclease-based gene drive system in the 45. Kistler KE, Vosshall LB, Matthews BJ. Genome engi-
human malaria mosquito. Nature. 2011;473(7346):212–5. neering with CRISPR-Cas9 in the mosquito Aedes aegypti.
36. Travanty EA, Adelman ZN, Franz AW, Keene KM, Beaty Cell Rep. 2015;11(1):51–60.
BJ, Blair CD, James AA, Olson KE. Using RNA interference 46. Adelman ZN, Tu Z. Control of mosquito-borne infectious
to develop dengue virus resistance in geneti­cally modified diseases: Sex and gene drive. Trends Parasitol. 2016;32(3):
Aedes aegypti. Insect Biochem Mol Biol. 2004;34(7):607–13. 219–29.

Critical Reviews™ in Eukaryotic Gene Expression

You might also like