INTRODUCTION
With the development of gene sequencing technology, the exploration of genome sequence function has recently become a new research direction [1,2]. The development of convenient and accurate gene editing tools has facilitated the investigation of gene function. To date, three types of gene editing tools have been developed: zinc-finger nuclease (ZFN) technology, transcription activator-like effector nuclease (TALEN) technology, and clustered regulatory interspaced short palindromic repeats (CRISPR) technology. The first gene editing tool used by researchers was ZFN, a chimeric fusion structure consisting of a zinc finger protein and the shearing domain of the Fok-I nucleic acid endonuclease. The zinc finger protein structural domain recognizes the target site and relies on the action of Fok-I to cut DNA double strands and form double-strand breaks. TALEN is derived from ZFN and likewise uses Fok-I nucleic acid endonuclease to cleave sequences. However, owing to the difficulty, complexity, and cost of design, neither gene editing method has been widely used in laboratory settings [3,4]. CRISPR-Cas, an immune system widely found in bacteria and most archaea, confers resistance to viral or exogenous sequences [5]. This technology quickly became the state of the art in the field of gene editing, because of its advantages of efficiency, convenience, speed, and affordability [6].
Currently, the CRISPR-Cas9 system most widely used in scientific research is that of Streptococcus pneumonia [7]. This system consists of three main parts: CRISPR-derived RNA (cr-RNA) [8], trans-activating cr-RNA (tracr-RNA) [9,10], and nucleic acid endonuclease 9, or the Cas9 protein [11]. The complete CRISPR-Cas9 system can achieve targeted cleavage of genes. After further exploration, researchers successfully chimerized cr-RNA and tracr-RNA, thus forming a complex called single-guide RNA (sg-RNA), which undergoes complementary base pairing with target DNA sequences. The use of sgRNA not only preserves the DNA cleavage function of Cas9 endonuclease but also simplifies the composition of the CRISPR system [6].
Double-strand breaks induce either nonhomologous end joining (NHEJ) or homologous directed repair (HDR) in vivo [12]. The NHEJ repair mechanism is the most frequent type of break repair at double-strand breaks. Because of its error-prone nature, random insertions or deletions of bases at the DNA break usually lead to DNA changes [13], which result in frameshift mutations in the original protein-coding strand, thereby disrupting the original protein composition. In contrast, HDR can be induced in vivo when a repair template is introduced. HDR consists primarily of the newly introduced target gene and homologous sequences at both ends of the cut site with the host template. HDR can be used to knock in a target fragment at the CRISPR cleavage site to achieve to expression of a specific gene [14], but it is much less efficient than NHEJ, and screening for an appropriate sgRNA and selecting the length of the homologous sequences are essential [15,16] (Fig 1).

CRISPR-Cas9 system cleavage and repair pathway. cr-RNA+tracr-RNA or sgRNA binds the Cas9 protein and forms the RNA-Cas9 RNP, which specifically recognizes the PAM in the gene sequence. Cleavage of double stranded DNA upstream of the PAM by the RuvC and HNH nuclease domains of the Cas9 protein forms double strand breaks. These double-strand breaks induce either NHEJ or HDR. NHEJ repair occurs through two main routes: random deletion or random insertion. HDR occurs through the introduction of repair templates containing homologous arms and a target sequence, and results in directed repair. Current mosquito gene editing strategies are based on embryo injection editing and ovary-targeted editing. This figure was created with BioRender.com.
In recent years, CRISPR-Cas9 technology has been applied to a variety of research fields, and has been widely used for gene editing in plants, including Oryza sativa [17,18], Triticum aestivum [19,20], and Arabidopsis thaliana [21]; in microorganisms, such as bacteria [22–24], fungi [25–27], and archaea [28]; and in animals, such as mosquitoes [29], zebrafish [30], mice [31], rabbits [32], and many others. This technology has even been used in human gene therapy [33].
CRISPR/CAS9 TECHNOLOGY IN MOSQUITOES
Mosquitoes may carry dengue fever, Zika, yellow fever, and chikungunya fever, and other flaviviruses of the arbovirus group [34], which cause widespread epidemics and morbidity, primarily in the tropics. With the intensification of global warming, epidemic trends have begun to emerge in some non-tropical countries [35], thus severely endangering human lives [36]. Suppressing mosquito populations or replacing wild populations with mosquitoes with diminished ability to act as vectors for viral pathogens would dramatically decrease the risk of insect-borne disease transmission. Since 1998, researchers have sought to genetically manipulate mosquitoes with ZFN and TALEN [37–39]. These research efforts have greatly accelerated with the maturation of CRISPR-Cas9 gene editing technology. Cas9 protein-nucleic acid complexes enable mosquito genes to be broken for gene editing. This process can result in random repair (i.e., NHEJ) or precision repair through the introduction of plasmids containing homologous sequences during the editing process (i.e., HDR); the latter technique is commonly used in gene-drive technology. Gene editing strategies have been developed for various mosquito species such as Aedes aegypti [40], Anopheles stephensi [41], Anopheles gambiae [29], and Culex burnetii [42]. Current directions in mosquito gene editing focus primarily on infectious disease prevention, population suppression, phenotype screening, and decreasing mosquito resistance to insecticides (Fig 2).

Application of CRISPR-Cas9 technology in mosquitoes and verification of gene functions. Illustrations of recent mosquito gene editing experiments with CRISPR-Cas9 technology. Currently, the main research directions for mosquito gene editing are as follows: ① infectious disease control, including anti-viral (AeRel1, Obp10, Obp22, GCTL-3, and CFAV-EVEs genes), anti-bacterial (OUT7B and Obp10 genes), and anti-plasmodium (CTL-4, Scorpine, SGS-1, LRIM-1, mosGILT, and FREP1 genes) targets; ② population suppression, including reproductive damage (CRVP379, Dsx, CYC, β-tubulin, Nix, GCTL-3, Fruitless, and miR-277/309 genes), pupation rate (5-HTR7A gene), and eclosion rate (E93, Myo-fem, AcAmt, Stretchin, and Act-4 genes) targets; ③ phenotypic screening, including olfaction (KMO, CYC, AcAmt, Gr22/23/24, and IR8a genes), appearance (Spn-F, White, Kh, DsRed, Op1/2, KMO, aaNAT1, Met, Ae-Forked, and 5-HTR2B genes), and activity (Stretchin, AeAct, PPK301, Myo-fem, and Spn-F genes) targets; and ④ insecticide resistance (V402L, L1014F, nAChR α6, mJHBP, NPYLR7, and CYP9M10 genes) targets. This figure was created with BioRender.com.
Prevention and control of mosquito-borne infectious diseases
The difficulty in developing vaccines for mosquito-borne pathogens and the increasing resistance of mosquitoes to insecticides have led to continued failure to effectively prevent the spread of mosquito-borne infectious diseases; therefore, a novel, safe, convenient, and efficient method to control mosquitoes is urgently needed [43–45]. With the advent of gene editing technology, researchers aim to regulate mosquito genes to prevent the reproduction and spread of infectious pathogens (arboviruses/parasites/bacteria). When a mosquito bites a pathogen-infected host, the pathogens enter the mosquito and reproduce, then further spread after the mosquito bites another healthy host [46,47]. Similarly to their mechanism of invasion in the human body, pathogens require receptor-mediated access to mosquito cells and the ability to escape the mosquito’s natural immune response to proliferate. On the basis of these characteristics, researchers have identified a series of key regulatory factors associated with pathogen replication and receptor-mediated access, through the analysis of the mosquito transcriptome and pathogenesis [48–51]. These findings can be applied to regulate immune effects in mosquitos through gene editing techniques. Currently, arboviruses and malaria remain the most critical mosquito-borne human health concerns. Major advances in this area are briefly reviewed below.
In the field of malaria research, Dong et al. have found that knocking out the fibrinogen-associated protein FREP1 gene in Anopheles gambiae inhibits Plasmodium falciparum and Plasmodium berghei infection. In addition, deletion of this gene significantly decreases blood-sucking, reproduction, and hatching efficiency; delays pupation; and shortens the mosquito lifespan, probably because the mosquito FREP1 gene functions similarly to fibrinogen proteins in other organisms, by directly affecting various developmental processes [52]. In another similar study, knockout of the immune response gene leucine-rich protein LRIM1 in Anopheles stephensi has been found to inhibit Plasmodium sporogony development via the melanization and phagocytosis of ookinetes. Plasmodium infection rates in mosquitoes subsequently decrease, together with the diversity and abundance of mosquito midgut microbiota. Additionally, deletion of the LRIM1 gene affects the reproductive capacity of mosquitoes, through a significant decrease in egg laying and hatching rates in females [53]. In 2022, the Simões research team reported that knockout of the CTL4 gene in Anopheles gambiae significantly increases Plasmodium falciparum suppression in mosquitoes, to a much greater extent than observed in gene-edited Anopheles mosquitoes with FREP1 host factor deletion and overexpression of the REL2 transcription factor [51,52]. Importantly, mosquitoes lacking CTL4 have elevated susceptibility to bacterial infection. Bacterial challenge assays have suggested that CTL4, in a melanization-independent manner, antagonizes systemic bacterial infections in mosquitoes [54]. Moreover, mutating the Anopheles gambiae lysosomal thiol reductase mosGILT34 [55] or inserting a known anti-microbial peptide, scorpion scorpine [56], has been shown to affect mosquito resistance to Plasmodium, thus enabling control of the spread of malaria epidemics. In addition, knockdown of the Aedes aegypti salivary gland surface protein SGS1 gene effectively attenuates the ability to carry Plasmodium gallinaceum, an avian malaria parasite. This discovery has provided new ideas for the prevention and treatment of human malaria diseases [57].
Dong’s research team has inhibited the transmission of arboviruses by disrupting the mosquito Obp10 and Obp22 genes, which encode odor-binding proteins that attenuate the secretion of virus particles into saliva; their exact mechanism of action is not understood and may work in conjunction with specific ligands [58]. The AaRel1 transcription factor mediates the antiviral effects of the mosquito Toll immune pathway. Overexpression of this gene in Aedes aegypti through CRISPR technology has been found to significantly suppress dengue virus serotype 2 titer in mosquitoes. Theoretically, this inhibition should also be effective against Zika virus [59]. Li’s team has found that mutation of GCTL-3 in Aedes aegypti also enhances suppression of both dengue II and Zika viruses [60]. A possible reason for this finding may be that GCTL-3 gene mutation causes a lack of Serratia marcescens, a bacterium that promotes viral transmission, in the mosquito midgut [61]. With advances in sequencing technology, host genomes have been discovered to frequently contain fragments of viral genomes known as endogenous viral elements (EVEs). EVEs integrated into mosquito genomes are believed to be closely associated with the corresponding viral infections. Suzuki’s team has used CRISPR-Cas9 technology to knock out the EVE gene of the cell fusion virus CFAV in Aedes aegypti, thus enhancing replication of CFAV in the ovaries, and has experimentally demonstrated that the interaction of RNAs from EVEs and viruses through the piRNA pathway inhibits viral replication, thereby suppressing viral transmission by mosquitoes [62]. Editing the non-retroviral EVE locus may provide new ideas for future prevention and control of mosquito-borne infectious diseases.
Mosquito population suppression
To control the risks inherent in artificially regulating mosquito populations, researchers have focused on altering mosquito motility, disrupting their sex-determining genes or reproductive functions, and then releasing the genetically edited mosquitoes into the wild to interfere with reproduction, through a method known as the sterile insect technique (SIT). The advent of gene drivers has altered the traditional Mendelian laws of inheritance by biasing the heritability of insects containing specific genes toward supra-Mendelian heritability (>50%), and has enabled the modification of populations, but their development has been slow, because of difficulties in application [63]. The emergence of CRISPR-Cas9, combined with gene-drive technology, has greatly decreased difficulties in population manipulation and opened new possibilities for mosquito population control. Consequently, researchers are continually searching for genes that regulate the physiological activity or sex determination of mosquitoes, to suppress their numbers or modify entire populations.
Numerous research teams are currently targeting male mosquitoes. Some have shown that knocking out the β2-tubulin (B2t) gene in Aedes aegypti significantly inhibits the fertility of male mosquitoes, most of which fail to produce progeny after mating with females [64], whereas knocking out the B2t gene in Anopheles gambiae causes X chromosome fragmentation in males and significantly increases the proportion of male mosquitoes in the population [65]. The presence of the fruitless gene in insects is a major driver of male courtship and mating behavior. The Basrur team has used CRISPR-Cas9 to knock out the fruitless gene in Aedes aegypti, thus preventing males from mating and reproducing. Interestingly, male mosquitoes lacking the fruitless gene are strongly attracted to human hosts and exhibit altered male feeding behavior; these findings warrant more intensive study [66]. Hall et al. have knocked out the male determinant Nix gene in Aedes aegypti, and subsequently observed incomplete male genital development and thus decreased reproductive capacity [67].
Studies are increasingly being conducted on female rather than male loci. Knocking out the tweedledee and tweedledum genes in Aedes aegypti effectively increases the activity and preservation time of eggs; females robustly retain eggs in their ovaries until freshwater is found, thus facilitating the search for areas suitable for egg-laying [68]. Female reproductive function is affected by mutations in the cysteine-rich toxin CRVP379 gene [69], β-Tubulin 85D gene [70], and core clock CYC gene [71] in Aedes aegypti; the kynurense hydroxylase kh gene in Anopheles stephensi [72]; and the dsx gene in Anopheles gambiae [73,74]. Mutations in miR-277 and miR-309 both affect ovarian development in Aedes aegypti, thus leading to population suppression [75,76], whereas mutations in the 5-hydroxytryptamine receptor 5-HTR7A gene [77] and myo-fem gene cause restricted motility in females, thereby affecting their normal reproductive function and decreasing their fertility [70].
Mosquito phenotypic screening
Genes determining the phenotypic characteristics of mosquitoes have been an additional research focus. These genes broadly regulate alterations in olfaction, motility, and appearance. Alterations in olfaction directly contribute to mosquitoes’ ability to discriminate between human hosts. CRISPR-based studies have shown that mutations in the odor-binding protein co-receptor Orco alter mosquito olfaction and attenuate mosquito localization to human hosts [78,79]. Disruption of the core clock gene CYC in Aedes aegypti eliminates the characteristic circadian patterns of locomotor activity and significantly decreases the response to human host odor and mating success [71], whereas the taste receptor gene Gr23/24 [80] and the pro-ionotropic receptor Ir8a gene [81] significantly alter olfactory ability in mosquitoes. Moreover, knocking out the ammonium transporter protein AcAmt gene, which is involved in multiple systems of mosquito olfaction, reproduction, and ammonia metabolism, severely affects motility [82]. Similarly, knockout of the muscle protein Stretchin gene [83], actin AeAct-4, and myo-fem gene all affect mosquitoes’ flight ability [84,85], thus further influencing physiological functions. Mutations in the White [86,87], Kh [86,88], Cardinal [86], Yellow [86,88], Ebony [86], DsRed [89], Kmo [90,91], Met [91,92], and ECFP genes [93] result in altered eye characteristics and pigmentation. Mutations in spn-F [94] affect the formation of bristle and falcate scales. Moreover, mutations in the aromatic amine-N-acetyltransferase aaNAT1 [95] result in an altered appearance, wherein the outer surface of the skeleton of mutant mosquitoes is rough, and the cuticle is dark in color. Nix [96] knockout males show feminization. The forked gene Ae-Forked [97] is required for bristle elongation. Finally, knockout of the 5-hydroxytryptamine receptor 5-HTR2B gene [98] leads to decreased body size, postponed development, shortened lifespan, retarded ovarian growth, and markedly diminished lipid accumulation. Together, these insights have demonstrated how CRISPR-Cas9 technology has enabled researchers to explore previously unknown functions of mosquito genes.
Mosquito insecticide resistance-related gene studies
Mutations at sites on the pyrethroid voltage-gated sodium channel VGSC can alter insecticide resistance. A study using CRISPR-Cas9 technology has demonstrated that mutations at both the V402L and L1014F sites in Anopheles gambiae can increase mosquito resistance to pyrethroids. Additionally, the L1014F mutation increases mosquito resistance to permethrin by 9.9-fold and to dichlorodiphenyltrichloroethane (DDT) by 24-fold [99,100]. In contrast, deletion of the cytochrome P450 gene CYP9M10 in Aedes aegypti results in a more than 100-fold decrease in mosquito resistance to pyrethroid [42]. The Lan research team has used CRISPR-Cas9 technology to mutate the nAChR α6 subunit of the nicotinic acetylcholine receptor in Anopheles gambiae, thus resulting in a 320-fold increase in mosquito resistance to spinosad [101]. Moreover, mutations in the mJHBP gene, encoding a juvenile-protective hormone-binding protein in Aedes aegypti, enhance mosquito susceptibility to bacteria, and significantly increase sepsis and mortality in mosquitoes attacked by bacteria [102].
Researchers are increasingly using CRISPR-Cas9 technology to target mosquito genes with gene editing to elucidate their functions. Table 1 summarizes the literature on mosquito gene editing based on CRISPR-Cas9, including the species of mosquito edited, the countries and research sponsors, the edited genes and their functions, and the survival rates of eggs after editing and the gene editing frequency. Notably, researchers use differing methods for calculating gene editing frequencies. These methods can be divided into two calculation modes: No. (mosquitoes with successful gene editing)/No. (injected mosquito eggs) and No. (mosquitoes with successful gene editing)/No. (mosquito eggs surviving after injection). The chosen calculation method leads to differences in the results of gene editing frequencies.
CRISPR/Cas9 in mosquito research.
Year | Species | Country and sponsor | Gene(s) | Function in mosquitoes | Survival and GEF a | Reference | ||||||
---|---|---|---|---|---|---|---|---|---|---|---|---|
Mosquito-borne Infectious Disease Control | ||||||||||||
2023 | Aedes aegypti | United States of America University of California San Diego | AaRel I | Suppression of DENV2 titer | Unknown | [59] | ||||||
2023 | Aedes aegypti | China Chinese Academy of Sciences | OTU7B | Enhanced resistance to fungal infection | Unknown | [103] | ||||||
2022 | Anopheles gambiae | United States of America Johns Hopkins University | CTL-4 | Suppression of Plasmodium falciparum | Unknown | [54] | ||||||
2021 | Anopheles gambiae | United Kingdom Imperial College London | Scorpine | Suppression of Plasmodium falciparum | 7.0% (62/889)/1.2% (11/889) b | [56] | ||||||
2021 | Aedes aegypti | United States of America Texas A&M University | SGS1 | Decreased Plasmodium sporozoite invasion of salivary glands | 19% (87/445)/1.1% (5/445) b | [57] | ||||||
2021 | Anopheles stephensi | United States of America Sanaria Inc. Rockville | LRIM I | Suppression of Plasmodium sporozoite development | 3.5% (19/549)/0.1% (1/549) b | [53] | ||||||
2021 | Aedes aegypti | United States of America Johns Hopkins University | Obp10/22 | Suppression of DENV2 and ZIKV transmission | 53.4% (103/193)/50% (3/6) c | [58] | ||||||
2020 | Anopheles gambiae | United States of America Yale University School of Medicine | mosGILT | Decreased oocyst numbers after Plasmodium infection | 15% (87/580)/4.5% (26/580) b | [55] | ||||||
2020 | Aedes aegypti | China National Tsing Hua University | GCTL-3 | Decreased DENV-2 infection rate | 26% (210/795)/1.1% (2/210) c | [60] | ||||||
2020 | Aedes aegypti | France Institut Pasteur | CFAV-EVEs | Enhanced CFAV replication in ovaries | Unknown | [62] | ||||||
2018 | Anopheles gambiae | United States of America Johns Hopkins University | FREP I | Suppression of Plasmodium infection | 10%/3.5% (28/800) b | [52] | ||||||
Mosquito Population Suppression | ||||||||||||
2023 | Aedes aegypti | United States of America Rockefeller University | Tweedledum | Enhanced egg retention | Unknown | [68] | ||||||
2022 | Aedes aegypti | China Hainan University | 5-HTR7A | Abnormal pupa development | 7.7% (14/180)/60% (6/10) c | [77] | ||||||
2022 | Aedes aegypti | United States of America Johns Hopkins Bloomberg School | CRVP379 | Decreased reproductive capacity in females | 33.87% (83/245)/5.7% (14/245) b | [69] | ||||||
2022 | Aedes aegypti | United States of America University of Kentucky | E93 | Completed pupal ecdysis, pupal death | 25% (150/600)/unknown | [104] | ||||||
2022 | Aedes aegypti | Pakistan Government College University Faisalabad | DsxF1/F2 | Decreased fecundity rate and hatching rate | 51.11% (230/450), 54.00% (243/450)/36.59%, 29.36% | [105] | ||||||
2022 | Aedes aegypti | United States of America Texas A&M University | CYC | Decreased egg hatching rate and adult survival rate | Unknown | [71] | ||||||
2021 | Aedes aegypti | United States of America University of California | Beta-tubulin | Suppression of fertility | Unknown 2.6% (18/700)/0.4% (3/700) b | [70,64] | ||||||
2021 | Aedes aegypti | United States of America University of California | Myo-fem | Decreased flight capacity | Unknown | [70] | ||||||
2021 | Anopheles gambiae | United States of America Vanderbilt University | AcAmt | Decreased insemination rate Increased mortality during eclosion | Unknown | [82] | ||||||
2021 | Aedes aegypti | United States of America Texas A&M University | Stretchin | Decreased flight capacity | 7.1% (57/800)/0.25% (2/800) b | [83] | ||||||
2020 | Aedes albopictus | China Southern Medical University | Nix | Feminization of males | 28.7% (286/996)/3.6% (36/996) b | [96] | ||||||
2020 | Anopheles stephensi | United States of America University of California | Kh | Decreased survival rate Impaired reproductive ability | 51.4% (259/504)/unknown | [72] | ||||||
2020 | Aedes aegypti | China National Tsing Hua University | GCTL-3 | Decreased reproductive ability | Unknown | [60] | ||||||
2020 | Anopheles gambiae | United Kingdom Imperial College London | Dsx | Production of unisex populations | Unknown | [73] | ||||||
2020 |
Aedes aegypti
Culex quinquefasciatus | United Kingdom The Pirbright Institute | Act-4 | Interference with flight muscle function | 0.7% (6/915)/ 17% (1/6) c | [84] | ||||||
2020 | Aedes aegypti | United States of America The Rockefeller University | Fruitless | Failure of males to mate | Unknown | [66] | ||||||
2018 | Anopheles gambiae | United Kingdom Imperial College London | Dsx | Intersex phenotype and complete sterility | Unknown | [74] | ||||||
2017 | Aedes aegypti | United States of America University of California | miR-277 | Suppression of ovarian development | 41%/unknown | [75] | ||||||
2016 | Anopheles gambiae | United Kingdom Imperial College London |
AGAP007280
AGAP011377 AGAP005958 | Recessive female sterility phenotype | 10.3% (56/540)/7.1% (4/56)
c
4.2% (21/500)/4.8% (1/21) c 12.3% (49/400)/2.0% (1/49) c | [29] | ||||||
2016 | Anopheles gambiae | United Kingdom Imperial College London | Beta-tubulin | Extreme male bias among progeny | Unknown | [65] | ||||||
2016 | Aedes aegypti | United States of America University of California | miR-309 | Impaired ovarian development | 51.5% (103/200)/64.1% (66/103) c | [76] | ||||||
2015 | Aedes aegypti | United States of America Virginia Polytechnic Institute and State University | Nix | Virilization of females | Unknown | [67] | ||||||
Mosquito Phenotypic Screening | ||||||||||||
2022 | Anopheles sinensis | China Chongqing Normal University | Orco | Impaired mosquito sensitivity to human hosts | Unknown | [78] | ||||||
2022 | Aedes aegypti | Israel Ben-Gurion University of the Negev | Spn-F | Tapered epithelial cellular extension structures | Unknown | [94] | ||||||
2022 | Aedes aegypti | United States of America Texas A&M University | CYC | Diminished response to host odor | Unknown | [71] | ||||||
2021 | Culex quinquefasciatus | United States of America University of California San Diego | White /Kh /Cardinal /Yellow /Ebony | Appearance change | 10.01% (34/337)/unknown 11.5% (46/400)/unknown 11.0% (32/290)/unknown 11.4% (25/220)/unknown 31.3% (90/288)/unknown | [86] | ||||||
2021 | Culex quinquefasciatus | United Kingdom The Pirbright Institute | DsRed | White eye phenotype | 14.8% (57/384)/0% | [89] | ||||||
2021 | Aedes aegypti | United States of America University of California | Op1/Op2 | Abolished vision-guided target attraction | Unknown | [106] | ||||||
2021 | Culex pipiens pallens | China Nanjing Medical University |
Kmo
| White and mosaic eye phenotypes | Unknown (ReMOT) | [90] | ||||||
2021 | Aedes aegypti | China Hainan University | aaNAT1 | Cuticular pigmentation formation | 5.6% (17/302)/unknown | [95] | ||||||
2021 | Anopheles gambiae | United States of America Vanderbilt University | AcAmt | Peripheral neuron sensitivity to ammonia | Unknown | [82] | ||||||
2021 | Aedes aegypti | United States of America Texas A&M University | Stretchin | Abnormal open wing posture | Unknown | [83] | ||||||
2021 | Aedes aegypti | United States of America University of Kentucky | Kmo/Met | Mosaic eye phenotypes | Unknown/ 0.6% (3/500) b | [91] | ||||||
2020 | Aedes aegypti | United States of America Texas A&M University | AeAct-4/Myo-fem | Flightlessness in females | 20.4% (83/407)/unknown | [85] | ||||||
2020 |
Aedes aegypti
Aedes albopictus | China Southern Medical University | Nix | Feminization and deformities in males | 34.9% (213/609)/14.1% (30/213)
c
34.7% (279/805)/24.0% (67/279) c 24.7% (246/996)/14.6% (36/246) c (Data from multiple experiments) | [96] | ||||||
2020 | Aedes aegypti | Israel Ben-Gurion University of the Negev | Ae-Forked | Required for bristle elongation | Unknown | [97] | ||||||
2020 | Anopheles coluzzii | United States of America Vanderbilt University | Gr22/23/24 | Required for CO2 sensitivity | Unknown | [80] | ||||||
2020 |
Aedes aegypti
Culex quinquefasciatus | United Kingdom The Pirbright Institute | Act-4 | Interference with flight muscle function | 0.7% (6/915)/unknown 4.0% (67/1802)/unknown 9.0% (68/736)/unknown (Data from multiple experiments) | [84] | ||||||
2020 | Anopheles coluzzii | United States of America Vanderbilt University | Orco | Decreased attraction to human odors | Unknown | [79] | ||||||
2019 | Aedes aegypti | United States of America The Rockefeller University | Ppk301 | Altered spawning capacity | Unknown | [107] | ||||||
2019 | Aedes aegypti | United States of America University of Kentucky |
Kmo-A
Kmo-B Kmo-C Kmo-D | Black larval phenotype | 48.2% (281/583)/0% (0/255)
c
27.4% (131/478)/8.2 (8/97) c 33.3% (72/216)/6% (4/65) c 22.1% (62/280)/0% (0/62) c | [92] | ||||||
2019 | Aedes albopictus | China Southern Medical University |
Kh
Yellow | Eye and body pigmentation defects | 72.91% (148/203)/51.35% (76/138)
c
87.03% (161/185)/32.92% (53/161) c | [88] | ||||||
2019 | Aedes aegypti | United States of America Florida International University | Ir8a | Decreased attraction to human odors | Unknown | [81] | ||||||
2018 | Anopheles gambiae | United States of America University of California | White | White and mosaic eye phenotypes | 37% (69/185)/46% (32/69)
c
32% (81/251)/59% (48/81) c 21% (47/219)/87% (41/47) c 19% (33/177)/88% (29/33) c 11% (24/228)/92% (22/24) c | [87] | ||||||
2018 | Aedes aegypti | United States of America University of California | 5-HTR2B | Decreased body size | 40%/unknown | [98] | ||||||
2015 | Aedes aegypti | United States of America University of Missouri | ECFP | Changes in eye characteristics | 11.7% (73/626)/5.5% (4/73) c | [93] | ||||||
Mosquito Resistance | ||||||||||||
2022 | Anopheles gambiae | United Kingdom Liverpool School of Tropical Medicine | V402L | Decreased mortality after exposure to pyrethroids and DDT | Unknown | [99] | ||||||
2021 | Anopheles gambiae | United Kingdom Liverpool School of Tropical Medicine | L1014F | Decreased mortality after exposure to pyrethroids and DDT | Unknown | [100] | ||||||
2021 | Anopheles gambiae | China Hainan University | nAChR alpha6 | Decreased mortality after exposure to spinosad | Unknown | [101] | ||||||
2020 | Aedes aegypti | United States of America NIH/NIAID Laboratory of Malaria and Vector Research | mJHBP | Decreased antimicrobial resistance | 16.7% (20/120)/unknown | [102] | ||||||
2016 | Culex quinquefasciatus | Japan National Institute of Infectious Diseases | CYP9M10 | Decreased mortality after exposure to pyrethroids | Unknown/3% (4/128) c | [42] |
aSurvival and GEF indicates egg survival rate/gene editing frequencies.
bNo. (mosquitoes with successful gene editing)/No. (injected mosquito eggs).
cNo. (mosquitoes with successful gene editing)/No. (mosquito eggs survived after injection).
DEVELOPMENT OF CRISPR-CAS9 DERIVED TECHNOLOGIES
New technologies in mosquitoes
Currently, CRISPR-Cas9-based mosquito editing techniques rely on the microinjection [15] of sgRNA and Cas9 protein or Cas9 mRNA into individual embryos within mosquito eggs [108–110]. This process is technically difficult and costly; requires rigorous experimental equipment and experimenter handling; and has extremely low injection success rates, thus causing irreparable damage to mosquito eggs [111]. To overcome these difficulties, Lule-Chávez has used an innovative particle inflow gun technique to transfer Cas9 protein and sgRNA into Aedes aegypti and Anopheles gambiae eggs via particle bombardment. This process not only increases transformation efficiency and survival rates, but also is faster, simpler, and less expensive than micro-injection [112].
The Chaverra-Rodriguez research team has pioneered the development of a technique called receptor-mediated ovary transduction of cargo (ReMOT Control), which bypasses the step of mosquito egg embryo injection (Fig 3). For introducing targeted heritable mutations, high concentrations of Cas9 sgRNA complexes are directly injected into the hemolymph in female Aedes aegypti 24 hours after blood feeding [113]. Notably, the Cas9 protein used in ReMOT Control is not a commercial protein but a Cas9 fusion protein with a Drosophila melanogaster yolk protein tag (DmYP), which has been shown to effectively target the ovaries of some insects. Compared with ordinary commercial Cas9 proteins, this fusion Cas9 protein has stronger targeting properties and therefore shows better gene editing performance. This technique has been used to edit the Kmo, Kh, ECFP, and DsRed genes of Anopheles stephensi. Given the differences in developmental characteristics between Anopheles stephensi and Aedes aegypti, injection 48 hours after blood feeding has been shown to efficiently induce mutations in Anopheles stephensi progeny. In addition, the use of saponin substantially increases the efficiency of gene knockout, achieving results similar to those with use of chloroquine; therefore, the agents used to mediate intranuclear endosomal escape may influence the effects of ReMOT [114]. Li’s team has used the ReMOT Control technique to knock out the Kmo gene in Culex paleus and have found that 24 h after blood feeding is a preferable injection time for this species [90].

Difference between ReMOT Control technology and traditional embryo Micro-injection. A Traditional CRISPR-Cas9-based gene editing in mosquitoes relies on micro-injection of sgRNA+Cas9 RNP into freshly laid eggs to achieve gene editing in mosquitoes. B ReMOT Control bypasses the step of mosquito egg injection and directly injects sgRNA+fusion Cas9 RNP into the hemolymph of female mosquitoes after blood feeding; the RNP circulates to the developing ovaries, thus achieving gene editing in offspring. This figure was created with BioRender.com.
New technologies in other vectors
Ticks carry and transmit a variety of viruses and microorganisms [115]. Researchers have attempted to genetically edit tick eggs with CRISPR-Cas9; however, the development of gene editing in ticks has been hindered by their hard chorionic membrane, high intra-egg pressure, and the female tick’s gene’s organ, which encases the eggs in a waxy substance [116,117]. The advent of ReMOT Control technology has substantially overcome these problems, thereby enabling successful editing of the genes of some tick progeny through direct injection of Cas9 protein complexes into female ticks. All injected ticks have been reported to survive. To date, researchers have successfully edited the genes of Nasonia vitripennis [118], the red flour beetle Tribolium castaneum [119], and the Silverleaf Whitefly [120] with ReMOT Control technology. Moreover, this technique has been effectively used in the treatment of the notable disease vector Rhodnius prolixus. The development of mosquito gene editing has been greatly advanced by the emergence of ReMOT Control technology, which overcomes many of the drawbacks of microscopic embryo injection and enables widespread use of the CRISPR system. However, current research is limited and requires continuous screening and optimization of injection conditions for different species to achieve higher editing efficiency.
Recently, Shirai’s research team has pioneered the development of direct parental CRISPR (DIPA-CRISPR), a new technology based on ReMOT Control. The primary difference between this method and ReMOT Control is that the DIPA technique uses commercial Cas9 proteins rather than fusion proteins with DmYP tags. Shirai’s team has discovered that co-injection of nontagged Cas9 protein with the sgRNA protein complex into female Blattella germanica effectively delivers the protein to developing oocytes and alters the progeny’s genes. This method may potentially be used to eradicate cockroaches at the genetic level [121]. The team has also successfully used this protocol to edit the cardinal gene of Tribolium castaneum and change its eye color, thus demonstrating its broad applicability to certain insects [120]. Given the lack of a requirement to express tagged fusion Cas9 proteins, the researchers believe that DIPA-CRISPR technology can be used in laboratory settings to expand the scope of gene editing in insects more easily and effectively. This technique could theoretically also be applied to mosquitoes, thereby further simplifying the experimental process and allowing more laboratories perform gene editing.
DISCUSSION
As global warming increases, the spread of mosquito-borne viruses is gradually increasing each year, and the task of preventing and controlling mosquitoes and mosquito-borne infectious diseases is critical. Mosquito-borne flaviviruses have similar sequences and structures, and extensive antibody cross-reactivity has slowed vaccine development. For example, no effective preventive measures are available for mosquito-borne dengue fever, which causes many infections and deaths every year. The emergence of gene editing has provided new hope for mosquito control and prevention.
The advent of CRISPR gene editing technology has intensified the modification of mosquito genes in recent years. Researchers have discovered a series of genes associated with mosquito phenotypes, resistance, and viral interaction. Several public databases in China and other countries contain transcriptomic data from mosquito-related experiments, with an aim to gather experimental data to discover more genetic loci associated with mosquito physiology and virus transmission, thus advancing the study of mosquito gene function. Gene editing for mosquitoes is focused primarily on physiological functions and vector efficacy. However, with the aid of genomic data on mosquitoes or other vector organisms, future research may encompass a more expansive range of topics, including a deeper exploration of gene function and its potential applications in the prevention and control of vector-borne diseases. Moreover, researchers have optimized experimental methods. Hsing-Han Li has summarized the various experimental methods used for mosquito-based CRISPR gene editing and evaluated their advantages and disadvantages, thus providing a valuable reference for researchers [122]. New technology for gene editing continues to emerge, to address the shortcomings of traditional embryo injections, simplify the difficulties experimental manipulation, and enable more laboratories to participate in modifying insects’ genes. In the future, the optimization and innovation of new technologies such as ReMOT Control and DIPA-CRISPR may be used to screen target genes applicable to mosquito oocytes, whereas precise yolk formation time may be considered for different mosquito species to achieve higher gene editing efficiency. Notably, these traditional and novel techniques have been successfully used for gene editing in other disease vectors, thus facilitating the investigation of gene function and regulation.
Although irradiated mosquito SIT technology has been implemented in many countries, its high cost and the mating difficulties between irradiated male mosquitoes and wild female mosquitoes have prevented this technology from achieving satisfactory results in mosquito control. With the advent of CRISPR technology, scientists have continued to attempt to create gene-drive mosquitoes, to achieve replacement of wild-type mosquitos with genetically modified mosquitoes by altering the sex genes and sterilizing offspring, or by altering the ability to transmit pathogens to decrease the spread of disease [123]. As with other CRISPR-Cas9 gene editing, the potential for off-target effects of genes must be considered. Mosquito gene knockout also necessitates the use of whole-genome sequencing and off-target site prediction software to mitigate the risk of off-target effects. Additionally, off-target site prediction based on AI modeling and bioinformatics is currently available to enhance the stability of gene editing. However, gene-drive methods, such as insect irradiation SIT and even Wolbachia, require the release of large numbers of mosquitoes to alter wild populations. Whether gene-drive mosquitoes might affect other populations or the environment, and whether they are safe, remains unknown. However, these gene-drive mosquitoes have been found to work satisfactorily in small-scale laboratory replacement [124], and several countries have established mosquito factories to release Wolbachia-carrying mosquitoes in the wild, which is indeed effective in decreasing mosquito populations. The release of mosquitoes in the wild that are unable to transmit the virus, thereby replacing wild populations, has been shown to limit viral transmission. Nonetheless, whether these viruses might infect other new vectors that might subsequently infect humans is an important consideration. Additionally, the absence of the virus in mosquitoes might possibly result in the evolution of new phenotypes and potentially lead to attenuation of gene-drive suppression. Whether the decrease in mosquito numbers achieved with gene-drives might increase the number of remaining vector organisms, thereby elevating the risk of other disease transmission, additionally remains unclear. How to regulate these engineered mosquitoes, and what policies and regulations should be put in place, will require further consideration [125–127]. Discussions are ongoing as to whether releasing gene-drive mosquitoes into the wild on a large scale is ethical. Finally, the release of gene-drive mosquitoes might raise fears among the public [128] (such as whether their release might change the diet of traditional mosquitoes, transmit new pathogens, or alter human genetics through bites). The release of gene-drive mosquitoes to replace wild populations, and further prevent and control the spread of arboviruses, will require implementation of robust regulatory frameworks and policies to oversee the process and assess its efficacy, to maintain a balance between the application and effects of this strategy. A positive balance in which the potential benefits of releasing gene-drive mosquitoes outweigh the potential harms might indicate that the latter are not significant, that the likelihood of their occurrence is tolerable, and that reliable mitigation strategies could address the potential harms. How to best address these questions warrants further in-depth study by researchers and government regulators.