Next Article in Journal
A Non-Destructive Method, Micro-CT, Supports the Identification of Three New Casmara Species from Sumatra and Taiwan (Lepidoptera: Ashinagidae)
Previous Article in Journal
Integrated Multi-Omics Analysis of the Developmental Stages of Antheraea pernyi Pupae: Dynamic Changes in Metabolite Profiles and Gene Expression
Previous Article in Special Issue
Effects of Trehalase on the Gene Expression of the Reproductive Regulation Pathway Network and Triglyceride Metabolism in Nilaparvata lugens (Stål)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Perspectives of RNAi, CUADb and CRISPR/Cas as Innovative Antisense Technologies for Insect Pest Control: From Discovery to Practice

1
Department of Entomology, Agriculture College, Garhwa, Birsa Agricultural University, Ranchi 834006, India
2
Department of General Biology and Genetics, V.I. Vernadsky Crimean Federal University, 295007 Simferopol, Russia
3
Division of Entomology, ICAR-Indian Agricultural Research Institute, New Delhi 110012, India
4
Division of Genomic Resources, National Bureau of Agricultural Insect Resources, Bengaluru 560024, India
5
Division of Vegetable Sciences, ICAR-Indian Agricultural Research Institute, New Delhi 110012, India
6
College of Plant Protection, Jilin Agricultural University, Changchun 130118, China
*
Author to whom correspondence should be addressed.
Insects 2025, 16(7), 746; https://doi.org/10.3390/insects16070746
Submission received: 10 July 2025 / Revised: 17 July 2025 / Accepted: 19 July 2025 / Published: 21 July 2025
(This article belongs to the Special Issue RNAi in Insect Physiology)

Simple Summary

RNA interference (RNAi), contact unmodified antisense DNA biotechnology (CUADb), and clustered regularly interspaced short palindromic repeats (CRISPR) along with CRISPR-associated proteins (CRISPR/Cas) represent three innovative cost-effective antisense technologies for insect pest control. Each method operates through the formation of specific unmodified nucleic acid complexes, RNAi involves guide RNA binding to messenger RNA (mRNA); CUADb: employs guide DNA interacting with ribosomal RNA (rRNA); and CRISPR/Cas: utilizes guide RNA targeting genomic DNA. These complexes function via nucleic acid-guided nucleases: Argonaute in RNAi; rRNase in CUADb, and CRISPR-associated proteins in CRISPR/Cas systems.

Abstract

Pest management is undergoing a transformative shift with the development of the cutting-edge antisense technologies: RNA interference (RNAi), contact unmodified antisense DNA biotechnology (CUADb), and the CRISPR-associated proteins (CRISPR/Cas). These approaches function by facilitating sequence-specific pairing of nucleic acids followed by nuclease-mediated cleavage, offering exceptional precision for targeted pest control. While RNA-guided mechanisms such as RNAi and CRISPR/Cas were initially characterized in non-insect systems, primarily as innate defenses against viral infections, the DNA-guided CUADb pathway was first identified in insect pests as a functional pest control strategy. Its broader role in ribosomal RNA (rRNA) biogenesis was recognized later. Together, these discoveries have revealed an entirely new dimension of gene regulation, with profound implications for sustainable pest management. Despite sharing a common principle of sequence-specific targeting RNAi, CUADb, and CRISPR/Cas differ in several key aspects, including their mechanisms of action, target specificity, and applicability. Rather than serving as universal solutions, each technology is likely to be optimally effective against specific pest groups. Moreover, these technologies allow for rapid adaptation of control strategies to overcome target-site resistance, ensuring long-term efficacy. This review summarizes the core functional characteristics, potential applications, and current limitations of each antisense technology, emphasizing their complementary roles in advancing environmentally sustainable pest control. By integrating foundational biological discoveries with applied innovations, this work provides a new perspectives on incorporating antisense-based strategies into next-generation integrated pest management systems.

1. Introduction

Nucleic acids, DNA and RNA, orchestrate cellular processes through precise complementary interactions [1]. The fundamental principles of Watson–Crick base pairing, combined with the action of specific enzymes, govern essential biological mechanisms such as replication, transcription, translation, and regulation of gene expression [2]. The specificity and fidelity of these processes arise from the unique combinations of nitrogenous bases, which constitute the molecular foundation of genetic control. Three innovative antisense technologies, RNA interference (RNAi) [3], contact unmodified antisense DNA biotechnology (CUADb) [4,5,6], and clustered regularly interspaced short palindromic repeats (CRISPR) along with CRISPR-associated proteins (CRISPR/Cas) [7,8,9], have harnessed these nucleic acid interactions to induce targeted genetic effects. These technologies operate through the formation of sequence-specific duplexes: RNAi (guide RNA–messenger RNA (mRNA)) [10], CUADb (guide DNA-ribosomal RNA (rRNA)) [5,11,12,13,14], and CRISPR/Cas (guide RNA-genomic DNA) [9,15,16], which subsequently recruit specialized nucleases such as Argonaute (Ago) [17], rRNase [5,6], and CRISPR-associated proteins [9].
The emergence of these technologies was made possible by significant advances in bioinformatics, in vitro nucleic acid synthesis, and the development of modern molecular tools. Typically, a pivotal insight or an experimental breakthrough reveals a pattern that can be reliably applied in practice. For RNAi, the key finding was the use of an antisense fragment within double-stranded RNA (dsRNA); for CUADb, it was the understanding that antisense oligodeoxyribonucleotides can be used as contact insecticides; and for CRISPR/Cas, it was the identification that genomic DNA could serve as the target for antisense-guided effects. These innovations have set new standards in molecular genetics and are now being widely explored for their applications in insect pest control. Each of these technologies emerged from fundamental research, evolving from uncertain beginnings into powerful tools with transformative potential. Since their core mechanism relies on the complementary binding of antisense molecule, either DNA or RNA, to a target nucleic acid, they are collectively referred to as antisense technologies (Figure 1). While RNA- and DNA-guided nucleases have been extensively studied, a targeted DNA-cleaving mechanism utilizing guide DNA to cut pest genomic DNA via a specific nuclease has not yet been developed, representing a potential direction for future research.
Despite the role of chemical insecticides is pest management [18], several factors drive the search for alternative control strategies. The most important of these is high economic cost of insect pest damage to agriculture and growing prevalence of insecticide resistance, which has increased dramatically and persistently since the mid-20th century [18,19,20]. Insecticide resistance generally arises through natural selection, whereby the frequency of specific alleles increases due to random mutations within pest populations [21]. Antisense technologies (RNAi, CUADb, and CRISPR/Cas) are able to counteract insecticide resistance by targeting conserved genes or conserved gene regions and by facilitating the rapid development of effective pest control agents when resistance to existing insecticides emerges. While CUADb [6] and RNAi [3] demonstrate promising potential as next-generation bioinsecticides, due to their fast biodegradability, selectivity, and low carbon footprint [12], CRISPR/Cas is primarily used to genetically attenuate insect pest populations through genetic engineering [7]. Nevertheless, these innovative antisense approaches, and their potential combinations, offer a broad and flexible toolkit for insect pest control. The primary challenge lies in selecting the most suitable strategy for each specific pest management scenario.
The development of antisense technologies was predicted on the discovery of the DNA double helix, followed by a formative period marked by pioneering studies, such as the site-specific modification of valine tRNA by Grineva and colleagues [22] and the work of Zamecnik and Stephenson using modified DNA against the Rous sarcoma virus [23]. However, it became critically important to develop methods employing unmodified (natural) nucleic acids and to transform them into cost-effective, environmentally safe antisense technologies. While the RNAi was discovered by a research group in the USA in 1998 [3], CUADb was introduced in 2008 and subsequently developed further in Crimea [4,6], the development of the CRISPR/Cas9 was established in 2012 through the contributions of several research groups, primarily from Lithuania, Sweden and the United States [7,24,25]. These efforts collectively lead to the establishment of the three main antisense technologies for pest control at the turn of the 21st century. Although CUADb offers a relatively straightforward framework for pesticide design, RNAi and CRISPR/Cas are still progressing toward streamlined, reliable strategies for creating selective and effective pest control agents. The objective of this review is to provide a concise historical overview of the emergence of antisense technologies and to assess their potential in insect pest control. Overall, we provide an evaluation of the current capabilities and limitations of antisense technologies in this field, and propose directions for future development.

2. RNAi

2.1. History of Discovery of RNAi

The discovery of RNAi was inspired by the pioneering work of Zamecnik and Stephenson (1978), who demonstrated that a short antisense sequence of modified nucleic acid could inhibit the replication of the Rous sarcoma virus [23]. In 1998, Mello and Fire investigated the effect of both antisense and sense RNA fragments on the development of the nematode Caenorhabditis elegans. Their aim was to elucidate the mechanisms underlying the efficacy of sense RNA fragments synthesized by bacteriophage RNA polymerase (specifically, DNA-dependent RNA polymerases), using dsRNA fragments as experimental controls. Bacteriophage polymerases, although highly specific, occasionally produce ectopic transcripts, and DNA transgene arrays, also known as extrachromosomal arrays, are known to generate aberrant RNA products. Mello and Fire hypothesized that the interfering RNA population might include molecules with double-stranded features. Unexpectedly, it was the dsRNA fragment that triggered a potent silencing effect on the endogenous mex-3 mRNA transcript, which is highly expressed in the gonads and early embryos of the nematode. The groundbreaking discovery, published in Nature in 1998 [3], established the foundational concept of RNAi: dsRNA molecules can initiate gene silencing by targeting homologous transcripts [26,27]. Subsequent research revealed that short antisense RNA fragments (21–23 nucleotides in length) are produced during RNAi and mediate the degradation of target mRNAs [28] (Figure 2).

2.2. How RNAi Works in Insect Pests

RNAi has been extensively studied for its potential in agricultural pest management, particularly through the silencing of essential insect genes to suppress pest populations. The small interfering RNA (siRNA) pathway, the most thoroughly characterized RNAi mechanism, consists of two branches: the exogenous siRNA (exo-siRNA) pathway, which mediates antiviral defense, and the endogenous siRNA (endo-siRNA) pathway, responsible for transposon suppression within the genome [29]. The exo-siRNA pathway is commonly utilized in experimental and applied contexts by introducing dsRNA to silence specific target genes [30,31]. For effective gene silencing, long dsRNA fragments (>200 bp) are typically used to trigger the production of short antisense siRNAs [26,27]. The RNAi process begins with cellular uptake of dsRNA, followed by cleavage into siRNAs (21–25 nucleotides) by the enzyme Dicer. These siRNAs are then incorporated into the RNA-induced silencing complex (RISC), where Argonaute (Ago) proteins serve as the catalytic core [32]. One strand of the siRNA duplex, known as the guide strand, directs RISC to the complementary mRNA target, leading to transcript degradation or translational repression [33].
Although much of the research has focused on the siRNA machinery, the RNAi pathway in insects is more complex and includes three sub-pathways: siRNA, micro (miRNA), and piwi-interacting RNA (piRNAs). miRNAs (18–24 nucleotides) are involved in diverse regulatory processes and interact with Ago proteins, while piRNAs (26–31 nucleotides) are primarily responsible for silencing transposable elements, particularly in germline cells, through interactions with PIWI proteins. Artificially synthesized dsRNA may influence not only the siRNA pathway but also genes associated with the miRNA pathway, suggesting a broader, interconnected regulatory network [34].
RNAi is particularly robust and systemic in Coleopteran insects [29]. However, systemic RNAi, defined as the environmental uptake of dsRNA followed by intercellular transport, is limited in Diptera, Lepidoptera [35,36,37], and sap-feeding Hemiptera [38,39], thereby constraining its utility in some agricultural contexts. Nonetheless, RNAi has demonstrated considerable potential for control of holometabolous and hemimetabolous insect pests, by targeting key genes involved in development, detoxification, and reproduction (Table 1).

2.3. Perspectives and Limitations of RNAi for Insect Pest Control

Despite significant advancements in RNAi research over the past 25 years, its practical management of insect pests and disease vectors remains limited. To date, only one commercial dsRNA-based biopesticide, Calantha®, has been developed for foliar application, with several others currently under development. Additionally, three transgenic crops expressing dsRNA are commercially available.
Calantha®, a sprayable dsRNA-based biopesticide targeting the Colorado potato beetle (CPB, Leptinotarsa decemlineata), was released commercially by GreenLight Biosciences in 2023 [68,69]. As with dsRNA-expressing crops, sprayable formulations are likely to be used in combination with other pest control tools. However, sprayable dsRNAs degrade rapidly in the environment [70], necessitating careful consideration of application timing, spray intervals and synchronization with insect life cycle to ensure that only a single generation is exposed to the dsRNA product. Subsequent insecticide treatments should then be rotated to a different mode of action [71,72].
Several challenges have hindered the widespread commercialization of RNAi-based products. These include variable efficacy across insect species, competition from transgenic Bt crops, and limited effectiveness against some sap-sucking pests. Another concern is the potential evolution of RNAi resistance. For example, studies have shown that western corn rootworm (Diabrotica virgifera virgifera) [73], willow leaf beetle (Plagiodera versicolora) [74], and CPB [75,76] can develop resistance to dsRNA-based control. Of note, Calantha® can be deployed in pesticide rotation programmes to mitigate selection for resistance in CPB [77]. Nonetheless, RNAi, when combined with precision agriculture and integrated pest management (IPM) strategies, could significantly enhance sustainability. Opportunities for synergy exist with advancing the technologies such as CUADb, CRISPR/Cas technology, microbial-based RNAi production, and nanoformulations for improved dsRNA delivery. Of note, RNA molecules are prone to degradation in the environment. To enhance their effectiveness, researchers are developing more stable RNA formulations and innovative delivery methods.
A clear understanding of dsRNA structure is critical for optimizing RNAi applications in pest control. This structural knowledge informs how dsRNA is processed into siRNAs and facilitates the precise targeting of pest genes for silencing [78,79]. Although the effects of dsRNA-mediated interference are highly potent and specific, there are several concerns that should be taken into account when designing RNAi-based experiments. If a sequence is shared among multiple closely related genes, RNAi may unintentionally silence several members of the gene family or homologous genes in related species [80]. Also, genes with low expression levels may exhibit resistance to RNAi, at least partially. For example, if the target protein is very stable, its depletion occurs much more slowly despite transcript degradation. Moreover, the function of a target protein can be compensated for by related proteins, which may even be upregulated by the cell to counteract the loss. Additionally, the transcription of the target gene itself could be upregulated by a regulatory gene network in response to RNAi mediated knock-down [81].
Importantly, insights into how Argonaute proteins bind to guide and target RNAs allow for the rational design of highly specific and effective RNAi tools. Structural analyses of the Argonaute active site and its interaction with target RNAs enable the optimization of siRNA sequences for enhanced gene silencing [82,83]. Furthermore, understanding the architecture of RISC components supports improvements in delivery strategies and silencing efficiency [84]. Overall, this structural knowledge is essential for developing RNAi-based pest control methods that are both species-specific and environmentally sustainable [85].
Currently, the implementation of RNAi-based pest control strategies is challenged by regulatory uncertainties and public apprehension. Gaining regulatory approval and public acceptance will depend on transparent scientific communication, rigorous safety evaluations, and evidence-based demonstrations of efficacy. A major practical limitation remains the high cost of dsRNA production; publicly accessible in vitro synthesis methods are still prohibitively expensive, exceeding $50 per milligram [86,87,88,89,90].
Despite these challenges, RNAi remains a promising strategy for insect pest control due to its selectively silence genes critical for insect development and survival. This specificity provides an opportunity to develop environmentally sustainable pest control solutions. However, the successful deployment of dsRNA-based insecticides will require coordinated efforts from researchers, policymakers, and industry stakeholders worldwide. Future advancements, particularly in dsRNA formulation, delivery systems, and cost-effective production, are expected to drive the broader adoption of RNAi in the coming decades, potentially transforming agricultural pest management practices [91].

3. CUADb

3.1. History of Discovery of CUADb

Unmodified DNA, as a programmable molecule and natural biopolymer, has long captured scientific interest. However, for many years it was believed that unmodified oligodeoxyribonucleotides were both cytotoxic and highly susceptible to degradation in eukaryotic cells due to nuclease activity [92], including in insects [11,93]. Some studies explicitly stated that unmodified (phosphodiester) antisense oligonucleotides should not be employed in experimental applications [94]. Additionally, it was widely assumed that ribosomal RNA (rRNA) was inherently resistant to degradation by antisense DNA oligonucleotides [95,96]. At the turn of the century, there was no indication in the scientific literature that DNA could be used as an insecticide.
The insecticidal properties of DNA were discovered serendipitously in the spongy moth (Lymantria dispar). In 2007, Oberemok initiated research on the transovarial transmission of L. dispar multiple nucleopolyhedrovirus (LdMNPV) as part of his doctoral work. Two specific primers were selected within the anti-apoptotic gene (IAP-3) of LdMNPV: a forward primer from the sense strand (5′-GCCGGCGGAACTGGCCCA-3; oligoBIR fragment) and a reverse primer from the antisense strand (5′-CGACGTGGTGGCACGGCG-3′; oligoRING fragment) [97]. These primers generated a 317 bp amplicon in PCR using purified viral DNA [98]. However, when applied to DNA extracted from virus-free L. dispar, the primers produced amplicons of varying lengths, suggesting non-specificity and indicating the presence of homologous sequences in the host genome—a phenomenon previously documented for other viruses [99]. Given this, it was hypothesized that the oligoBIR and oligoRING fragments could modulate the expression of homologous L. dispar IAP genes, potentially inducing apoptosis. In April 2008, Oberemok tested this hypothesis unconventionally by topically applying drops of aqueous solutions of the primers to the surface of L. dispar larvae [4,100]. Remarkably, after 3–5 days, significant larval mortality was observed, attributable to the applied DNA fragments derived from the viral genome. This experiment marked the inception of a new class of contact DNA insecticides, termed oligonucleotide insecticides, or briefly olinscides, and the CUADb (Contact Unmodified Antisense DNA biotechnology) platform. The first results were reported in a Ukrainian patent (No. 36445) in 2008, followed by articles in Pesticide Biochemistry and Physiology [101,102]. The earliest oligonucleotide insecticides (18–20 nt) targeting anti-apoptotic genes were effective in both virus-free and LdMNPV-infected larvae [97]. Importantly, unique antisense DNA sequences of 11–20 nt offer high target selectivity, although their efficacy depends on the abundance of target RNA. Consequently, CUADb was optimized to target rRNA—which constitutes approximately 80% of total cellular RNA—via a DNA containment (DNAc) mechanism, resulting in a robust algorithm for insect pest control (Figure 3). This concept later was used by RNAi researchers. Notably, three years after Oberemok’s discovery, Wang et al. [103] successfully demonstrated, for the first time, the use of dsRNA fragments as contact insecticides in pest control.

3.2. How CUADb Works on Insect Pests

In 2019, Oberemok and colleagues introduced several major refinements to the CUADb platform aimed at enhancing its efficacy and cost-effectiveness. A key advancement was the selection of insect ribosomal RNA (rRNA) as the primary target for oligonucleotide insecticides [11]. Targeting pre-rRNA and mature rRNA significantly enhances efficacy because these RNA molecules account for approximately 80% of total cellular RNA [104], in contrast, mRNAs, which constitute only around 5%. This strategic targeting substantially increases the signal-to-noise ratio, up to approximately 100,000:1 when compared to random mRNA sequences. Insects possess both nuclear and mitochondrial rRNA, including 28S (~3900 nucleotides (nt)), 18S (~1920 nt), 5.8S (~160 nt), and 5S (~120 nt), 16S (~1140 nt) and 12S (~600 nt) making rRNA an ideal and abundant molecular target.
A second major refinement involved reducing the length of oligonucleotide insecticides to 10–12 nt to lower production costs. Shorter sequences result in higher yields during phosphoramidite-based DNA synthesis increasing product mass per synthesis cycle. Despite their brevity, 11-nt oligonucleotides retain sufficient specificity, with a uniqueness frequency of approximately 1 in 4.19 million sequences, adequate for most agricultural systems [105]. In more complex ecosystems, such as forest, longer sequences (15–20 nt) may be used to improve selectivity [12].
A third key advancement was the identification of susceptibility to unmodified antisense oligonucleotides in insects of the suborder Sternorrhyncha (Hemiptera) [6]. Since then, CUADb-based insecticides have been successfully tested against a wide range of sap-feeding insect pests. These include 28S rRNA targets in Unaspis euonymi, Dynaspidiotus britannicus, Icerya purchasi, Ceroplastes japonicus, Aonidia lauri, Coccus hesperidum [5,12,13,106,107,108,109]; 18S rRNA in Pseudococcus viburni [110]; and internal transcribed spacer 2 (ITS2) of pre-rRNA in Macrosiphoniella sanborni, Schizolachnus pineti [14,111], and Trioza alacris [112] (Table 2). ITS2-targeting oligonucleotides have also demonstrated acaricidal potential against the spider mite Tetranychus urticae [113,114]. Typically, a single contact treatment with oligonucleotide insecticides at 100 ng/μL results in mortality rates of approximately 80% within 3 to 14 days [12,109]. The first successful application of these insecticides within Sternorrhyncha was conducted on U. euonymi in 2019 [107,115].
It is important to note that studies on Sternorrhynchan insects demonstrated that unmodified oligodeoxyribonucleotides (also known as oligonucleotide insecticides or briefly olinscides) can induce both upregulation and downregulation of target genes through a mechanism known as DNA containment (DNAc). This mechanism operates in two sequential steps: the first involves the arrest of target rRNA and/or pre-rRNA (formation of DNA–rRNA duplex), leading to a functional block of ribosomes and triggering a compensatory overexpression of rRNA via rDNA transcription. The second step of DNA containment mechanism involves degradation of the arrested rRNA and/or pre-rRNA by DNA-guided rRNase [5,6,117]. In the presence of rRNase, DNA–rRNA duplex is processed via a mechanism analogous to a zipper, referred to as the ‘genetic zipper’ method [112]. Our recent studies on C. hesperidum (4th day after contact application of oligonucleotide insecticide Coccus-11) show that almost all ribosomal proteins of 40S and 60S ribosomal subunits are also significantly upregulated during DNAc promoting the formation of new ribosomes together with hypercompensated rRNA. Ribosome biogenesis proteins involved in the formation of ribosomal subunits (NOP53, UTP30, NSA2, MAK21, BRX1, WDR12) are upregulated as well. Also during DNAc, most of ATP-dependent enzymes are downregulated (including mTOR, serine/threonine protein kinase playing crucial role in ribosome biogenesis through mTORC1 complex) while proteins from mitochondrial ATP synthase complex are upregulated indicating deficiency of cellular energy caused by oligonucleotide insecticide Coccus-11. RNase H1 is also significantly upregulated during DNAc. RNase H1 functions independently of cell cycle and cleaves RNA-DNA hybrids, including those formed between DNA and rRNA. Of note, the innovative ‘genetic zipper’ method not only facilitates accumulation of rRNA and its subsequent degradation as a core process during DNAc but also serves as a predictive algorithm capable of estimating the efficacy of a specific olinscide against the intended target species and closely related organisms possessing identical rRNA target sequences [14,112].
Oligonucleotide insecticides can be designed using DNAInsector program (available at dnainsector.com) or manually, based on pre-rRNA and rRNA sequences retrieved from the GenBank database. In practical terms, this means that individuals with basic sequence knowledge can design an olinscide complementary to the pre-rRNA or rRNA of a Sternorrhynchan insect with a high probability of success. However, to ensure species specificity, it is essential to compare homologous sites in non-target organisms to prevent off-target effects. Synthesis of oligonucleotide insecticides is typically performed using the phosphoramidite method, in either liquid-phase or solid-phase formats. Commonly used solid-phase synthesizers include the ASM-800 (BIOSSET, Novosibirsk, Russia), OligoPilotTM (Cytiva, Uppsala, Sweden), and the 10-Column DNA Synthesizer (PolyGen, Langen, Germany) [108]. The standard preparation involves dissolving the olinscide in nuclease-free water at a concentration of 1 mg per 10 mL and applying it to approximately 1 m2 of infested plant foliage.

3.3. Perspectives and Limitations of CUADb for Insect Pest Control

DNA-based technologies are currently transforming plant protection by enabling the development of novel control agents with advanced characteristics. DNA, being a natural biopolymer, offers enhanced compatibility with ecological systems, including agroecosystems. Its programmability allows the design of highly specific treatments, including insecticidal and acaricidal agents. Moreover, if resistance arises, adaptive strategies can be implemented by simply redesigning olinscides to the target upstream or downstream regions of the resistance-conferring site in pre-rRNA and rRNA sequences [5]. Oligonucleotide insecticides offer several distinct advantages: they exhibit a low carbon footprint, high specificity to target pests, rapid biodegradability in ecosystems, and minimal risk of target-site resistance. Furthermore, their efficacy can now be predicted across different pest species, particularly in phylogenetically related taxa with conserved target sequences [112]. Given these benefits, it is likely only a matter of time before such DNA-based products become mainstream tools in crop protection. The mechanism of action, known as the DNA containment mechanism, is well understood. The phosphoramidite synthesis method has been optimized, and delivery routes (primarily contact-based, as oral delivery remains less effective) have been established. Although cost remains a barrier, the price of DNA insecticides is expected to decrease with further technological advancements. In fact, CUADb-based insecticides are already becoming cost-competitive with conventional chemical alternatives for some pest species [12].
For instance, CUADb has significantly reduced costs in conifer aphid management by adopting liquid-phase DNA synthesis [13,108]. Sumitomo Chemical Co., Ltd. (Tokyo, Japan), a leading developer of liquid-phase synthesis, reportedly offers 1 kg of unmodified 11-nucleotide oligonucleotides for $25,000 [13]. By contrast, synthesizing the same quantity using standard solid-phase methods, more widely used in research laboratories, can cost up to $1 million. At an application rate of 200 L per hectare with an active ingredient concentration of 0.1 mg/L (0.1 ng/mL), the cost per hectare is approximately $0.50 using liquid-phase synthesis [13]. This level of affordability enables frequent field applications. However, for many Sternorrhynchan pests, effective control requires concentrations around 0.05 g/L, raising the cost to approximately $250 per hectare, even with cost-efficient synthesis. Consequently, achieving an optimal balance between cost and efficacy remains essential for widespread adoption of nucleic acid-based insecticides. Current estimates suggest that CUADb-based approaches could be applicable to 10–15% of all insect pest species.
While oligonucleotide insecticides have demonstrated strong efficacy against hemipteran and moderate efficacy against lepidopteran pests, they have shown much lower activity against coleopterans such as L. decemlineata [118]. Incorporating adjuvants (e.g., spreaders, adhesives, penetrants, and UV protectants) into formulations may improve performance, though their environmental safety must be thoroughly assessed. Another critical design consideration is the potential for non-canonical base pairing, such as A:C (C:A) and G:U (T:G) interactions [119,120], which may compromise specificity and increase the risk to non-target organisms. Thus, such interactions must be carefully accounted for during olinscide development [13,15]
Although the CUAD-based ‘genetic zipper’ method presents a promising and cost-effective platform for DNA insecticide development, further optimization of production processes and cost-reduction strategies will be necessary for large-scale implementation. Current research is focused on increasing target specificity by designing olinscides for unique rRNA regions, thereby minimizing off-target effects. Additionally, greater insight into olinscide uptake mechanisms by insect cells is needed to enhance efficacy. In conclusion, the ‘genetic zipper’ holds substantial promise for advancing insect pest control. However, its commercialization will depend on overcoming challenges related to delivery efficiency, production cost, resistance management, and environmental safety. Addressing these limitations will be essential for CUADb-based oligonucleotide insecticides to become a sustainable and widely adopted component of modern pest management.

4. CRISPR/Cas

4.1. History of Discovery of CRISPR/Cas

While the development of RNA interference (RNAi) technology and the CUADb database followed a relatively linear and well-documented trajectory, the emergence of CRISPR/Cas technology was shaped by the cumulative efforts of numerous research groups. This collaborative history makes it difficult to attribute the discovery to any single individual or team. This section provides an overview of the widely accepted scientific milestones in the development of the CRISPR/Cas system, while acknowledging that many contributors may not be named individually.
In 1987, Ishino and colleagues identified a previously unknown repeat sequence in Escherichia coli, although its significance was not fully understood at the time [121]. In 1989, Spanish scientist Mojica, while studying the archaeal microbe Haloferax mediterranei, observed similar palindromic repeats of approximately 30 base pairs, separated by unique spacer sequences of roughly 36 base pairs. These did not resemble any known repeat families in prokaryotes at the time [122]. Later Mojica and Jansen introduced the term CRISPR to describe these structures [25]. In 2002, Jansen et al. [123] identified Cas (CRISPR-associated) genes located adjacent to CRISPR loci by suggesting a functional relationship. In 2007, Horvath and colleagues provided the first experimental evidence of CRISPR-Cas system’s role in adaptive immunity. They demonstrated that Streptococcus thermophilus required the Cas7 protein for spacer acquisition and that resistance to phages was mantained by the phage-derived spacer, even in the absence of Cas7, indicating its role in spacer acquisition rather than the immunity process itself [124]. Cas9, previously referred to as Cas5, Csn1, or Csx12, which contains two nuclease motifs, HNH and RuvC, and was shown to be essential for phage resistance, confirming its direct role in DNA cleavage and adaptive immunity [125,126,127]. In 2011, Charpentier identified a small RNA in Streptococcus pyogenes known as trans-activating CRISPR RNA (tracrRNA), which is essential for the processing of CRISPR RNAs (crRNAs) and for guiding the Cas9 complex to cleave target DNA [7]. In 2012, Šikšnys et al. published a seminal study in demontsrating that Cas9 could be programmed with custom-designed spacers to cleave specific DNA targets in vitro [24]. Independently, Charpentier and Jennifer Doudna published in Science, that Cas9 could be guided by synthetic crRNAs and tracrRNAs to cleave dsDNA, with each domains cleaving a complementary DNA strand [25]. These foundational studies established the CRISPR/Cas9 system as a robust genome-editing tool. Although Charpentier, Doudna, and Šikšnys are often credited with demonstrating the system’s genome-editing potential, the technology’s development is the product of decades of collaborative scientific progress.

4.2. How CRISPR/Cas Works on Insect Pests

The CRISPR/Cas9 system is the predominant tool in gene editing used for insect pest control, characterized by its simplicity and efficiency. It consists of two key components: a sgRNA (single-guide RNA) and the Cas9 nuclease. The sgRNA is an engineered fusion of CRISPR RNA (crRNA), which defines the target DNA sequence, and trans-activating CRISPR RNA (tracrRNA), which facilitates Cas9 binding. This complex directs Cas9 to specific genomic loci via base pairing. Cas9, a multi-domain DNA endonuclease derived from Streptococcus pyogenes, recognizes a protospacer adjacent motif (PAM) sequence—typically 5′-NGG-3′—located near the target DNA site. Upon recognition, Cas9 induces a double-stranded break (DSB) three base pairs upstream of the PAM site. The HNH and RuvC nuclease domains cleave the target and complementary DNA strands, respectively [128,129,130,131,132]. Cellular repair of DSBs occurs via two pathways: non-homologous end joining (NHEJ), which often results in small insertions or deletions that disrupt gene function, or homology-directed repair (HDR), which enables precise gene editing using a repair template) [132,133]. Targeting a gene’s coding sequence with CRISPR/Cas9 and allowing repair via NHEJ often results in a nonfunctional protein. Alternatively, HDR allows for gene knock-ins through integration of exogenous sequences [133,134] (Figure 4).
Since its initial use in Drosophila, CRISPR/Cas9 has been widely adopted in diverse insect species. Genome editing aims to introduce engineered traits into wild pest populations, providing a promising strategy to manage invasive pests [135,136]. In the context of insect pests, CRISPR/Cas can be applied in several keyways (Table 3). This approach has been employed to disrupt genes involved in insecticide resistance. A comprehensive review by Xu et al. [137] utlines the application of CRISPR/Cas9 across various insect orders and editing types. Beyond resistance, CRISPR/Cas9 has also been used to manipulate genes regulating reproduction and sex determination. For example, targeting the Astra-2 (a sex determination gene) in Anastrepha ludens results in sterility in males and intersexual phenotypes in females [138]. The Doublesex (Dsx) gene, a key regulator of sexual differentiation in insects. has also been edited in species such as diamondback, producing gender-specific sterility through isoform disruption [139]. In addition to these applications, CRISPR/Cas9 has been used to suppress vector competence in disease-transmitting insects. By editing genes essential to pathogen transmission, researchers have successfully reduced the capacity of mosquitoes to spread diseases such as malaria, dengue, and Zika [140,141,142]. Moreover, the system has facilitated the development of gene drives designed to spread deleterious traits through populations, leading to sterility or reduced fitness and, ultimately, population suppression [132,143]. These gene drives bypass Mendelian inheritance through homing endonuclease activity and rely on Cas9 and guide RNAs to propagate engineered alleles during DNA repair [132,144,145,146]. Notably, Meccariello et al. [147] demonstrated the effectiveness of CRISPR/Cas9 gene drives in the Mediterranean fruit fly (Ceratitis capitata) by targeting the transformer gene to convert genetic females into fertile XX males. Similar strategies have been explored in beetles, moths and grasshoppers [148,149,150]. In some insect species, such as silkworms, CRISPR-based editing has proven more efficient than RNAi. For instance, knockdown of the homeobox gene Scr led to developmental abnormalities and malformed adult structures, showcasing the system’s potential in Lepidoptera [151]. Furthermore, CRISPR/Cas9 has been used synergistically with RNAi technologies. Chloroplast-engineered dsRNA targeting Frankliniella occidentalis has been combined with CRISPR/Cas to enhance pest control efficacy [152,153]. In addition to pest management, the technology has significantly advanced functional studies in insects. In S. frugiperda, genome editing has expanded our understanding of insect physiology, development, morphology, and behavior [133].

4.3. Perspectives and Limitations of CRISPR/Cas for Insect Pest Control

CRISPR/Cas technology presents a promising avenue for insect pest control, offering gene-editing capabilities that may contribute to the development of sustainable pest management strategies, including gene drives and insect-resistant plants. The application of molecular genetic engineering and insect transformation using CRISPR/Cas9 across multiple insect species has helped to overcome numerous limitations associated with traditional approaches, which largely relied on naturally occurring genetic mutations or transposable elements. CRISPR/Cas genome editing is widely favored for its efficiency, simplicity, and adaptability. In addition to Cas9, other Cas proteins, such as Cas3, Cas12a, and Cas13a, have expanded the toolbox for both natural and engineered gene editing, enabling precise modifications, base editing, prime editing, and gene regulation. Recent advancements have also facilitated DNA-free editing, allowing for genome modification without inducing double-stranded breaks [178,179,180].
Despite these developments, several significant challenges remain. It is currently difficult to accurately predict the outcomes of gene editing in insect pests or to determine which gene targets will yield the most effective results. Moreover, CRISPR/Cas systems may introduce unintended edits at off-target genomic sites, which can result in unforeseen biological consequences. Delivering CRISPR/Cas components into insect cells presents another major hurdle, as the efficiency of delivery methods can vary depending on species, developmental stage, or tissue type. Environmental concerns also persist regarding the use of gene drives and insect-resistant organisms, particularly in terms of their potential impacts on non-target species and broader ecosystem dynamics. Gene drives require successful mating between genetically modified and wild-type insects to facilitate the spread of edited traits; however, reproductive barriers, both prezygotic and postzygotic, may restrict gene flow and thus compromise the drive’s efficiency. In addition, genetically modified insects may suffer from reduced fitness, which could limit their ability to compete with wild populations and reduce the long-term success of the strategy. The introduction of precise genetic changes through HDR is also constrained by the technical challenge of delivering sufficient quantities of donor DNA templates [181]. Achieving complex genome modifications remains technically demanding, and issues such as mosaicism, genetic variability among cells in the same organism, may further complicate phenotypic analysis and stable trait inheritance. Although population replacement strategies offer potential benefits, ensuring the long-term sustainability and ecological containment of gene drive systems is essential. Particular attention must be paid to the possibility of unintended spread into non-target species or closely related taxa [133,182]. Furthermore, the long-term effects of CRISPR/Cas-mediated genetic modifications on insect development, behavior, and ecological interactions are not yet fully understood and remain an active area of investigation [137,183].
As research in insect molecular biology continues to progress, CRISPR/Cas technology is poised to become a transformative tool in modern pest management. Its capacity for targeted, species-specific interventions offers a refined and potentially more sustainable alternative to conventional chemical insecticides. Incorporating CRISPR/Cas-based strategies into IPM programs could significantly enhance the precision and resilience of crop protection across diverse agricultural systems, especially when integrated with broader principles of synthetic biology [133].

5. Conclusions

Antisense technologies are grounded in natural mechanisms that regulate essential life processes. Complementary interactions between nucleic acids underpin key biological functions such as cell division, metabolism and defense. The development of practical tools based on these antisense mechanisms represents a highly relevant and promising area of scientific research. We believe that sustained, long-term investment in this field is warranted to achieve effective and environmentally friendly solutions for pest management. RNAi, CUADb, and CRISPR/Cas systems are powerful gene silencing tools that operate at distinct molecular levels and offer varying degrees of permanence. RNAi and CUADb transiently reduce gene expression by targeting mRNA and rRNA of pests, respectively, whereas CRISPR/Cas induces permanent gene silencing through targeting alterations of genomic DNA. The CUADb system is unique among these technologies due to its DNA-guided mechanism and involvement in rRNA biogenesis, although it shares certain functional similarities with RNAi and CRISPR/Cas. Collectively, these systems hold considerable promise in overcoming insecticide resistance by targeting conserved genes and supporting the development of effective pest control agents.
The application of these technologies in insect pest control has already demonstrated significant potential. Notably, RNAi, CUADb and CRISPR/Cas exhibit optimal efficacy against specific insect pest groups, highlighting the potential for combining these approaches to enhance their effectiveness across broader pest populations. Although there is some level of competition among these modern antisense technologies, the physiological and genetic diversity of insect pests indicates that no single approach will provide a universal solution. Currently, antisense-based insect pest control technologies remain in the development phase. As data on their successes and limitations continue to accumulate, major companies are actively pursuing commercialization. Meanwhile, legislative framework governing the implementation of antisense technologies are gradually evolving in various countries. Although rapid breakthroughs may not be imminent, steady progress is inevitable, and will ultimately contribute to improvements in both human health and environmental sustainability.
In conclusion, RNAi, CUADb, and CRISPR/Cas represent three complementary ‘worlds’ of antisense technologies with promising future applications in diverse fields. These technologies hold potential for improving agriculture and developing new pest control strategies. CRISPR/Cas can create mutations for long-term changes, while RNAi and CUADb provide temporary gene silencing to block certain gene function or quickly adapt to resistance of insect pests. This combination of antisense tools offers versatility in addressing various aspects of pest control, including resistance management, population suppression, and developing novel and eco-friendly pest management strategies. However, challenges related to delivery, affordability, and environmental considerations need to be addressed for these technologies to reach their full potential.

Author Contributions

Conceptualization, V.O.; software, H.K., N.G. and V.O.; validation, V.O. and N.G.; formal analysis, V.O.; resources, V.O.; writing—original draft preparation, H.K., N.G., V.S., N.N., R.F., A.C., J.A., V.O. and K.L.; writing—review and editing, N.G., J.A. and V.O.; visualization, H.K., N.G. and V.O.; supervision, N.G. and V.O.; project administration, N.G. and V.O.; funding acquisition, V.O. All authors have read and agreed to the published version of the manuscript.

Funding

The research obtained funding from the Russian Science Foundation No. 25-16-20070, https://rscf.ru/project/25-16-20070/ (accessed on 27 Juny 2025) (Section 1 and Section 3) and obtained funding within the framework of a state assignment V.I. Vernadsky Crimean Federal University for 2024 and the planning period of 2024–2026 No. FZEG-2024–0001 (Section 2, Section 4 and Section 5).

Data Availability Statement

No new data were created.

Acknowledgments

We thank our many colleagues, too numerous to name, for the technical advances and lively discussions that prompted us to write this brief research report. We apologize to the many colleagues whose work has not been cited. Experiments were carried out at the Molecular Genetics and Biotechnologies Lab created within the framework of a state assignment V.I. Vernadsky Crimean Federal University for 2024 and the planning period of 2024–2026 No. FZEG-2024–0001. We are very much indebted to all reviewers and our colleagues from the lab on DNA technologies, PCR analysis, and creation of DNA insecticides (V.I. Vernadsky Crimean Federal University, Department of General Biology and Genetics), and OLINSCIDE BIOTECH LLC for valuable comments on our manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Minchin, S.; Lodge, J. Understanding biochemistry: Structure and function of nucleic acids. Essays Biochem. 2019, 63, 433–456. [Google Scholar] [CrossRef] [PubMed]
  2. Westhof, E.; Yusupov, M.; Yusupova, G. Recognition of Watson-Crick base pairs: Constraints and limits due to geometric selection and tautomerism. F1000Prime Rep. 2014, 6, 19. [Google Scholar] [CrossRef] [PubMed]
  3. Fire, A.; Xu, S.; Montgomery, M.K.; Kostas, S.A.; Driver, S.E.; Mello, C.C. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature 1998, 391, 806–811. [Google Scholar] [CrossRef] [PubMed]
  4. Oberemok, V.V. Method of Elimination of Phyllophagous Insects from Order Lepidoptera. UA Patent 36445 19 May 2008. [Google Scholar]
  5. Gal’chinsky, N.V.; Yatskova, E.V.; Novikov, I.A.; Sharmagiy, A.K.; Plugatar, Y.V.; Oberemok, V.V. Mixed insect pest populations of Diaspididae species under control of oligonucleotide insecticides: 3′-end nucleotide matters. Pestic. Biochem. Physiol. 2024, 200, 105838. [Google Scholar] [CrossRef] [PubMed]
  6. Oberemok, V.V.; Laikova, K.V.; Gal’chinsky, N.V. Contact unmodified antisense DNA (CUAD) biotechnology: List of pest species successfully targeted by oligonucleotide insecticides. Front. Agron. 2024, 6, 1415314. [Google Scholar] [CrossRef] [PubMed]
  7. Jinek, M.; Chylinski, K.; Fonfara, I.; Hauer, M.; Doudna, J.A.; Charpentier, E.A. Programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 2012, 337, 816–821. [Google Scholar] [CrossRef] [PubMed]
  8. Gasiunas, G.; Barrangou, R.; Horvath, P.; Siksnys, V. Cas9-crRNA ribonucleoprotein complex mediates specific DNA cleavage for adaptive immunity in bacteria. Proc. Natl. Acad. Sci. USA 2012, 109, E2579–E2586. [Google Scholar] [CrossRef] [PubMed]
  9. Doudna, J.A.; Charpentier, E. Genome editing. The new frontier of genome engineering with CRISPR-Cas9. Science 2014, 346, 1258096. [Google Scholar] [CrossRef] [PubMed]
  10. Li, Z.; Rana, T.M. Molecular mechanisms of RNA-triggered gene silencing machineries. Acc. Chem. Res. 2012, 45, 1122–1131. [Google Scholar] [CrossRef] [PubMed]
  11. Oberemok, V.V.; Laikova, K.V.; Gal’chinsky, N.V.; Useinov, R.Z.; Novikov, I.A.; Temirova, Z.Z.; Shumskykh, M.N.; Krasnodubets, A.M.; Repetskaya, A.I.; Dyadichev, V.V.; et al. DNA insecticide developed from the Lymantria dispar 5.8S ribosomal RNA gene provides a novel biotechnology for plant protection. Sci. Rep. 2019, 9, 6197. [Google Scholar] [CrossRef] [PubMed]
  12. Oberemok, V.V.; Laikova, K.V.; Andreeva, O.A.; Gal’chinsky, N.V. Oligonucleotide insecticides and RNA-based insecticides: 16 years of experience in contact using of the next generation pest control agents. J. Plant. Dis. Prot. 2024, 131, 1837–1852. [Google Scholar] [CrossRef]
  13. Oberemok, V.V.; Puzanova, Y.V.; Gal’chinsky, N.V. The ‘genetic zipper’ method offers a cost-effective solution for aphid control. Front. Insect Sci. 2024, 4, 1467221. [Google Scholar] [CrossRef] [PubMed]
  14. Oberemok, V.V.; Gal’chinsky, N.V.; Novikov, I.A.; Sharmagiy, A.K.; Yatskova, E.V.; Laikova, E.V.; Plugatar, Y. rRNA-specific antisense DNA and dsDNA trigger rRNA biogenesis and cause potent insecticidal effect on insect pest hesperidum L. biorXiv 2024, 10, 618468. [Google Scholar] [CrossRef]
  15. Wiles, M.V.; Qin, W.; Cheng, A.W.; Wang, H. CRISPR-Cas9-mediated genome editing and guide RNA design. Mamm. Genome 2015, 26, 501–510. [Google Scholar] [CrossRef] [PubMed]
  16. Li, T.; Yang, Y.; Qi, H.; Cui, W.; Zhang, L.; Fu, X.; He, X.; Liu, M.; Li, P.-f.; Yu, T. CRISPR/Cas9 therapeutics: Progress and prospects. Sig. Transduct. Target. Ther. 2023, 8, 36. [Google Scholar] [CrossRef] [PubMed]
  17. Ma, X.; Zuo, Z.; Shao, W.; Jin, Y.; Meng, Y. The expanding roles of Argonautes: RNA interference, splicing and beyond. Brief. Funct. Genom. 2018, 17, 191–197. [Google Scholar] [CrossRef] [PubMed]
  18. Araújo, M.F.; Castanheira, E.M.S.; Sousa, S.F. The Buzz on Insecticides: A Review of Uses, Molecular Structures, Targets, Adverse Effects, and Alternatives. Molecules 2023, 28, 3641. [Google Scholar] [CrossRef] [PubMed]
  19. Siddiqui, J.A.; Fan, R.; Naz, H.; Bamisile, B.S.; Hafeez, M.; Ghani, M.I.; Wei, Y.; Xu, Y.; Chen, X. Insights into insecticide-resistance mechanisms in invasive species: Challenges and control strategies. Front. Physiol. 2023, 13, 1112278. [Google Scholar] [CrossRef] [PubMed]
  20. Gul, H.; Gadratagi, B.G.; Güncan, A.; Tyagi, S.; Ullah, F.; Desneux, N.; Liu, X. Fitness costs of resistance to insecticides in insects. Front. Physiol. 2023, 14, 1238111. [Google Scholar] [CrossRef] [PubMed]
  21. Hawkins, N.J.; Bass, C.; Dixon, A.; Neve, P. The evolutionary origins of pesticide resistance. Biol. Rev. Camb. Philos. Soc. 2019, 94, 135–155. [Google Scholar] [CrossRef] [PubMed]
  22. Belikova, A.; Zarytova, V.; Grineva, N. Synthesis of ribonucleosides and diribonucleoside phosphates containing 2-chloroethylamine and nitrogen mustard residues. Tetrahedron Lett. 1967, 37, 3557–3562. [Google Scholar] [CrossRef] [PubMed]
  23. Zamecnik, P.C.; Stephenson, M.L. Inhibition of Rous sarcoma virus replication and cell transformation by a specific oligodeoxynucleotide. Proc. Natl. Acad. Sci. USA 1978, 75, 280–284. [Google Scholar] [CrossRef] [PubMed]
  24. Lander, E.S. The Heroes of CRISPR. Cell 2016, 164, 18–28. [Google Scholar] [CrossRef] [PubMed]
  25. Shmakova, A.A.; Shmakova, O.P.; Karpukhina, A.A.; Vassetzky, Y.S. CRISPR/Cas: History and Perspectives. Russ. J. Dev. Bio. 2022, 53, 272–282. [Google Scholar] [CrossRef]
  26. Tomoyasu, Y.; Miller, S.C.; Tomita, S.; Schoppmeier, M.; Grossmann, D.; Bucher, G. Exploring systemic RNA interference in insects: A genome-wide survey for RNAi genes in Tribolium. Genome Biol. 2008, 9, R10. [Google Scholar] [CrossRef] [PubMed]
  27. Svoboda, P. Key mechanistic principles and considerations concerning RNA interference. Front. Plant Sci. 2020, 11, 1237. [Google Scholar] [CrossRef] [PubMed]
  28. Zhao, Y.; Shu, R.; Liu, J. The development and improvement of ribonucleic acid therapy strategies. Mol. Ther. Nucleic Acids. 2021, 26, 997–1013. [Google Scholar] [CrossRef] [PubMed]
  29. Zhu, K.Y.; Palli, S.R. Mechanisms, applications, and challenges of insect RNA interference. Annu. Rev. Entomol. 2020, 65, 293–311. [Google Scholar] [CrossRef] [PubMed]
  30. Cooper, A.M.; Silver, K.; Zhang, J.; Park, Y.; Zhu, K.Y. Molecular mechanisms influencing efficiency of RNA interference in insects. Pest Manag. Sci. 2019, 75, 18–28. [Google Scholar] [CrossRef] [PubMed]
  31. Cooper, A.M.; Song, H.; Yu, Z.; Biondi, M.; Bai, J.; Shi, X.; Weerasekara, S.M.; Hua, D.H.; Silver, K.; Zhang, J.; et al. Comparison of strategies for enhancing RNA interference efficiency in Ostrinia nubilalis. Pest Manag. Sci. 2021, 77, 635–645. [Google Scholar] [CrossRef] [PubMed]
  32. Vogel, E.; Santos, D.; Mingels, L.; Verdonckt, T.; Broeck, J.V. RNA Interference in Insects: Protecting Beneficials and Controlling Pests. Front. Physiol. 2019, 9, 1912. [Google Scholar] [CrossRef] [PubMed]
  33. Girard, C.; Budin, K.; Boisnard, S.; Zhang, L.; Debuchy, R.; Zickler, D.; Espagne, E. RNAi-Related Dicer and Argonaute Proteins Play Critical Roles for Meiocyte Formation, Chromosome-Axes Lengths and Crossover Patterning in the Fungus Sordaria macrospora. Front. Cell Dev. Biol. 2021, 9, 684108. [Google Scholar] [CrossRef] [PubMed]
  34. Silver, K.; Cooper, A.M.; Zhu, K.Y. Strategies for enhancing the efficiency of RNA interference in insects. Pest Manag. Sci. 2021, 77, 2645–2658. [Google Scholar] [CrossRef] [PubMed]
  35. Terenius, O.; Papanicolaou, A.; Garbutt, J.S.; Eleftherianos, I.; Huvenne, H.; Kanginakudru, S.; Albrechtsen, M.; An, C.; Aymeric, J.L.; Barthel, A.; et al. RNA interference in Lepidoptera: An overview of successful and unsuccessful studies and implications for experimental design. J. Insect Physiol. 2011, 57, 231–245. [Google Scholar] [CrossRef] [PubMed]
  36. Lucena-Leandro, V.S.; Abreu, E.F.A.; Vidal, L.A.; Torres, C.R.; Junqueira, C.I.C.V.F.; Dantas, J.; Albuquerque, É.V.S. Current Scenario of Exogenously Induced RNAi for Lepidopteran Agricultural Pest Control: From dsRNA Design to Topical Application. Int. J. Mol. Sci. 2022, 23, 15836. [Google Scholar] [CrossRef] [PubMed]
  37. Christiaens, O.; Tardajos, M.G.; Martinez Reyna, Z.L.; Dash, M.; Dubruel, P.; Smagghe, G. Increased RNAi Efficacy in via the Formulation of dsRNA With Guanylated Polymers. Front. Physiol. 2018, 9, 316. [Google Scholar] [CrossRef] [PubMed]
  38. Jain, R.G.; Robinson, K.E.; Asgari, S.; Mitter, N. Current scenario of RNAi-based hemipteran control. Pest Manag. Sci. 2021, 77, 2188–2196. [Google Scholar] [CrossRef] [PubMed]
  39. Kaplanoglu, E.; Kolotilin, I.; Menassa, R.; Donly, C. Plastid Transformation of Micro-Tom Tomato with a Hemipteran Double-Stranded RNA Results in RNA Interference in Multiple Insect Species. Int. J. Mol. Sci. 2022, 23, 3918. [Google Scholar] [CrossRef] [PubMed]
  40. Tian, H.; Peng, H.; Yao, Q.; Chen, H.; Xie, Q.; Tang, B.; Zhang, W. Developmental control of a lepidopteran pest Spodoptera exigua by ingestion of bacteria expressing dsRNA of a non-midgut gene. PLoS ONE 2009, 4, e6225. [Google Scholar] [CrossRef] [PubMed]
  41. Zha, W.; Peng, X.; Chen, R.; Du, B.; Zhu, L.; He, G. Knockdown of midgut genes by dsRNA-transgenic plant-mediated RNA interference in the hemipteran insect Nilaparvata lugens. PLoS ONE 2011, 6, e20504. [Google Scholar] [CrossRef] [PubMed]
  42. Khan, A.M.; Ashfaq, M.; Khan, A.A.; Naseem, M.T.; Mansoor, S. Evaluation of potential RNA-interference-target genes to control cotton mealybug, Phenacoccus solenopsis (Hemiptera: Pseudococcuidae). Insect Sci. 2018, 25, 778–786. [Google Scholar] [CrossRef] [PubMed]
  43. Prentice, K.; Christiaens, O.; Pertry, I.; Bailey, A.; Niblett, C.; Ghislain, M.; Gheysen, G.; Smagghe, G. RNAi-based gene silencing through dsRNA injection or ingestion against the African sweet potato weevil Cylas puncticollis (Coleoptera: Brentidae). Pest Manag. Sci. 2017, 73, 44–52. [Google Scholar] [CrossRef] [PubMed]
  44. Camargo, R.A.; Barbosa, G.O.; Possignolo, I.P.; Peres, L.E.; Lam, E.; Lima, J.E.; Figueira, A.; Marques-Souza, H. RNA interference as a gene silencing tool to control Tuta absoluta in tomato (Solanum lycopersicum). PeerJ 2016, 4, e2673. [Google Scholar] [CrossRef] [PubMed]
  45. Sun, Z.; Liu, J.; Chen, Y.; Zhang, J.; Zhong, G. RNAi-mediated knockdown of α-Spectrin depresses reproductive performance in female Bactrocera dorsalis. Pestic. Biochem. Physiol. 2023, 196, 105611. [Google Scholar] [CrossRef] [PubMed]
  46. Arya, S.K.; Singh, S.; Upadhyay, S.K.; Tiwari, V.; Saxena, G.; Verma, P.C. RNAi-based gene silencing in Phenacoccus solenopsis and its validation by in planta expression of a double-stranded RNA. Pest Manag. Sci. 2021, 77, 1796–1805. [Google Scholar] [CrossRef] [PubMed]
  47. Chen, J.; Zhang, D.; Yao, Q.; Zhang, J.; Dong, X.; Tian, H.; Chen, J.; Zhang, W. Feeding-based RNA interference of a trehalose phosphate synthase gene in the brown planthopper, Nilaparvata lugens. Insect Mol. Biol. 2010, 19, 777–786. [Google Scholar] [CrossRef] [PubMed]
  48. Chen, J.Z.; Jiang, Y.X.; Li, M.W.; Li, J.W.; Zha, B.H.; Yang, G. Double-stranded RNA-degrading enzymes reduce the efficiency of RNA interference in Plutella xylostella. Insects 2021, 12, 712. [Google Scholar] [CrossRef] [PubMed]
  49. Li, N.; Xu, X.; Li, J.; Hull, J.J.; Chen, L.; Liang, G. A spray-induced gene silencing strategy for Spodoptera frugiperda oviposition inhibition using nanomaterial-encapsulated dsEcR. Int. J. Biol. Macromol. 2024, 281, 136503. [Google Scholar] [CrossRef] [PubMed]
  50. Bera, P.; Suby, S.B.; Dixit, S.; Vijayan, V.; Kumar, N.; Sekhar, J.C.; Vadassery, J. Identification of novel target genes for RNAi mediated management of the pest, Fall Armyworm (Spodoptera frugiperda, JE Smith). Crop Prot. 2025, 187, 106972. [Google Scholar] [CrossRef]
  51. Ma, Y.F.; Liu, T.T.; Zhao, Y.Q.; Luo, J.; Feng, H.Y.; Zhou Gong, L.L.; Zhang, M.Q.; He, Y.Y.; Hull, J.J.; Dewer, Y.; et al. RNA interference-screening of potentially lethal gene targets in the white-backed planthopper Sogatella furcifera via a spray-induced and nanocarrier-delivered gene silencing system. J. Agric. Food Chem. 2024, 72, 1007–1016. [Google Scholar] [CrossRef] [PubMed]
  52. Li, S.; Yang, H.; Wang, Y.; Wei, L.; Lyu, J.; Shan, Z.; Zhang, X.; Fan, D. RNA Interference Reveals the Impacts of CYP6CY7 on Imidacloprid Resistance in Aphis glycines. Insects 2024, 15, 188. [Google Scholar] [CrossRef] [PubMed]
  53. Saberi, E.; Mondal, M.; Paredes-Montero, J.R.; Nawaz, K.; Brown, J.K.; Qureshi, J.A. Optimal dsRNA Concentration for RNA Interference in Asian Citrus Psyllid. Insects 2024, 15, 58. [Google Scholar] [CrossRef] [PubMed]
  54. Wang, C.X.; Bao, H.Q.; Yan, Z.C.; Wang, J.; Wang, S.; Li, Y.X. Knockdown of vitellogenin receptor based on minute insect RNA interference methods affects the initial mature egg load in the pest natural enemy Trichogramma dendrolimi. Insect Sci. 2024, 32, 487–500. [Google Scholar] [CrossRef] [PubMed]
  55. Liu, J.; Yang, Y.; Yang, Q.; Lin, X.; Liu, Y.; Li, Z.; Swevers, L. Successful oral RNA interference efficiency in the silkworm Bombyx mori through nanoparticle-shielded dsRNA delivery. J. Insect Physiol. 2025, 161, 104749. [Google Scholar] [CrossRef] [PubMed]
  56. Liu, J.; Chen, W.; Chen, S.; Li, S.; Swevers, L. Similarly to BmToll9-1, BmToll9-2 Is a Positive Regulator of the Humoral Immune Response in the Silkworm, Bombyx mori. Insects 2024, 15, 1005. [Google Scholar] [CrossRef] [PubMed]
  57. Liu, J.; Chen, W.; Situ, J.; Li, J.; Chen, J.; Lai, M.; Huang, F.; Li, B. BmToll9-1 Is a Positive Regulator of the Immune Response in the Silkworm Bombyx mori. Insects 2024, 15, 643. [Google Scholar] [CrossRef] [PubMed]
  58. Liang, Y.; Wang, T.; Yang, W.; Chen, Z.; Li, Q.; Swevers, L.; Liu, J. Silencing of the immune gene BmPGRP-L4 in the midgut affects the growth of silkworm (Bombyx mori) larvae. Insect Mol. Biol. 2023, 32, 340–351. [Google Scholar] [CrossRef] [PubMed]
  59. Darweesh, A.F.; Fahmy, I.; Ali, M.; Elwahy, A. RNA interference of cysteine protease genes for the management of whitefly (Bemisia tabaci) by oral route. Egypt. J. Bot. 2025, 65, 43–56. [Google Scholar] [CrossRef]
  60. Shimomura, K.; Sakita, K.; Terajima, T.; Tomizawa, M. Gene Silencing of Olfactory Receptor Coreceptor by Systemic RNA Interference in Callosobruchus maculatus. J. Chem. Ecol. 2025, 51, 5. [Google Scholar] [CrossRef] [PubMed]
  61. Guo, P.P.; Yang, X.B.; Yang, H.; Zhou, C.; Long, G.Y.; Jin, D.C. Knockdown of the β-N-acetylhexosaminidase genes by RNA interference inhibited the molting and increased the mortality of the white-backed planthopper, Sogatella furcifera. Pestic. Biochem. Physiol. 2025, 207, 106216. [Google Scholar]
  62. Schellens, S.; Lenaerts, C.; Pérez Baca, M.D.R.; Cools, D.; Peeters, P.; Marchal, E.; Vanden Broeck, J. Knockdown of the Halloween genes Spook, Shadow and Shade influences oocyte development, egg shape, oviposition and hatching in the desert locust. Int. J. Mol. Sci. 2022, 23, 9232. [Google Scholar] [CrossRef] [PubMed]
  63. Mun, S.; Young Noh, M.; Dittmer, N.T.; Muthukrishnan, S.; Kramer, K.J.; Kanost, M.R.; Arakane, Y. Cuticular protein with a low complexity sequence becomes cross-linked during insect cuticle sclerotization and is required for the adult molt. Sci. Rep. 2015, 5, 10484. [Google Scholar] [CrossRef] [PubMed]
  64. Fu, D.; Dai, L.; Gao, H.; Sun, Y.; Liu, B.; Chen, H. Identification, expression patterns and RNA interference of aquaporins in Dendroctonus armandi (Coleoptera: Scolytinae) larvae during overwintering. Front. Physiol. 2019, 10, 967. [Google Scholar] [CrossRef] [PubMed]
  65. Araujo, R.N.; Santos, A.; Pinto, F.S.; Gontijo, N.F.; Lehane, M.J.; Pereira, M.H. RNA interference of the salivary gland nitrophorin 2 in the triatomine bug Rhodnius prolixus (Hemiptera: Reduviidae) by dsRNA ingestion or injection. Insect Biochem. Mol. Biol. 2006, 36, 683–693. [Google Scholar] [CrossRef] [PubMed]
  66. Shang, F.; Xiong, Y.; Xia, W.K.; Wei, D.D.; Wei, D.; Wang, J.J. Identification, characterization and functional analysis of a chitin synthase gene in the brown citrus aphid, Toxoptera citricida (Hemiptera, Aphididae). Insect Mol. Biol. 2016, 25, 422–430. [Google Scholar] [CrossRef] [PubMed]
  67. Lu, J.; Shen, J. Target genes for RNAi in pest control: A comprehensive overview. Entomol. Gen. 2024, 44, 95–114. [Google Scholar] [CrossRef]
  68. Pallis, S.; Alyokhin, A.; Manley, B.; Rodrigues, T.; Barnes, E.; Narva, K. Effects of Low Doses of a Novel dsRNA-based Biopesticide (Calantha) on the Colorado Potato Beetle. J. Econ. Entomol. 2023, 116, 456–461. [Google Scholar] [CrossRef] [PubMed]
  69. GreenLight Biosciences. Available online: https://www.greenlightbiosciences.com/calantha-for-cpb (accessed on 19 July 2025).
  70. Bachman, P.; Fischer, J.; Song, Z.; Urbanczyk-Wochniak, E.; Watson, G. Environmental fate and dissipation of applied dsRNA in soil, aquatic systems, and plants. Front. Plant Sci. 2020, 11, 21. [Google Scholar] [CrossRef] [PubMed]
  71. Sparks, T.C.; Nauen, R. IRAC: Mode of action classification and insecticide resistance management. Pestic. Biochem. Physiol. 2015, 121, 122–128. [Google Scholar] [CrossRef] [PubMed]
  72. Sparks, T.C.; Storer, N.; Porter, A.; Slater, R.; Nauen, R. Insecticide resistance management and industry: The origins and evolution of the insecticide resistance action committee (IRAC) and the mode of action classification scheme. Pest Manag. Sci. 2021, 77, 2609–2619. [Google Scholar] [CrossRef] [PubMed]
  73. Khajuria, C.; Ivashuta, S.; Wiggins, E.; Flagel, L.; Moar, W.; Pleau, M.; Miller, K.; Zhang, Y.; Ramaseshadri, P.; Jiang, C. Development and characterization of the first dsRNA-resistant insect population from western corn rootworm, Diabrotica virgifera virgifera LeConte. PLoS ONE 2018, 13, e0197059. [Google Scholar] [CrossRef] [PubMed]
  74. Liao, C.; Zhang, M.; Zhang, J. Characterization and potential mechanism of resistance to double-stranded RNA in willow leaf beetle, Plagiodera versicolora. J. Pest Sci. 2024, 97, 2217–2226. [Google Scholar] [CrossRef]
  75. Mishra, S.; Dee, J.; Moar, W.; Dufner-Beattie, J.; Baum, J.; Dias, N.P.; Alyokhin, A.; Buzza, A.; Rondon, S.I.; Clough, M.; et al. Selection for high levels of resistance to double-stranded RNA (dsRNA) in Colorado potato beetle (Leptinotarsa decemlineata Say) using non-transgenic foliar delivery. Sci. Rep. 2021, 11, 6523. [Google Scholar] [CrossRef] [PubMed]
  76. Mishra, S.; Moar, W.; Jurat-Fuentes, J.L. Larvae of Colorado potato beetle (Leptinotarsa decemlineata Say) resistant to double stranded RNA (dsRNA) remain susceptible to small-molecule pesticides. Pest Manag. Sci. 2024, 80, 905–909. [Google Scholar] [CrossRef] [PubMed]
  77. Narva, K.; Toprak, U.; Alyokhin, A.; Groves, R.; Jurat-Fuentes, J.L.; Moar, W.; Nauen, R.; Whipple, S.; Head, G. Insecticide resistance management scenarios differ for RNA-based sprays and traits. Insect Mol. Biol. 2025, 34, 518–526. [Google Scholar] [CrossRef] [PubMed]
  78. Christiaens, O.; Whyard, S.; Vélez, A.M.; Smagghe, G. Double-Stranded RNA Technology to Control Insect Pests: Current Status and Challenges. Front. Plant Sci. 2020, 11, 451. [Google Scholar] [CrossRef] [PubMed]
  79. Cedden, D.; Güney, G.; Rostás, M.; Bucher, G. Optimizing dsRNA sequences for RNAi in pest control and research with the dsRIP web platform. BMC Biol. 2025, 23, 114. [Google Scholar] [CrossRef] [PubMed]
  80. Chen, J.; Peng, Y.; Zhang, H.; Wang, K.; Zhao, C.; Zhu, G.; Reddy Palli, S.; Han, Z. Off-target effects of RNAi correlate with the mismatch rate between dsRNA and non-target mRNA. RNA Biol. 2021, 18, 1747–1759. [Google Scholar] [CrossRef] [PubMed]
  81. Cedden, D.; Bucher, G. The quest for the best target genes for RNAi-mediated pest control. Insect Mol. Biol. 2024, 34, 505–517. [Google Scholar] [CrossRef] [PubMed]
  82. Yang, J.; Cho, W.C.; Zheng, Y. Argonaute proteins: Structural features, functions and emerging roles. J. Adv. Res. 2020, 24, 317–324. [Google Scholar] [CrossRef] [PubMed]
  83. Höck, J.; Meister, G. The Argonaute protein family. Genome Biol. 2008, 9, 210. [Google Scholar] [CrossRef] [PubMed]
  84. Okamura, K.; Ishizuka, A.; Siomi, H.; Siomi, M.C. Distinct roles for Argonaute proteins in small RNA-directed RNA cleavage pathways. Genes Dev. 2004, 18, 1655–1666. [Google Scholar] [CrossRef] [PubMed]
  85. Hoang, B.T.L.; Fletcher, S.J.; Brosnan, C.A.; Ghodke, A.B.; Manzie, N.; Mitter, N. RNAi as a Foliar Spray: Efficiency and Challenges to Field Applications. Int. J. Mol. Sci. 2022, 23, 6639. [Google Scholar] [CrossRef] [PubMed]
  86. Socha, W.; Kwasnik, M.; Larska, M.; Rola, J.; Rozek, W. Vector-borne viral diseases as a current threat for human and animal health—One Health perspective. J. Clin. Med. 2022, 11, 3026. [Google Scholar] [CrossRef] [PubMed]
  87. Yang, W.; Wang, B.; Lei, G.; Chen, G.; Liu, D. Advances in nanocarriers to improve the stability of dsRNA in the environment. Front. Bioeng. Biotechnol. 2022, 10, 974646. [Google Scholar] [CrossRef] [PubMed]
  88. Nitnavare, R.B.; Bhattacharya, J.; Singh, S.; Kour, A.; Hawkesford, M.J.; Arora, N. Next generation dsRNA-based insect control: Success so far and challenges. Front. Plant Sci. 2021, 12, 673576. [Google Scholar] [CrossRef] [PubMed]
  89. Rank, A.P.; Koch, A. Lab-to-Field Transition of RNA Spray Applications—How Far Are We? Front. Plant Sci. 2021, 12, 755203. [Google Scholar] [CrossRef] [PubMed]
  90. Verdonckt, T.W.; Vanden Broeck, J. Methods for the Cost-Effective Production of Bacteria-Derived Double-Stranded RNA for in vitro Knockdown Studies. Front. Physiol. 2022, 13, 836106. [Google Scholar] [CrossRef] [PubMed]
  91. Palli, S.R. RNAi turns 25: Contributions and challenges in insect science. Front. Insect Sci. 2023, 3, 1209478. [Google Scholar] [CrossRef] [PubMed]
  92. Egli, M.; Manoharan, M. Chemistry, structure and function of approved oligonucleotide therapeutics. Nucleic Acids Res. 2023, 51, 2529–2573. [Google Scholar] [CrossRef] [PubMed]
  93. Peng, Y.; Wang, K.; Fu, W.; Sheng, C.; Han, Z. Biochemical comparison of dsRNA degrading nucleases in four different insects. Front. Physiol. 2018, 9, 624. [Google Scholar] [CrossRef] [PubMed]
  94. Dias, N.; Stein, C.A. Antisense oligonucleotides: Basic concepts and mechanisms. Mol. Cancer Ther. 2002, 1, 347–355. [Google Scholar] [PubMed]
  95. Bachellerie, J.P.; Cavaillé, J.; Hüttenhofer, A. The expanding snoRNA world. Biochimie 2002, 84, 775–790. [Google Scholar] [CrossRef] [PubMed]
  96. Will, C.L.; Lührmann, R. Spliceosomal UsnRNP biogenesis, structure and function. Curr. Opin. Cell Biol. 2001, 13, 290–301. [Google Scholar] [CrossRef] [PubMed]
  97. Oberemok, V.V.; Laikova, K.V.; Zaitsev, A.S.; Shumskykh, M.N.; Kasich, I.N.; Gal’chinsky, N.V.; Bekirova, V.V.; Makarov, V.V.; Agranovsky, A.A.; Gushchin, V.A.; et al. Molecular Alliance of Lymantria dispar Multiple Nucleopolyhedrovirus and a Short Unmodified Antisense Oligonucleotide of Its Anti-Apoptotic IAP-3 Gene: A Novel Approach for Gypsy Moth Control. Int. J. Mol. Sci. 2017, 18, 2446. [Google Scholar] [CrossRef] [PubMed]
  98. Oberemok, V.V. DNA Markers in the Study of the Relationship Between Nuclear Polyhedrosis Virus and Its Host Lymantria dispar. Ph.D. Thesis, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine, 2011; 22p. [Google Scholar]
  99. Cerio, R.J.; Vandergaast, R.; Friesen, P.D. Host insect inhibitor-of-apoptosis SfIAP functionally replaces baculovirus IAP but is differentially regulated by its N-terminal leader. J. Virol. 2010, 84, 11448–11460. [Google Scholar] [CrossRef] [PubMed]
  100. Manju, M.; Nirosha, V.; Tullika, T.; Mankhanniang, G. DNA insecticides: An emerging tool in pest management. Agriallis 2022, 4, 29–35. [Google Scholar]
  101. Oberemok, V.V.; Skorokhod, O.A. Single-stranded DNA fragments of insect-specific nuclear polyhedrosis virus act as selective DNA insecticides for gypsy moth control. Pestic. Biochem. Physiol. 2014, 113, 1–7. [Google Scholar] [CrossRef] [PubMed]
  102. Oberemok, V.V.; Laikova, K.V.; Zaitsev, A.S.; Gushchin, V.A.; Skorokhod, O.A. The RING for gypsy moth control: Topical application of fragment of its nuclear polyhedrosis virus anti-apoptosis gene as insecticide. Pestic. Biochem. Physiol. 2016, 131, 32–39. [Google Scholar] [CrossRef] [PubMed]
  103. Wang, Y.; Zhang, H.; Li, H.; Miao, X. Second-generation sequencing supply an effective way to screen RNAi targets in large scale for potential application in pest insect control. PLoS ONE 2011, 6, e18644. [Google Scholar] [CrossRef] [PubMed]
  104. Warner, J.R. The economics of ribosome biosynthesis in yeast. Trends Biochem. Sci. 1999, 24, 437–440. [Google Scholar] [CrossRef] [PubMed]
  105. Oberemok, V.V.; Useinov, R.Z.; Skorokhod, O.A.; Gal’chinsky, N.V.; Novikov, I.A.; Makalish, T.P.; Yatskova, E.V.; Sharmagiy, A.K.; Golovkin, I.O.; Gninenko, Y.I.; et al. Oligonucleotide insecticides for green agriculture: Regulatory role of contact DNA in plant- insect interactions. Int. J. Mol. Sci. 2022, 23, 15681. [Google Scholar] [CrossRef] [PubMed]
  106. Useinov, R.Z.; Galchinsky, N.; Yatskova, E.; Novikov, I.; Puzanova, Y.; Trikoz, N.; Sharmagiy, A.; Plugatar, Y.; Laikova, K.; Oberemok, V. To bee or not to bee: Creating DNA insecticides to replace non-selective organophosphate insecticides for use against the soft scale insect Ceroplastes Japonicus green. J. Plant. Prot. Res. 2020, 60, 406–409. [Google Scholar]
  107. Gal’chinsky, N.; Useinov, R.; Yatskova, E.; Laikova, K.; Novikov, I.; Gorlov, M.; Trikoz, N.; Sharmagiy, A.; Plugatar, Y.; Oberemok, V. A breakthrough in the efficiency of contact DNA insecticides: Rapid high mortality rates in the sap-sucking insects Dynaspidiotus britannicus Comstock and Unaspis euonymi Newstead. J. Plant. Prot. Res. 2020, 60, 220–223. [Google Scholar] [CrossRef]
  108. Gal’chinsky, N.V.; Yatskova, E.V.; Novikov, I.A.; Useinov, R.Z.; Kouakou, N.J.; Kouame, K.F.; Kra, K.D.; Sharmagiy, A.K.; Plugatar, Y.V.; Laikova, K.V.; et al. Icerya purchasi Maskell (Hemiptera: Monophlebidae) control using low carbon footprint oligonucleotide insecticides. Int. J. Mol. Sci. 2023, 24, 11650. [Google Scholar] [CrossRef] [PubMed]
  109. Oberemok, V.V.; Gal’chinsky, N.V.; Useinov, R.Z.; Novikov, I.A.; Puzanova, Y.V.; Filatov, R.I.; Kouakou, N.J.; Kouame, K.F.; Kra, K.D.; Laikova, K.V. Four Most Pathogenic Superfamilies of Insect Pests of Suborder Sternorrhyncha: Invisible Superplunderers of Plant Vitality. Insects 2023, 14, 462. [Google Scholar] [CrossRef] [PubMed]
  110. Novikov, A.; Yatskova, E.; Bily, A.; Puzanova, Y.; Sharmagiy, A.; Oberemok, V. Efficient control of the obscure mealybug Pseudococcus viburni with DNA insecticides. In Vitro Cellular & Developmental Biology-Animal; Springer: New York, NY, USA, 2023; pp. 92–108. [Google Scholar]
  111. Puzanova, Y.V.; Novikov, I.A.; Bilyk, A.I.; Sharmagiy, A.K.; Plugatar, Y.V.; Oberemok, V.V. Perfect complementarity mechanism for aphid control: Oligonucleotide insecticide macsan-11 selectively causes high mortality rate for Macrosiphoniella sanborni Gillette. Int. J. Mol. Sci. 2023, 24, 11690. [Google Scholar] [CrossRef] [PubMed]
  112. Oberemok, V.V.; Novikov, I.A.; Yatskova, E.V.; Bilyk, A.I.; Sharmagiy, A.K.; Gal’chinsky, N.V. Potent and selective ‘genetic zipper’ method for DNA-programmable plant protection: Innovative oligonucleotide insecticides against Trioza alacris Flor. Chem. Biol. Technol. Agric. 2024, 11, 144. [Google Scholar] [CrossRef]
  113. Gavrilova, D.; Grizanova, E.; Novikov, I.; Laikova, E.; Zenkova, A.; Oberemok, V.; Dubovskiy, I. Antisense DNA acaricide targeting pre-rRNA of two-spotted spider mite Tetranychus urticae as efficacy-enhancing agent of fungus Metarhizium robertsii. J. Invertebr. Pathol. 2025, 211, 108297. [Google Scholar] [CrossRef] [PubMed]
  114. IZ. Available online: https://en.iz.ru/en/1870413/maria-neduk-denis-gricenko/agrarian-evolution-dna-drug-will-destroy-main-enemy-greenhouse-crops (accessed on 16 April 2025).
  115. Oberemok, V.V.; Laikova, K.V.; Useinov, R.Z.; Gal’chinsky, N.V.; Novikov, I.A.; Gorlov, M.V.; Balykina, E.V.; Trikoz, N.N.; Yatskova, E.V.; Sharmagiy, A.K. High mortality of sap-sucking insects one week after topical application of DNA insecticides. Vitr. Cell. Dev. Biol. Anim. 2020, 56, 31–39. [Google Scholar]
  116. Plugatar, Y.V.; Chichkanova, E.S.; Yatskova, E.V.; Sharmagii, A.K.; Oberemok, V.V. An innovative method of Diaspis echinocacti Bouche control using DNA insecticide on Opuntia ficus-indica (L.) Mill. in the Nikitsky Botanical Garden, Crimea. South Russ. Ecol. Dev. 2021, 16, 119–128. [Google Scholar] [CrossRef]
  117. Oberemok, V.V.; Gal’chinsky, N.V. Oligonucleotide insecticides (contact unmodified antisense DNA biotechnology) and RNA biocontrols (double-stranded RNA technology): Newly born fraternal twins in plant protection. bioRxiv 2024, 3, 584797. [Google Scholar] [CrossRef]
  118. Oberemok, V.; Laikova, K.; Shumskykh, M.; Kenyo, I.; Kasich, I.; Deri, K.; Seidosmanova, E.; Krasnodubets, A.; Bekirova, V.; Gal’chinsky, N. A primary attempt of Leptinotarsa decemlineata control using contact DNA insecticide based on short antisense oligonucleotide of its CYP6B gene. J. Plant Prot. Res. 2018, 58, 106–108. [Google Scholar] [CrossRef]
  119. Du, Q.; Thonberg, H.; Wang, J.; Wahlestedt, C.; Liang, Z.A. A systematic analysis of the silencing effects of an active siRNA at all single-nucleotide mismatched target sites. Nucleic Acids Res. 2005, 33, 1671–1677. [Google Scholar] [CrossRef] [PubMed]
  120. Luige, O.; Karalė, K.; Bose, P.P.; Bollmark, M.; Tedebark, U.; Murtola, M.; Strömberg, R. Influence of sequence variation on the RNA cleavage activity of Zn2+-dimethyl-dppz-PNA-based artificial enzymes. RSC Adv. 2022, 12, 5398–5406. [Google Scholar] [CrossRef] [PubMed]
  121. Ishino, Y.; Shinagawa, H.; Makino, K.; Amemura, M.; Nakata, A. Nucleotide sequence of the iap gene, responsible for alkaline phosphatase isozyme conversion in Escherichia coli, and identification of the gene product. J. Bacteriol. 1987, 169, 5429–5433. [Google Scholar] [CrossRef] [PubMed]
  122. Mojica, M.J.; Juez, G.; Rodríguez-Valera, F. Transcription at different salinities of Haloferax mediterranei sequences adjacent to partially modified PstI sites. Mol. Microbiol. 1993, 9, 613–621. [Google Scholar] [CrossRef] [PubMed]
  123. Jansen, R.; Embden, J.D.A.; Gaastra, W.; Schouls, L.M. Identification of genes that are associated with DNA repeats in prokaryotes. Mol. Microbiol. 2002; 43, 1565–1575. [Google Scholar]
  124. Barrangou, R.; Fremaux, C.; Deveau, H.; Richards, M.; Boyaval, P.; Moineau, S.; Romero, D.A.; Horvath, P. CRISPR provides acquired resistance against viruses in prokaryotes. Science 2007, 315, 1709–1712. [Google Scholar] [CrossRef] [PubMed]
  125. Bolotin, A.; Quinquis, B.; Sorokin, A.; Ehrlich, S.D. Clustered regularly interspaced short palindrome repeats (CRISPRs) have spacers of extrachromosomal origin. Microbiology 2005, 151, 2551–2561. [Google Scholar] [CrossRef] [PubMed]
  126. Makarova, K.S.; Grishin, N.V.; Shabalina, S.A.; Wolf, Y.I.; Koonin, E.V. A putative RNA-interference-based immune system in prokaryotes: Computational analysis of the predicted enzymatic machinery, functional analogies with eukaryotic RNAi, and hypothetical mechanisms of action. Biol. Direct. 2006, 1, 7. [Google Scholar] [CrossRef] [PubMed]
  127. Aljabali, A.A.A.; El-Tanani, M.; Tambuwala, M.M. Principles of CRISPR-Cas9 technology: Advancements in genome editing and emerging trends in drug delivery. J. Drug Deliv. Sci. Technol. 2024, 92, 105338. [Google Scholar] [CrossRef]
  128. Asokan, R.; Rai, A.; Dash, S.; Manamohan, M.; Ashok, K.; Bhargava, C.N.; Wishard, R.; Pradhan, S.K.; Parvathy, M.S. Application of genome editing in entomology. Indian J. Entomol. 2022, 84, 6–103. [Google Scholar]
  129. Khan, M.D.; Ahmad, B.; Ahmed, S.F.; Ijaz, M.; Abdin, Z.U.; Ghafoor, I.; Siddiqui, G.M.; Adrees, T.; Ali, S.; Shahzadi, N.; et al. CRISPR-Cas genome editing in crops: A promising frontier for climate-smart agriculture. Phytopathogenom. Dis. Control. 2023, 2, 71–77. [Google Scholar] [CrossRef]
  130. Wang, J.Y.; Doudna, J.A. CRISPR technology: A decade of genome editing is only the beginning. Science 2023, 379, eadd8643. [Google Scholar] [CrossRef] [PubMed]
  131. Wang, J.Y.; Pausch, P.; Doudna, J.A. Structural biology of CRISPR–Cas immunity and genome editing enzymes. Nat. Rev. Microbiol. 2022, 20, 641–656. [Google Scholar] [CrossRef] [PubMed]
  132. Zhang, F. Development of CRISPR-Cas systems for genome editing and beyond. Q. Rev. Biophys. 2019, 52, e6. [Google Scholar] [CrossRef]
  133. Salum, Y.M.; Yin, A.; Zaheer, U.; Liu, Y.; Guo, Y.; He, W. CRISPR/Cas9-Based Genome Editing of Fall Armyworm (Spodoptera frugiperda): Progress and Prospects. Biomolecules 2024, 14, 1074. [Google Scholar] [CrossRef] [PubMed]
  134. Yan, Y.; Aumann, R.A.; Häcker, I.; Schetelig, M.F. CRISPR-based genetic control strategies for insect pests. J. Integr. Agric. 2023, 22, 651–668. [Google Scholar] [CrossRef]
  135. Ranian, K.; Zahoor, M.K.; Zulhussnain, M.; Ahmad, A. CRISPR/Cas9 mediated sex-ratio distortion by sex specific gene editing in Aedes aegypti. Saudi J. Biol. Sci. 2022, 29, 3015–3022. [Google Scholar] [CrossRef] [PubMed]
  136. Zulhussnain, M.; Zahoor, M.K.; Ranian, K.; Ahmad, A.; Jabeen, F. CRISPR Cas9 mediated knockout of sex determination pathway genes in Aedes aegypti. Bull. Entomol. Res. 2023, 113, 243–252. [Google Scholar] [CrossRef] [PubMed]
  137. Xu, Q.; Wang, M.; Zeng, J.; Sun, H.; Wei, X.; Jiang, H.; Shentu, X.; Sun, D. CRISPR/Cas Technology in Insect Insecticide Resistance. Insects 2025, 16, 345. [Google Scholar] [CrossRef] [PubMed]
  138. Li, J.; Handler, A.M. CRISPR/Cas9-mediated gene editing in an exogenous transgene and an endogenous sex determination gene in the Caribbean fruit fly, Anastrepha suspensa. Gene 2019, 691, 160–166. [Google Scholar] [CrossRef] [PubMed]
  139. Wang, Y.; Chen, X.E.; Liu, Z.; Xu, J.; Li, X.; Bi, H.; Andongma, A.A.; Niu, C.; Huang, Y. Mutation of doublesex induces sex-specific sterility of the diamondback moth Plutella xylostella. Insect Biochem. Mol. Biol. 2019, 112, 103180. [Google Scholar] [CrossRef] [PubMed]
  140. Nourani, L.; Mehrizi, A.A.; Pirahmadi, S.; Pourhashem, Z.; Asadollahi, E.; Jahangiri, B. CRISPR/Cas advancements for genome editing, diagnosis, therapeutics, and vaccine development for Plasmodium parasites, and genetic engineering of Anopheles mosquito vector. Infect. Genet. Evol. 2023, 109, 105419. [Google Scholar] [CrossRef] [PubMed]
  141. Zamanian, M.; Andersen, E.C. Prospects and challenges of CRISPR/Cas genome editing for the study and control of neglected vector-borne nematode diseases. FEBS J. 2016, 283, 3204–3221. [Google Scholar] [CrossRef] [PubMed]
  142. Bimal Kumar Sahoo, B.M.; Mahesh Pathak, M.; Sushruta Boruah, S.; Kasturi Sarmah, K. Bio-engineered defenders: Crispr-cas9 unleashing disease-resistant insects to curb vector-borne diseases. In Futuristic Trends in Agriculture Engineering & Food Sciences; IIP Series: Novi, MI, USA, 2024; Volume 3. [Google Scholar] [CrossRef]
  143. Xu, J.; Xu, X.; Zhan, S.; Huang, Y. Genome editing in insects: Current status and challenges. Natl. Sci. Rev. 2019, 6, 399–401. [Google Scholar] [CrossRef] [PubMed]
  144. Courtier-Orgogozo, V.; Morizot, B.; Boëte, C. Using CRISPR-based gene drive for agriculture pest control. EMBO Rep. 2017, 18, 1481. [Google Scholar] [CrossRef] [PubMed]
  145. Wang, G.H.; Du, J.; Chu, C.Y.; Madhav, M.; Hughes, G.L.; Champer, J. Symbionts and gene drive: Two strategies to combat vector-borne disease. Trends Genet. 2022, 38, 708–723. [Google Scholar] [CrossRef] [PubMed]
  146. Zhao, Y.; Li, L.; Wei, L.; Wang, Y.; Han, Z. Advancements and Future Prospects of CRISPR-Cas-Based Population Replacement Strategies in Insect Pest Management. Insects 2024, 15, 653. [Google Scholar] [CrossRef] [PubMed]
  147. Meccariello, A.; Hou, S.; Davydova, S.; Fawcett, J.D.; Siddall, A.; Leftwich, P.T.; Krsticevic, F.; Papathanos, P.A.; Windbichler, N. Gene drive and genetic sex conversion in the global agricultural pest Ceratitis capitata. Nat. Commun. 2024, 15, 372. [Google Scholar] [CrossRef] [PubMed]
  148. Oye, K.A.; Esvelt, K.; Appleton, E.; Catteruccia, F.; Church, G.; Kuiken, T.; Lightfoot, S.B.Y.; McNamara, J.; Smidler, A.; Collins, J.P. Regulating gene drives. Science 2014, 345, 626–628. [Google Scholar] [CrossRef] [PubMed]
  149. Shukla, J.N.; Palli, S.R. Tribolium castaneum Transformer-2 regulates sex determination and development in both males and females. Insect Biochem. Mol. Biol. 2013, 43, 1125–1132. [Google Scholar] [CrossRef] [PubMed]
  150. Scott, M.J.; Gould, F.; Lorenzen, M.; Grubbs, N.; Edwards, O.; O’Brochta, D. Agricultural production: Assessment of the potential use of Cas9-mediated gene drive systems for agricultural pest control. J. Responsible Innov. 2018, 5 (Suppl. 1), S98–S120. [Google Scholar] [CrossRef]
  151. Tang, Y.H.; Zhang, X.; Dai, Z.C.; Li, H.; Yang, Y.; Zhao, T.J.; Yuan, D.Q.; Qian, W.L.; Cheng, D.J. CRISPR-Cas13-mediated RNA editing in the silkworm Bombyx mori. Zool. Res. 2024, 45, 1249–1260. [Google Scholar] [CrossRef] [PubMed]
  152. Bulle, M.; Sheri, V.; Aileni, M.; Zhang, B. Chloroplast Genome Engineering: A Plausible Approach to Combat Chili Thrips and Other Agronomic Insect Pests of Crops. Plants 2023, 12, 3448. [Google Scholar] [CrossRef] [PubMed]
  153. Han, W.K.; Yang, Y.L.; Si, Y.X.; Wei, Z.Q.; Liu, S.R.; Liu, X.L.; Yan, Q.; Dong, S.L. Involvement of GOBP2 in the perception of a sex pheromone component in both larval and adult Spodoptera litura revealed using CRISPR/Cas9 mutagenesis. Insect Biochem. Mol. Biol. 2022, 141, 103719. [Google Scholar] [CrossRef] [PubMed]
  154. Gantz, V.M.; Jasinskiene, N.; Tatarenkova, O.; Fazekas, A.; Macias, V.M.; Bier, E.; James, A.A. Highly efficient Cas9-mediated gene drive for population modification of the malaria vector mosquito Anopheles stephensi. Proc. Natl. Acad. Sci. USA 2015, 112, E6736–E6743. [Google Scholar] [CrossRef] [PubMed]
  155. Zhu, G.H.; Chereddy, S.C.; Howell, J.L.; Palli, S.R. Genome editing in the fall armyworm, Spodoptera frugiperda: Multiple sgRNA/Cas9 method for identification of knockouts in one generation. Insect Biochem. Mol. Biol. 2020, 122, 103373. [Google Scholar] [CrossRef] [PubMed]
  156. Gu, J.; Wang, J.; Bi, H.; Li, X.; Merchant, A.; Zhang, P.; Zhang, Q.; Zhou, X. CRISPR/Cas9-mediated mutagenesis of sex-specific doublesex splicing variants leads to sterility in Spodoptera frugiperda, a global invasive pest. Cells 2022, 11, 3557. [Google Scholar] [CrossRef] [PubMed]
  157. Wang, C.; Zhao, T.; Liu, X.; Li, T.; He, L.; Wang, Q.; Wang, L.; Zhou, L. CRISPR/Cas9-Mediated Mutagenesis of Antennapedia in Spodoptera frugiperda. Insects 2023, 15, 16. [Google Scholar] [CrossRef] [PubMed]
  158. Anu, C.N.; Ashok, K.; Bhargava, C.N.; Dhawane, Y.; Manamohan, M.; Jha, G.K.; Asokan, R. CRISPR/Cas9 mediated validation of spermatogenesis-related gene, tssk2 as a component of genetic pest management of fall armyworm, Spodoptera frugiperda (JE Smith) (Lepidoptera: Noctuidae). Arch. Insect Biochem. Physiol. 2024, 116, e22121. [Google Scholar] [CrossRef] [PubMed]
  159. Wang, X.; Ma, Y.; Wang, F.; Yang, Y.; Wu, S.; Wu, Y. Disruption of nicotinic acetylcholine receptor α6 mediated by CRISPR/Cas9 confers resistance to spinosyns in Plutella xylostella. Pest Manag. Sci. 2020, 76, 1618–1625. [Google Scholar] [CrossRef] [PubMed]
  160. Xu, X.; Harvey-Samuel, T.; Yang, J.; You, M.; Alphey, L. CRISPR/Cas9-based functional characterization of the pigmentation gene ebony in Plutella xylostella. Insect Mol. Biol. 2021, 30, 615–623. [Google Scholar] [CrossRef] [PubMed]
  161. Nie, H.Y.; Liang, L.Q.; Li, Q.F.; Zhu, Y.N.; Guo, Y.K.; Zheng, Q.L.; Lin, Y.; Yang, D.L.; Li, Z.G.; Su, S.K. CRISPR/Cas9 mediated knockout of Amyellow-y gene results in melanization defect of the cuticle in adult Apis mellifera. J. Insect Physiol. 2021, 132, 104264. [Google Scholar] [CrossRef] [PubMed]
  162. Ahmed, S.; Roy, M.C.; Al Baki, M.A.; Jung, J.K.; Lee, D.; Kim, Y. CRISPR/Cas9 mutagenesis against sex pheromone biosynthesis leads to loss of female attractiveness in Spodoptera exigua, an insect pest. PLoS ONE 2021, 16, e0259322. [Google Scholar] [CrossRef] [PubMed]
  163. Chen, J.X.; Li, W.X.; Lyu, J.; Hu, Y.T.; Huang, G.; Zhang, W.Q. CRISPR/Cas9-mediated knockout of the NlCSAD gene results in darker cuticle pigmentation and a reduction in female fecundity in Nilaparvata lugens (Hemiptera: Delphacidae). Comp. Biochem. Physiol. Part A Mol. Integr. Physiol. 2021, 256, 110921. [Google Scholar] [CrossRef] [PubMed]
  164. Fu, X.; Li, R.; Qiu, Q.; Wang, M.; Zhao, T.; Zhou, L. Study on the function of Helicoverpa armigera Wnt1 gene using CRISPR/Cas9 system. J. Asia-Pac. Entomol. 2022, 25, 101869. [Google Scholar] [CrossRef]
  165. Bi, H.; Merchant, A.; Gu, J.; Li, X.; Zhou, X.; Zhang, Q. CRISPR/Cas9-mediated mutagenesis of abdominal-A and ultrabithorax in the Asian corn borer, Ostrinia furnacalis. Insects 2022, 13, 384. [Google Scholar] [CrossRef] [PubMed]
  166. Konu, M.; Kulmuni, J.; Viljakainen, L. Genetic modification of the ant Lasius niger using CRISPR-Cas9 technology. Insect Mol. Biol. 2023, 32, 11–25. [Google Scholar] [CrossRef] [PubMed]
  167. Bi, H.; Xu, X.; Li, X.; Wang, Y.; Zhou, S.; Huang, Y. CRISPR/Cas9-mediated Serine protease 2 disruption induces male sterility in Spodoptera litura. Front. Physiol. 2022, 13, 931824. [Google Scholar] [CrossRef] [PubMed]
  168. Shirk, B.D.; Shirk, P.D.; Furlong, R.B.; Scully, E.D.; Wu, K.; Siegfried, B.D. Gene editing of the ABC Transporter/White locus using CRISPR/Cas9-mediated mutagenesis in the Indian Meal Moth. J. Insect Physiol. 2023, 145, 104471. [Google Scholar] [CrossRef] [PubMed]
  169. Ashok, K.; Bhargava, C.N.; Babu, K.P.; Rohan, W.; Manamohan, M.; Rai, A.; Sanjay, K.P.; Parvathy, M.S.; Kennedy, J.S.; Asokan, R. First report on CRISPR/Cas9 mediated editing of the eye colour gene, tryptophan 2, 3-dioxygenase in egg plant shoot and fruit borer Leucinodes orbonalis Guenée (Lepidoptera: Crambidae). J. Asia-Pac. Entomol. 2023, 26, 102031. [Google Scholar] [CrossRef]
  170. Ashok, K.; Bhargava, C.N.; Venkatesh, R.; Balasubramani, V.; Murugan, M.; Geethalakshmi, V.; Manamohan, M.; Jha, G.K.; Asokan, R. Molecular characterization and CRISPR/Cas9 validation of the precursor of egg yolk protein gene, vitellogenin of Leucinodes orbonalis Guenée (Lepidoptera: Crambidae). Gene 2025, 933, 148925. [Google Scholar] [CrossRef] [PubMed]
  171. Ashok, K.; Bhargava, C.N.; Asokan, R.; Pradeep, C.; Kennedy, J.S.; Rai, A.; Manamohan, M. First report on the utility of pupal case for early determination of CRISPR/Cas9 ribonucleoprotein mediated genomic edits in the oriental fruit fly, Bactrocera dorsalis (Hendel) (Tephritidae: Diptera). Arch. Insect Biochem. Physiol. 2023, 113, e22024. [Google Scholar] [CrossRef] [PubMed]
  172. Bhargava, C.N.; Ashok, K.; Asokan, R.; Prasad Babu, K.; Parvathy, M.S.; Yogi, D.; Shashikala, T.; Chiranth, R.K.; Ashok, U.; Harsha, C.G. CRISPR/Cas9 Mediated Editing of the white (wh) locus Affects Body Size and Reproduction of the Oriental Fruit Fly, Bactocera dorsalis (Hendel). Agric. Res. 2024, 13, 733–740. [Google Scholar] [CrossRef]
  173. Pradhan, S.K.; Karuppannasamy, A.; Sujatha, P.M.; Nagaraja, B.C.; Narayanappa, A.C.; Chalapathi, P.; Dhawane, Y.; Bynakal, S.; Riegler, M.; Maligeppagol, M.; et al. Embryonic microinjection of ribonucleoprotein complex (Cas9+ sgRNA) of white gene in melon fly, Zeugodacus cucurbitae (Coquillett) (Diptera: Tephritidae) produced white eye phenotype. Arch. Insect Biochem. Physiol. 2023, 114, e22059. [Google Scholar] [CrossRef] [PubMed]
  174. Sujatha, P.M.; Karuppannasamy, A.; Chalapathi, P.; Dhawane, Y.; Narasimhappa, N.S.; Narayanappa, A.C.; Munikrishnappa, V.K.T.; Chikmagalur Nagaraja, B.; Thalooru, S.; Kesavan, S.; et al. CRISPR/Cas9 Ribo nucleoprotein complex-mediated editing of the OBP13 gene affected the response of male Bactrocera dorsalis (Diptera: Tephritidae) to methyl eugenol. J. Appl. Entomol. 2024, 148, 760–771. [Google Scholar] [CrossRef]
  175. Yadav, A.K.; Butler, C.; Yamamoto, A.; Patil, A.A.; Lloyd, A.L.; Scott, M.J. CRISPR/Cas9-based split homing gene drive targeting doublesex for population suppression of the global fruit pest Drosophila suzukii. Proc. Natl. Acad. Sci. USA 2023, 120, e2301525120. [Google Scholar] [CrossRef] [PubMed]
  176. Fang, C.; James, B.; Williams, M.; Bachler, A.; Tay, W.T.; Walsh, T.; Frese, M. Cry1 resistance in a CRISPR/Cas9-mediated HaCad1 gene knockout strain of the Australian cotton bollworm Helicoverpa armigera conferta (Lepidoptera: Noctuidae). Pest Manag. Sci. 2025, 81, 959–965. [Google Scholar] [CrossRef] [PubMed]
  177. Matsuoka, Y.; Nakamura, T.; Watanabe, T.; Barnett, A.A.; Tomonari, S.; Ylla, G.; Whittle, C.A.; Noji, S.; Mito, T.; Extavour, C.G. Establishment of CRISPR/Cas9-based knock-in in a hemimetabolous insect: Targeted gene tagging in the cricket Gryllus bimaculatus. Development 2025, 152, dev199746. [Google Scholar] [CrossRef] [PubMed]
  178. Tao, J.; Bauer, D.E.; Chiarle, R. Assessing and advancing the safety of CRISPR-Cas tools: From DNA to RNA editing. Nat. Commun. 2023, 14, 212. [Google Scholar] [CrossRef] [PubMed]
  179. Richardson, C.; Kelsh, R.N.; J Richardson, R. New advances in CRISPR/Cas-mediated precise gene-editing techniques. Dis. Model. Mech. 2023, 16, dmm049874. [Google Scholar] [CrossRef] [PubMed]
  180. Ahmad, A.; Munawar, N.; Khan, Z.; Qusmani, A.T.; Khan, S.H.; Jamil, A.; Ashraf, S.; Ghouri, M.Z.; Aslam, S.; Mubarik, M.S.; et al. An Outlook on Global Regulatory Landscape for Genome-Edited Crops. Int. J. Mol. Sci. 2021, 22, 11753. [Google Scholar] [CrossRef] [PubMed]
  181. Asad, M.; Chang, Y.; Liao, J.; Yang, G. CRISPR/Cas9 Genome Editing in the Diamondback Moth: Current Progress, Challenges, and Prospects. Int. J. Mol. Sci. 2025, 26, 1515. [Google Scholar] [CrossRef] [PubMed]
  182. Komal, J.; Desai, H.R.; Samal, I.; Mastinu, A.; Patel, R.D.; Kumar, P.V.D.; Majhi, P.K.; Mahanta, D.K.; Bhoi, T.K. Unveiling the Genetic Symphony: Harnessing CRISPR-Cas Genome Editing for Effective Insect Pest Management. Plants 2023, 12, 3961. [Google Scholar] [CrossRef] [PubMed]
  183. Moon, T.T.; Maliha, I.J.; Khan, A.A.M.; Chakraborty, M.; Uddin, M.S.; Amin, M.R.; Islam, T. CRISPR-Cas Genome Editing for Insect Pest Stress Management in Crop Plants. Stresses 2022, 2, 493–514. [Google Scholar] [CrossRef]
Figure 1. Antisense technologies based on unmodified nucleic acids and used for insect pest control: guide nucleic acid (RNA or DNA) forms duplex with target nucleic acid (RNA or DNA) and the target nucleic acid is cleaved by specific nuclease.
Figure 1. Antisense technologies based on unmodified nucleic acids and used for insect pest control: guide nucleic acid (RNA or DNA) forms duplex with target nucleic acid (RNA or DNA) and the target nucleic acid is cleaved by specific nuclease.
Insects 16 00746 g001
Figure 2. Main route of RNAi used for creation of dsRNA insecticides (exogenous siRNA pathway): 1–dsRNA cleavage by Dicer; 2–silencing complex formation and silencing complex activation; 3–target mRNA degradation by Ago.
Figure 2. Main route of RNAi used for creation of dsRNA insecticides (exogenous siRNA pathway): 1–dsRNA cleavage by Dicer; 2–silencing complex formation and silencing complex activation; 3–target mRNA degradation by Ago.
Insects 16 00746 g002
Figure 3. DNA containment (DNAc) is the main mechanism of action of oligonucleotide insecticides (olinscides): 1—antisense DNA sequences complementary to rRNA and/or pre-rRNA of pests are used as oligonucleotide insecticides; 2—formation of duplex between DNA (oligonucleotide insecticide) and RNA (rRNA and/or pre-rRNA) of the pests; 3—at first step of DNAc, antisense DNA oligonucleotide (oligonucleotide insecticide) ‘arrests’ target rRNA and/or pre-rRNA followed by its hypercompensation and interferes with normal functioning and biogenesis of ribosomes (‘arrested’ ribosomes); 4—at second step of DNAc, rRNase cleaves target rRNA and/or pre-rRNA and substantial decrease in its concentration occurs.
Figure 3. DNA containment (DNAc) is the main mechanism of action of oligonucleotide insecticides (olinscides): 1—antisense DNA sequences complementary to rRNA and/or pre-rRNA of pests are used as oligonucleotide insecticides; 2—formation of duplex between DNA (oligonucleotide insecticide) and RNA (rRNA and/or pre-rRNA) of the pests; 3—at first step of DNAc, antisense DNA oligonucleotide (oligonucleotide insecticide) ‘arrests’ target rRNA and/or pre-rRNA followed by its hypercompensation and interferes with normal functioning and biogenesis of ribosomes (‘arrested’ ribosomes); 4—at second step of DNAc, rRNase cleaves target rRNA and/or pre-rRNA and substantial decrease in its concentration occurs.
Insects 16 00746 g003
Figure 4. CRISPR/Cas9 system used for insect pest control: 1—sgRNA-Cas9 complex formation; 2—formation of RNA-DNA duplex with target DNA; 3—cleavage of target double-stranded DNA by Cas9; 4—DNA repair through non-homologous end joining (NHEJ) or homologous-dependent repair (HDR); * crRNA—CRISPR RNA; tracrRNA—trans-activating crRNA.
Figure 4. CRISPR/Cas9 system used for insect pest control: 1—sgRNA-Cas9 complex formation; 2—formation of RNA-DNA duplex with target DNA; 3—cleavage of target double-stranded DNA by Cas9; 4—DNA repair through non-homologous end joining (NHEJ) or homologous-dependent repair (HDR); * crRNA—CRISPR RNA; tracrRNA—trans-activating crRNA.
Insects 16 00746 g004
Table 1. List of pest species successfully targeted by RNAi.
Table 1. List of pest species successfully targeted by RNAi.
Sl. No.Names of Model InsectsTargeted Gene(s)Affected ProcessesReferences
1Beet armyworm, Spodoptera exigua (Lepidoptera)Chitin synthase gene AChitin synthesis[40]
2Brown planthopper, Nilaparvata lugens (Hemiptera)NlHT1, Nlcar, Nltry
NlTPS
Digestive system
Enzymatic activity
[41,42]
3African sweet potato weevil, Cylas puncticollis (Coleoptera)Snf7Digestive system[43]
4Tomato pinworm, Tuta absoluta (Lepidoptera)Vacuolar ATPase-A and Arginine kinaseHigh mortality[44]
5Oriental fruit fly, Bactrocera dorsalis (Diptera)α-SpectrinOviposition and ovary size[45]
6Cotton mealybug, Phenacoccus solenopsis (Hemiptera)Krüppel homologue-1, ADP-ATP/Translocase, IDGF-1
Bursicon, V-ATPase
Not specified
Cuticle hardening and V-ATPases act as proton pumps
[46,47]
7Diamondback moth, Plutella xylostella (Lepidoptera)PxChtChitin synthesis[48]
8Fall armyworm, Spodoptera frugiperda (Lepidoptera)Met, EcR, USP
COPIα, COPIβ, GSTU1
Reproductive system,
fertility
Insect reproduction
[49,50]
9White-backed planthopper, Sogatella furcifera (Hemiptera)hsc70-3, PP-αInsect metamorphosis[51]
10Soybean aphid, Aphis glycines (Hemiptera)Cytochrome P450 monooxygenases (CYP450s)Insect resistance[52]
11Asian citrus psyllid, Diaphorina citri (Hemiptera)CHC, vATPase-A, Snf7Transmembrane system[53]
12Trichogramma dendrolimi (Lepidoptera)Vitellogenin receptor (VgR)Female reproductive system[54]
13Domestic silk moth, Bombyx mori (Lepidoptera)BmToll9-1, BmToll9-2, PGRP-L4Toll and immune deficiency signaling pathways[55,56,57,58]
14Silverleaf whitefly, Bemisia tabaci (Hemiptera)Cysteine proteaseDigestive system[59]
15Cowpea weevil, Callosobruchus maculatus (Coleoptera)Olfactory receptor coreceptor (Cmac\Orco)Insect sensory system[60]
16White-backed planthopper, S. furcifera (Hemiptera)β-N-acetylhexosaminidase genesInsect metamorphosis[61]
17Desert locust, Schistocerca gregaria (Orthoptera)Cytochrome P450Ecdysteroid pathway[62]
18Red flour beetle, Tribolium castaneum (Coleoptera)CPAPsCuticular proteins[63]
19Chinese white pine beetle, Dendroctonus armandi (Coleoptera)AquaporinOsmoregulation[64]
20Kissing bug, Rhodnius prolixus (Hemiptera)Nitrophorin 2 (NP2)Anticoagulant and apyrase activities in saliva[65]
21Citrus aphid, Toxoptera citricida (Hemiptera)TCiCHSChitin synthesis[66]
22Potato psyllid, Bactericera cockerelli (Hemiptera)SUC1, ST4Osmoregulatory[67]
Table 2. List of pest species successfully targeted by CUADb.
Table 2. List of pest species successfully targeted by CUADb.
Sl. No.Names of Model InsectsTargeted Gene(s)Affected ProcessesReferences
1Euonymous scale, U. euonymi (Hemiptera)28S rRNAProtein biosynthesis [107,115]
2Holly scale, D. britannicus (Hemiptera)28S rRNAProtein biosynthesis[5,107]
3Japanese wax scale, C. japonicus (Hemiptera)28S rRNAProtein biosynthesis[106]
4Cactus scale, Diaspis echinocacti (Hemiptera)28S rRNAProtein biosynthesis[116]
5Bay sucker, T. alacris (Hemiptera)ITS2 of pre-rRNA and 28S rRNAProtein biosynthesis[112]
6Cottony cushion scale, I. purchasi (Hemiptera)28S rRNAProtein biosynthesis[108]
7Chrysanthemum aphid, M. sanborni (Hemiptera)ITS2 of pre-rRNAProtein biosynthesis[111]
8Mealybug, P. viburni (Hemiptera)5.8S, 18S and 28S rRNAProtein biosynthesis[110]
9Laureal scale, A. lauri (Hemiptera)28S rRNAProtein biosynthesis[5]
10Soft scale, C. hesperidum (Hemiptera)28S rRNAProtein biosynthesis[105]
11Two-spotted spider mite, T. urticae (Trombidiformes)ITS2 of pre-rRNAProtein biosynthesis[113]
12Grey pine aphid, S. pineti (Hemiptera)ITS2 of pre-rRNAProtein biosynthesis[13]
13Large pine aphid,
Cinara pinea (Hemiptera)
ITS2 of pre-rRNAProtein biosynthesis[13]
14Pine needle aphid, Eulachnus rileyi (Hemiptera)ITS2 of pre-rRNAProtein biosynthesis[13]
Table 3. List of insect species successfully targeted by CRISPR/Cas system.
Table 3. List of insect species successfully targeted by CRISPR/Cas system.
Sl. No.Names of Model InsectsTarget Gene(s)Affected ProcessesReference
1Mosquito, Anopheles stephensi (Diptera)Kynurenine hydroxylaseParasite-resistance[154]
2Fall armyworm, S. frugiperda (Lepidoptera)Ebony
Doublesex (dsx) (Sfdsx)
Antennapedia (Antp)
Spermatogenesis-related, tssk2
Melanin biosynthesis
Sex differentiation
Insect thorax and wing development
Male reproductive system
[155,156,157,158]
3Diamondback moth, P. xylostella (Lepidoptera)Yellow
Ebony
LW-opsin
Body pigmentation
Body pigmentation
Efficiency of phototaxis
[46,159,160]
4European bee, Apis mellifera (Hymenoptera)Amyellow-yMelanization in cuticle[161]
5Beet armyworm, S. exigua (Lepidoptera)Desaturase (SexiDES5)Sex pheromone biosynthesis[162]
6Brown planthopper, N. lugens (Hemiptera)Cysteine sulfinic acid decarboxylase (CSAD)Melanin metabolism[163]
7Chickpea pod borer, H. armigera (Lepidoptera)Wnt1Segmentation, appendage development, and pigmentation[164]
8Asian corn borer, Ostrinia furnacalis (Lepidoptera)Abdominal-A (Abd-A) and Ultrabithorax (Ubx)Anatomical structure formation[165]
9Black garden ant, Lasius niger (Hymenoptera)CinnabarEye pigmentation[166]
10Common cutworm, S. litura (Lepidoptera)Serine protease 2
Odorant-binding proteins
Male sterility
Perception of a sex pheromone
[153,167]
11Indian meal moth, Plodia interpunctella (Lepidoptera)ATP binding cassette (ABC) proteinsEye pigmentation[168]
12Eggplant shoot and fruit borer, Leucinodes orbonalis (Lepidoptera)Tryptophan 2, 3-dioxygenase
Vitellogenin (Vg)
Eye pigmentation
Female reproductive system
[169,170]
13Mango fruit fly, B. dorsalis (Diptera)White
White locus
OBP13
Eye pigmentation
Eye pigmentation
Methyl eugenol
[171,172,173,174]
14Pomace fly, Drosophila suzukii (Diptera)DoublesexPopulation suppression[175]
15Australian cotton bollworm, H. armigera conferta (Lepidoptera)CadherinCry1Ac resistance[176]
16Cricket, Gryllus bimaculatus (Orthoptera)Laccase 2 (Gb-lac2)Cuticle system pigmentation[177]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kumar, H.; Gal’chinsky, N.; Sweta, V.; Negi, N.; Filatov, R.; Chandel, A.; Ali, J.; Oberemok, V.; Laikova, K. Perspectives of RNAi, CUADb and CRISPR/Cas as Innovative Antisense Technologies for Insect Pest Control: From Discovery to Practice. Insects 2025, 16, 746. https://doi.org/10.3390/insects16070746

AMA Style

Kumar H, Gal’chinsky N, Sweta V, Negi N, Filatov R, Chandel A, Ali J, Oberemok V, Laikova K. Perspectives of RNAi, CUADb and CRISPR/Cas as Innovative Antisense Technologies for Insect Pest Control: From Discovery to Practice. Insects. 2025; 16(7):746. https://doi.org/10.3390/insects16070746

Chicago/Turabian Style

Kumar, Hemant, Nikita Gal’chinsky, Verma Sweta, Nikita Negi, Roman Filatov, Anamika Chandel, Jamin Ali, Vol Oberemok, and Kate Laikova. 2025. "Perspectives of RNAi, CUADb and CRISPR/Cas as Innovative Antisense Technologies for Insect Pest Control: From Discovery to Practice" Insects 16, no. 7: 746. https://doi.org/10.3390/insects16070746

APA Style

Kumar, H., Gal’chinsky, N., Sweta, V., Negi, N., Filatov, R., Chandel, A., Ali, J., Oberemok, V., & Laikova, K. (2025). Perspectives of RNAi, CUADb and CRISPR/Cas as Innovative Antisense Technologies for Insect Pest Control: From Discovery to Practice. Insects, 16(7), 746. https://doi.org/10.3390/insects16070746

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop