We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
ReportsOpen Accesscc iconby iconnc iconnd icon

Comparing robotic and manual injection methods in zebrafish embryos for high-throughput RNA silencing using CRISPR-RfxCas13d

    Joaquin Abugattas-Nuñez Del Prado‡

    Institute of Biology Leiden, Animal Science & Health, Leiden University, Einsteinweg 55, Leiden, 2333CC, The Netherlands

    Department of Biology, Universidad Peruana Cayetano Heredia, Av. Honorio Delgado 430, Lima, 15102, Perú

    ,
    Yi Ding‡

    Life Science Methods BV, JH Oortweg 19, Leiden, 2333CH, The Netherlands

    ,
    Jan de Sonneville

    Life Science Methods BV, JH Oortweg 19, Leiden, 2333CH, The Netherlands

    ,
    Kees-Jan van der Kolk

    Life Science Methods BV, JH Oortweg 19, Leiden, 2333CH, The Netherlands

    ,
    Miguel A Moreno-Mateos

    Andalusian Center for Developmental Biology (CABD), Pablo de Olavide University/CSIC/Junta de Andalucía, Ctra. Utrera Km.1, Seville, 41013, Spain

    Department of Molecular Biology & Biochemical Engineering, Pablo de Olavide University, Ctra. Utrera Km.1, Seville, 41013, Spain

    ,
    Edward Málaga-Trillo

    Department of Biology, Universidad Peruana Cayetano Heredia, Av. Honorio Delgado 430, Lima, 15102, Perú

    &
    Herman P Spaink

    *Author for correspondence:

    E-mail Address: h.p.spaink@biology.leidenuniv.nl

    Institute of Biology Leiden, Animal Science & Health, Leiden University, Einsteinweg 55, Leiden, 2333CC, The Netherlands

    Published Online:https://doi.org/10.2144/btn-2023-0062

    Abstract

    In this study, the authors compared the efficiency of automated robotic and manual injection methods for the CRISPR-RfxCas13d (CasRx) system for mRNA knockdown and Cas9-mediated DNA targeting in zebrafish embryos. They targeted the no tail (TBXTA) gene as a proof-of-principle, evaluating the induced embryonic phenotypes. Both Cas9 and CasRx systems caused loss of function phenotypes for TBXTA. Cas9 protein exhibited a higher percentage of severe phenotypes compared with mRNA, while CasRx protein and mRNA showed similar efficiency. Both robotic and manual injections demonstrated comparable phenotype percentages and mortality rates. The findings highlight the potential of RNA-targeting CRISPR effectors for precise gene knockdown and endorse automated microinjection at a speed of 1.0 s per embryo as a high-throughput alternative to manual methods.

    METHOD SUMMARY

    The authors compared the efficiency of automated robotic and manual injection methods for the CRISPR-RfxCas13d system for mRNA knockdown and Cas9-mediated DNA targeting in zebrafish embryos. Both robotic and manual injections demonstrated comparable phenotype percentages and mortality rates. The findings show that automated microinjection is highly suitable as a high-throughput alternative to manual methods of gene silencing in zebrafish embryos.

    The zebrafish is a vertebrate model organism with many experimental advantages, such as large progeny, embryo transparency and amenability to genetic manipulation by microinjection. However, microinjection of the small zebrafish embryos is laborious and time-consuming. Therefore, better high-throughput methods for the rapid microinjection of zebrafish embryos would significantly facilitate studies of gene function, pharmacological screens and disease models.

    The authors' laboratory has shown that a robotic system allows for high-throughput injections of CRISPR-Cas9 and DNA constructs into zebrafish zygotes with an efficiency comparable to that of manual injections [1]. Moreover, similar types of embryo injection robots were efficiently used for zebrafish tuberculosis infection studies and compound screens [2–5].

    In addition to DNA modifications via CRISPR-Cas9 to generate F0 knockouts [6,7], robotic injection also offers the possibility of generating and screening phenotypes resulting from various RNA depletion methods. For instance, RNA interference technologies and morpholinos offer an approach to RNA knockdown in several organisms. However, RNA interference technologies have failed to be established in a zebrafish model [8,9] and morpholinos have shown toxicity, off-target effects, innate immune responses and discordant phenotypes with mutant animals [10–13].

    Furthermore, CRISPR-Cas systems were first described as bacterial adaptive immune systems against phages [14–19]. They are organized in two main classes: class 1 with Cas effectors composed of multiple subunits and class 2 with monomeric large proteins; on top of that, specific subtypes within classes depend on the Cas endonuclease and its mechanism of action (Table 1). From class 2, Cas9 and Cas12 are used for many genomic engineering applications [20–24] and have been applied in the zebrafish model [6,25–27]. Moreover, the recently discovered class 2 type VI Cas13 systems are RNA-guided ribonucleases applied for transcriptomic engineering (Cas13a–Cas13d) [28–33]; in particular, the CRISPR-RfxCas13d (CasRx) system induces efficient knockdown of both zygotically and maternally provided RNAs in animal embryos [34].

    Table 1. Classification of different CRISPR-Cas systems by classes and types known alongside with their number of subtypes, different Cas endonucleases and their target molecule.
    ClassTypeSubtypes (n)Cas endonucleaseTarget
    Class 1Type I7Cas3DNA
    Type III2Cas10DNA/RNA
    Type IV2
    Class 2Type II3Cas9DNA
    Type V7Cas12DNA
    Type VI3Cas13RNA

    “–” indicates that the mechanism of action and biological function are yet not confirmed for this type IV system.

    Data taken from [33].

    The authors used automation to quantitatively evaluate the relative efficiencies of CRISPR-CasRx and CRISPR-Cas9 to inactivate the function of the TBXTA gene at the RNA or DNA level, respectively, and generate the resulting no-tail phenotype [35]. They show that high-speed robotic injection of the CRISPR-CasRx achieves survival rates and knockdown efficiencies comparable with manual injections, thus opening the possibility of high-throughput interrogation of the transcriptome in a tunable manner.

    Materials & methods

    Zebrafish maintenance & embryo production

    Leiden University researchers follow the European rules for the use of animals. Zebrafish wild-type strains ABTL are maintained and bred according to standard conditions [36]. One-cell stage ABTL embryos were used in this study.

    CasRx TBXTA gRNA production

    The gRNAs for CasRx targeting TBXTA [34,37] gene were produced by fill-in polymerase chain reaction (PCR) with Phire HotStart II DNA polymerase (Thermo Fisher Scientific, F122S; primers in Supplementary Table 1, Integrated DNA Technologies (IDT) in a thermal cycler (BioRad, T100) with 10 uM final concentration of each primer and the following program: 3 min at 95°C, 35 cycles of 30 s at 95°C, 30 s at 60°C and 30 s at 72°C, and a final step at 72°C for 5 min. The PCR product was in vitro transcribed (Epicentre, ASF3507) under the control of promoter T7 for 12 h at 37°C and DNAse treated with TURBO-DNAse for 20 min at 37°C. Finally, the gRNA was precipitated with 3 M ammonium acetate and resuspended in nuclease-free water. gRNA integrity was verified with electrophoresis (Supplementary Figure 3), quantified by Nanodrop 2000 (Thermo Fisher Scientific) and titrated until high phenotypical penetrance was obtained. The authors observe that quantifying gRNAs after in vitro transcription can yield inaccuracies, which may lead to an overestimation of the amount of gRNAs being utilized. However, they mitigated this issue by titration of the gRNA employed.

    Cas9 & CasRx mRNA production

    Plasmids containing Cas9 and CasRx genes, pT3Ts-nCas9n (Addgene 46757) and pT3Ts-RfxCas13d-HA (Addgene 141320), respectively, were linearized with XbaI (NEB R0145L) for 3 h at 37°C and then in vitro transcribed under control of promoter T3 (Ambion AM1348) for 3 h at 37°C and DNAse treated with TURBO-DNAse for 20 min at 37°C. Capped mRNA from these reactions was purified with RNAeasy MiniKit (Qiagen 74104). RNA integrity was verified by electrophoresis (Supplementary Figures 1 & 2) and quantified with Nanodrop 2000 (Thermo Fisher Scientific).

    CasRx protein expression & purification

    Competent Rosetta DE3 pLysS (Novagen 70956) was transformed with the plasmid pET28b-RfxCas13d-His (Addgene 141322), which contains the recombinant CasRx gene. Cells were grown for 3 h at 37°C and then induced with 0.1 mM isopropyl β-D-1-thiogalactopyranoside for another 3 h. Cells were washed once with 20 mM Tris-HCl pH 7.5 and the pellet was frozen at -80°C before the next steps.

    Pellets were resuspended with lysis buffer (50 mM HEPES*KOH pH 7.5, 500 mM KCl, 10% v/v glycerol, 1 mM dithiothreitol and 10 mM imidazole) and sonicated (QSonica, Q125) using the following program: 3 s on, 10 s off, 120 cycles, 30% amplitude. Lysate was centrifuged and then filtered using a 0.2 μm cellulose acetate filter. CasRx protein was then purified using a HisPur™ Ni-NTA spin column (Thermo Fisher Scientific, 88226) and washed with 20 column volumes of lysis buffer by gravity. Elution was performed in five steps with elution buffer (50 mM HEPES*KOH pH 7.5, 500 mM KCl, 10% v/v glycerol, 1 mM dithiothreitol and 500 mM imidazole) diluted with lysis buffer to get increasing concentrations of imidazole (10 mM, 30 mM, 50 mM, 100 mM and 200 mM). Fractions containing purified CasRx were pooled and then concentrated and dialyzed with dialysis buffer (50 mM HEPES-KOH pH 7.5, 250 mM KCl, 1 mM dithiothreitol and 10% glycerol) using a 50K Amicon Ultra-15 (Millipore UFC905024) until the concentration of 3 µg/ml was reached. Protein concentration was estimated with Nanodrop 2000 (Thermo Fisher Scientific) and extraction quality was verified by SDS-PAGE (Supplementary Figure 4). Single-use aliquots of 4 μl were stored at -80°C.

    Cas9 protein & gRNAs

    Alt-R™ – Cas9 protein, Alt-R™ crRNA and Alt-R™ tracrRNA were purchased from IDT. The same gRNAs (Supplementary Table 1) were used in both mRNA and protein injections of Cas9. Alt-R CRISPR systems from IDT are optimized genome editing tools with predesigned gRNAs, to produce on-target double-strand breaks using high-purity solutions.

    Production of mRNAs, gRNA & protein

    CasRx and Cas9 (see methods) showed a high rate of capped mRNA production from linearized DNA template (∼100-fold ng of RNA α ng of DNA, data not shown), with little degradation after purifying, as shown in the agarose gels (Supplementary Figures 1 & 2). Similarly, the gRNA generation showed higher efficiency (∼800-fold ng of RNA α ng of DNA, data not shown), having a reaction time four-times longer than mRNA reaction and with low degradation (Supplementary Figure 3). The CasRx protein production was induced and purified with a Ni-NTA column followed by concentration with an Amicon ultracentrifugation column. The authors obtained a concentration of 3 μg/ml with a purity of approximately 85% (Supplementary Figure 4).

    Microinjection solutions & injection quantities

    Cas9 gRNAs were prepared by annealing crRNA with tracrRNA using duplex buffer IDT and incubating at 95°C for 5 min. Three different gRNAs were pooled and then mixed with Cas9 mRNA. The final quantity injected with 1 nl per embryo was 200 pg of Cas9 mRNA and 1070 pg of gRNA (357 pg each). Control groups were only injected with 200 pg Cas9 mRNA. To produce the Cas9 ribonucleoprotein, the same gRNAs were mixed with Cas9 protein IDT and incubated at 37°C for 5 min. The final quantity injected with 1 nl per embryo was 5029 pg of Alt-R Cas9 protein and 1070 pg of gRNA (357 pg each). Control groups were only injected with 5029 pg Cas9 protein. In the case of CasRx, the mRNA was mixed with the gRNA targeting TBXTA transcript. The final quantity injected with 1 nl per embryo was 200 pg of mRNA and 300 pg of gRNA. Control groups were only injected with 200 pg CasRx mRNA. CasRx protein was mixed with gRNA targeting TBXTA transcript. The final quantity injected with 1 nl per embryo was 3 ng of protein with 300 pg of gRNA. Control groups were only injected with 3 ng CasRx protein.

    Manual microinjection

    Manual microinjection was performed using a Pneumatic Pico-Pump (PV 820, WPI) and all embryos were injected into the yolk during one-cell stage with ~5–8 psi depending on the needle pore size. Commercial needles with the tip size within 6–7 μm (Clunbury Scientific LLC) were used. All experiments were performed with at least three replicates.

    Automated robot microinjection & software updates

    The detailed procedure of using the upgraded automated microinjection system (Life Science Methods BV, The Netherlands) is described in Figure 1. The same type of commercial needles from manual injection was used for robotic injection. Some software updates were carried out to improve the automated injection throughput and efficiency. A deep learning network was updated to recognize the yolk center and the first cell interface. Another deep learning network was added to recognize the shape of the droplet and compute to volume. The speed increased from 1.8 s per injection in the previous version to 1.0 s per injection on average in the current system.

    Figure 1. Overview of the zebrafish egg injection robot and experimental outline.

    (A) A representative image of the integrated egg injector. The needle holder is used to load needles and the motorized stage enables the movement of plates containing embryos during injections. A 96-well plate is for droplet calibration and the agarose gel grid plate is for the placement of zebrafish eggs. A touch screen allows for the operation and visualization of all the injection processes. (B–H) The procedure of zebrafish embryo injection using the robot. (B) Step1: preparation of an agarose grid plate using a mold. Different molds (e.g., 9 × 100, 1024, 360 grid wells) can be used based on research purposes. (C) Step 2: placement of zebrafish eggs on the agarose grid. Zebrafish embryos can be placed randomly at any wells of the grid, as empty wells can be recognized and skipped by the robot. (D) Step 3: needle calibration for both xy direction and z direction, as shown on the screenshots from the touch screen. (E) Step 4: droplet calibration is performed by injection of suspension into mineral oil and droplet size can be determined automatically by the robot. (F) Step 5: injection positions should be chosen, either yolk center or close to the first cell; subsequently, the robot can do injections automatically for wells that users have selected. Different injection distances (e.g., 0 μm, 30 μm and 50 μm) can be chosen for close to the first cell injection. (G) Step 6: noninjected eggs can be killed by loading a blunt needle. (H) Step 7: after the injection is finished, the injected zebrafish eggs can be transferred to petri dishes for culture and dead eggs should be removed.

    Results & discussion

    Establishment of robotic injection procedures

    In this study, the authors developed and upgraded a robot based on previous versions [1,3,5]. The new version of the robot is a one-box fully integrated device with touch screen and provides optimal injection speed into early-stage zebrafish embryos (Figure 1A). An agarose grid plate is prepared using a 9 × 100 well mold (Figure 1B & Supplementary Video 1). Zebrafish embryos can be placed in each grid well (Figure 1C & Supplementary Video 2). It takes approximately 2 min to position the eggs within the 9 × 100 grid wells. Subsequently, a needle is mounted into the needle holder, and needle tip (xy direction) and height (z direction) are calibrated (Figure 1D & Supplementary Video 3). For droplet calibration, a machine learning algorithm is added to recognize the shape of the droplet in a well filled with mineral oil and the volume of the droplet is given automatically (Figure 1E & Supplementary Video 4). Injections can be chosen to be either into the middle of the yolk (Figure 1F& Supplementary Video 5) or close to the first cell (Figure 1F & Supplementary Video 6). Statistics on the top left of the screen give an overview of the number of embryos in each category during injection (Figure 1F & Supplementary Videos 5 & 6). Eggs where a first cell cannot be recognized (such as “no-cell” type) can be chosen to be injected or skipped. The skipped embryos are registered, and these eggs can be killed using a blunt needle (Figure 1G & Supplementary Video 7). After the killing is finished, the embryos can be easily transferred to petri dishes for culture (Figure 1H & Supplementary Video 8). The count of the destroyed eggs can be verified, and this count should match the total number of eggs that were not injected, displayed at the top left corner of the screen.

    In this study, the authors presented a detailed procedure for utilizing an advanced zebrafish embryo injection robot and conducted a comparative analysis of injection outcomes between robotic and manual methods. The robotic system offered several advantages. The implementation of this robot allows individuals without any prior zebrafish egg injection experience to perform large-scale injections effortlessly. Additionally, the inclusion of a user-friendly touch screen interface enhances the operational convenience and provides a visually appealing display of the injection processes, ensuring a comfortable experience for the operator. Moreover, the robot offers the advantage of precise injection volumes by automatically calculating droplet sizes using two-dimensional image analysis. This automated computation significantly reduces errors associated with manual estimation, minimizing the potential translation of such errors into inaccuracies in injected volume. Finally, the robotic system incorporates a feature whereby it records the positions of noninjected eggs and subsequently performs selective euthanasia, ensuring that only the injected embryos remain alive for further analysis.

    The results indicated that both the robotic and manual injection approaches yielded similar percentages of phenotypes and mortality rates, specifically regarding the knockdown of the TBXTA gene. However, it is noteworthy that the robotic method exhibited superior speed, being three-times faster than manual injections. This improvement in speed is attributed to the expanded region of interest, which allows for simultaneous evaluation of the next well during the ongoing injection process. On average, the robot achieved an injection speed of 1.0 s per injection, as demonstrated in Supplementary Videos 5 & 6.

    Comparison of manual & automated robotic injection

    To compare injection results between manual and robotic injections, both CRISPR-Cas9 and CRISPR-CasRx systems targeting the TBXTA gene were used. The Cas9 gRNAs achieved more than 90% of F0 biallelic knockouts, which were validated by DNA sequencing [6]. Additionally, the CasRx gRNA used to reproduce this phenotype was previously validated by reverse transcription–quantitative PCR, resulting in an average transcript level decrease of 60% [34]. Knockdown of the TBXTA gene using both systems resulted in different levels of tail and notochord defect phenotypes at 30 hpi (Figure 2A). Moreover, the knockdown efficiency of both Cas9 and CasRx when administered through a robot was very similar to that achieved using a manual injector (Figure 2B & C). Embryos graded with phenotype 1 were more frequent with CasRx treatment. A higher percentage of embryos with severe phenotype (type III) and a lower percentage with mild phenotypes (type I and type II) were observed in Cas9 protein than in Cas9 mRNA groups (Figure 2B). However, there were no clear differences in the percentages of embryos displaying different levels of phenotypes between the groups treated with CasRx protein and those treated with CasRx mRNA (Figure 2C). Since in previous papers it has been shown that the position of the injection in the zebrafish embryos results in different penetrance in gene silencing technologies, we compared the effect of the position of injection in the embryo. Injection into the cell interface and yolk center of zebrafish embryos showed no difference in the phenotype (Figure 2B & C). The mortality rate of injected embryos was similar between manual and robotic injection (Table 2). However, the speed of the robot (59 ± 3 embryos/min, 2169 injected embryos) was three-times faster compared with the manual injections (20 ± 3 embryos/min, 990 injected embryos; Table 2).

    Figure 2. Percentage of zebrafish embryos with different grades of no tail phenotype caused by TBXTA gene knockdown.

    (A) Knockdown of the TBXTA gene using both Cas9 and RfxCas13d (CasRx) systems results in different levels of no tail phenotype. Representative phenotype images are acquired from CasRx injections. Scale bar = 485.6 μm. (B) Percentage of embryos of each grade of no tail phenotype caused by Cas9 protein or mRNA targeting TBXTA gene using automatic robot or manual injections. As all the control groups injected either with Cas9 protein or mRNA only show the wild-type phenotype, the authors combined all the control groups to one bar, namely “All controls” (Cas9 protein robot, n = 434; Cas9 protein manual, n = 213; Cas9 mRNA robot, n = 447; Cas9 mRNA manual, n = 203). (C) Percentage of embryos of each grade of no tail phenotype caused by CasRx protein or mRNA targeting TBXTA gene using automatic robot or manual injections. As all the control groups injected with either CasRx protein or mRNA only showed the wild-type phenotype, the authors combined all the control groups to one bar, namely “All controls” (CasRx protein robot, n = 334; Cas13d protein manual, n = 173; CasRx mRNA robot, n = 432; CasRx mRNA manual, n = 249). Type I: Short tail and normal notochord; type II: Notochord development failure and short tail; type III: Notochord development failure and shorter tail; WT: Normal tail and notochord.

    Table 2. Different characteristics of manual and robot injection methods.
     ManualRobotNoninjected
    Dead rate 6 hpi3.9 ± 0.83.3 ± 1.11.0 ± 1.1
    Dead rate 30 hpi4.3 ± 1.64.7 ± 1.32.2 ± 1.6
    Dead rate overall4.1 ± 1.34.0 ± 1.41.6 ± 1.5
    Injection speed (embryos/min)19.7 ± 3.158.9 ± 2.6Not applicable
    Total embryos9902169710

    Mortality rate 6 hpi 30 hpi are expressed in percentage ± standard deviation. Injection speed is expressed in injected embryos per minute ± standard deviation. Total embryos is the number of injected or noninjected embryos. Noninjected embryos belong to the same batch of embryos used for both robotic and manual injections. At least three independent experiments were performed.

    It has been observed that Cas9 protein injections lead to more pronounced phenotypic effects compared with Cas9 mRNA injections in terms of TBXTA gene knockout. This disparity may be attributed to the ability of Cas9 protein to perform DNA double-strand breaks from one-cell stage at the moment of injection; on the other hand, the mRNA would have to be translated for the protein to cause a DNA double-strand break. Moreover, this study also highlighted the potential of the CasRx system at the RNA level. The CasRx system for knockdown has undergone significant improvements through upgraded protocols and platforms in recent years [37]. In the present study, the authors utilized previously validated CasRx gRNAs targeting TBXTA mRNA [34,37], taking advantage of the enhanced CasRx system. Given that CasRx protein is not commercially available, the authors expressed and purified the protein in their own laboratory. Both the purified CasRx protein and CasRx mRNA injections resulted in high phenotypic penetrance. The percentage of phenotypes observed in the CasRx protein group was similar to that in the mRNA group. This low difference between mRNA and protein injection of CasRx may be because the target gene, TBXTA, is involved in embryo notochord formation, which occurs relatively late in zebrafish development. Nevertheless, if an early development gene is studied, the CasRx protein might have a higher phenotypic penetrance. Moreover, after the CasRx mRNA injection, protein translation in the embryo has not yet been studied in different developmental stages and the phenotypic penetrance would also depend on the target mRNA levels in these stages. Another interesting observation is that grade I phenotypes can be seen with the CasRx experiments. This could be explained by the previous validation for the used gRNA, which showed a reduction of mRNA levels of about 60% of the TBXTA transcript by reverse transcription–quantitative PCR [34,37], as the authors expect the same efficiency in their experiments. This reduction would still allow for a low expression of this gene in the embryo. Additionally, it was demonstrated that disrupting a single nucleotide in the tail of the gRNA (CasRx binding domain) and using a 23-nucleotide binding sequence significantly improves the phenotypic penetrance [38]. Specifically, the percentage of grade III no tail cases doubles from 35% to 70% with this chemically synthetized, highly efficient gRNA structure [37].

    In the context of gene editing, traditional practices have involved injecting DNA/mRNAs/gRNAs into the first cell of zebrafish embryos. However, recent studies, such as the work by Kroll et al. [6], have demonstrated that injection directly into the yolk is adequate for achieving F0 knockouts using the highly efficient CRISPR-Cas9 technique. In the present study, the authors utilized a combination of multiple gRNAs targeting the TBXTA gene and observed that yolk injections resulted in a substantial occurrence of tail and notochord defects. Moreover, they found that the proportion of phenotypes caused by yolk injection closely resembled that resulting from cell interface injection using the robotic system. It is important to note that these injections were administered at the one-cell stage, shortly after fertilization, minimizing the time elapsed before the cell started to inflate. Consequently, the authors propose that the timing of injection, specifically immediately following fertilization and prior to cellular expansion, holds greater significance than injecting into the zygotic cell itself [6].

    Conclusion

    In conclusion, this study establishes the efficacy and advantages of robotic automated injection methods in zebrafish embryos for gene silencing experiments. The developed robotic system, with its user-friendly touch screen interface and automated droplet size calculation, streamlines large-scale injections, making it accessible even to individuals without prior zebrafish egg injection experience. The precision in injection volumes, error reduction and the unique feature of recording and selectively euthanizing noninjected eggs contribute to the system's efficiency. Our results demonstrate comparable phenotypes and mortality rates between manual and robotic injections, particularly in the context of the knockdown of the TBXTA gene. On the other hand, the robotic method surpasses manual injections in speed, being three-times faster. Additionally, we observed that Cas9 protein injections result in more pronounced phenotypic effects compared with Cas9 mRNA injections for TBXTA gene knockout. Furthermore, our study highlights the potential of the CasRx system at the RNA level, emphasizing its efficacy in achieving high phenotypic penetrance, particularly in early developmental genes. Overall, our study contributes to the understanding of the benefits and feasibility of robotic automated injection methods in zebrafish embryos for gene silencing experiments. The combination of precise injection control and increased efficiency provided by the robotic system holds promise for accelerating research in early embryonic development and other areas that require large-scale injections. Furthermore, the exploration of the CasRx system showcases the potential of RNA-targeting CRISPR effectors for precise and potent gene knockdown at high throughput.

    Future perspective

    Zebrafish automation is expected to gain increasing importance in screening research requiring high throughput, such as drug screening, gene and transcript function analysis, toxicity assessments and pathogen studies. We anticipate that our approach can serve as a foundation for further technical advancements. These could include improvements in throughput levels; enhanced injection accuracy, particularly in specific embryo or larval body regions; and increased ease of needle loading in the case of blockages. Such technologies may also find application in other models, such as organoid cultures. It is evident that the CRISPR-Cas toolbox will undergo further refinement, unlocking new possibilities for modifying DNA, RNA and even protein functions. The swift integration of this technology into our developed robotic pipeline is feasible. In particular, the exploration of the CasRx system highlights the potential of RNA-targeting CRISPR effectors for precise and potent gene knockdown at high throughput and with higher accuracies, as high-fidelity versions are being developed.

    Executive summary

    Robotic injection procedures

    • The development of an advanced robotic system with an integrated touch screen, automated droplet calculation and selective euthanasia has enhanced the efficiency and precision of zebrafish injection.

    • Here we present a stepwise procedure showcasing the robot's efficiency of speed of 59 embryos/min and high precision in zebrafish embryo injection procedures.

    Comparison of manual & robotic injections

    • The robotic injections demonstrated a three-times faster injection speed, achieving a rate of 59 embryos per minute, compared to manual injections, which managed only 20 embryos per minute.

    • Exploration of injection positions, wether at the yolk center or cell interface, was conducted to assess their impact on phenotypes; demonstating that the position of injection has not efect in the phenotype penetrance.

    RfxCas13d system & RNA-level manipulation

    • RfxCas13d system demonstrated efficacy at the RNA level, exhibiting high phenotypic penetrance in TBXTA gene knockdown in both robotic and manual injections.

    • This study contributes to understanding the potential of RNA-targeting CRISPR effectors for precise gene knockdown in a highthroughput manner.

    Conclusion

    • The study establishes the efficacy and advantages of robotic automated injection methods for gene silencing in zebrafish embryos, compared to manual injection methods.

    • The robotic system offers user-friendly features, precision and unique capabilities, streamlining large-scale injections.

    • Overall, the study advances understanding in the field, providing a promising approach for accelerating research in early embryonic development and large-scale injections; including, the use of CRISPR effectors for highthroughput genetic and transcriptomic screenings.

    Supplementary data

    To view the supplementary data that accompany this paper please visit the journal website at: www.future-science.com/doi/suppl/10.2144/btn-2023-0062

    Author contributions

    J Abugattas-Nuñez Del Prado expressed and purified CasRx protein and produced CasRx and Cas9 mRNA and RfxCas13d gRNAs. J Abugattas-Nuñez Del Prado and Y Ding performed the injection experiments and wrote the first version of the manuscript. J de Sonneville designed the robotic injection equipment. K-J van der Kolk designed the software for robot control and machine learning. J de Sonneville, E Málaga-Trillo, MA Moreno-Mateos and HP Spaink supervised the study and provided materials. All authors contributed to writing this article.

    Acknowledgments

    The authors would like to thank Guus van der Velden, Ulrike Nehrdich and Natasha Montiadi for helping us take care of the zebrafish. They are also grateful to BEV Koch for helping with the discussion and suggestions during RfxCas13d protein production standardization and manuscript submission.

    Financial disclosure

    Work in MA Moreno-Mateos' lab was supported by grants PID2021-127535NB-I00, CEX2020-001088-M and RyC-2017-23041 funded by MCIN/AEI/ 10.13039/501100011033 by “ERDF A way of making Europe” and by ESF Investing in your future, grant UPO1380590 funded by “ERDF A way of making Europe”, University Pablo de Olavide and Junta de Andalucía. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

    Competing interests disclosure

    Y Ding, K-J van der Kolk and J de Sonneville work for a company that commercially exploits the robotic system that is used in this publication. The authors have no other competing interests or relevant affiliations with any organization or entity with the subject matter or materials discussed in the manuscript apart from those disclosed.

    Writing disclosure

    No writing assistance was utilized in the production of this manuscript.

    Ethical conduct of research

    The animal study and the breeding of adult fish were approved by the local animal welfare committee (DEC) of the University of Leiden. All experiments were done on 1-day-postfertilization fish, therefore prior to the free-feeding stage, and did not fall under animal experimentation law according to the EU Animal Protection Directive 2010/63/EU.

    Open access

    This work is licensed under the Attribution-NonCommercial-NoDerivatives 4.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/4.0/

    References

    • 1. Cordero-Maldonado ML, Perathoner S, Van Der Kolk KJ et al. Deep learning image recognition enables efficient genome editing in zebrafish by automated injections. PLOS ONE 14(1), e0202377 (2019).
    • 2. Habjan E, Ho VQT, Gallant J et al. An anti-tuberculosis compound screen using a zebrafish infection model identifies an aspartyl-tRNA synthetase inhibitor. Dis. Model Mech. 14(12), dmm049145 (2021).
    • 3. Carvalho R, De Sonneville J, Stockhammer OW et al. A high-throughput screen for tuberculosis progression. PLOS ONE 6(2), e16779 (2011).
    • 4. Ordas A, Raterink RJ, Cunningham F et al. Testing tuberculosis drug efficacy in a zebrafish high-throughput translational medicine screen. Antimicrob. Agents Chemother. 59(2), 753–762 (2015).
    • 5. Spaink HP, Cui C, Wiweger MI et al. Robotic injection of zebrafish embryos for high-throughput screening in disease models. Methods 62(3), 246–254 (2013).
    • 6. Kroll F, Powell GT, Ghosh M et al. A simple and effective F0 knockout method for rapid screening of behaviour and other complex phenotypes. Elife 10 (2021).
    • 7. Gaj T, Gersbach CA, Barbas CF 3rd. ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering. Trends Biotechnol. 31(7), 397–405 (2013).
    • 8. Chen GR, Sive H, Bartel DP. A seed mismatch enhances argonaute2-catalyzed cleavage and partially rescues severely impaired cleavage found in fish. Mol. Cell. 68(6), 1095–1107 e1095 (2017).
    • 9. Kelly A, Hurlstone AF. The use of RNAi technologies for gene knockdown in zebrafish. Brief Funct. Genomics 10(4), 189–196 (2011).
    • 10. Schulte-Merker S, Stainier DY. Out with the old, in with the new: reassessing morpholino knockdowns in light of genome editing technology. Development 141(16), 3103–3104 (2014).
    • 11. Kok FO, Shin M, Ni CW et al. Reverse genetic screening reveals poor correlation between morpholino-induced and mutant phenotypes in zebrafish. Dev. Cell. 32(1), 97–108 (2015).
    • 12. Lai JKH, Gagalova KK, Kuenne C, El-Brolosy MA, Stainier DYR. Induction of interferon-stimulated genes and cellular stress pathways by morpholinos in zebrafish. Dev. Biol. 454(1), 21–28 (2019).
    • 13. Joris M, Schloesser M, Baurain D, Hanikenne M, Muller M, Motte P. Number of inadvertent RNA targets for morpholino knockdown in Danio rerio is largely underestimated: evidence from the study of Ser/Arg-rich splicing factors. Nucleic Acids Res. 45(16), 9547–9557 (2017).
    • 14. Barrangou R, Fremaux C, Deveau H et al. CRISPR provides acquired resistance against viruses in prokaryotes. Science 315(5819), 1709–1712 (2007).
    • 15. Garneau JE, Dupuis ME, Villion M et al. The CRISPR/Cas bacterial immune system cleaves bacteriophage and plasmid DNA. Nature 468(7320), 67–71 (2010).
    • 16. Marraffini LA, Sontheimer EJ. CRISPR interference limits horizontal gene transfer in staphylococci by targeting DNA. Science 322(5909), 1843–1845 (2008).
    • 17. Mojica FJ, Diez-Villasenor C, Garcia-Martinez J, Soria E. Intervening sequences of regularly spaced prokaryotic repeats derive from foreign genetic elements. J. Mol. Evol. 60(2), 174–182 (2005).
    • 18. Sapranauskas R, Gasiunas G, Fremaux C, Barrangou R, Horvath P, Siksnys V. The Streptococcus thermophilus CRISPR/Cas system provides immunity in Escherichia coli. Nucleic Acids Res. 39(21), 9275–9282 (2011).
    • 19. 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. U S A 109(39), e2579–e2586 (2012).
    • 20. Hsu PD, Lander ES, Zhang F. Development and applications of CRISPR-Cas9 for genome engineering. Cell 157(6), 1262–1278 (2014).
    • 21. Mali P, Esvelt KM, Church GM. Cas9 as a versatile tool for engineering biology. Nat. Methods 10(10), 957–963 (2013).
    • 22. Mali P, Yang L, Esvelt KM et al. RNA-guided human genome engineering via Cas9. Science 339(6121), 823–826 (2013).
    • 23. Liu L, Chen P, Wang M et al. C2c1-sgRNA complex structure reveals RNA-guided DNA cleavage mechanism. Mol. Cell. 65(2), 310–322 (2017).
    • 24. Yang H, Gao P, Rajashankar KR, Patel DJ. PAM-dependent target DNA recognition and cleavage by C2c1 CRISPR-Cas endonuclease. Cell 167(7), 1814–1828.e1812 (2016).
    • 25. Moreno-Mateos MA, Vejnar CE, Beaudoin JD et al. CRISPRscan: designing highly efficient sgRNAs for CRISPR-Cas9 targeting in vivo. Nat. Methods 12(10), 982–988 (2015).
    • 26. Vejnar CE, Moreno-Mateos MA, Cifuentes D, Bazzini AA, Giraldez AJ. Optimized CRISPR-Cas9 system for genome editing in zebrafish. Cold Spring Harb. Protoc. 2016(10), (2016).
    • 27. Liu P, Luk K, Shin M et al. Enhanced Cas12a editing in mammalian cells and zebrafish. Nucleic Acids Res. 47(8), 4169–4180 (2019).
    • 28. Abudayyeh OO, Gootenberg JS, Konermann S et al. C2c2 is a single-component programmable RNA-guided RNA-targeting CRISPR effector. Science 353(6299), aaf5573 (2016).
    • 29. Abudayyeh OO, Gootenberg JS, Essletzbichler P et al. RNA targeting with CRISPR-Cas13. Nature 550(7675), 280–284 (2017).
    • 30. Cox DBT, Gootenberg JS, Abudayyeh OO et al. RNA editing with CRISPR-Cas13. Science 358(6366), 1019–1027 (2017).
    • 31. Hale CR, Zhao P, Olson S et al. RNA-guided RNA cleavage by a CRISPR RNA–Cas protein complex. Cell 139(5), 945–956 (2009).
    • 32. Konermann S, Lotfy P, Brideau NJ, Oki J, Shokhirev MN, Hsu PD. Transcriptome engineering with RNA-targeting type VI-D CRISPR effectors. Cell 173(3), 665–676 e614 (2018).
    • 33. Makarova KS, Wolf YI, Iranzo J et al. Evolutionary classification of CRISPR-Cas systems: a burst of class 2 and derived variants. Nat. Rev. Microbiol. 18(2), 67–83 (2020).
    • 34. Kushawah G, Hernandez-Huertas L, Abugattas-Nunez Del Prado J et al. CRISPR-Cas13d induces efficient mRNA knockdown in animal embryos. Dev. Cell. 54(6), 805–817 e807 (2020).
    • 35. Halpern ME, Ho RK, Walker C, Kimmel CB. Induction of muscle pioneers and floor plate is distinguished by the zebrafish no tail mutation. Cell 75(1), 99–111 (1993).
    • 36. Westerfield M. The Zebrafish Book: A Guide for the Laboratory Use of Zebrafish (Danio Rerio). University of Oregon Press, OR, USA (2000).
    • 37. Hernandez-Huertas L, Kushawah G, Diaz-Moscoso A et al. Optimized CRISPR-RfxCas13d system for RNA targeting in zebrafish embryos. STAR Protoc. 3(1), 101058 (2022).
    • 38. Wessels HH, Mendez-Mancilla A, Guo X, Legut M, Daniloski Z, Sanjana NE. Massively parallel Cas13 screens reveal principles for guide RNA design. Nat. Biotechnol. 38(6), 722–727 (2020).