Next Article in Journal
The Utility of Novel Kidney Injury Biomarkers in Early Detection of CSA-AKI
Next Article in Special Issue
CRISPR/Cas9-Mediated Gene Editing in Salmonids Cells and Efficient Establishment of Edited Clonal Cell Lines
Previous Article in Journal
Deep Brain Stimulation in the Subthalamic Nucleus Can Improve Skilled Forelimb Movements and Retune Dynamics of Striatal Networks in a Rat Stroke Model
Previous Article in Special Issue
Structure- and Content-Dependent Efficiency of Cas9-Assisted DNA Cleavage in Genome-Editing Systems
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Decade of CRISPR-Cas Gnome Editing in C. elegans

Division of Natural and Applied Sciences, Duke Kunshan University, Kunshan 215316, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2022, 23(24), 15863; https://doi.org/10.3390/ijms232415863
Submission received: 11 November 2022 / Revised: 5 December 2022 / Accepted: 5 December 2022 / Published: 14 December 2022

Abstract

:
CRISPR-Cas allows us to introduce desired genome editing, including mutations, epitopes, and deletions, with unprecedented efficiency. The development of CRISPR-Cas has progressed to such an extent that it is now applicable in various fields, with the help of model organisms. C. elegans is one of the pioneering animals in which numerous CRISPR-Cas strategies have been rapidly established over the past decade. Ironically, the emergence of numerous methods makes the choice of the correct method difficult. Choosing an appropriate selection or screening approach is the first step in planning a genome modification. This report summarizes the key features and applications of CRISPR-Cas methods using C. elegans, illustrating key strategies. Our overview of significant advances in CRISPR-Cas will help readers understand the current advances in genome editing and navigate various methods of CRISPR-Cas genome editing.

1. Introduction

CRISPR-Cas9 (clustered regularly interspaced short palindromic repeats-associated) is the current choice for genome editing. It is an RNA-guided system where a crRNA and a trRNA, together called a guide RNA (sgRNA), direct a Cas9 nuclease to the target gene of interest (Figure 1, [1]). The crRNA consists of a 20-nucleotide sequence from the spacer of the CRISPR locus and corresponds to a target DNA. The trRNA is complementary to a pre-crRNA, thus forming an RNA duplex later cleaved by RNase III to form a crRNA–trRNA hybrid, thereby directing the Cas9 RGN to make a double-stranded break (DSB) at the target site. The DSB can then be repaired by non-homologous end joining (NHEJ) or homologous recombination (HR)/homology-directed repair (HDR). In C. elegans, CRISPR-Cas9 technology was first adopted in 2013 (Figure 2), and since then, the nematode-research community has produced increasingly sophisticated strategies for genome editing. Here, we introduce the significant discoveries of CRISPR-Cas genome editing achieved in the last decade (Figure 2).

2. History of C. elegans CRISPR-Cas

Since the first publication of the effects of the CRISPR-Cas9 nuclease in vitro in 2012 [3], parallel studies in different organisms have proven the role of molecular scissors approaches in vivo. The first demonstration of CRISPR-Cas9 in C. elegans involved the simple generation of loss-of-function mutants via a non-homologous end-joining (NHEJ) pathway [6]. This study targeted unc-119 and dpy-13, which enabled screening of the transgenic mutants by screening uncoordinated (unc) or dumpy (dpy) phenotypes. Additionally, it demonstrated that a gene (klp-12, Y61A9LA.1) exhibiting no distinct phenotypes can also be identified by PCR screening methods. Single guide RNA (sgRNA), which combines crRNA and trRNA, can cleave target sites, thus eliminating the use of crRNA and trRNA separately, as demonstrated in S. pyogenes [3]. However, another report revealed that chimeric sgRNA is not as efficient as individual crRNA and trRNA [7].
Previously, homologous recombination in C. elegans was neither an efficient nor a major recombination pathway for double-stranded breaks [22,23]. However, this idea was no longer valid after the appearance of CRISPR-Cas. Soon after the first publication of NHEJ, multiple articles were released, with swiftly varying strategies. Three back-to-back reports demonstrated that CRISPR-Cas9 could edit the C. elegans genome via homologous recombination [7,8,9]. Hence, CRISPR-Cas-mediated homologous recombination (HR) allows a promising path for customized precise modifications.

3. Screening of Transgenic Worms

3.1. PCR and Drug-Based Selection

Whereas CRISPR induces DNA double-strand break (DSB) at the target genome, and the cell’s DSB repair system completes genome editing, the screening of the corrected genome still remains for scientists to complete. PCR has been the breakthrough technique for screening targeted mutations, and it is still helpful for most small-scale screenings [1]. However, for a large-scale genetic screening approach, it is often not time- and cost-efficient. Drug selection provides a new path, in addition to phenotype-based screening, to efficiently identify homologous recombination. Chen et al. incorporated the hygromycin resistance gene at the ben-1 locus, endowing both hygromycin and benomyl resistance [8]. Dpy-3 was also proposed as a new CRISPR-Cas tool, since its homozygous and hemizygous phenotypes are distinct [10].

3.2. Co-CRISPR and Co-Conversion: A Mutation in an Endogenous Marker Presents A Visible Phenotype

In 2014, C. elegans genome editing entered a new phase by adopting a second endogenous marker. Kim et al. introduced the Co-CRISPR strategy to detect genome editing. In addition to the gene of interest, unc-22 was targeted and selected by identifying twitching worms. The unc-22 indicates active Cas9 expression and a twitcher-based indel frequency via NHEJ mutations (Figure 3, [11]). Similarly, Arribere et al. proposed targeting a gene of interest by hitting second markers, such as dpy-10 and sqt-1 [12]. Since this strategy relies on homologous recombination events, it was named co-conversion. Co-CRISPR and co-conversion are similar in the sense that both methods target the gene of interest and an additional marker gene that confers phenotypic changes, thus ensuring that genome editing is being processed. Co-CRISPR uses NHEJ for targeting marker genes, whereas co-conversion relies on homologous recombination (HR) via template DNA injected into the germline.

3.3. Streamlined Genome Editing

In 2015, another set of new strategies was introduced to minimize hands-on labor and screening procedures. The streamlined methods incorporated three key features: a drug-resistance gene, a fluorescence marker, and Cre-Lox recombinase technology for seamless genome editing (Figure 4). The essential advantage of these methods is that minimal PCR screening is required to identify recombination by employing antibiotic resistance and a fluorescence reporter.
Notably, Norris and Kim et al. reported strategies to find recombinant versus extrachromosomal arrays by observing the mosaic expression of GFP and mCherry markers [13]. Studies from two independent labs described seamless genome editing by removing scars with Cre recombinase [13,14]. While both methods required Cre recombinase expression for seamless editing, Dickinson et al. eliminated the second microinjection of Cre by incorporating inducible Cre recombinase in the repair template [14].

4. Advances in Genome Editing Components

4.1. Cas9 Variants

Over the past decade, several CRISPR nuclease variants have been developed, and the range of its targets has been expanded (Figure 5). Nickases create a single-strand rather than a double-strand break. Targeting with nickase and two adjacent sgRNAs creates double-strand breaks with overhang, with reduces the off-target effects compared to the canonical/wild-type Cas9 system [4]. Dead Cas9 (dCas9) is an inactive catalytic nuclease. While the native Cas9 induces double-strand breaks on target DNA [5], dCas9 provides a binding site for activators or enhancers, promoting gene expression.
Since the C. elegans genome is an AT-rich genome, targeting conventional NGG near DSBs is frequently challenging. Therefore, having alternative sets of PAM sequences would expand the regions capable of being genome-edited. Kleinstiver et al. reported that Cas9 VQR (which possesses the amino acid substitutions D1135V, R1335Q, and T1337R) recognizes NGA PAM sequences instead of canonical NGG [24]. VQR recognizes NGAG as efficiently as wild-type Cas9 targets NGG [15], thus broadening the range of targets. Additionally, SpG and SpRY are two modified versions of Cas9, with more relaxed PAM requirements than Cas9 [25]. Thus, they target more portions of the genome. Of note, SpG and SpRY performed as efficiently as wild-type Cas9 at an increased concentration of CRISPR-Cas reagents (8 µM in the injection mix).

4.2. Guide RNA

Since guide RNA must designate the unique site of a gene of interest, multiple factors must be considered to achieve a high efficacy and specificity regarding genome editing. Studies have investigated strategies for designing guide RNA to enhance the genome editing frequency. For example, Farboud et al. reported that a GG motif at the 3′ end of the target sequences induced a high frequency of mutagenesis via NHEJ and HR pathways [16]. The median frequency at the targets was 10-fold higher than those reported in previous studies with multiple guide RNAs [6,11,26].
Later, the same group explored the location of DSB repair events in CRISPR-Cas editing. Interestingly, NHEJ induces asymmetric insertions and deletions (indels) preferentially in regions of 5′ of PAM [27]. Notably, a similar propensity for repair has been found in mammals, advocating the use of C. elegans as a model to understand the universal rules underlying genome editing [28,29].
Since variants of Cas nucleases have expanded the range of genome targets, identifying a proper target may now involve additional effort. Currently, a web-based database of guide RNA can help to identify guide RNAs for target genes and minimize off-target sites. Several database services can identify potential guide RNA for the entire C. elegans genome (genome.sfu.ca/gexplore (accessed on 4 December 2022), www.crisprscan.org (accessed on 4 December 2022), crispr.dbcls.jp (accessed on 4 December 2022), crispor.tefor.net (accessed on 4 December 2022) [30,31,32,33]). The user can search the guides present in the database by entering multiple factors, including genomic interval, GC content, gene name, and the presence of GG at the 3′ end of the guides. Notably, CRISPRscan can identify targets for the Cas9 variants SpG and SpRY.

4.3. Dual sgRNA/Dual DSBs

While Cas9 nuclease induces a single DSB at a target site, dual or triple DSBs offer advantages for genome editing. For example, deletion mutants can be achieved by adopting two sgRNAs via NHEJ or HR pathways (Figure 6A, [17]). Likewise, dual sgRNA can generate long chromosome deletion between two sgRNAs [18]. Chen et al. also demonstrated that dual DSBs generate reciprocal chromosomal translocation, thereby providing a practical approach to studying genome rearrangement [34].
Zhang et al. reported that dual cutting of the repair template, rather than genomic DNA, enhanced precise genome editing efficacy (Figure 6). The dual-cut repair template, flanked by two sgRNA at both ends of the plasmid, increased HR efficiency by between two and fivefold compared to a conventional uncut circular template. They observed a proportional increase in HR efficiency, with a more extended homologous arm in either circular or linear DNA, suggesting that a longer flanking sequence improves dual-cut-mediated HR efficacy [35]. Similarly, dual DSBs promoted the insertion of large (9300 bp) DNA fragments, in combination with a dpy-10 co-conversion strategy, where single DSBs failed (5% vs. 0%, respectively) [27]. It is worth noting that, when the dsDNA template was cleaved by two DSBs, orienting both PAMs outward resulted in efficient homologous recombination (21% vs. 12% in an out/out and in/in configuration, respectively, Figure 6).

4.4. Oligonucleotide as a Repair Template for Homologous Recombination

Since linear DNA is prone to degradation by nucleases present in the cells, the conventional way of delivering a repair template is to supply double-stranded DNA as a part of circular DNA. Therefore, plasmid DNA is desirable, especially for large-sized repair templates. However, a growing number of studies have found that single-stranded oligonucleotides can often be good alternatives for HR-mediated genome editing. Specifically, oligonucleotides offer a few advantages over plasmid DNA: they are cloning-free and can be rapidly synthesized via commercially available resources. Therefore, adopting linear DNA reduces the amount of time required for the entire genome editing process.
The first demonstration of oligonucleotides as repair templates was performed in 2014, substantiating their simplicity and efficacy [19]. An oligonucleotide, ~100 bp long, serves as a template to repair DSBs occurring at four different genes via homologous recombination (Figure 7). This strategy has demonstrated its efficiency in several labs [12,36,37]. It is worth noting, however, that it appears that smaller flanking homology requires the DSB/cleavage site to be near the site of genome editing [12].

4.5. Modified Repair Template

Previous research investigated whether modification of the donor template improves HR efficacy. Although controversial at first, a growing number of studies have reported that the modification of repair donors enhances genome editing proficiency (Figure 7). In 2014, Zhao et al. employed a phosphorothioate-modified oligonucleotide as a repair template in C. elegans for the first time. Although the modified oligonucleotide resulted in the desired genome editing, it was unclear whether its efficacy was enhanced compared that resulting from to non-modification [19]. In mammalian cultured cells, however, phosphorothioate-modified oligonucleotides enhanced the genome editing efficiency of single-stranded oligonucleotide donors. In addition, modified oligonucleotides allow for insertions > 100 bp long, providing design flexibility [37]. In contrast, fluorescent and amine modifications to the 5′- and 3′-termini of single-stranded oligodeoxynucleotide (ssODN) donors did not alter HR frequency compared to nonmodified donors in human cells [38].
With this controversy, recent studies have explored donors with 5′-end modifications that enhance genome editing efficacy. Gutierrez-Triana et al. showed that adding biotin at the 5′ ends of dsDNA leads to an increase in HR efficiency of up to 60% in the injected generation of medaka fish embryos. Provocatively, the authors demonstrated that biotin and SpC3 5′ modifications prevent donor multimerization/NHEJ of dsDNA, thereby providing optimal conditions for HR-mediated CRISPR-Cas genome editing [39]. Similarly, Yu et al. reported that 5′ C6-PEG10-modified dsDNA increased knockin frequency up to fivefold, in combination with Cas9 ribonucleoprotein (RNP) in human cells [40]. In line with these reports in fish and mammalian cells, the C. elegans study demonstrated that 5′ modifications of the donor improved the efficacy of HR frequency roughly twofold [41]. Interestingly, TEG (triethylene glycol) and RNA::TEG modifications performed with similar efficacy in C. elegans, whereas RNA::TEG was superior to TEG in human cells and zebrafish. They demonstrated that these 5′ modifications suppress donor ligation reactions in a similar way to that of biotin and SpC3 5′ modifications [39].
It is worth noting that C. elegans studies from the Mello Lab reported additional alterations that enhance HR efficiency dramatically by modifying repair template DNA. First, a single-stranded overhang containing dsDNA donors yielded higher integration rates at three loci [36]. Second, denaturing and cooling the dsDNA donor template increased the HR frequency by up to 50% [20].

5. Other Developments

5.1. Off-Target Effects

Given that genome editing relies on the creation of DSB, potential off-target effects have been one of the major concerns. The off-target effects of CRISPR-Cas9 have been addressed a few times in C. elegans studies. Chiu et al. performed high-throughput sequencing after CRISPR-Cas genome editing and found no distinct evidence of off-target genomic lesions using GATK pipeline or split-read analysis [42]. Second, whole-genome sequencing of five strains revealed no distinct mutations at predicted off-target sites [17]. These suggest that off-target effects can be avoided by the accurate designing of guide RNA, with the help of computational tools.

5.2. CRISPR-Cas Genome Editing Protocols

We have listed some of the C. elegans genome editing protocols. Please see the list of major C. elegans CRISP-Cas publications for more information (Table S1).
  • Efficient Genome Editing in Caenorhabditis elegans with a Toolkit of Dual-Marker Selection Cassettes [13].
  • Precision genome editing using CRISPR-Cas9 and linear repair templates in C. elegans [43].
  • An Efficient Genome Editing Strategy to Generate Putative Null Mutants in Caenorhabditis elegans Using CRISPR/Cas9 [44].
  • CRISPR-Cas9-Guided Genome Engineering in Caenorhabditis elegans [1].
  • An affordable plasmid miniprep suitable for proficient microinjection in Caenorhabditis elegans [45].
  • Design of Repair Templates for CRISPR-Cas9-Triggered Homologous Recombination in Caenorhabditis elegans [46].
  • Microinjection for precision genome editing in Caenorhabditis elegans [47].
  • High-efficiency CRISPR gene editing in C. elegans using Cas9 integrated into the genome [48].
  • Approaches for CRISPR/Cas9 Genome Editing in C. elegans [49].
  • CRISPR/Cas9 Methodology for the Generation of Knockout Deletions in Caenorhabditis elegans [50].
  • CRISPR-Based Methods for Caenorhabditis elegans Genome Engineering [51].

6. Perspectives

6.1. Pros and Impact

CRISPR represents the greatest revolution in gene editing to date. With the advance of CRISPR-Cas genome editing technology, scientists have continued to expand this revolution for over a decade. The impact of precise and robust genome editing is broad and potent and has thus been able to influence the plant industry, scientific research, livestock improvement, and biomedical engineering, as well as human diseases and fertility. Furthermore, it has not just been limited to microbes, but has also been applied to mammalian cells, animal models, and plants, as well as for the modification of secondary metabolites, such as antibiotics [52], and medicinally bioactive compounds, including morphine and thebaine in opium. The unprecedented potential and impact of CRISPR-Cas have also produced concerns. Misuse of gene editing could trigger risks and dangers beyond imagination; therefore, urgent ethical concerns need to be addressed. In this regard, C. elegans is an ethical-issue-free animal model that can substitute for some portion of the conventional animal models used in research labs.

6.2. C. elegans for CRISPR-Cas Genome Editing

C. elegans has served as the leading animal model of CRISPR-Cas. The success of C. elegans as a CRISPR-Cas genome editing model lies in its large progeny (∼300 eggs), rapid life cycle (~3 days), self-fertilization, and easy maintenance [53,54]. Strategies established in C. elegans were successively adopted in other nematode species. The strategies work in C. remanei, C. briggsae, and the parasitic nematodes Strongyloides spp. and A. freiburgensis [55,56,57,58,59,60]. Specifically, CRISPR-Cas9 in C. briggsae can utilize C. elegans promoters to express sgRNAs and Cas9 and, similar to C. elegans, can use the 3′ GG motif to increase genome editing efficacy [61]. Thus, applying CRISPR-Cas discovered in C. elegans to other nematodes would provide a powerful tool to investigate the functions of orthologs, thereby leading to a better understanding of the molecular mechanisms. In contrast, there are intrinsic hurdles in C. elegans genome editing. Although some studies developed a quantity drug screening in C. elegans, a high-throughput CRISPR screening approach for identifying genes or pathways is not feasible. This is mainly because genome editing relies on a slow, multi-step microinjection process. Given that a recent study demonstrated the successful delivery of sgRNA [21], feeding-based CRISPR may be an alternative method for genome editing.
Studies over the last decade have demonstrated that improvements in genome editing can unlock the enormous potential that CRISPR offers. However, CRISPR-Cas is still far from perfect. Consequently, we are expecting various aspects of progress. For example, while conventional strategies rely on type II Cas9, the remaining five CRISPR systems have not been well investigated. Studies on the remaining types expand the targets of CRISPR-Cas application. For example, Cas12 of type V induced staggered breaks in dsDNA, unlike blunt breaks created by Cas9 [62]. Cas13 of type VI targets RNA, not DNA [63]. Second, improved delivery systems may enhance genome editing efficacy. Since CRISPR-Cas9 requires a large Cas9 complex (RNA-Protein) to cleave a target DNA in the nucleus, the low delivery efficiency restricts its successful editing. CRISPR-Cas combined with viruses, lipids, and synthetic molecules may enhance the delivery of CRISPR-Cas9 to the target DNA (Figure 6, [64,65]).
Advances in new screening methods, secondary endogenous markers, dual DSBs, modified repair templates, computational tools, and Cas variants have extended the efficiency and specificity of genome editing. This further strengthens overall genome editing strategies and incentivizes their use in science, medicine, pharmaceuticals, and industry. Furthermore, given the high level of conservation between mammals and C. elegans—60–80% of C. elegans genes are conserved between the two groups of organisms [66,67]—the mechanism of genome editing and the DNA repair pathway found in C. elegans may also be applicable to mammalian systems.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms232415863/s1.

Author Contributions

Y.H.: literature survey, figure preparation (Figure 3 and Supplementary Table S1), proofreading; J.C.: literature survey, figure preparation (Figure 1 and Figure 5, and Supplementary Table S1); H.-M.K.: writing—original manuscript, review and editing; figure preparation (Figure 2, Figure 4, Figure 6 and Figure 7), reference collection, project administration, funding acquisition, conceptualization, literature survey, and supervision. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the National Natural Science Foundation of China (NSFC No. 31972876) and a Synear and Wang-Cai grant from Duke Kunshan University to H-MK.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank members of Kim Laboratory for discussions and proofreading.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Kim, H.M.; Colaiacovo, M.P. CRISPR-Cas9-Guided Genome Engineering in Caenorhabditis elegans. Curr. Protoc. Mol. Biol. 2019, 129, e106. [Google Scholar] [CrossRef] [PubMed]
  2. 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] [Green Version]
  3. 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]
  4. Mali, P.; Aach, J.; Stranges, P.B.; Esvelt, K.M.; Moosburner, M.; Kosuri, S.; Yang, L.; Church, G.M. CAS9 transcriptional activators for target specificity screening and paired nickases for cooperative genome engineering. Nat. Biotechnol. 2013, 31, 833–838. [Google Scholar] [CrossRef]
  5. Qi, L.S.; Larson, M.H.; Gilbert, L.A.; Doudna, J.A.; Weissman, J.S.; Arkin, A.P.; Lim, W.A. Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell 2013, 152, 1173–1183. [Google Scholar] [CrossRef] [Green Version]
  6. Friedland, A.E.; Tzur, Y.B.; Esvelt, K.M.; Colaiacovo, M.P.; Church, G.M.; Calarco, J.A. Heritable genome editing in C. elegans via a CRISPR-Cas9 system. Nat. Methods 2013, 10, 741–743. [Google Scholar] [CrossRef] [Green Version]
  7. Lo, T.W.; Pickle, C.S.; Lin, S.; Ralston, E.J.; Gurling, M.; Schartner, C.M.; Bian, Q.; Doudna, J.A.; Meyer, B.J. Precise and heritable genome editing in evolutionarily diverse nematodes using TALENs and CRISPR/Cas9 to engineer insertions and deletions. Genetics 2013, 195, 331–348. [Google Scholar] [CrossRef] [Green Version]
  8. Chen, C.; Fenk, L.A.; de Bono, M. Efficient genome editing in Caenorhabditis elegans by CRISPR-targeted homologous recombination. Nucleic Acids Res. 2013, 41, e193. [Google Scholar] [CrossRef] [Green Version]
  9. Tzur, Y.B.; Friedland, A.E.; Nadarajan, S.; Church, G.M.; Calarco, J.A.; Colaiacovo, M.P. Heritable custom genomic modifications in Caenorhabditis elegans via a CRISPR-Cas9 system. Genetics 2013, 195, 1181–1185. [Google Scholar] [CrossRef] [Green Version]
  10. Cho, S.W.; Lee, J.; Carroll, D.; Kim, J.S.; Lee, J. Heritable gene knockout in Caenorhabditis elegans by direct injection of Cas9-sgRNA ribonucleoproteins. Genetics 2013, 195, 1177–1180. [Google Scholar] [CrossRef]
  11. Kim, H.; Ishidate, T.; Ghanta, K.S.; Seth, M.; Conte, D., Jr.; Shirayama, M.; Mello, C.C. A co-CRISPR strategy for efficient genome editing in Caenorhabditis elegans. Genetics 2014, 197, 1069–1080. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Arribere, J.A.; Bell, R.T.; Fu, B.X.; Artiles, K.L.; Hartman, P.S.; Fire, A.Z. Efficient marker-free recovery of custom genetic modifications with CRISPR/Cas9 in Caenorhabditis elegans. Genetics 2014, 198, 837–846. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Norris, A.D.; Kim, H.M.; Colaiacovo, M.P.; Calarco, J.A. Efficient Genome Editing in Caenorhabditis elegans with a Toolkit of Dual-Marker Selection Cassettes. Genetics 2015, 201, 449–458. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Dickinson, D.J.; Pani, A.M.; Heppert, J.K.; Higgins, C.D.; Goldstein, B. Streamlined Genome Engineering with a Self-Excising Drug Selection Cassette. Genetics 2015, 200, 1035–1049. [Google Scholar] [CrossRef] [Green Version]
  15. Bell, R.T.; Fu, B.X.; Fire, A.Z. Cas9 Variants Expand the Target Repertoire in Caenorhabditis elegans. Genetics 2016, 202, 381–388. [Google Scholar] [CrossRef] [Green Version]
  16. Farboud, B.; Meyer, B.J. Dramatic enhancement of genome editing by CRISPR/Cas9 through improved guide RNA design. Genetics 2015, 199, 959–971. [Google Scholar] [CrossRef] [Green Version]
  17. Paix, A.; Wang, Y.; Smith, H.E.; Lee, C.Y.; Calidas, D.; Lu, T.; Smith, J.; Schmidt, H.; Krause, M.W.; Seydoux, G. Scalable and versatile genome editing using linear DNAs with microhomology to Cas9 Sites in Caenorhabditis elegans. Genetics 2014, 198, 1347–1356. [Google Scholar] [CrossRef] [Green Version]
  18. Chen, X.; Xu, F.; Zhu, C.; Ji, J.; Zhou, X.; Feng, X.; Guang, S. Dual sgRNA-directed gene knockout using CRISPR/Cas9 technology in Caenorhabditis elegans. Sci. Rep. 2014, 4, 7581. [Google Scholar] [CrossRef] [Green Version]
  19. Zhao, P.; Zhang, Z.; Ke, H.; Yue, Y.; Xue, D. Oligonucleotide-based targeted gene editing in C. elegans via the CRISPR/Cas9 system. Cell Res. 2014, 24, 247–250. [Google Scholar] [CrossRef] [Green Version]
  20. Ghanta, K.S.; Mello, C.C. Melting dsDNA Donor Molecules Greatly Improves Precision Genome Editing in Caenorhabditis elegans. Genetics 2020, 216, 643–650. [Google Scholar] [CrossRef]
  21. Fischer, F.; Benner, C.; Goyala, A.; Grigolon, G.; Vitiello, D.; Wu, J.; Zarse, K.; Ewald, C.Y.; Ristow, M. Ingestion of single guide RNAs induces gene overexpression and extends lifespan in Caenorhabditis elegans via CRISPR activation. J. Biol. Chem. 2022, 298, 102085. [Google Scholar] [CrossRef] [PubMed]
  22. Plasterk, R.H.; Groenen, J.T. Targeted alterations of the Caenorhabditis elegans genome by transgene instructed DNA double strand break repair following Tc1 excision. EMBO J. 1992, 11, 287–290. [Google Scholar] [CrossRef]
  23. Berezikov, E.; Bargmann, C.I.; Plasterk, R.H. Homologous gene targeting in Caenorhabditis elegans by biolistic transformation. Nucleic Acids Res. 2004, 32, e40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Kleinstiver, B.P.; Prew, M.S.; Tsai, S.Q.; Topkar, V.V.; Nguyen, N.T.; Zheng, Z.; Gonzales, A.P.; Li, Z.; Peterson, R.T.; Yeh, J.R.; et al. Engineered CRISPR-Cas9 nucleases with altered PAM specificities. Nature 2015, 523, 481–485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Vicencio, J.; Sánchez-Bolaños, C.; Moreno-Sánchez, I.; Brena, D.; Vejnar, C.E.; Kukhtar, D.; Ruiz-López, M.; Cots-Ponjoan, M.; Rubio, A.; Melero, N.R.; et al. Genome editing in animals with minimal PAM CRISPR-Cas9 enzymes. Nat. Commun. 2022, 13, 2601. [Google Scholar] [CrossRef]
  26. Waaijers, S.; Portegijs, V.; Kerver, J.; Lemmens, B.B.; Tijsterman, M.; van den Heuvel, S.; Boxem, M. CRISPR/Cas9-targeted mutagenesis in Caenorhabditis elegans. Genetics 2013, 195, 1187–1191. [Google Scholar] [CrossRef] [Green Version]
  27. Farboud, B.; Severson, A.F.; Meyer, B.J. Strategies for Efficient Genome Editing Using CRISPR-Cas9. Genetics 2019, 211, 431–457. [Google Scholar] [CrossRef] [Green Version]
  28. Mali, P.; Yang, L.; Esvelt, K.M.; Aach, J.; Guell, M.; DiCarlo, J.E.; Norville, J.E.; Church, G.M. RNA-guided human genome engineering via Cas9. Science 2013, 339, 823–826. [Google Scholar] [CrossRef] [Green Version]
  29. Richardson, C.D.; Kazane, K.R.; Feng, S.J.; Zelin, E.; Bray, N.L.; Schafer, A.J.; Floor, S.N.; Corn, J.E. CRISPR-Cas9 genome editing in human cells occurs via the Fanconi anemia pathway. Nat. Genet. 2018, 50, 1132–1139. [Google Scholar] [CrossRef]
  30. Moreno-Mateos, M.A.; Vejnar, C.E.; Beaudoin, J.D.; Fernandez, J.P.; Mis, E.K.; Khokha, M.K.; Giraldez, A.J. CRISPRscan: Designing highly efficient sgRNAs for CRISPR-Cas9 targeting in vivo. Nat. Methods 2015, 12, 982–988. [Google Scholar] [CrossRef]
  31. Naito, Y.; Hino, K.; Bono, H.; Ui-Tei, K. CRISPRdirect: Software for designing CRISPR/Cas guide RNA with reduced off-target sites. Bioinformatics 2015, 31, 1120–1123. [Google Scholar] [CrossRef] [Green Version]
  32. Concordet, P.J.; Haeussler, M. CRISPOR: Intuitive guide selection for CRISPR/Cas9 genome editing experiments and screens. Nucleic Acids Res. 2018, 46, W242–W245. [Google Scholar] [CrossRef] [Green Version]
  33. Hutter, H.; Ng, M.P.; Chen, N. GExplore: A web server for integrated queries of protein domains, gene expression and mutant phenotypes. BMC Genom. 2009, 10, 529. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Chen, X.; Li, M.; Feng, X.; Guang, S. Targeted Chromosomal Translocations and Essential Gene Knockout Using CRISPR/Cas9 Technology in Caenorhabditis elegans. Genetics 2015, 201, 1295–1306. [Google Scholar] [CrossRef] [Green Version]
  35. Zhang, J.P.; Li, X.L.; Li, G.H.; Chen, W.; Arakaki, C.; Botimer, G.D.; Baylink, D.; Zhang, L.; Wen, W.; Fu, Y.W.; et al. Efficient precise knockin with a double cut HDR donor after CRISPR/Cas9-mediated double-stranded DNA cleavage. Genome Biol. 2017, 18, 35. [Google Scholar] [CrossRef] [Green Version]
  36. Dokshin, G.A.; Ghanta, K.S.; Piscopo, K.M.; Mello, C.C. Robust Genome Editing with Short Single-Stranded and Long, Partially Single-Stranded DNA Donors in Caenorhabditis elegans. Genetics 2018, 210, 781–787. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Renaud, J.B.; Boix, C.; Charpentier, M.; De Cian, A.; Cochennec, J.; Duvernois-Berthet, E.; Perrouault, L.; Tesson, L.; Edouard, J.; Thinard, R.; et al. Improved Genome Editing Efficiency and Flexibility Using Modified Oligonucleotides with TALEN and CRISPR-Cas9 Nucleases. Cell. Rep. 2016, 14, 2263–2272. [Google Scholar] [CrossRef] [Green Version]
  38. Lee, K.; Mackley, V.A.; Rao, A.; Chong, A.T.; Dewitt, M.A.; Corn, J.E.; Murthy, N. Synthetically modified guide RNA and donor DNA are a versatile platform for CRISPR-Cas9 engineering. Elife 2017, 6, e25312. [Google Scholar] [CrossRef] [PubMed]
  39. Gutierrez-Triana, J.A.; Tavhelidse, T.; Thumberger, T.; Thomas, I.; Wittbrodt, B.; Kellner, T.; Anlas, K.; Tsingos, E.; Wittbrodt, J. Efficient single-copy HDR by 5′ modified long dsDNA donors. Elife 2018, 7, e39468. [Google Scholar] [CrossRef] [PubMed]
  40. Yu, Y.; Guo, Y.; Tian, Q.; Lan, Y.; Yeh, H.; Zhang, M.; Tasan, I.; Jain, S.; Zhao, H. An efficient gene knock-in strategy using 5’-modified double-stranded DNA donors with short homology arms. Nat. Chem. Biol. 2020, 16, 387–390. [Google Scholar] [CrossRef]
  41. Ghanta, K.S.; Chen, Z.; Mir, A.; Dokshin, G.A.; Krishnamurthy, P.M.; Yoon, Y.; Gallant, J.; Xu, P.; Zhang, X.-O.; Ozturk, A.R.; et al. 5′-Modifications improve potency and efficacy of DNA donors for precision genome editing. eLife 2021, 10, e72216. [Google Scholar] [CrossRef] [PubMed]
  42. CChiu, H.; Schwartz, H.T.; Antoshechkin, I.; Sternberg, P.W. Transgene-free genome editing in Caenorhabditis elegans using CRISPR-Cas. Genetics 2013, 195, 1167–1171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Paix, A.; Folkmann, A.; Seydoux, G. Precision genome editing using CRISPR-Cas9 and linear repair templates in C. elegans. Methods 2017, 121–122, 86–93. [Google Scholar] [CrossRef] [PubMed]
  44. Wang, H.; Park, H.; Liu, J.; Sternberg, P.W. An Efficient Genome Editing Strategy To Generate Putative Null Mutants in Caenorhabditis elegans Using CRISPR/Cas9. G3 (Bethesda) 2018, 8, 3607–3616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Kim, H.M.; Tian, S.; Wang, S. An affordable plasmid miniprep suitable for proficient microinjection in Caenorhabditis elegans. 3 Biotech 2020, 10, 350. [Google Scholar] [CrossRef]
  46. Kim, H.M.; Zhang, X. Design of Repair Templates for CRISPR-Cas9-Triggered Homologous Recombination in Caenorhabditis elegans. In Springer Protocols Handbooks; Humana: New York, NY, USA, 2021. [Google Scholar]
  47. Ghanta, K.S.; Ishidate, T.; Mello, C.C. Microinjection for precision genome editing in Caenorhabditis elegans. STAR Protoc. 2021, 2, 100748. [Google Scholar] [CrossRef]
  48. Schwartz, M.L.; Davis, M.W.; Rich, M.S.; Jorgensen, E.M. High-efficiency CRISPR gene editing in C. elegans using Cas9 integrated into the genome. PLoS Genet. 2021, 17, e1009755. [Google Scholar] [CrossRef]
  49. Martin, C.J.; Calarco, J.A. Approaches for CRISPR/Cas9 Genome Editing in C. elegans. Methods Mol. Biol. 2022, 2468, 215–237. [Google Scholar] [CrossRef]
  50. Au, V.; Li-Leger, E.; Raymant, G.; Flibotte, S.; Chen, G.; Martin, K.; Fernando, L.; Doell, C.; Rosell, F.I.; Wang, S.; et al. CRISPR/Cas9 Methodology for the Generation of Knockout Deletions in Caenorhabditis elegans. G3 (Bethesda) 2019, 9, 135–144. [Google Scholar] [CrossRef] [Green Version]
  51. Dickinson, D.J.; Goldstein, B. CRISPR-Based Methods for Caenorhabditis elegans Genome Engineering. Genetics 2016, 202, 885–901. [Google Scholar] [CrossRef]
  52. Lim, Y.H.; Wong, F.T.; Yeo, W.L.; Ching, K.C.; Lim, Y.W.; Heng, E.; Chen, S.; Tsai, D.J.; Lauderdale, T.L.; Shia, K.S.; et al. Auroramycin: A Potent Antibiotic from Streptomyces roseosporus by CRISPR-Cas9 Activation. Chembiochem 2018, 19, 1716–1719. [Google Scholar] [CrossRef]
  53. Meneely, P.M.; Dahlberg, C.L.; Rose, J.K. Working with Worms: Caenorhabditis elegans as a Model Organism. Curr. Protoc. Essent. Lab. Tech. 2019, 19, e35. [Google Scholar] [CrossRef] [Green Version]
  54. Corsi, A.K. A biochemist’s guide to Caenorhabditis elegans. Anal. Biochem. 2006, 359, 1–17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Yin, D.; Schwarz, E.M.; Thomas, C.G.; Felde, R.L.; Korf, I.F.; Cutter, A.D.; Schartner, C.M.; Ralston, E.J.; Meyer, B.J.; Haag, E.S. Rapid genome shrinkage in a self-fertile nematode reveals sperm competition proteins. Science 2018, 359, 55–61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. LLok, J.B. CRISPR/Cas9 Mutagenesis and Expression of Dominant Mutant Transgenes as Functional Genomic Approaches in Parasitic Nematodes. Front. Genet. 2019, 10, 656. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Gang, S.S.; Castelletto, M.L.; Bryant, A.S.; Yang, E.; Mancuso, N.; Lopez, J.B.; Pellegrini, M.; Hallem, E.A. Targeted mutagenesis in a human-parasitic nematode. PLoS Pathog. 2017, 13, e1006675. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Adams, S.; Pathak, P.; Shao, H.; Lok, J.B.; Pires-daSilva, A. Liposome-based transfection enhances RNAi and CRISPR-mediated mutagenesis in non-model nematode systems. Sci. Rep. 2019, 9, 483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. O’Halloran, D.M. Genome aware CRISPR gRNA target prediction for parasitic nematodes. Mol. Biochem. Parasitol. 2019, 227, 25–28. [Google Scholar] [CrossRef]
  60. Cohen, S.; Sternberg, P. Genome editing of Caenorhabditis briggsae using CRISPR/Cas9 co-conversion marker dpy-10. MicroPubl. Biol. 2019, 2019. [Google Scholar] [CrossRef]
  61. Culp, E.; Richman, C.; Sharanya, D.; Jhaveri, N.; van den Berg, W.; Gupta, B.P. Genome editing in the nematode Caenorhabditis briggsae using the CRISPR/Cas9 system. Biol. Methods Protoc. 2020, 5, bpaa003. [Google Scholar] [CrossRef]
  62. Yan, W.X.; Hunnewell, P.; Alfonse, L.E.; Carte, J.M.; Keston-Smith, E.; Sothiselvam, S.; Garrity, A.J.; Chong, S.; Makarova, K.S.; Koonin, E.V.; et al. Functionally diverse type V CRISPR-Cas systems. Science 2019, 363, 88–91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Abudayyeh, O.O.; Gootenberg, J.S.; Essletzbichler, P.; Han, S.; Joung, J.; Belanto, J.J.; Verdine, V.; Cox, D.B.T.; Kellner, M.J.; Regev, A.; et al. RNA targeting with CRISPR-Cas13. Nature 2017, 550, 280–284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Yan, J.; Kang, D.D.; Dong, Y. Harnessing lipid nanoparticles for efficient CRISPR delivery. Biomater. Sci. 2021, 9, 6001–6011. [Google Scholar] [CrossRef] [PubMed]
  65. Yang, S.; Wu, Q.; Wei, Y.; Gong, C. CRISPR-Cas9 Delivery by Artificial Virus (RRPHC). Methods Mol. Biol. 2019, 1961, 81–91. [Google Scholar] [CrossRef]
  66. Kaletta, T.; Hengartner, M.O. Finding function in novel targets: C. elegans as a model organism. Nat. Rev. Drug Discov. 2006, 5, 387–398. [Google Scholar] [CrossRef]
  67. Leung, M.C.; Williams, P.L.; Benedetto, A.; Au, C.; Helmcke, K.J.; Aschner, M.; Meyer, J.N. Caenorhabditis elegans: An emerging model in biomedical and environmental toxicology. Toxicol. Sci. 2008, 106, 5–28. [Google Scholar] [CrossRef]
Figure 1. Overview of the CRISPR-Cas9 genome editing approach in C. elegans. The gonads of adult hermaphrodites are injected with the CRISPR-Cas9-containing DNA mixture. Injected Cas9 (grey color) forms a sequence-specific endonuclease when complexed with crRNA and trRNA (together called a guide RNA, or single guide RNA). The Cas9-guide RNA complex recognizes a target DNA sequence containing a PAM sequence (NGG) and induces a double-strand break (DSB), which will be repaired by non-homologous end joining (NHEJ) or homologous recombination (HR/HDR). Transgenic worms will be identified via marker-free (PCR) or marker-dependent strategies [1].
Figure 1. Overview of the CRISPR-Cas9 genome editing approach in C. elegans. The gonads of adult hermaphrodites are injected with the CRISPR-Cas9-containing DNA mixture. Injected Cas9 (grey color) forms a sequence-specific endonuclease when complexed with crRNA and trRNA (together called a guide RNA, or single guide RNA). The Cas9-guide RNA complex recognizes a target DNA sequence containing a PAM sequence (NGG) and induces a double-strand break (DSB), which will be repaired by non-homologous end joining (NHEJ) or homologous recombination (HR/HDR). Transgenic worms will be identified via marker-free (PCR) or marker-dependent strategies [1].
Ijms 23 15863 g001
Figure 2. Fundamental discoveries and advances in C. elegans CRISPR-Cas genome editing. Timeline highlighting major events of C. elegans CRISPR-Cas9 (green circle) and other species (red circle). CRISPR were first identified in E. coli in 1987 [2,3,4,5,6,7,8,9,10,11,12,13,14,15,16,17,18,19,20,21].
Figure 2. Fundamental discoveries and advances in C. elegans CRISPR-Cas genome editing. Timeline highlighting major events of C. elegans CRISPR-Cas9 (green circle) and other species (red circle). CRISPR were first identified in E. coli in 1987 [2,3,4,5,6,7,8,9,10,11,12,13,14,15,16,17,18,19,20,21].
Ijms 23 15863 g002
Figure 3. Co-CRISPR and co-conversion. Both methods employ an endogenous marker gene that exhibits phenotypic changes, thus ensuring that proper genome editing is processed. While Co-CRISPR uses NHEJ for targeting marker genes, co-conversion relies on homologous recombination (HR) via a repair template.
Figure 3. Co-CRISPR and co-conversion. Both methods employ an endogenous marker gene that exhibits phenotypic changes, thus ensuring that proper genome editing is processed. While Co-CRISPR uses NHEJ for targeting marker genes, co-conversion relies on homologous recombination (HR) via a repair template.
Ijms 23 15863 g003
Figure 4. Diagram of streamlined CRISPR-Cas. The streamlined strategies integrate a drug-resistance gene (yellow), a fluorescence marker (green), and a Cre recombinase to facilitate smooth genome editing by removing these two markers.
Figure 4. Diagram of streamlined CRISPR-Cas. The streamlined strategies integrate a drug-resistance gene (yellow), a fluorescence marker (green), and a Cre recombinase to facilitate smooth genome editing by removing these two markers.
Ijms 23 15863 g004
Figure 5. Diagram illustrating different types of Cas9 variants. (A), wild-type Cas9 nuclease. (B), Cas9 nickases that introduce a single-strand break at the target DNA. (C), catalytically inactive (dead) dCas9, which induces no breaks.
Figure 5. Diagram illustrating different types of Cas9 variants. (A), wild-type Cas9 nuclease. (B), Cas9 nickases that introduce a single-strand break at the target DNA. (C), catalytically inactive (dead) dCas9, which induces no breaks.
Ijms 23 15863 g005
Figure 6. Dual DSBs enhance deletions, homologous recombination, and insertions. (A) Single versus dual DSBs at targeted sites. (B) Dual DSBs (right) in the repair template enhances HR efficacy compared to single DSB (left). (C) Locating both PAMs outward of the DSB sites enhances insertion frequency.
Figure 6. Dual DSBs enhance deletions, homologous recombination, and insertions. (A) Single versus dual DSBs at targeted sites. (B) Dual DSBs (right) in the repair template enhances HR efficacy compared to single DSB (left). (C) Locating both PAMs outward of the DSB sites enhances insertion frequency.
Ijms 23 15863 g006
Figure 7. Oligonucleotides can serve as a template to repair DSBs via HR. At the site of CRISPR-Cas9-induced DSB, Single-stranded DNA (ssDNA), double-stranded DNA (dsDNA), and modified repair templates serve as a repair template. (A) ssDNA, (B) dsDNA, and (C) modified nucleotides as a repair template.
Figure 7. Oligonucleotides can serve as a template to repair DSBs via HR. At the site of CRISPR-Cas9-induced DSB, Single-stranded DNA (ssDNA), double-stranded DNA (dsDNA), and modified repair templates serve as a repair template. (A) ssDNA, (B) dsDNA, and (C) modified nucleotides as a repair template.
Ijms 23 15863 g007
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kim, H.-M.; Hong, Y.; Chen, J. A Decade of CRISPR-Cas Gnome Editing in C. elegans. Int. J. Mol. Sci. 2022, 23, 15863. https://doi.org/10.3390/ijms232415863

AMA Style

Kim H-M, Hong Y, Chen J. A Decade of CRISPR-Cas Gnome Editing in C. elegans. International Journal of Molecular Sciences. 2022; 23(24):15863. https://doi.org/10.3390/ijms232415863

Chicago/Turabian Style

Kim, Hyun-Min, Yebin Hong, and Jiani Chen. 2022. "A Decade of CRISPR-Cas Gnome Editing in C. elegans" International Journal of Molecular Sciences 23, no. 24: 15863. https://doi.org/10.3390/ijms232415863

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