CRISPR modified cell lines

CRISPR modified cell lines

We are using CRISPR gene editing technologies to generate new cell lines as part of the funded project NIH ORIP R24 OD019847 "Next-generation Drosophila cell lines to elucidate the cellular basis of human diseases" (N. Perrimon, PI; A. Simcox, Co-PI).

GFP-tagged knock-in cell lines

As part of the ORIP-funded project, we are making GFP-tagged cell lines, with an emphasis on visualization of various organelles and sub-cellular compartments. The following GFP knock-in cell lines made at the DRSC in collaboration with the Bellne lab are available for distribution by the DGRC in Bloomington, IN. 

Note that the parental cell line is positive for an mCherry fusion and Cas9, as the parental cell line is S2R+-MT::Cas9 (DGRC cell catalog #268), which is described in Viswanatha et al. 2018 (PubMed ID 30051818). This parental cell line was itself derived from DRSC cell line S2R+ NPT005 (DGRC cell catalog #229), which is described in Neumuller et al. 2012 (PubMed ID 22174071).

S2R+ with GFP::Cnx99a. Ordering information: DGRC cell catalog ID #273
S2R+ with GFP::Rab11. Ordering information: DGRC cell catalog ID #274
S2R+ with GFP::Polo. Ordering information: DGRC cell catalog ID #275
S2R+ with GFP::Gmap (clone #4). Ordering information: DGRC cell catalog ID #276
S2R+ with GFP::Gmap (clone $7). Ordering information: DGRC cell catalog ID #277
S2R+ with GFP::Fib (clone #11). Ordering information: DGRC cell catalog ID #278
S2R+ with GFP::Fib (clone #12). Ordering information: DGRC cell catalog ID #279
S2R+ with GFP::Golgin. Ordering information: DGRC cell catalog ID #280
S2R+ with GFP::Arl8. Ordering information: DGRC cell catalog ID #291
S2R+ with GFP::Lam. Ordering information: DGRC cell catalog ID #292
S2R+ with GFP::Spin. Ordering information: DGRC cell catalog ID #293
S2R+ with GFP::Sec23. Ordering information: DGRC cell catalog ID #294
S2R+ with GFP::Tom20. Ordering information: DGRC cell catalog ID #302

These cell lines were made using constructs designed and provided by Kanca and Bellen (Baylor College of Medicine). The cell lines were engineered, isolated, and validated at the DRSC. Validation testing included live-cell imaging, fixed-cell imaging (co-stained with an antibody, when possible), and molecular characterization of the insertion endpoints.

See the file attached below for a slide presentation with images of live and fixed cells, as well as other relevant information about the approach and the resulting GFP-tagged cell lines.

In addition, C-terminal GFP knock-in cell lines were generated as described in a BioRxiv preprint from Bosch et al. (2019) using an 'armless' donor approach. Act5c::GFP, Tub84B::GFP, His2Av::GFP, and Lamin::GFP fusion cell lines were made using this approach and are being shared with the DGRC for distribution to the community.

Knockout cell lines

S2R+-ZnT63C-KO, NHEJ-mediated knockout of ZnT63C. As described in PMID: 29223976. Ordering information: DGRC cell catalog #265.
S2R+-IA2-KO, NHEJ-mediated knockout of ia2. As described in PMID: 29223976. Ordering information: DGRC cell catalog #266.
S2R+-Apc-KO, two independent cell lines made with one method, DGRC cell catalog #271 and DGRC cell catalog #272
S2R+-Apc-KO, one additional cell line made with a different method, DGRC cell catalog #270
S2R+-gig-KO, DGRC cell catalog #297
S2R+-hairy-KO, DGRC cell catalog #298
S2R+-Moe-KO, DGRC cell catalog #303
S2R+-Pex19-KO, DGRC cell catalog #306
S2R+-Pten-KO, DGRC cell catalog #307
S2R+-Slik-KO, DGRC cell catalog #308
S2R+-Tnks-KO, three independent cell lines, DGRC cell catalog #299, DGRC cell catalog #300, and DGRC cell catalog #301
S2R+-Tnks-KO, one additional cell line made with a different method, DGRC cell catalog #304
S2R+-TSC1-KO, DGRC cell catalog #305
S2R+-Yki-KO, DGRC cell catalog #309

These cell lines have been sequence verified as containing only knockout alleles by PCR amplification of the target region followed by next-generation sequencing of the PCR amplicon, contig assembly, and comparison with the wild-type reference sequence (or, for KO alleles generated via knock-in, verified using PCR validation of the insertion allele). We wanted to make sure that the distribution copies of the cell lines are correct. To do this, for a subset of these cell lines, (1) the DGRC prepared genomic DNA from their distribution copies of the cell lines, (2) they shipped that gDNA to the DRSC/TRiP, and (3) we used that gDNA as template for PCR and NGS, and validated that all alleles are predicted to be gene knockout alleles. Information relevant to re-validation in your own lab is provided at the DGRC website.

Did you request GFP-tagged or KO cells from the DGRC and use them in a study? If so, please acknowledge both the cell line developers (DRSC) and the distribtors (DGRC) by citing NIH Grant 5R24OD019847, which supported production of the resource at DRSC/TRiP, and the Drosophila Genome Resource Center, NIH grant 2P40OD010949, as well as relevant pulications.

Slide presentation file -- GFP-tagged cell lines:

smohr_drsc_gfp-tag_websharecopy.pdf12.23 MB

Publications

Nisha Singh, Agustin Rolandelli, Anya J O’Neal, Rainer L. Butler, Sourabh Samaddar, Hanna J Laukaitis-Yousey, Matthew Butnaru, Stephanie E Mohr, Norbert Perrimon, and Joao HF Pedra. 9/8/2023. “Genetic manipulation of an Ixodes scapularis cell line.” bioRxiv, Pp. 9/8/2023. 09.08.556855. Publisher's VersionAbstract
Although genetic manipulation is one of the hallmarks in model organisms, its applicability to non-model species has remained difficult due to our limited understanding of their fundamental biology. For instance, manipulation of a cell line originated from the blacklegged tick Ixodes scapularis, an arthropod that serves as a vector of several human pathogens, has yet to be established. Here, we demonstrate the successful genetic modification of the commonly used tick ISE6 line through ectopic expression and clustered regularly interspaced palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) genome editing. We performed ectopic expression using nucleofection and attained CRISPR-Cas9 editing via homology dependent recombination. Targeting the E3 ubiquitin ligase X-linked inhibitor of apoptosis (xiap) and its substrate p47 led to alteration in molecular signaling within the immune deficiency (IMD) network and increased infection of the rickettsial agent Anaplasma phagocytophilum in I. scapularis ISE6 cells. Collectively, our findings complement techniques for genetic engineering of ticks in vivo and aid in circumventing the long-life cycle of I. scapularis, of which limits efficient and scalable molecular genetic screens.Importance Genetic engineering in arachnids has lagged compared to insects, largely because of substantial differences in their biology. This study unveils the implementation of ectopic expression and CRISPR-Cas9 gene editing in a tick cell line. We introduced fluorescently tagged proteins in ISE6 cells and edited its genome via homology dependent recombination. We ablated the expression of xiap and p47, two signaling molecules present in the immune deficiency (IMD) pathway of I. scapularis. Impairment of the tick IMD pathway, an analogous network of the tumor necrosis factor receptor in mammals, led to enhanced infection of the rickettsial agent A. phagocytophilum. Altogether, our findings provide a critical technical resource to the scientific community to enable a deeper understanding of biological circuits in the blacklegged tick Ixodes scapularis.Competing Interest StatementThe authors have declared no competing interest.
Agustin Rolandelli, Hanna J Laukaitis-Yousey, Haikel N Bogale, Nisha Singh, Sourabh Samaddar, Anya J O’Neal, Camila R Ferraz, Matthew Butnaru, Enzo Mameli, Baolong Xia, Tays M. Mendes, Rainer L. Butler, Liron Marnin, Francy ECabrera Paz, Luisa M Valencia, Vipin S Rana, Ciaran Skerry, Utpal Pal, Stephanie E Mohr, Norbert Perrimon, David Serre, and Joao HF Pedra. 2023. “Tick hemocytes have pleiotropic roles in microbial infection and arthropod fitness.” bioRxiv, Pp. 2023.08.31.555785. Publisher's VersionAbstract
Uncovering the complexity of systems in non-model organisms is critical for understanding arthropod immunology. Prior efforts have mostly focused on Dipteran insects, which only account for a subset of existing arthropod species in nature. Here, we describe immune cells or hemocytes from the clinically relevant tick Ixodes scapularis using bulk and single cell RNA sequencing combined with depletion via clodronate liposomes, RNA interference, Clustered Regularly Interspaced Short Palindromic Repeats activation (CRISPRa) and RNA-fluorescence in situ hybridization (FISH). We observe molecular alterations in hemocytes upon tick infestation of mammals and infection with either the Lyme disease spirochete Borrelia burgdorferi or the rickettsial agent Anaplasma phagocytophilum. We predict distinct hemocyte lineages and reveal clusters exhibiting defined signatures for immunity, metabolism, and proliferation during hematophagy. Furthermore, we perform a mechanistic characterization of two I. scapularis hemocyte markers: hemocytin and astakine. Depletion of phagocytic hemocytes affects hemocytin and astakine levels, which impacts blood feeding and molting behavior of ticks. Hemocytin specifically affects the c-Jun N-terminal kinase (JNK) signaling pathway, whereas astakine alters hemocyte proliferation in I. scapularis. Altogether, we uncover the heterogeneity and pleiotropic roles of hemocytes in ticks and provide a valuable resource for comparative biology in arthropods.Competing Interest StatementThe authors have declared no competing interest.
Baolong Xia, Raghuvir Viswanatha, Yanhui Hu, Stephanie E Mohr, and Norbert Perrimon. 2023. “Pooled genome-wide CRISPR activation screening for rapamycin resistance genes in cells.” Elife, 12.Abstract

Loss-of-function and gain-of-function genetic perturbations provide valuable insights into gene function. In cells, while genome-wide loss-of-function screens have been extensively used to reveal mechanisms of a variety of biological processes, approaches for performing genome-wide gain-of-function screens are still lacking. Here, we describe a pooled CRISPR activation (CRISPRa) screening platform in cells and apply this method to both focused and genome-wide screens to identify rapamycin resistance genes. The screens identified three genes as novel rapamycin resistance genes: a member of the SLC16 family of monocarboxylate transporters (), a member of the lipocalin protein family (), and a zinc finger C2H2 transcription factor (). Mechanistically, we demonstrate that overexpression activates the RTK-Akt-mTOR signaling pathway and that activation of insulin receptor (InR) by requires cholesterol and clathrin-coated pits at the cell membrane. This study establishes a novel platform for functional genetic studies in cells.

Ying Xu, Raghuvir Viswanatha, Oleg Sitsel, Daniel Roderer, Haifang Zhao, Christopher Ashwood, Cecilia Voelcker, Songhai Tian, Stefan Raunser, Norbert Perrimon, and Min Dong. 2022. “CRISPR screens in Drosophila cells identify Vsg as a Tc toxin receptor.” Nature, 610, 7931, Pp. 349-355.Abstract
Entomopathogenic nematodes are widely used as biopesticides1,2. Their insecticidal activity depends on symbiotic bacteria such as Photorhabdus luminescens, which produces toxin complex (Tc) toxins as major virulence factors3-6. No protein receptors are known for any Tc toxins, which limits our understanding of their specificity and pathogenesis. Here we use genome-wide CRISPR-Cas9-mediated knockout screening in Drosophila melanogaster S2R+ cells and identify Visgun (Vsg) as a receptor for an archetypal P. luminescens Tc toxin (pTc). The toxin recognizes the extracellular O-glycosylated mucin-like domain of Vsg that contains high-density repeats of proline, threonine and serine (HD-PTS). Vsg orthologues in mosquitoes and beetles contain HD-PTS and can function as pTc receptors, whereas orthologues without HD-PTS, such as moth and human versions, are not pTc receptors. Vsg is expressed in immune cells, including haemocytes and fat body cells. Haemocytes from Vsg knockout Drosophila are resistant to pTc and maintain phagocytosis in the presence of pTc, and their sensitivity to pTc is restored through the transgenic expression of mosquito Vsg. Last, Vsg knockout Drosophila show reduced bacterial loads and lethality from P. luminescens infection. Our findings identify a proteinaceous Tc toxin receptor, reveal how Tc toxins contribute to P. luminescens pathogenesis, and establish a genome-wide CRISPR screening approach for investigating insecticidal toxins and pathogens.
Hans M Dalton, Raghuvir Viswanatha, Roderick Brathwaite, Jae Sophia Zuno, Alexys R Berman, Rebekah Rushforth, Stephanie E Mohr, Norbert Perrimon, and Clement Y Chow. 2022. “A genome-wide CRISPR screen identifies DPM1 as a modifier of DPAGT1 deficiency and ER stress.” PLoS Genet, 18, 9, Pp. e1010430.Abstract
Partial loss-of-function mutations in glycosylation pathways underlie a set of rare diseases called Congenital Disorders of Glycosylation (CDGs). In particular, DPAGT1-CDG is caused by mutations in the gene encoding the first step in N-glycosylation, DPAGT1, and this disorder currently lacks effective therapies. To identify potential therapeutic targets for DPAGT1-CDG, we performed CRISPR knockout screens in Drosophila cells for genes associated with better survival and glycoprotein levels under DPAGT1 inhibition. We identified hundreds of candidate genes that may be of therapeutic benefit. Intriguingly, inhibition of the mannosyltransferase Dpm1, or its downstream glycosylation pathways, could rescue two in vivo models of DPAGT1 inhibition and ER stress, even though impairment of these pathways alone usually causes CDGs. While both in vivo models ostensibly cause cellular stress (through DPAGT1 inhibition or a misfolded protein), we found a novel difference in fructose metabolism that may indicate glycolysis as a modulator of DPAGT1-CDG. Our results provide new therapeutic targets for DPAGT1-CDG, include the unique finding of Dpm1-related pathways rescuing DPAGT1 inhibition, and reveal a novel interaction between fructose metabolism and ER stress.
Jonathan Zirin, Justin Bosch, Raghuvir Viswanatha, Stephanie E Mohr, and Norbert Perrimon. 2022. “State-of-the-art CRISPR for in vivo and cell-based studies in Drosophila.” Trends Genet, 38, 5, Pp. 437-453.Abstract
For more than 100 years, the fruit fly, Drosophila melanogaster, has served as a powerful model organism for biological and biomedical research due to its many genetic and physiological similarities to humans and the availability of sophisticated technologies used to manipulate its genome and genes. The Drosophila research community quickly adopted CRISPR technologies and, in the 8 years since the first clustered regularly interspaced short palindromic repeats (CRISPR) publications in flies, has explored and innovated methods for mutagenesis, precise genome engineering, and beyond. Moreover, the short lifespan and ease of genetics have made Drosophila an ideal testing ground for in vivo applications and refinements of the rapidly evolving set of CRISPR-associated (CRISPR-Cas) tools. Here, we review innovations in delivery of CRISPR reagents, increased efficiency of cutting and homology-directed repair (HDR), and alternatives to standard Cas9-based approaches. While the focus is primarily on in vivo systems, we also describe the role of Drosophila cultured cells as both an indispensable first step in the process of assessing new CRISPR technologies and a platform for genome-wide CRISPR pooled screens.
Hans M. Dalton, Raghuvir Viswanatha, Ricky Brathwaite Jr., Jae Sophia Zuno, Stephanie E Mohr, Norbert Perrimon, and Clement Y. Chow. 12/4/2021. “A genome-wide CRISPR screen identifies the glycosylation enzyme DPM1 as a modifier of DPAGT1 deficiency and ER stress.” BioRxiv. Publisher's VersionAbstract
Partial loss-of-function mutations in glycosylation pathways underlie a set of rare diseases called Congenital Disorders of Glycosylation (CDGs). In particular, DPAGT1 CDG is caused by mutations in the gene encoding the first step in N-glycosylation, DPAGT1, and this disorder currently lacks effective therapies. To identify potential therapeutic targets for DPAGT1-CDG, we performed CRISPR knockout screens in Drosophila cells for genes associated with better survival and glycoprotein levels under DPAGT1 inhibition. We identified hundreds of candidate genes that may be of therapeutic benefit. Intriguingly, inhibition of the mannosyltransferase Dpm1, or its downstream glycosylation pathways, could rescue two in vivo models of DPAGT1 inhibition and ER stress, even though impairment of these pathways alone usually cause CDGs. While both in vivo models ostensibly cause ER stress (through DPAGT1 inhibition or a misfolded protein), we found a novel difference in fructose metabolism that may indicate glycolysis as a modulator of DPAGT1-CDG. Our results provide new therapeutic targets for DPAGT1-CDG, include the unique finding of Dpm1-related pathways rescuing DPAGT1 inhibition, and reveal a novel interaction between fructose metabolism and ER stress.
Raghuvir Viswanatha, Enzo Mameli, Jonathan Rodiger, Pierre Merckaert, Fabiana Feitosa-Suntheimer, Tonya M Colpitts, Stephanie E Mohr, Yanhui Hu, and Norbert Perrimon. 11/24/2021. “Bioinformatic and cell-based tools for pooled CRISPR knockout screening in mosquitos.” Nat Commun, 12, 1, Pp. 6825.Abstract
Mosquito-borne diseases present a worldwide public health burden. Current efforts to understand and counteract them have been aided by the use of cultured mosquito cells. Moreover, application in mammalian cells of forward genetic approaches such as CRISPR screens have identified essential genes and genes required for host-pathogen interactions, and in general, aided in functional annotation of genes. An equivalent approach for genetic screening of mosquito cell lines has been lacking. To develop such an approach, we design a new bioinformatic portal for sgRNA library design in several mosquito genomes, engineer mosquito cell lines to express Cas9 and accept sgRNA at scale, and identify optimal promoters for sgRNA expression in several mosquito species. We then optimize a recombination-mediated cassette exchange system to deliver CRISPR sgRNA and perform pooled CRISPR screens in an Anopheles cell line. Altogether, we provide a platform for high-throughput genome-scale screening in cell lines from disease vector species.
Raghuvir Viswanatha, Enzo Mameli, Jonathan Rodiger, Pierre Merckaert, Fabiana Feitosa-Suntheimer, Tonya M. Colpitts, Stephanie E. Mohr, Yanhui Hu, and Norbert Perrimon. 3/30/2021. “Bioinformatic and cell-based tools for pooled CRISPR knockout screening in mosquitos [NOTE: A modified final version was published in Nat Comm and is now available.].” bioRxiv. Publisher's VersionAbstract
Mosquito-borne diseases present a worldwide public health burden. Genome-scale screening tools that could inform our understanding of mosquitos and their control are lacking. Here, we adapt a recombination-mediated cassette exchange system for delivery of CRISPR sgRNA libraries into cell lines from several mosquito species and perform pooled CRISPR screens in an Anopheles cell line. To implement this method, we engineered modified mosquito cell lines, validated promoters and developed bioinformatics tools for multiple mosquito species.Competing Interest StatementThe authors have declared no competing interest.
J. A. Bosch, G. Birchak, and N. Perrimon. 2021. “Precise genome engineering in Drosophila using prime editing.” Proc Natl Acad Sci U S A, 118.Abstract
Precise genome editing is a valuable tool to study gene function in model organisms. Prime editing, a precise editing system developed in mammalian cells, does not require double-strand breaks or donor DNA and has low off-target effects. Here, we applied prime editing for the model organism Drosophila melanogaster and developed conditions for optimal editing. By expressing prime editing components in cultured cells or somatic cells of transgenic flies, we precisely introduce premature stop codons in three classical visible marker genes, ebony, white, and forked Furthermore, by restricting editing to germ cells, we demonstrate efficient germ-line transmission of a precise edit in ebony to 36% of progeny. Our results suggest that prime editing is a useful system in Drosophila to study gene function, such as engineering precise point mutations, deletions, or epitope tags.
R. Viswanatha, M. Zaffagni, J. Zirin, N. Perrimon, and S. Kadener. 11/1/2020. “CRISPR-Cas13 mediated Knock Down in Drosophila cultured cells.” BioRxiv.Abstract
Manipulation of gene expression is one of the best approaches for studying gene function in vivo. CRISPR-Cas13 has the potential to be a powerful technique for manipulating RNA expression in diverse animal systems in vivo, including Drosophila melanogaster. Studies using Cas13 in mammalian cell lines for gene knockdown showed increased on-target efficiency and decreased off-targeting relative to RNAi. Moreover, catalytically inactive Cas13 fusions can be used to image RNA molecules, install precise changes to the epitranscriptome, or alter splicing. However, recent studies have suggested that there may be limitations to the deployment of these tools in Drosophila, so further optimization of the system is required. Here, we report a new set of PspCas13b and RfxCas13d expression constructs and use these reagents to successfully knockdown both reporter and endogenous transcripts in Drosophila cells. As toxicity issues have been reported with high level of Cas13, we effectively decreased PspCas13b expression without impairing its function by tuning down translation. Furthermore, we altered the spatial activity of both PspCas13b and RfxCas13d by introducing Nuclear Exportation Sequences (NES) and Nuclear Localization Sequences (NLS) while maintaining activity. Finally, we generated a stable cell line expressing RfxCas13d under the inducible metallothionein promoter, establishing a useful tool for high-throughput genetic screening. Thus, we report new reagents for performing RNA CRISPR-Cas13 experiments in Drosophila, providing additional Cas13 expression constructs that retain activity.
Yanhui Hu, Aram Comjean, Jonathan Rodiger, Yifang Liu, Yue Gao, Verena Chung, Jonathan Zirin, Norbert Perrimon, and Stephanie E Mohr. 2020. “FlyRNAi.org-the database of the Drosophila RNAi screening center and transgenic RNAi project: 2021 update.” Nucleic Acids Res.Abstract
The FlyRNAi database at the Drosophila RNAi Screening Center and Transgenic RNAi Project (DRSC/TRiP) provides a suite of online resources that facilitate functional genomics studies with a special emphasis on Drosophila melanogaster. Currently, the database provides: gene-centric resources that facilitate ortholog mapping and mining of information about orthologs in common genetic model species; reagent-centric resources that help researchers identify RNAi and CRISPR sgRNA reagents or designs; and data-centric resources that facilitate visualization and mining of transcriptomics data, protein modification data, protein interactions, and more. Here, we discuss updated and new features that help biological and biomedical researchers efficiently identify, visualize, analyze, and integrate information and data for Drosophila and other species. Together, these resources facilitate multiple steps in functional genomics workflows, from building gene and reagent lists to management, analysis, and integration of data.
More