An official website of the United States government
Here's how you know
Official websites use .gov
A
.gov website belongs to an official
government organization in the United States.
Secure .gov websites use HTTPS
A lock (
) or https:// means you've safely
connected to the .gov website. Share sensitive
information only on official, secure websites.
As a library, NLM provides access to scientific literature. Inclusion in an NLM database does not imply endorsement of, or agreement with,
the contents by NLM or the National Institutes of Health.
Learn more:
PMC Disclaimer
|
PMC Copyright Notice
. Author manuscript; available in PMC: 2018 Nov 14.
Published in final edited form as: Nature. 2017 Sep 27;550(7674):114–118. doi: 10.1038/nature24027
A reversible haploid murine embryonic stem cell biobank resource for functional genomics
The publisher's version of this article is available at Nature
Summary
The ability to directly uncover the contributions of genes to a given phenotype is fundamental for biology research. However, ostensibly homogeneous cell populations exhibit large clonal variance1,2 that can confound analyses and undermine reproducibility3. Here, we used genome-saturated mutagenesis to create a biobank of over 100,000 individual haploid murine embryonic stem cell (mESC) lines targeting 16,950 genes with genetically bar-coded, conditional and reversible mutations. This Haplobank is the largest resource of hemi-/homozygous mutant mESCs to date and is available to all researchers. Reversible mutagenesis overcomes clonal variance by permitting functional annotation of the genome directly in sister cells. We utilize Haplobank in reverse genetic screens to investigate the temporal resolution of essential genes in mESCs, and to identify novel genes that control sprouting angiogenesis and blood vessel lineage specification. Further, a genome-wide forward screen with Haplobank identified PLA2G16 as a host factor required for cytotoxicity by rhinoviruses, which cause the common cold. Thus, Haplobank clones and revertible technologies enable high-throughput, reproducible functional annotation of the genome.
Approaches to functionally analyze the mammalian genome include ENU mutagenesis4, gene targeting5, RNA interference6,7, and CRISPR-mediated genome editing8. Although powerful, these approaches suffer from various caveats, such as poor knockdown efficiency and off-target effects9–11. Additionally, clonal variability within populations can compromise comparisons and reproducibility3,12,13,14. Thus, reversibile mutations enabling direct comparison of phenotypes within a single clone are pivotal to study genetic dependencies.
To generate a conditional mutagenesis system at a genome-wide scale and at the clonal level, we applied insertional mutagenesis with genetically bar-coded lentiviral-, retroviral- 15,16 and transposon [Tol217 and Sleeping beauty (SB)18]-based vectors in haploid murine embryonic stem cells (mESCs, Fig. 1a), which enable recessive genetics. We analyzed several parthenogenic haploid mESC lines (Extended Data Fig. 1a,b) and chose to pursue AN3-12 cells (Extended Data Figs. 1c-f, 2 and 3). Insertional mutagenesis permits the integration of invertable splice acceptors, resulting in conditional alleles, as well as high-throughput direct identification of integration sites. To map insertion sites and complex internal bar-codes (>107) by inverse PCR, an optimized universal sequencing strategy was established (Extended Data Fig. 4a,b). A combination of strategies was used to avoid genomic biases of the insertional mutagenesis systems (Fig. 1a,b; Extended Data Figs. 4a-f and 5a, reviewed in19,20), yielding genome-wide mutagenesis (Fig. 1c). Of note, Tol2 outperformed the classical viral delivery systems and even in silico mutagenesis at <1 million integrations (Fig. 1c). The mutagenesis systems also generated anti-sense and intergenic integrations at high frequency due to cryptic transcriptional start sites (Extended Data Fig. 5b,c), however this is not expected to affect disruption of transcription in sense oriantation. Thus, we utilized various delivery systems to obtain unbiased, genome-saturated, and conditional mutagenesis.
Next, we developed a high-throughput pipeline to create a biobank of reversible mutations in AN3-12 haploid mESCs. Starting from haploid mESC pools that contained between 107-108 distinct mutations for each mutagen, we arrayed, processed, banked, and mapped over 100,000 individual mESC clones (Extended Data Fig. 6). In total, we generated sense and anti-sense clones that target 16,970 of ~ 24,000 annotated mouse genes (genome release mm10), covering over 70% of the protein-coding genome (Extended Data Table 1 and Extended Data Fig. 7). Integrations display a 5’ bias in genes and in coding sequence (Extended Data Fig. 8), resulting in truncations that are likely to generate loss-of-function alleles. All pools of mutated cells and clones were deposited to Haplobank, and all mapping information, expression levels, and quality control measures can be accessed at www.haplobank.at. A complete collection of cell culture and molecular protocols was submitted to Nature Protocol Exchange and is available as Supplementary file. This resource represents a comprehensive library of mESC clones carrying hemi/homozygous, twice reversible, bar-coded integrations, combining the power of stem cells with tunable mutagenesis.
A key advantage of Haplobank is the ability to compare each mESC clone with its sister clone carrying the conditionally inverted splice acceptor. As a proof-of-principle, we analyzed two mESC clones containing sense integrations within Ctnna1, which encodes alphaE-catenin and is critical for mESC adhesion21. mESCs with sense integrations exhibited reduced Ctnna1 expression and impaired cell adhesion, which were both restored by FlpO-mediated reversal of the integrated mutagenesis vectors. Further, Cre-mediated reversal back into the sense orientation again disrupted alphaE catenin-mediated cell adhesion (Fig. 1d,e). Additionally, we evaluated mESC lines carrying non-disruptive, anti-sense integrations in presumed essential genes. We infected these mESC lines with a pool of retroviruses that encode Cre and mCherry, or GFP only. If a gene is essential, Cre-mediated reversion of the integration to the disruptive sense orientation should specifically deplete mCherry+ cells from the mCherry+/GFP+ cell pool over time, as detected by flow cytometry. Indeed, we confirmed the essential role of several genes for mESC survival (Fig. 2a, b). Thus, our system allows for a direct functional annotation of essentiality, instead of screening for the absence of mutations; moreover, one can directly examine the penetrance and timing of lethal phenotypes.
Next, we performed a genome-wide screen to uncover novel hits for resistance against infection with a common cold virus. We chose the rhinovirus serotype RV-A1a, which replicates in mouse cells22. A pool of mESCs carrying gene trap insertions was exposed to rhinovirus every other day for 3 weeks. The surviving, virus-resistant cells displayed an enrichment for multiple disruptive insertions in the low density lipoprotein receptor (Ldlr), a known entry portal for this virus (p-value=2.9*10-12), and in the phospholipase Pla2g16 (p-value=1.4*10-11) (Fig. 2c). We confirmed that these genes are required for viral killing using three different sister clones with revertible integrations (Fig. 2d). Next, we used CRISPR-Cas9 technology to disrupt these genes in human embryonic kidney (HEK293T) cells and monitored competitive proliferation with and without RV-A1a infection. In this assay, LDLR was not required for RV-A1a-mediated killing (Fig. 2e), presumably because the virus can enter via other receptors in the absence of functional LDLR in HEK293T cells23. Importantly, we confirmed that inactivation of PLA2G16 with different sgRNAs confers a selective survival advantage to HEK293T cells exposed to RV-A1a (Fig. 2e). Thus, Haplobank enables genome-saturated forward screening and validation to discover novel genes underlying specific phenotypes.
PLA2G16 can catalyze the rate limiting step of arachidonic acid synthesis, and thus couples to cyclooxygenases (COX) and prostaglandin synthesis24,25. However, different COX inhibitory drugs did not block RV-A1a-mediated cell death of control or repaired Pla2g16 sister mESCs, nor did arachidonic acid enhance RV-A1a toxicity (Extended Data Fig. 9a). Selective survival of Pla2g16 or Ldlr mutant mES cells was not affected by COX inhibitors (Extended Data Fig. 9b). Of note, the COX inhibitory drugs ibuprofen and indomethacin conferred partial resistance to RV-A1a in HEK293T cells (Extended Data Fig. 10), albeit at concentrations that also affect other pathways26. Structurally, the short C-terminal vesicular domain of PLA2G16 extends into the endosomal lumen where the virion is located prior to releasing its RNA (Fig. 2f)27. To test the relevance of this domain, we edited the Pla2g16 coding region in the endosomal C-terminus and 3’UTR using CRISPR/Cas9 and selected for RV-A1a resistant cells. Mutations conferring resistance to RV-A1a were enriched in the transmembrane domain and the vesicular domains (Fig. 2f; Extended Data Fig. 11), consistent with a recent, independent haploid screen that identified PLA2G16 in picorna viridae infection28. Our results identify the C-terminal domain of Pla2g16 is a target to block rhinoviral infections.
As a third application of Haplobank, we investigated pathways required for angiogenesis29. Multiple candidate angiogenesis genes have been proposed, but few have been functionally validated30,31. We adapted embryoid body (EB) blood vessel sprouting, which recapitulates key features of in vivo angiogenesis32, to our haploid mESCs. Sprouts stained positive for the endothelial marker CD31 and the basal membrane protein collagen IV, and formed lumens (Fig. 3b). Similar to in vivo blood vessel formation, the cells fronting the vascular structures exhibited characteristic features of tip cells, such as CD31-positive filopodia protrusions followed by stalk cells (Fig. 3b). Tip cells express Delta like ligand 4 (Dll4), which activates the Notch1 pathway on stalk cells to suppress their conversion into tip cells33. We used Notch1 anti-sense non-disruptive clones from Haplobank and, via Cre recombination, created stable sister, sense clones that knock-out Notch1 expression (Extended Data Fig. 12a). EBs derived from multiple sense, knock-out sister clones displayed significantly increased vessel density (Extended Data Fig. 12b and not shown). Gene trap integration upstream of the Notch1 gene did not alter vessel density (Extended Data Fig. 12c). Thus, our EB sprouting assay recapitulates normal blood vessel development.
To identify novel genes, we selected candidates that are more highly expressed in tip versus stalk cells30,31 and were associated with human vascular disease (Extended Data Fig. 13a). We focused on 32 genes,represented in Haplobank. To ensure that differences in angiogenesis are directly linked to inactivation of the respective target genes, and not genetic background noise or clonal effects, we performed color tracing. We infected selected mESCs with GFP- or mCherry-Cre expressing retroviruses to generate sister clones with reverted orientation of the integration (Fig. 3c). We observed large variability between independent clones (Extended Data Fig. 13b,c), highlighting the importance of comparing mutant to genetically repaired sister clones for each gene. Genetic inactivation of Myst3, Mecom, Gja1, Gabrb3, Tnfrsf1a, and Dlg2 reduced sprouting angiogenesis and decreased vessel formation at least 2-fold compared to anti-sense clones; gene trapping of Enpp3, Smarca1, Ndufs4, Plcb1, or Hck promoted blood vessel growth (Fig. 3d,e; Extended Data Fig. 14). Thus, Haplobank enabled rapid, functional and reproducible validation of candidate angiogenesis genes in engineered blood vessels.
To assess the in vivo role of these novel angiogenesis genes, we generated EBs from mESCs stably expressing mCherry and injected them into immuno-compromised mice. The resulting teratomas were assessed for mESC-derived mCherry+ IB4+ blood vessels (Fig. 4a, Extended Data Fig. 15a). To control for teratoma growth rates, we injected 1:1 mixed mosaic EBs of sense (mutant, GFP+) and their respective anti-sense clones (repaired, mCherry+). The contribution of GFP- or mCherry-expressing cells to the endothelial lineage as well as non-endothelial tissues was assessed by cytometry (Extended Data Fig. 15b). Mutated clones of Myst3, Gja1 and Grin2b displayed a decreased contribution to IB4+ vasculature (Fig. 4a; Extended Data Fig. 15b). In contrast, mutant clones that showed increased vessel density in vitro exhibited a greater capacity to form blood vessels in vivo than their anti-sense sister clones (Fig. 4a). These data were confirmed using in situ blood vessel analysis of mixed teratomas (Fig. 4b). To test whether the identified genes modulate angiogenesis via specification of tip cell fate, we performed mosaic tip cell competition assays (Extended Data Fig. 15c,d). As a positive control, we assessed genetic modulation of the Notch1 pathway. As expected, mCherry+ (sense knock-out) Notch1 mutant cells preferentially localized to the tip position compared to GFP+ anti-sense Notch1-expressing sister clones (Fig. 4c,d). Most clones carrying gene mutations that increased sprouting activity displayed a significant increase to the tip cell position, and vice versa (Fig. 4c-e). Therefore, the newly identified angiogenesis genes can control the tip cell fate.
We tested whether one of our novel angiogenesis genes, Gja1, encoding the gap junction protein Connexin43 34, is involved in physiological vascularization of the mouse retina, which begins at birth and progresses until postnatal day 7 35. At postnatal day 6, we observed high Gja1/Connexin43 expression at the angiogenic front where Gja1/Connexin43 localized to endothelial junctions, with the highest intensity in tip cells (Extended Data Fig. 16a). At the vascular plexus, Gja1/Connexin43 expression was predominantly detected in perivascular cells, not endothelial cells (Extended Data Fig. 16b). Moreover, Gja1/Connexin43 was primarily detected at the tip of the developing vascular sprout in 3D blood vessels (Extended Data Fig. 16c). Newborn mice injected i.v. with a Gja1/Connexin43 blocking peptide displayed a delay in vascular network progression and complexity in the retina compared to those injected with a scrambled control peptide36 (Fig. 4f). The number of tip cells at the angiogenic front was decreased, together with decreased numbers of branch points in the vascular plexus (Fig. 4f). Thus, Gja1/Connexin43 is a key regulator of tip cell fate and physiological angiogenesis in vivo.
In summary, the Haplobank resource contains over 100,000 individually mutagenized and bar-coded mESC lines targeting 16,970 protein coding genes. Haplobank complements a collection of 3,396 reversibley targeted genes in a near haploid human leukemia cell line45. Our proof-of-principle experiments uncovering genes required for rhinovirus infection and angiogenesis establish the power of Haplobank in forward and reverse genetic screens, respectively. The strong variability between independent clones revealed the importance of assessing mutant and repaired clones side-by-side, and addresses an increased demand for rigor and reproductibility37. Thus, Haplobank clones and revertible technologies enable high-throughput, reproducible functional annotation of the genome.
Extended Data
Extended Data Table 1. Haplobank clones.
Numbers of clones available with respect to different mutagens, orientation of the inserted gene trap to gene transcription, as well as the number of different genes hit. A gene is defined as the genomic region between the transcriptional start and stop sites. www.haplobank.at (Please access as User: BetaForReviewer; password: ready2GoLive).
We thank all members of our laboratories, IMBA/IMP and VBCF services for support and Life Science Editors for assistance. We thank Britta Knapp, Ireos Filipuzzi, and Thomas Aust for clone picking, N. Rao Movva and Tewis Bouwmeester (NIBR) for support, and Kristina Handler for the differentiation protocols. Haplobank is generously funded by Nestlé Institute of Health Science NIHS, the Austrian National Bank (OeNB), an Advanced ERC grant and Era of Hope/National Coalition against Breast Cancer/DoD (to J.M.P.). U.E. is a Wittgenstein Prize fellow. D.B is supported by FWF P23308-B13. A.S. is supported by an ERC Consolidator Grant, Boehringer Ingelheim, and FFG.
Footnotes
Author Contributions. U.E. generated the haploid library with technical support from An.L., C.H., J.L., M.H., A.-M.H., M.K., N.S., E.W., N.V.D.L, J.M., P.D., D.H., R.R., D.E.. U.E., R.AW and Al.L. characterized cell line. An.L., G.M., U.E., D.B., T.D., T.B., and T.M. performed Rhinovirus work. A.S. wrote bioinformatics algorithms and set-up the Haplobank webpage. S.Z. performed RACE experiments, F.Y. and B.F. karyotyping, C.A.A. supported standardization. J.A.Z.M. and O.B. performed ATACseq. Z.I. advised on mutagenesis vectors. R.A.W., I.A., D.A., Al.L, and H.G. performed blood vessel experiments. U.E. and J.M.P. coordinated the project.
The authors declare no competing financial interests.
5.Robertson E, Bradley A, Kuehn M, Evans M. Germ-line transmission of genes introduced into cultured pluripotential cells by retroviral vector. Nature. 1986;323:445–8. doi: 10.1038/323445a0. [DOI] [PubMed] [Google Scholar]
6.Elbashir SM, et al. Duplexes of 21 ± nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature. 2001;411:494–498. doi: 10.1038/35078107. [DOI] [PubMed] [Google Scholar]
7.Brummelkamp TR, Bernards R, Agami R. A system for stable expression of short interfering RNAs in mammalian cells. Science. 2002;296:550–553. doi: 10.1126/science.1068999. [DOI] [PubMed] [Google Scholar]
8.Jinek M, et al. A Programmable Dual-RNA – Guided. Science. 2012;337:816–822. doi: 10.1126/science.1225829. [DOI] [PMC free article] [PubMed] [Google Scholar]
9.Fu Y, et al. High-frequency off-target mutagenesis induced by CRISPR-Cas nucleases in human cells. Nat Biotechnol. 2013;31:822–6. doi: 10.1038/nbt.2623. [DOI] [PMC free article] [PubMed] [Google Scholar]
10.Morgens DW, Deans RM, Li A, Bassik MC. Systematic comparison of CRISPR / Cas9 and RNAi screens for essential genes. Nat Biotechnol. 2016;34:1–4. doi: 10.1038/nbt.3567. [DOI] [PMC free article] [PubMed] [Google Scholar]
11.Evers B, et al. CRISPR knockout screening outperforms shRNA and CRISPRi in identifying essential genes. Nat Biotechnol. 2016;34:11–14. doi: 10.1038/nbt.3536. [DOI] [PubMed] [Google Scholar]
12.Rouhani F, et al. Genetic Background Drives Transcriptional Variation in Human Induced Pluripotent Stem Cells. PLoS Genet. 2014;10 doi: 10.1371/journal.pgen.1004432. [DOI] [PMC free article] [PubMed] [Google Scholar]
13.Fedorov LM, Haegel-Kronenberger H, Hirchenhain J. A comparison of the germline potential of differently aged ES cell lines and their transfected descendants. Transgenic Res. 1997;6:223–231. doi: 10.1023/a:1018446325492. [DOI] [PubMed] [Google Scholar]
14.Echeverri CJ, et al. Minimizing the risk of reporting false positives in large-scale RNAi screens. Nat Methods. 2006;3:777–9. doi: 10.1038/nmeth1006-777. [DOI] [PubMed] [Google Scholar]
15.Schnutgen F, et al. Genomewide production of multipurpose alleles for the functional analysis of the mouse genome. Proc Natl Acad Sci U S A. 2005;102:7221–7226. doi: 10.1073/pnas.0502273102. [DOI] [PMC free article] [PubMed] [Google Scholar]
17.Ika Mayasari N, et al. Mixture of differentially tagged Tol2 transposons accelerates conditional disruption of a broad spectrum of genes in mouse embryonic stem cells. Nucleic Acids Res. 2012;40 doi: 10.1093/nar/gks262. [DOI] [PMC free article] [PubMed] [Google Scholar]
18.Ivics Z, Hackett PB, Plasterk RH, Izsvák Z. Molecular Reconstruction of Sleeping Beauty, a Tc1-like Transposon from Fish, and Its Transposition in Human Cells. Cell. 1997;91:501–510. doi: 10.1016/s0092-8674(00)80436-5. [DOI] [PubMed] [Google Scholar]
19.Bellen HJ, et al. The Drosophila gene disruption project: Progress using transposons with distinctive site specificities. Genetics. 2011;188:731–743. doi: 10.1534/genetics.111.126995. [DOI] [PMC free article] [PubMed] [Google Scholar]
21.Torres M, et al. An alpha-E-catenin gene trap mutation defines its function in preimplantation development. Proc Natl Acad Sci U S A. 1997;94:901–6. doi: 10.1073/pnas.94.3.901. [DOI] [PMC free article] [PubMed] [Google Scholar]
22.Reithmayer M, Reischl A, Snyers L, Blaas D. Species-specific receptor recognition by a minor-group human rhinovirus (HRV): HRV serotype 1A distinguishes between the murine and the human low-density lipoprotein receptor. J Virol. 2002;76:6957–6965. doi: 10.1128/JVI.76.14.6957-6965.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
23.Hofer F, et al. Members of the low density lipoprotein receptor family mediate cell entry of a minor-group common cold virus. Proc Natl Acad Sci. 1994;91:1839–1842. doi: 10.1073/pnas.91.5.1839. [DOI] [PMC free article] [PubMed] [Google Scholar]
24.Jaworski K, et al. AdPLA ablation increases lipolysis and prevents obesity induced by high-fat feeding or leptin deficiency. Nat Med. 2009;15:159–168. doi: 10.1038/nm.1904. [DOI] [PMC free article] [PubMed] [Google Scholar]
25.Duncan RE, Sarkadi-Nagy E, Jaworski K, Ahmadian M, Hei SS. Identification and functional characterization of adipose-specific phospholipase A2 (AdPLA) J Biol Chem. 2008;283:25428–25436. doi: 10.1074/jbc.M804146200. [DOI] [PMC free article] [PubMed] [Google Scholar]
26.Tsutsumi S, et al. Endoplasmic reticulum stress response is involved in nonsteroidal anti-inflammatory drug-induced apoptosis. Cell Death Differ. 2004;11:1009–1016. doi: 10.1038/sj.cdd.4401436. [DOI] [PubMed] [Google Scholar]
27.Uyama T, et al. Interaction of Phospholipase A/acyltransferase-3 with Pex19p: A possible involvement in the down-regulation of peroxisomes. J Biol Chem. 2015;290:17520–17534. doi: 10.1074/jbc.M114.635433. [DOI] [PMC free article] [PubMed] [Google Scholar]
28.Staring J, et al. PLA2G16 represents a switch between entry and clearance of Picornaviridae. Nature. 2017;541:412–416. doi: 10.1038/nature21032. [DOI] [PubMed] [Google Scholar]
29.Potente M, Gerhardt H, Carmeliet P. Basic and therapeutic aspects of angiogenesis. Cell. 2011;146:873–887. doi: 10.1016/j.cell.2011.08.039. [DOI] [PubMed] [Google Scholar]
30.Del Toro R, et al. Identification and functional analysis of endothelial tip cell-enriched genes. Blood. 2010;116:4025–4033. doi: 10.1182/blood-2010-02-270819. [DOI] [PMC free article] [PubMed] [Google Scholar]
31.Strasser GA, Kaminker JS, Tessier-Lavigne M. Microarray analysis of retinal endothelial tip cells identifies CXCR4 as a mediator of tip cell morphology and branching. Blood. 2010;115:5102–5110. doi: 10.1182/blood-2009-07-230284. [DOI] [PubMed] [Google Scholar]
32.Jakobsson L, et al. Endothelial cells dynamically compete for the tip cell position during angiogenic sprouting. Nat Cell Biol. 2010;12:943–953. doi: 10.1038/ncb2103. [DOI] [PubMed] [Google Scholar]
33.Hellström M, et al. Dll4 signalling through Notch1 regulates formation of tip cells during angiogenesis. Nature. 2007;445:776–780. doi: 10.1038/nature05571. [DOI] [PubMed] [Google Scholar]
34.Valdimarsson G, De Sousa PA, Beyer EC, Paul DL, Kidder GM. Zygotic expression of the connexin43 gene supplies subunits for gap junction assembly during mouse preimplantation development. Mol Reprod Dev. 1991;30:18–26. doi: 10.1002/mrd.1080300103. [DOI] [PubMed] [Google Scholar]
35.Pitulescu ME, Schmidt I, Benedito R, Adams RH. Inducible gene targeting in the neonatal vasculature and analysis of retinal angiogenesis in mice. Nat Protoc. 2010;5:1518–34. doi: 10.1038/nprot.2010.113. [DOI] [PubMed] [Google Scholar]
36.Chaytor AT, Martin PE, Edwards DH, Griffith TM. Gap junctional communication underpins EDHF-type relaxations evoked by ACh in the rat hepatic artery. Am J Physiol Heart Circ Physiol. 2001;280:H2441–H2450. doi: 10.1152/ajpheart.2001.280.6.H2441. [DOI] [PubMed] [Google Scholar]
37.Baker M. 1,500 Scientists Lift the Lid on Reproducibility. Nature. 2016;533:452–454. doi: 10.1038/533452a. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.