High-throughput Functional Genomics Using CRISPR–Cas9

2y ago
14 Views
2 Downloads
694.86 KB
13 Pages
Last View : 11d ago
Last Download : 3m ago
Upload by : Gideon Hoey
Transcription

REVIEWSHigh-throughput functional genomicsusing CRISPR–Cas9Ophir Shalem, Neville E. Sanjana and Feng ZhangAbstract Forward genetic screens are powerful tools for the discovery and functionalannotation of genetic elements. Recently, the RNA-guided CRISPR (clustered regularlyinterspaced short palindromic repeat)-associated Cas9 nuclease has been combined withgenome-scale guide RNA libraries for unbiased, phenotypic screening. In this Review, wedescribe recent advances using Cas9 for genome-scale screens, including knockoutapproaches that inactivate genomic loci and strategies that modulate transcriptionalactivity. We discuss practical aspects of screen design, provide comparisons with RNAinterference (RNAi) screening, and outline future applications and challenges.Small interfering RNA(siRNA). RNA molecules thatare 21–23 nucleotides longand that are processed fromlong double-stranded RNAs;they are functionalcomponents of theRNA-induced silencingcomplex (RISC). siRNAstypically target and silencemRNAs by binding perfectlycomplementary sequences inthe mRNA and causing theirdegradation and/ortranslational inhibition.Broad Institute of MIT andHarvard, 7 Cambridge Center,Cambridge, Massachusetts02142, USA; McGovernInstitute for Brain Research,Department of Brainand Cognitive Sciences, andDepartment of BiologicalEngineering, MassachusettsInstitute of Technology,77 Massachusetts Avenue,Cambridge, Massachusetts02139, USA.Correspondence to O.S. andF.Z. e-mails: ophir.shalem@gmail.com; ed online 9 April 2015A key goal in genetic analysis is to identify which genescontribute to specific biological phenotypes and diseases. Hypothesis-driven, reverse genetic methods takea ‘genotype-to-phenotype’ approach by using priorknowledge to test the causal role of specific geneticperturbations. By contrast, forward genetic screens are‘phenotype-to-genotype’ approaches that involve modifying or modulating the expression of many genes,selecting for the cells or organisms with a phenotypeof interest, and then characterizing the mutations thatresult in those phenotypic changes.Initial forward genetic experiments carried out onmodel organisms such as yeast, flies, plants, zebrafish,nematodes and rodents1–9 relied on the use of chemical DNA mutagens followed by the isolation of individuals with an aberrant phenotype. These screens haveuncovered many basic biological mechanisms, suchas RAS and NOTCH signalling pathways10, as well asmolecular mechanisms of embryonic patterning11,12 anddevelopment13,14.A major shortcoming of DNA-mutagen-basedscreens is that the causal mutations in the selectedclones are initially unknown. Identifying the causalmutations can be costly and labour intensive, requiring linkage analysis through crosses with characterized lines. These challenges can now be more easilyaddressed by mapping mutations using next-generationsequencing (NGS)15 and by replacing chemical mutagens with viruses and transposons, which use definedinsertion sequences that are amenable to sequencingbased analysis16–18. An additional limitation of randommutagenesis approaches is that the resulting mutantsare typically heterozygotes, which can mask recessivephenotypes. In model organisms, homozygosity canbe achieved by intercrossing progeny derived from theinitial heterozygous mutant. In mammalian cell culture,recessive screens have been limited to near-haploidcell lines19,20 or to cell lines that are deficient in Bloomhelicase (BLM), which have an increased rate of mitoticrecombination21.Over the past decade, forward genetic screens havebeen revolutionized by the development of tools thatuse the RNA interference (RNAi) pathway for geneknockdown. RNAi is a conserved endogenous pathway in which mRNA molecules are targeted for degradation on the basis of sequence complementarity22,23,thus facilitating design and scalability of the tools.Several RNAi reagents have been developed, includinglong double-stranded RNA (dsRNA)24, synthetic smallinterfering RNA (siRNA)25, short hairpin RNA (shRNA)26and shRNAs embedded in microRNA (miRNA) precursors (shRNAmirs)27,28. Screens using RNAi toolshave provided a wealth of information on gene function1,26,29–32, but their utility has been hindered by incomplete gene knockdown and extensive off-target activity,making it difficult to interpret phenotypic changes33–35.Sequence-specific programmable nucleases haveemerged as an exciting new genetic perturbation system that enables the targeted modification of the DNAsequence itself. In particular, the RNA-guided endo nuclease Cas9 (REFS 36–41) from the microbial adaptiveimmune system CRISPR (clustered regularly interspaced short palindromic repeat) provides a convenientsystem for achieving targeted mutagenesis in eukaryotic cells42,43. Cas9 is targeted to specific genomic loci viaa guide RNA, which recognizes the target DNA throughNATURE REVIEWS GENETICSVOLUME 16 MAY 2015 299 2015 Macmillan Publishers Limited. All rights reserved

REVIEWSWatson–Crick base pairing. Therefore, Cas9 combinesthe permanently mutagenic nature of classical mutagenswith the programmability of RNAi.In this Review, we discuss recent Cas9‑based functional genetic screening tools, including genome-wideknockout approaches and related strategies using modified forms of Cas9 to cause gene knockdown or transcriptional activation in a non-mutagenic manner44–49.We discuss how these newer approaches comparewith and complement existing RNAi-based screeningtechnologies. We also present some practical considerations for designing Cas9‑based screens and potential future directions for targeted screening technologydevelopment.Short hairpin RNA(shRNA). Small RNAs forminghairpins that can inducesequence-specific silencingin mammalian cells throughRNA interference, both whenexpressed endogenously andwhen produced exogenouslyand transfected into the cell.microRNA(miRNA). Small RNAmolecules processed fromhairpin-containing RNAprecursors that areproduced from endogenousmiRNA-encodinggenes. mi RNAs are 21–23nucleotides in length and,through the RNA-inducedsilencing complex (RISC), theytarget and silence mRNAscontaining imperfectlycomplementary sequences.Indel(Insertion and deletion).Mutations due to smallinsertions or deletions ofDNA sequences.Single guide RNA(sgRNA). An artificial fusion ofCRISPR (clustered regularlyinterspaced short palindromicrepeat) RNA (crRNA) andtransactivating crRNA(tracrRNA) with criticalsecondary structures forloading onto Cas9 for genomeediting. It functionallysubstitutes the complex ofcrRNA and tracrRNA thatoccurs in natural CRISPRsystems. It uses RNA–DNAhybridization to guide Cas9to the genomic target.Nonsense-mediated decay(NMD). An mRNA surveillancemechanism that degradesmRNAs containing nonsensemutations to prevent theexpression of truncated orerroneous proteins.Mechanisms of perturbationLoss‑of‑function perturbations mediated by Cas9 andRNAi. Cas9 nuclease is a component of the type IICRISPR bacterial adaptive immune system that hasrecently been adapted for genome editing in manyeukaryotic models (reviewed in REFS 50,51). Targetedgenome engineering with Cas9 and other nucleasesexploits endogenous DNA double-strand break (DSB)repair pathways to create mutations at specific locationsin the genome. Although there is a large diversity of DSBrepair mechanisms, genome editing in mammalian cellsprimarily relies on homology-directed repair (HDR),in which an exogenous DNA template can facilitateprecise repair, as well as non-homologous end-joining(NHEJ), which is an error-prone repair mechanism thatintroduces indel mutations at the repair site52. To induceDSBs, Cas9 can be targeted to specific locations in thegenome by specifying a short single guide RNA (sgRNA)41to complement the target DNA. For the commonly usedStreptococcus pyogenes Cas9, the sgRNA contains a 20-bpguide sequence. The target DNA needs to contain the20-bp target sequence followed by a 3-bp protospaceradjacent motif (PAM), although some mismatches canbe tolerated (see below).Loss‑of‑function mutations mediated by Cas9 nuclease are achieved by targeting a DSB to a constitutivelyspliced coding exon. When a DSB is repaired by NHEJ, itcan introduce an indel mutation. This frequently causesa coding frameshift, resulting in a premature stop codonand the initiation of nonsense-mediated decay (NMD) ofthe transcript (FIG. 1). NMD might not be active for allgenes and is not necessarily required for Cas9‑mediatedknockout, as an early frameshift mutation or large indelsmight be sufficient to produce a non-functional protein. Early exons are preferred for targeting, as indelsin these exons have a higher probability of introducingan early stop codon or a frameshift of a larger portionof the protein53. As DSB induction and NHEJ-mediatedrepair occur independently at each allele in diploid cells,targeting by Cas9 results in a range of biallelic and heterozygous target gene lesions in different cells. We andothers44–47 have used the simple, RNA-mediated programmability of Cas9 and its nuclease function to conduct genome-scale knockout screens in mammalian cellcultures. These initial screens uncovered both knownand novel insights into gene essentiality and resistance to Figure 1 Molecular mechanisms underlying geneperturbation via lentiviral delivery of RNA interferencereagents, Cas9 nuclease and dCas9 transcriptionaleffectors. a Lentiviral transduction begins with thefusion of virus particles with the cell membrane andthe insertion of the single-stranded RNA (ssRNA) viralgenome into the cell cytoplasm. A reverse transcriptasethen converts the ssRNA genome into double-strandedDNA (dsDNA) that is imported into the nucleus andintegrates into the host cell genome. Short hairpin RNA(shRNA) or single guide RNA (sgRNA) transgenes are thenexpressed from an RNA polymerase III (Pol III) or Pol IIpromoter. b For shRNA transgenes, maturation involvesa series of nucleolytic processing steps that result incytoplasmic small interfering RNA (siRNA) with sequencecomplementarity to the target mRNA. Drosha processingis required for reagents consisting of shRNAs embedded inmicroRNA precursors (shRNAmirs) but is usually bypassedfor simple stem–loop shRNA reagents. Gene silencing isachieved by siRNA recruitment to the RNA-inducedsilencing complex (RISC) for mRNA degradation andtranslational inhibition. c,d By contrast, both the Cas9nuclease and catalytically inactive Cas9 (dCas9)-mediatedtranscriptional modulation act in the nucleus. Thetransgene-encoded Cas9–sgRNA complex targets agenomic locus through sequence complementarity to the20-bp sgRNA spacer sequence (part c). For Cas9nuclease-mediated knockout, double-strand break (DSB)formation is followed by non-homologous end-joining(NHEJ) DNA repair that can introduce an indel mutationand a coding frameshift. For dCas9‑mediatedtranscriptional modulation, the modification of expression(white arrows) depends on the exact type of fusion ofeither dCas9 or sgRNA (part d) (FIG. 2). These inducednuclear events, together with endogenous transcriptdegradation and dilution through cell division, will resultin a new steady-state expression level in the cytoplasm.drugs and toxins. Most importantly, Cas9‑based screensdisplayed high reagent consistency, strong phenotypiceffects and high validation rates, demonstrating thepromise of this approach.Although the application of Cas9 to targeted screening is relatively recent, similar approaches based on RNAitechnologies have been extensively used over the pastdecade in mammalian cell culture and in vivo1,3,26,29,30,54–58.RNAi is a conserved natural pathway that is triggered byvarious types of dsRNAs (often single-stranded RNAsfolded into hairpin structures) and that results in theselective downregulation of transcripts with sequencecomplementarity to one strand of the dsRNA23. Naturalsources of dsRNAs include endogenous mi RNAs59 andexogenous linear dsRNAs that are typically introducedinto cells by invading viruses60–62. Artificial targeted geneknockdown is achieved by the delivery of a wide rangeof designed RNAi reagents55,63, including long dsRNAs24,siRNAs25, shRNAs26 and miRNA-embedded shRNAs27,28.The delivery of RNAi reagents is achieved by transfectionof pre-synthesized RNA (for siRNAs and dsRNAs), bytransfection of DNA (which encodes a promoter-drivenshRNA or shRNAmir) or by viral transduction methods using lentiviral, retroviral or transposon constructs300 MAY 2015 VOLUME 16www.nature.com/reviews/genetics 2015 Macmillan Publishers Limited. All rights reserved

REVIEWSaFusion of virus particlesand insertion of viral genomeinto the cellssRNAReverse transcriptiondsDNANuclear importGenomicintegrationTransgene RNARISCAAAAAActive mRNAdegradation andtranslational inhibitionNATURE REVIEWS GENETICSPrematurestop codonDepletion oftarget mRNAby naturaldegradationand dilutionsduring celldivisionAAAAAAAAAAAAAAAAAAAAAdjustmentto a newexpression levelAAAAAAAAAAAAAAAAAAAAwith a cloned shRNA or shRNAmir cassette (FIG. 1). Incontrast to RNA polymerase III (Pol III)-driven expression of shRNAs or sgRNAs, Pol II‑driven expression ofshRNAmirs can be temporally controlled and genetically restricted across tissues63. Most RNAi reagents arenucleo lytically processed by the enzyme Dicer into functional siRNAs. Before processing by Dicer, shRNAmirsrequire nuclear processing by Drosha–DGCR8, but thisstep is usually bypassed with other reagents63. Regardlessof the reagent type, the resultant siRNAs are then loadedinto the RNA-induced silencing complex (RISC), whichis guided to the target mRNA molecule by the siRNA toinitiate mRNA degradation or translational inhibition23.Catalytically inactive Cas9 for transcriptional modu‑lation. In addition to gene knockout that is mediatedby the error-prone repair of targeted DSBs and RNAibased gene knockdowns, catalytically inactive Cas9(dCas9) and various fusions of either dCas9 or sgRNAswith transcriptional activator, repressor and recruitmentdomains have been used to modulate gene expressionat targeted loci without introducing irreversible mutations to the genome. The dCas9‑based transcriptionalinhibition and activation systems are commonly referredto as CRISPRi and CRISPRa, respectively (FIG. 2). dCas9 byitself can have a repressive effect on gene expression,which is probably due to steric hindrance of the components of the transcription initiation and elongationmachinery64,65 (FIG. 2Aa). Although this approach hasbeen successful in Escherichia coli, the degree of repression achieved in mammalian cells has been modest64–68.Chromatin-modifying repressor domains have beenfused to dCas9 in an attempt to improve repression inmammalian cells66 (FIG. 2Ab). However, the magnitude ofrepression displayed high variability across sgRNAs evenwith these fusion proteins66. To achieve a more robusteffect, sgRNA libraries tiling the upstream regions ofgenes were constructed, and the variability in the measured effect on transcription was used to infer rules forthe design of more-potent repressive sgRNAs48. Theserules included the sgRNA target location relative to thetranscription start site, the length of the protospacer andthe spacer nucleotide composition features48. AlthoughdCas9‑mediated repression and RNAi-based tools seemto result in a similar molecular effect, dCas9 repressionoccurs by inhibiting transcription, whereas RNAi acts onthe mRNAs in the cytoplasm. These differences mightresult in varying cellular responses.Whereas loss‑of‑function screens can be conductedusing a variety of both established and new Cas9‑basedtools, gain‑of‑function screens have been limited tocDNA overexpression libraries69. The coverage of suchlibraries is incomplete owing to the difficulty of cloning or expressing large cDNA constructs. Furthermore,these libraries often do not capture the full complexityof transcript isoforms, and they express genes independently of the endogenous regulatory context. Tofacilitate Cas9‑based gain‑of‑function screens, synthetic activators were constructed by fusing dCas9with transcriptional activation domains such as VP64or p65 (REFS 68,70–73) (FIG. 2Ba). However, these fusionsNature Reviews Genetics 2015 Macmillan Publishers Limited. All rights reservedVOLUME 16 MAY 2015 301

REVIEWSCRISPRiAn engineered transcriptionalsilencing complex based oncatalytically inactive Cas9(dCas9) fusions and/or singleguide RNA (sgRNA)modification.CRISPRaAn engineered transcriptionalactivation complex based oncatalytically inactive Cas9(dCas9) fusions and/or singleguide RNA (sgRNA)modification.only led to modest activation when delivered with asingle sgRNA in mammalian cells. The delivery ofmultiple sgRNAs targeting the same promoter regionimproved target gene activation70–72, but this was still notreliable enough to implement genome-wide activationscreens. To amplify the signal of dCas9 fusion effectordomains, a repeating peptide array of epitopes fused todCas9 was developed together with activation effector domains fused to a single-chain variable fragment(ScFv) antibody74 (FIG. 2Bb). Similar to the repressionscreen, a tiling approach was then used to infer rules forpotent sgRNAs, followed by the design of a genome-widelibrary and the implementation of an activation screen48.We recently took advantage of a crystal structure ofCas9 in complex with a guide RNA and target singlestranded DNA (ssDNA)75 to rationally design an efficient Cas9 activation complex composed of a dCas9fusion protein and modified sgRNA49 (FIG. 2Bc). Thisdesign was guided by the following principles: the useof alternative attachment positions to recruit endogenous transcription machineries more effectively; themimicking of natural transcriptional activation mechanisms by recruiting multiple distinct activators that actin synergy to drive transcription; and the identificationof design rules for efficient positioning of the Cas9 activation complex on the promoter. We used this designA Transcriptional repression (CRISPRi)Aato implement a genome-wide gain‑of‑function screen49to identify genes that confer vemurafenib resistance inmelanoma cells when upregulated.Modified scaffolds with different RNA-bindingmotifs were recently developed for both activation andrepression of gene expression76 (FIG. 2C). A combinationof these scaffolds enabled the execution of complex synthetic transcriptional programmes with the simultaneousactivation and repression of different genes.The most apparent advantage of dCas9‑mediatedtranscriptional activation is that induction originates fromthe endogenous gene locus (unlike expression from anexogenous cDNA construct). Yet, the extent to whichsynthetic transcriptional modulators preserve the complexity of transcript isoforms and different types of feedback regulation remains to be tested77,78. In one testedcase49, two transcript isoforms were expressed at equallevels, suggesting that transcript complexity can be preserved. One important advantage of cDNA expressionvectors is the ability to easily express mutated geneswithout modifying the endogenous genomic loci.Libraries and screening strategiesFunctional screens in cultured cells are conducted intwo general formats: arrayed or pooled (FIG. 3). In anarrayed format, individual reagents are arranged inC Multiplexed activation and A 300MS2MCPKRABPP7PCPVP64comComB Transcriptional activation antibodyBcVP64dCas9p65 HSF1MCPsgRNAsgRNA–400sgRNA–50Figure 2 dCas9‑mediated transcriptional modulation. The differentways in which catalytically inactive Cas9 (dCas9) fusions have been used tosynthetically repress (CRISPRi) or activate (CRISPRa) expression are shown.All approaches use a single guide RNA (sgRNA) to direct dCas9 to a

initial heterozygous mutant. In mammalian cell culture, . using CRISPR–Cas9 Ophir Shalem, Neville E. Sanjana and Feng Zhang . genes and is not necessarily required for Cas9 -mediated knockout, as an early frameshift mutation or large indels might be sufficient to produce a non-functio

Related Documents:

Cancer Genomics Techniques used in genomics Structural Genomics Functional genomics Epigenomics and Epigenetics Pharmacogenomics Personalized and Regenerative medicine Clinical Genomics . different advantages of being in a noteworthy metropolitan territory.

So, what is functional genomics? Where sequence-based genomics looks at the structure and components of genomes, and analyses the similarities and differences between genomes Functional genomics looks at how genomes result in cellular phenotypes , and analyses di

3 New disciplines Genomics Genome: complete set of gene or DNA of a species. Genomics: understanding / study of the genome: use of the full genetic information (study not only individual genes or groups of genes) Functional genomics: assigning function to genes by genomic methods (experimental and computational [in silico] procedures)Structural genomics: exploring the spatial structure .

Functional genomics! mics!Metabolomics! Next generation sequencing! Mass spectrometry! Bioinformatics! Knowledge management! Ontology! Pathway! Network! High-dimensionality! Curse of dimensionality!Clustering!Feature selection!Prediction analysis!Text-mining S. Ballereau (&) ! A. Chaiboonchoe .

Table 1. Cisco ACE to Avi Networks Cisco CSP 2100 Existing Cisco Model Migration to Cisco CSP Avi Vantage Ace 4710 Throughput: 0.5, 1, 2, 4 Gbps SSL Throughput: 1 Gbps SSL TPS: 7,500 Cisco CSP 4-core Avi SE Throughput: 20 Gbps SSL Throughput: 4 Gbps SSL TPS: 8,000 Ace 30 Service Module Throughput: 4, 8, 16 Gbps

NSA 5600 firewall only 01-SSC-3830 NSA 5600 TotalSecure (1-year) 01-SSC-3833 Firewall NSA 6600 Firewall throughput 12.0 Gbps IPS throughput 4.5 Gbps Anti-malware throughput 3.0 Gbps Full DPI throughput 3.0 Gbps IMIX throughput 3.5 Gbps Maximum DPI connections 500,000 New connections/sec 90,000/sec Description SKU NSA 6600 firewall only 01-SSC-3820

Significance of the Genomics Revolution data driven biology – functional genomics – comparative genomics – systems biology molecular medicine – identification of genetic components of various maladies – diagnosis/prognosis f

Figure 13. iPerf3 throughput test. Note the measured throughput now is approximately 7.99 Gbps, which is different than the value assigned in the tbf rule (10 Gbps). In the next section, the test is repeated but using a higher MSS. Step 6. In order to stop the server, press Ctrl c in host h2's terminal. The user can see the throughput results .