You are currently viewing Pooled CRISPR Screen & Arrayed CRISPR Screen

Pooled CRISPR Screen & Arrayed CRISPR Screen

  • Post category:Knowledge
  • Post last modified:December 30, 2024
  • Reading time:29 mins read

https://pmc.ncbi.nlm.nih.gov/articles/PMC10200264


Pooled CRISPR Screen

Overview

Pooled genome-wide CRISPR–Cas9 knockout screening is a powerful technique that enables researchers to systematically disrupt (knock out) individual genes in a large population of cells, all in a single experiment. By introducing a “library” of thousands to tens of thousands of distinct single-guide RNAs (sgRNAs) targeting different genes, one can identify which gene knockouts lead to specific phenotypic changes—such as increased or decreased cell proliferation, altered drug sensitivity, or changes in gene expression.

Below is a step-by-step look at the key components and workflow of a pooled genome-wide CRISPR–Cas9 screen, along with considerations and common applications.



1. Designing the sgRNA Library

  1. Genome-wide coverage
    • A typical library includes multiple sgRNAs (3–10 or more) per gene to ensure robust gene knockout and reduce off-target effects.
    • Genome annotations (e.g., from Ensembl or RefSeq) guide the selection of sgRNA target sites.
  2. Targeting considerations
    • sgRNAs are usually designed to target coding exons in early regions of the gene to ensure a high likelihood of loss-of-function.
    • Optimizing on-target specificity: Tools like CRISPick (Broad Institute), CHOPCHOP, and sgRNA Scorer can help design sgRNAs that minimize off-target binding.
  3. Library format
    • Libraries can be purchased as pooled oligonucleotide libraries or synthesized in-house.
    • They are typically cloned into a lentiviral or retroviral vector to facilitate delivery into cells.



2. Delivery of the CRISPR–Cas9 Components

  1. Cas9 expression
    • Cas9 can be expressed stably in the cell line of interest (via integration in the genome or long-term expression vectors), or it can be co-delivered with the sgRNA library.
    • Using cells that stably express Cas9 often simplifies the workflow, as you only need to introduce the sgRNA library.
  2. Library transduction
    • The pooled sgRNA library is often packaged into lentiviral particles and used to infect the Cas9-expressing cells at low multiplicity of infection (MOI).
    • A low MOI ensures that most cells receive only one sgRNA, linking each cell’s phenotype to a single gene knockout.
  3. Antibiotic or fluorescent selection
    • Vectors typically contain a selectable marker (e.g., puromycin resistance).
    • After infection, you can apply antibiotic selection to ensure cells that remain in the population carry the sgRNA construct.



3. Phenotypic Selection or Enrichment

Once the library has been stably integrated, researchers apply a selection pressure or measure a phenotype of interest. Common types of selection/phenotype readouts include:

  1. Survival or proliferation
    • For example, treat cells with a drug to identify genes conferring resistance or sensitivity.
    • Genes that, when knocked out, allow increased survival become enriched in the surviving population.
  2. Surface marker sorting (FACS-based)
    • If the screening goal is to identify regulators of a surface antigen or a fluorescent reporter, you can use fluorescence-activated cell sorting (FACS).
    • Cells with desired marker expression can be sorted out for further analysis.
  3. Reporter-based readouts
    • A reporter system (luciferase, fluorescent proteins) linked to a specific pathway or target gene expression can identify sgRNA knockouts that modulate that pathway.
  4. In vivo studies
    • Cells transduced with the sgRNA library can be introduced into model organisms (e.g., mice) to screen for genes that influence tumor growth, metastasis, or other in vivo phenotypes.



4. sgRNA Quantification (Next-Generation Sequencing)

  1. DNA extraction
    • After the selection step (or after a designated time point), genomic DNA is extracted from the cell population.
  2. PCR amplification of sgRNA barcodes
    • The integrated sgRNA sequences (or their barcode regions) are PCR-amplified from the genomic DNA.
  3. Deep sequencing
    • Amplicons are subjected to next-generation sequencing (e.g., Illumina) to measure the abundance of each sgRNA in the population.
    • Changes in sgRNA abundance (relative to control/reference time point) reflect changes in survival or expansion of the corresponding knockout cells.



5. Data Analysis and Hit Identification

  1. Data normalization
    • Normalize sequencing counts across samples and time points to account for differences in sequencing depth.
  2. Statistical approaches
    • Several tools and methods are available (e.g., MAGeCK, DESeq2, edgeR) to identify significantly enriched or depleted sgRNAs.
    • Enrichment: sgRNAs that become more abundant under selection suggest that knockouts give cells a growth or survival advantage.
    • Depletion: sgRNAs that disappear or decrease under selection suggest that knockouts are lethal or disadvantageous under the given condition.
  3. Gene-level scoring
    • Combine the effects of multiple sgRNAs per gene to rank “hit” genes.
    • Apply false-discovery rate (FDR) corrections to prioritize high-confidence targets.
  4. Pathway-level analysis
    • Often, multiple genes from the same pathway will emerge as hits.
    • Bioinformatic tools (e.g., GSEA, DAVID, Enrichr) can help identify enriched biological processes or pathways.



6. Validation and Follow-Up

  1. Individual sgRNA validation
    • Re-clone individual sgRNAs into a separate vector, transduce cells, and confirm the phenotype.
    • Alternatively, use orthogonal gene-knockdown or knockout methods (e.g., independent CRISPR sgRNAs, RNAi) to confirm on-target effects.
  2. Functional assays
    • Conduct mechanistic studies to determine how the identified hits mediate the observed phenotype.
    • Assess protein expression, pathway activation, or interactions with other genes.
  3. Therapeutic or translational relevance
    • Hits identified in a screen may suggest new drug targets or strategies for drug combinations.
    • Follow-up experiments in more physiologically relevant models (3D cultures, in vivo models) are often used to validate translational potential.



Key Considerations

  1. Library complexity and coverage
    • Ensuring that each sgRNA is represented adequately in the cell population (good coverage) is critical for data reliability.
    • Typical coverage targets: 200–500× (or more) cells per sgRNA.
  2. Cas9 activity and off-target effects
    • Off-target cleavages can complicate the interpretation of results. Choosing high-specificity Cas9 variants or optimizing sgRNA design can help.
  3. Phenotypic assay sensitivity
    • The success of a screen depends on having a robust assay that cleanly separates “hit” phenotypes from the rest of the population.
  4. Time course
    • The dynamics of knockout effects vary; lethal phenotypes may manifest quickly, while subtle phenotypes (e.g., changes in differentiation) may require longer culture.
  5. Data analysis pitfalls
    • Noise from sequencing depth, PCR amplification biases, and cell growth rates can skew results.
    • A well-designed analysis pipeline and biological replicates are essential.



Common Applications

  • Cancer biology: Identifying tumor suppressors or oncogenes that drive proliferation, metastasis, or drug resistance.
  • Drug target discovery: Pinpointing genes essential for cancer cell survival under specific drug treatments.
  • Functional genomics: Systematically examining gene function in processes like cell cycle regulation, apoptosis, immunity, or metabolism.
  • Synthetic lethality screens: Identifying gene pairs where the combined knockout is lethal, guiding combination therapies in oncology.
  • Host-pathogen interactions: Determining host factors critical for viral or bacterial infection.



Summary

A pooled genome-wide CRISPR–Cas9 knockout screen is an invaluable tool to map gene function on a large scale, discover novel regulators of cellular processes, and identify potential therapeutic targets. By employing carefully designed sgRNA libraries, robust phenotypic assays, and thorough data analysis pipelines, researchers can rapidly pinpoint genes whose loss confers a selectable advantage or disadvantage. Subsequent validation and characterization of these candidate genes/targets lead to deeper biological insights and can pave the way for new translational strategies.



Arrayed CRISPR Screen

An arrayed CRISPR screen is a functional genomics approach in which CRISPR-based gene editing reagents (e.g., sgRNAs) are delivered to cells in a well-by-well or sample-by-sample manner—rather than mixing them in a single pooled population (as done in a pooled CRISPR screen). Below is an overview of how arrayed CRISPR screens work, why they are used, and the key considerations involved.



1. Overview and Key Differences from Pooled Screens

Pooled CRISPR Screen

  • Single mixed population: All sgRNAs are introduced into a single pool of cells simultaneously.
  • Complex phenotypic readouts often require selection/enrichment: For instance, cells might be subjected to drug treatment, and surviving cells are collected for sgRNA analysis.
  • High-throughput, cost-effective: Large gene sets can be interrogated at once.
  • Deconvolution: The frequency of sgRNAs in the final population is typically read out by next-generation sequencing (NGS).

Arrayed CRISPR Screen

  • Individual sgRNA per well (or per sample): Each gene perturbation is separated, allowing phenotypes to be measured independently.
  • Direct phenotypic readouts: In an arrayed format, one can observe the phenotype of each gene knockdown (or knockout) in a dedicated well, making it straightforward to link the phenotype to the specific sgRNA.
  • Greater control and specificity: No need for deconvolution via NGS counts; you already know which sgRNA is in each well.
  • More labor-intensive and higher cost: The step-by-step nature of arrayed screens is more expensive and requires more effort to scale.



2. How an Arrayed CRISPR Screen Works

  1. Plate Setup
    • Typically, CRISPR constructs (often lentiviral or plasmid-based) are pre-arrayed into microtiter plates (e.g., 96-, 384-, or 1536-well plates).
    • Each well contains a defined sgRNA targeting one gene (or a non-targeting control).
  2. Cell Seeding
    • The target cells of interest are dispensed into each well.
    • Depending on the experimental design, the cells may be transduced (lentiviral) or transfected (plasmid, ribonucleoprotein complexes) in that well.
  3. Gene Editing
    • The CRISPR system (Cas9 + sgRNA) induces double-strand breaks in the target gene.
    • After a suitable incubation period, the effect of the knockout can manifest at the phenotype or molecular level (e.g., loss of protein expression).
  4. Phenotypic Assay
    • Because each well has a unique gene knockout, a variety of phenotypic readouts can be performed, such as:
    • Cell viability or proliferation (e.g., ATP-based assays like CellTiter-Glo).
    • Microscopy-based assays (cell morphology, organelle structure, protein localization).
    • Reporter gene or fluorescence readouts (e.g., flow cytometry for surface markers).
    • Omics-level measurements (e.g., transcriptomics or proteomics in each well).
  5. Data Analysis
    • Because each sgRNA is assigned to a well, the hit identification and downstream analysis are relatively straightforward compared to pooled approaches.
    • Hits may be confirmed or validated in secondary assays (often also arrayed).



3. Advantages of Arrayed CRISPR Screens

  1. Well-Resolved Phenotypes
    • Each gene knockout is in a separate well. This allows for clear, direct measurement of cell response for that specific gene perturbation.
  2. Assay Versatility
    • Enables high-content or complex phenotypic assays, such as microscopy or flow cytometry, which can be more challenging in pooled screens.
  3. No Deconvolution
    • In pooled screens, sgRNA representation has to be read out by sequencing, and subtle differences in sgRNA abundance may complicate data interpretation. In arrayed screens, every well already has a single gene targeted.
  4. Robust Quality Control
    • Each well can contain built-in positive controls (sgRNAs known to produce specific phenotypic outcomes) or negative controls (non-targeting sgRNAs), ensuring high data quality.
  5. High Sensitivity
    • There is no competition between sgRNA-edited cell populations, as can occur in pooled screens. This can be especially important for phenotypes that benefit from being assayed on a well-by-well basis.



4. Disadvantages and Considerations

  1. Higher Cost and Effort
    • Generating and handling arrayed libraries—where each sgRNA is in a separate well—requires a larger operational footprint.
    • More reagents (plates, media, transfection or transduction reagents) and more time to prepare and handle individual wells.
  2. Scalability
    • Pooled approaches can simultaneously screen tens of thousands of sgRNAs in one experiment. Arrayed screens are more resource-intensive to scale up to large gene sets.
  3. Potential Heterogeneity in Editing Efficiencies
    • While each well only has one sgRNA, editing efficiency can vary from well to well. Good experimental design (using multiple sgRNAs per gene, replicates, and controls) is crucial.
  4. Delivery Method Optimization
    • Transfection or transduction must be optimized for each cell line in an arrayed format to ensure consistent gene editing efficiencies across all wells.



5. Typical Applications

  1. High-Content Imaging Screens
    • Allows for microscopy-based readouts such as changes in cell shape, subcellular localization, or protein aggregation.
  2. Elucidating Pathways in Detail
    • With arrayed screens, it is easier to link each gene perturbation to downstream molecular changes since each well can be analyzed independently (e.g., by qPCR, western blot, or proteomic methods).
  3. Drug Synergy Studies
    • In oncology drug discovery or other therapeutics research, arrayed CRISPR knockouts can be combined with treatments in parallel to systematically assess gene-drug interactions.
  4. Precision Medicine and Target Validation
    • For validating candidate drug targets, arrayed CRISPR screens provide a precise readout of which gene manipulations have the desired therapeutic effect.



6. Best Practices and Experimental Design Tips

  1. Use Multiple sgRNAs per Gene
    • At least 2–4 sgRNAs per gene can help confirm that observed phenotypes are on-target.
    • mitigate the risk of off-target effects from any single sgRNA.
  2. Include Proper Controls
    • Positive controls: sgRNAs targeting housekeeping genes (leading to measurable phenotypic changes, e.g., viability loss) or well-characterized “benchmark” genes.
    • Negative controls: Non-targeting sgRNAs or sgRNAs targeting safe-harbor loci.
  3. Replicates
    • Biological and technical replicates are essential for robust statistical analysis and to manage well-to-well variability.
  4. Automation & Robotics
    • Large-scale arrayed CRISPR screens often rely on liquid-handling robots for reproducibility and scale.
  5. Validation of Gene Editing
    • Confirm editing at the DNA or protein level in select wells (e.g., by Sanger sequencing, TIDE analysis, or Western blot).



Summary

An arrayed CRISPR screen offers high-resolution phenotypic data by targeting genes in a well-by-well manner, making it an excellent choice for high-content or specialized assays that require direct observation of each individual gene’s effect. While more resource-intensive and less scalable than pooled screens, arrayed CRISPR screens are invaluable for detailed phenotypic profiling, functional validation of drug targets, and in-depth mechanistic studies. By carefully optimizing the experimental design (multiple sgRNAs per gene, robust controls, automation), researchers can harness the power of arrayed CRISPR screens for precise, high-throughput functional genomics.