Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 251
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 186(14): 2977-2994.e23, 2023 07 06.
Article in English | MEDLINE | ID: mdl-37343560

ABSTRACT

Comparative studies of great apes provide a window into our evolutionary past, but the extent and identity of cellular differences that emerged during hominin evolution remain largely unexplored. We established a comparative loss-of-function approach to evaluate whether human cells exhibit distinct genetic dependencies. By performing genome-wide CRISPR interference screens in human and chimpanzee pluripotent stem cells, we identified 75 genes with species-specific effects on cellular proliferation. These genes comprised coherent processes, including cell-cycle progression and lysosomal signaling, which we determined to be human-derived by comparison with orangutan cells. Human-specific robustness to CDK2 and CCNE1 depletion persisted in neural progenitor cells and cerebral organoids, supporting the G1-phase length hypothesis as a potential evolutionary mechanism in human brain expansion. Our findings demonstrate that evolutionary changes in human cells reshaped the landscape of essential genes and establish a platform for systematically uncovering latent cellular and molecular differences between species.


Subject(s)
Hominidae , Neural Stem Cells , Pluripotent Stem Cells , Stem Cells , Animals , Humans , Pan troglodytes/genetics
2.
Cell ; 185(14): 2559-2575.e28, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35688146

ABSTRACT

A central goal of genetics is to define the relationships between genotypes and phenotypes. High-content phenotypic screens such as Perturb-seq (CRISPR-based screens with single-cell RNA-sequencing readouts) enable massively parallel functional genomic mapping but, to date, have been used at limited scales. Here, we perform genome-scale Perturb-seq targeting all expressed genes with CRISPR interference (CRISPRi) across >2.5 million human cells. We use transcriptional phenotypes to predict the function of poorly characterized genes, uncovering new regulators of ribosome biogenesis (including CCDC86, ZNF236, and SPATA5L1), transcription (C7orf26), and mitochondrial respiration (TMEM242). In addition to assigning gene function, single-cell transcriptional phenotypes allow for in-depth dissection of complex cellular phenomena-from RNA processing to differentiation. We leverage this ability to systematically identify genetic drivers and consequences of aneuploidy and to discover an unanticipated layer of stress-specific regulation of the mitochondrial genome. Our information-rich genotype-phenotype map reveals a multidimensional portrait of gene and cellular function.


Subject(s)
Genomics , Single-Cell Analysis , CRISPR-Cas Systems/genetics , Chromosome Mapping , Genotype , Phenotype , Single-Cell Analysis/methods
3.
Cell ; 185(11): 1905-1923.e25, 2022 05 26.
Article in English | MEDLINE | ID: mdl-35523183

ABSTRACT

Tumor evolution is driven by the progressive acquisition of genetic and epigenetic alterations that enable uncontrolled growth and expansion to neighboring and distal tissues. The study of phylogenetic relationships between cancer cells provides key insights into these processes. Here, we introduced an evolving lineage-tracing system with a single-cell RNA-seq readout into a mouse model of Kras;Trp53(KP)-driven lung adenocarcinoma and tracked tumor evolution from single-transformed cells to metastatic tumors at unprecedented resolution. We found that the loss of the initial, stable alveolar-type2-like state was accompanied by a transient increase in plasticity. This was followed by the adoption of distinct transcriptional programs that enable rapid expansion and, ultimately, clonal sweep of stable subclones capable of metastasizing. Finally, tumors develop through stereotypical evolutionary trajectories, and perturbing additional tumor suppressors accelerates progression by creating novel trajectories. Our study elucidates the hierarchical nature of tumor evolution and, more broadly, enables in-depth studies of tumor progression.


Subject(s)
Neoplasms , Animals , Genes, ras , Mice , Neoplasms/genetics , Phylogeny , Exome Sequencing
4.
Cell ; 185(4): 690-711.e45, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35108499

ABSTRACT

Single-cell (sc)RNA-seq, together with RNA velocity and metabolic labeling, reveals cellular states and transitions at unprecedented resolution. Fully exploiting these data, however, requires kinetic models capable of unveiling governing regulatory functions. Here, we introduce an analytical framework dynamo (https://github.com/aristoteleo/dynamo-release), which infers absolute RNA velocity, reconstructs continuous vector fields that predict cell fates, employs differential geometry to extract underlying regulations, and ultimately predicts optimal reprogramming paths and perturbation outcomes. We highlight dynamo's power to overcome fundamental limitations of conventional splicing-based RNA velocity analyses to enable accurate velocity estimations on a metabolically labeled human hematopoiesis scRNA-seq dataset. Furthermore, differential geometry analyses reveal mechanisms driving early megakaryocyte appearance and elucidate asymmetrical regulation within the PU.1-GATA1 circuit. Leveraging the least-action-path method, dynamo accurately predicts drivers of numerous hematopoietic transitions. Finally, in silico perturbations predict cell-fate diversions induced by gene perturbations. Dynamo, thus, represents an important step in advancing quantitative and predictive theories of cell-state transitions.


Subject(s)
Single-Cell Analysis , Transcriptome/genetics , Algorithms , Female , Gene Expression Regulation , HL-60 Cells , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Humans , Kinetics , Models, Biological , RNA, Messenger/metabolism , Staining and Labeling
5.
Cell ; 184(22): 5635-5652.e29, 2021 10 28.
Article in English | MEDLINE | ID: mdl-34653350

ABSTRACT

While prime editing enables precise sequence changes in DNA, cellular determinants of prime editing remain poorly understood. Using pooled CRISPRi screens, we discovered that DNA mismatch repair (MMR) impedes prime editing and promotes undesired indel byproducts. We developed PE4 and PE5 prime editing systems in which transient expression of an engineered MMR-inhibiting protein enhances the efficiency of substitution, small insertion, and small deletion prime edits by an average 7.7-fold and 2.0-fold compared to PE2 and PE3 systems, respectively, while improving edit/indel ratios by 3.4-fold in MMR-proficient cell types. Strategic installation of silent mutations near the intended edit can enhance prime editing outcomes by evading MMR. Prime editor protein optimization resulted in a PEmax architecture that enhances editing efficacy by 2.8-fold on average in HeLa cells. These findings enrich our understanding of prime editing and establish prime editing systems that show substantial improvement across 191 edits in seven mammalian cell types.


Subject(s)
Gene Editing , CRISPR-Cas Systems/genetics , Cell Line , DNA/metabolism , DNA Mismatch Repair/genetics , Female , Genes, Dominant , Genome, Human , Humans , Male , Models, Biological , MutL Protein Homolog 1/genetics , Mutation/genetics , RNA/metabolism , Reproducibility of Results
6.
Cell ; 184(22): 5653-5669.e25, 2021 10 28.
Article in English | MEDLINE | ID: mdl-34672952

ABSTRACT

Cells repair DNA double-strand breaks (DSBs) through a complex set of pathways critical for maintaining genomic integrity. To systematically map these pathways, we developed a high-throughput screening approach called Repair-seq that measures the effects of thousands of genetic perturbations on mutations introduced at targeted DNA lesions. Using Repair-seq, we profiled DSB repair products induced by two programmable nucleases (Cas9 and Cas12a) in the presence or absence of oligonucleotides for homology-directed repair (HDR) after knockdown of 476 genes involved in DSB repair or associated processes. The resulting data enabled principled, data-driven inference of DSB end joining and HDR pathways. Systematic interrogation of this data uncovered unexpected relationships among DSB repair genes and demonstrated that repair outcomes with superficially similar sequence architectures can have markedly different genetic dependencies. This work provides a foundation for mapping DNA repair pathways and for optimizing genome editing across diverse modalities.


Subject(s)
DNA Breaks, Double-Stranded , Genomics , CRISPR-Associated Protein 9/metabolism , Cell Line , Cluster Analysis , DNA Repair/genetics , Gene Editing , Gene Expression Regulation , Genome, Human , Humans , Phenotype , RNA, Guide, Kinetoplastida/metabolism , Reproducibility of Results
7.
Cell ; 184(9): 2503-2519.e17, 2021 04 29.
Article in English | MEDLINE | ID: mdl-33838111

ABSTRACT

A general approach for heritably altering gene expression has the potential to enable many discovery and therapeutic efforts. Here, we present CRISPRoff-a programmable epigenetic memory writer consisting of a single dead Cas9 fusion protein that establishes DNA methylation and repressive histone modifications. Transient CRISPRoff expression initiates highly specific DNA methylation and gene repression that is maintained through cell division and differentiation of stem cells to neurons. Pairing CRISPRoff with genome-wide screens and analysis of chromatin marks establishes rules for heritable gene silencing. We identify single guide RNAs (sgRNAs) capable of silencing the large majority of genes including those lacking canonical CpG islands (CGIs) and reveal a wide targeting window extending beyond annotated CGIs. The broad ability of CRISPRoff to initiate heritable gene silencing even outside of CGIs expands the canonical model of methylation-based silencing and enables diverse applications including genome-wide screens, multiplexed cell engineering, enhancer silencing, and mechanistic exploration of epigenetic inheritance.


Subject(s)
CRISPR-Cas Systems , Cellular Reprogramming , Epigenesis, Genetic , Epigenome , Gene Editing , Induced Pluripotent Stem Cells/cytology , Neurons/cytology , Cell Differentiation , CpG Islands , DNA Methylation , Gene Silencing , Histone Code , Humans , Induced Pluripotent Stem Cells/metabolism , Neurons/metabolism , Protein Processing, Post-Translational
8.
Cell ; 174(4): 953-967.e22, 2018 08 09.
Article in English | MEDLINE | ID: mdl-30033366

ABSTRACT

Seminal yeast studies have established the value of comprehensively mapping genetic interactions (GIs) for inferring gene function. Efforts in human cells using focused gene sets underscore the utility of this approach, but the feasibility of generating large-scale, diverse human GI maps remains unresolved. We developed a CRISPR interference platform for large-scale quantitative mapping of human GIs. We systematically perturbed 222,784 gene pairs in two cancer cell lines. The resultant maps cluster functionally related genes, assigning function to poorly characterized genes, including TMEM261, a new electron transport chain component. Individual GIs pinpoint unexpected relationships between pathways, exemplified by a specific cholesterol biosynthesis intermediate whose accumulation induces deoxynucleotide depletion, causing replicative DNA damage and a synthetic-lethal interaction with the ATR/9-1-1 DNA repair pathway. Our map provides a broad resource, establishes GI maps as a high-resolution tool for dissecting gene function, and serves as a blueprint for mapping the genetic landscape of human cells.


Subject(s)
Biomarkers/metabolism , Cholesterol/metabolism , Epistasis, Genetic , Gene Regulatory Networks , Clustered Regularly Interspaced Short Palindromic Repeats , High-Throughput Nucleotide Sequencing , Humans , Jurkat Cells , K562 Cells , Protein Interaction Mapping
9.
Cell ; 173(6): 1398-1412.e22, 2018 05 31.
Article in English | MEDLINE | ID: mdl-29731168

ABSTRACT

Noncoding mutations in cancer genomes are frequent but challenging to interpret. PVT1 encodes an oncogenic lncRNA, but recurrent translocations and deletions in human cancers suggest alternative mechanisms. Here, we show that the PVT1 promoter has a tumor-suppressor function that is independent of PVT1 lncRNA. CRISPR interference of PVT1 promoter enhances breast cancer cell competition and growth in vivo. The promoters of the PVT1 and the MYC oncogenes, located 55 kb apart on chromosome 8q24, compete for engagement with four intragenic enhancers in the PVT1 locus, thereby allowing the PVT1 promoter to regulate pause release of MYC transcription. PVT1 undergoes developmentally regulated monoallelic expression, and the PVT1 promoter inhibits MYC expression only from the same chromosome via promoter competition. Cancer genome sequencing identifies recurrent mutations encompassing the human PVT1 promoter, and genome editing verified that PVT1 promoter mutation promotes cancer cell growth. These results highlight regulatory sequences of lncRNA genes as potential disease-associated DNA elements.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Genes, myc , RNA, Long Noncoding/genetics , Animals , Breast Neoplasms/metabolism , CRISPR-Cas Systems , Carcinogenesis/genetics , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic , Chromatin , DNA, Neoplasm/genetics , Enhancer Elements, Genetic , Female , Gene Expression Profiling , Humans , Mice , Mice, Inbred NOD , Mutation , Neoplasm Transplantation , Promoter Regions, Genetic , RNA, Long Noncoding/metabolism , Transcription, Genetic
10.
Mol Cell ; 84(6): 1101-1119.e9, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38428433

ABSTRACT

Mitochondrial outer membrane ⍺-helical proteins play critical roles in mitochondrial-cytoplasmic communication, but the rules governing the targeting and insertion of these biophysically diverse proteins remain unknown. Here, we first defined the complement of required mammalian biogenesis machinery through genome-wide CRISPRi screens using topologically distinct membrane proteins. Systematic analysis of nine identified factors across 21 diverse ⍺-helical substrates reveals that these components are organized into distinct targeting pathways that act on substrates based on their topology. NAC is required for the efficient targeting of polytopic proteins, whereas signal-anchored proteins require TTC1, a cytosolic chaperone that physically engages substrates. Biochemical and mutational studies reveal that TTC1 employs a conserved TPR domain and a hydrophobic groove in its C-terminal domain to support substrate solubilization and insertion into mitochondria. Thus, the targeting of diverse mitochondrial membrane proteins is achieved through topological triaging in the cytosol using principles with similarities to ER membrane protein biogenesis systems.


Subject(s)
Mitochondrial Membranes , Saccharomyces cerevisiae Proteins , Animals , Mitochondrial Membranes/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Mutation , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Protein Transport , Saccharomyces cerevisiae Proteins/metabolism , Mammals/metabolism
12.
Cell ; 167(7): 1853-1866.e17, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27984732

ABSTRACT

Genetic screens help infer gene function in mammalian cells, but it has remained difficult to assay complex phenotypes-such as transcriptional profiles-at scale. Here, we develop Perturb-seq, combining single-cell RNA sequencing (RNA-seq) and clustered regularly interspaced short palindromic repeats (CRISPR)-based perturbations to perform many such assays in a pool. We demonstrate Perturb-seq by analyzing 200,000 cells in immune cells and cell lines, focusing on transcription factors regulating the response of dendritic cells to lipopolysaccharide (LPS). Perturb-seq accurately identifies individual gene targets, gene signatures, and cell states affected by individual perturbations and their genetic interactions. We posit new functions for regulators of differentiation, the anti-viral response, and mitochondrial function during immune activation. By decomposing many high content measurements into the effects of perturbations, their interactions, and diverse cell metadata, Perturb-seq dramatically increases the scope of pooled genomic assays.


Subject(s)
Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Animals , Cell Cycle , Clustered Regularly Interspaced Short Palindromic Repeats , Feedback , Gene Expression Profiling , Gene Knockdown Techniques , Humans , K562 Cells , Mice , Mice, Transgenic , Transcription Factors/metabolism
13.
Cell ; 167(7): 1867-1882.e21, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27984733

ABSTRACT

Functional genomics efforts face tradeoffs between number of perturbations examined and complexity of phenotypes measured. We bridge this gap with Perturb-seq, which combines droplet-based single-cell RNA-seq with a strategy for barcoding CRISPR-mediated perturbations, allowing many perturbations to be profiled in pooled format. We applied Perturb-seq to dissect the mammalian unfolded protein response (UPR) using single and combinatorial CRISPR perturbations. Two genome-scale CRISPR interference (CRISPRi) screens identified genes whose repression perturbs ER homeostasis. Subjecting ∼100 hits to Perturb-seq enabled high-precision functional clustering of genes. Single-cell analyses decoupled the three UPR branches, revealed bifurcated UPR branch activation among cells subject to the same perturbation, and uncovered differential activation of the branches across hits, including an isolated feedback loop between the translocon and IRE1α. These studies provide insight into how the three sensors of ER homeostasis monitor distinct types of stress and highlight the ability of Perturb-seq to dissect complex cellular responses.


Subject(s)
Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Animals , Clustered Regularly Interspaced Short Palindromic Repeats , Endoribonucleases , Feedback , Humans , Models, Molecular , Protein Serine-Threonine Kinases , RNA, Guide, Kinetoplastida/metabolism , Transcription, Genetic , Unfolded Protein Response
14.
Cell ; 165(6): 1493-1506, 2016 Jun 02.
Article in English | MEDLINE | ID: mdl-27238023

ABSTRACT

Essential gene functions underpin the core reactions required for cell viability, but their contributions and relationships are poorly studied in vivo. Using CRISPR interference, we created knockdowns of every essential gene in Bacillus subtilis and probed their phenotypes. Our high-confidence essential gene network, established using chemical genomics, showed extensive interconnections among distantly related processes and identified modes of action for uncharacterized antibiotics. Importantly, mild knockdown of essential gene functions significantly reduced stationary-phase survival without affecting maximal growth rate, suggesting that essential protein levels are set to maximize outgrowth from stationary phase. Finally, high-throughput microscopy indicated that cell morphology is relatively insensitive to mild knockdown but profoundly affected by depletion of gene function, revealing intimate connections between cell growth and shape. Our results provide a framework for systematic investigation of essential gene functions in vivo broadly applicable to diverse microorganisms and amenable to comparative analysis.


Subject(s)
Bacillus subtilis/genetics , Genes, Bacterial , Genes, Essential , CRISPR-Cas Systems , Gene Knockdown Techniques , Gene Library , Gene Regulatory Networks , Molecular Targeted Therapy
15.
Cell ; 160(1-2): 339-50, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25533786

ABSTRACT

Eukaryotic cells execute complex transcriptional programs in which specific loci throughout the genome are regulated in distinct ways by targeted regulatory assemblies. We have applied this principle to generate synthetic CRISPR-based transcriptional programs in yeast and human cells. By extending guide RNAs to include effector protein recruitment sites, we construct modular scaffold RNAs that encode both target locus and regulatory action. Sets of scaffold RNAs can be used to generate synthetic multigene transcriptional programs in which some genes are activated and others are repressed. We apply this approach to flexibly redirect flux through a complex branched metabolic pathway in yeast. Moreover, these programs can be executed by inducing expression of the dCas9 protein, which acts as a single master regulatory control point. CRISPR-associated RNA scaffolds provide a powerful way to construct synthetic gene expression programs for a wide range of applications, including rewiring cell fates or engineering metabolic pathways.


Subject(s)
CRISPR-Cas Systems , Gene Expression , Genetic Techniques , HEK293 Cells , Humans , Metabolic Engineering , RNA, Guide, Kinetoplastida/genetics , Saccharomyces cerevisiae/genetics , Streptococcus pyogenes/genetics
16.
Nature ; 627(8003): 389-398, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38253266

ABSTRACT

The human blood system is maintained through the differentiation and massive amplification of a limited number of long-lived haematopoietic stem cells (HSCs)1. Perturbations to this process underlie diverse diseases, but the clonal contributions to human haematopoiesis and how this changes with age remain incompletely understood. Although recent insights have emerged from barcoding studies in model systems2-5, simultaneous detection of cell states and phylogenies from natural barcodes in humans remains challenging. Here we introduce an improved, single-cell lineage-tracing system based on deep detection of naturally occurring mitochondrial DNA mutations with simultaneous readout of transcriptional states and chromatin accessibility. We use this system to define the clonal architecture of HSCs and map the physiological state and output of clones. We uncover functional heterogeneity in HSC clones, which is stable over months and manifests as both differences in total HSC output and biases towards the production of different mature cell types. We also find that the diversity of HSC clones decreases markedly with age, leading to an oligoclonal structure with multiple distinct clonal expansions. Our study thus provides a clonally resolved and cell-state-aware atlas of human haematopoiesis at single-cell resolution, showing an unappreciated functional diversity of human HSC clones and, more broadly, paving the way for refined studies of clonal dynamics across a range of tissues in human health and disease.


Subject(s)
Cell Lineage , Hematopoiesis , Hematopoietic Stem Cells , Humans , Chromatin/genetics , Chromatin/metabolism , Clone Cells/classification , Clone Cells/cytology , Clone Cells/metabolism , DNA, Mitochondrial/genetics , Hematopoietic Stem Cells/classification , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Mutation , Single-Cell Analysis , Transcription, Genetic , Aging
17.
Cell ; 157(1): 52-64, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-24679526

ABSTRACT

Proteins are notorious for their unpleasant behavior-continually at risk of misfolding, collecting damage, aggregating, and causing toxicity and disease. To counter these challenges, cells have evolved elaborate chaperone and quality control networks that can resolve damage at the level of the protein, organelle, cell, or tissue. On the smallest scale, the integrity of individual proteins is monitored during their synthesis. On a larger scale, cells use compartmentalized defenses and networks of communication, capable sometimes of signaling between cells, to respond to changes in the proteome's health. Together, these layered defenses help protect cells from damaged proteins.


Subject(s)
Cell Physiological Phenomena , Proteins/metabolism , Animals , Endoplasmic Reticulum/metabolism , Homeostasis , Humans , Protein Folding , Proteins/chemistry , Proteolysis , Proteome
18.
Cell ; 157(3): 624-35, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24766808

ABSTRACT

Quantitative views of cellular functions require precise measures of rates of biomolecule production, especially proteins-the direct effectors of biological processes. Here, we present a genome-wide approach, based on ribosome profiling, for measuring absolute protein synthesis rates. The resultant E. coli data set transforms our understanding of the extent to which protein synthesis is precisely controlled to optimize function and efficiency. Members of multiprotein complexes are made in precise proportion to their stoichiometry, whereas components of functional modules are produced differentially according to their hierarchical role. Estimates of absolute protein abundance also reveal principles for optimizing design. These include how the level of different types of transcription factors is optimized for rapid response and how a metabolic pathway (methionine biosynthesis) balances production cost with activity requirements. Our studies reveal how general principles, important both for understanding natural systems and for synthesizing new ones, emerge from quantitative analyses of protein synthesis.


Subject(s)
Escherichia coli/metabolism , Protein Biosynthesis , Bacterial Proteins/metabolism , Genome-Wide Association Study , Methionine/biosynthesis , Multiprotein Complexes/metabolism , Ribosomes/metabolism , Saccharomyces cerevisiae/metabolism , Transcription Factors/metabolism
19.
Cell ; 159(3): 635-46, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25307933

ABSTRACT

Signals in many biological processes can be amplified by recruiting multiple copies of regulatory proteins to a site of action. Harnessing this principle, we have developed a protein scaffold, a repeating peptide array termed SunTag, which can recruit multiple copies of an antibody-fusion protein. We show that the SunTag can recruit up to 24 copies of GFP, thereby enabling long-term imaging of single protein molecules in living cells. We also use the SunTag to create a potent synthetic transcription factor by recruiting multiple copies of a transcriptional activation domain to a nuclease-deficient CRISPR/Cas9 protein and demonstrate strong activation of endogenous gene expression and re-engineered cell behavior with this system. Thus, the SunTag provides a versatile platform for multimerizing proteins on a target protein scaffold and is likely to have many applications in imaging and controlling biological outputs.


Subject(s)
Molecular Imaging/methods , Optical Imaging/methods , Protein Multimerization , Proteins/chemistry , Animals , CRISPR-Cas Systems , Genetic Techniques , Humans , Single-Chain Antibodies/chemistry
20.
Cell ; 159(3): 647-61, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25307932

ABSTRACT

While the catalog of mammalian transcripts and their expression levels in different cell types and disease states is rapidly expanding, our understanding of transcript function lags behind. We present a robust technology enabling systematic investigation of the cellular consequences of repressing or inducing individual transcripts. We identify rules for specific targeting of transcriptional repressors (CRISPRi), typically achieving 90%-99% knockdown with minimal off-target effects, and activators (CRISPRa) to endogenous genes via endonuclease-deficient Cas9. Together they enable modulation of gene expression over a ∼1,000-fold range. Using these rules, we construct genome-scale CRISPRi and CRISPRa libraries, each of which we validate with two pooled screens. Growth-based screens identify essential genes, tumor suppressors, and regulators of differentiation. Screens for sensitivity to a cholera-diphtheria toxin provide broad insights into the mechanisms of pathogen entry, retrotranslocation and toxicity. Our results establish CRISPRi and CRISPRa as powerful tools that provide rich and complementary information for mapping complex pathways.


Subject(s)
CRISPR-Cas Systems , Genetic Techniques , Transcription, Genetic , Cell Line , Cholera Toxin/metabolism , Diphtheria Toxin/metabolism , Genome, Human , Humans
SELECTION OF CITATIONS
SEARCH DETAIL