Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Elife ; 82019 08 27.
Article in English | MEDLINE | ID: mdl-31452512

ABSTRACT

Only a subset of cancer patients respond to T-cell checkpoint inhibitors, highlighting the need for alternative immunotherapeutics. We performed CRISPR-Cas9 screens in a leukemia cell line to identify perturbations that enhance natural killer effector functions. Our screens defined critical components of the tumor-immune synapse and highlighted the importance of cancer cell interferon-γ signaling in modulating NK activity. Surprisingly, disrupting the ubiquitin ligase substrate adaptor DCAF15 strongly sensitized cancer cells to NK-mediated clearance. DCAF15 disruption induced an inflamed state in leukemic cells, including increased expression of lymphocyte costimulatory molecules. Proteomic and biochemical analysis revealed that cohesin complex members were endogenous client substrates of DCAF15. Genetic disruption of DCAF15 was phenocopied by treatment with indisulam, an anticancer drug that functions through DCAF15 engagement. In AML patients, reduced DCAF15 expression was associated with improved survival. These findings suggest that DCAF15 inhibition may have useful immunomodulatory properties in the treatment of myeloid neoplasms.


Subject(s)
Intracellular Signaling Peptides and Proteins/genetics , Killer Cells, Natural/immunology , Leukemia, Myeloid, Acute/pathology , Cell Line, Tumor , Gene Expression Profiling , Gene Knockout Techniques , Humans , Leukemia, Myeloid, Acute/mortality , Survival Analysis
2.
Science ; 355(6320)2017 01 06.
Article in English | MEDLINE | ID: mdl-27980086

ABSTRACT

The human genome produces thousands of long noncoding RNAs (lncRNAs)-transcripts >200 nucleotides long that do not encode proteins. Although critical roles in normal biology and disease have been revealed for a subset of lncRNAs, the function of the vast majority remains untested. We developed a CRISPR interference (CRISPRi) platform targeting 16,401 lncRNA loci in seven diverse cell lines, including six transformed cell lines and human induced pluripotent stem cells (iPSCs). Large-scale screening identified 499 lncRNA loci required for robust cellular growth, of which 89% showed growth-modifying function exclusively in one cell type. We further found that lncRNA knockdown can perturb complex transcriptional networks in a cell type-specific manner. These data underscore the functional importance and cell type specificity of many lncRNAs.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Genome, Human , RNA, Long Noncoding/genetics , Cell Growth Processes/genetics , Cell Line , Gene Knockdown Techniques , Gene Regulatory Networks , Genetic Loci , Genetic Testing , Humans , Induced Pluripotent Stem Cells , Machine Learning , RNA Interference , Transcription, Genetic , Transcriptome
3.
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
4.
Elife ; 52016 09 23.
Article in English | MEDLINE | ID: mdl-27661255

ABSTRACT

We recently found that nucleosomes directly block access of CRISPR/Cas9 to DNA (Horlbeck et al., 2016). Here, we build on this observation with a comprehensive algorithm that incorporates chromatin, position, and sequence features to accurately predict highly effective single guide RNAs (sgRNAs) for targeting nuclease-dead Cas9-mediated transcriptional repression (CRISPRi) and activation (CRISPRa). We use this algorithm to design next-generation genome-scale CRISPRi and CRISPRa libraries targeting human and mouse genomes. A CRISPRi screen for essential genes in K562 cells demonstrates that the large majority of sgRNAs are highly active. We also find CRISPRi does not exhibit any detectable non-specific toxicity recently observed with CRISPR nuclease approaches. Precision-recall analysis shows that we detect over 90% of essential genes with minimal false positives using a compact 5 sgRNA/gene library. Our results establish CRISPRi and CRISPRa as premier tools for loss- or gain-of-function studies and provide a general strategy for identifying Cas9 target sites.


Subject(s)
Bacterial Proteins/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats , Endonucleases/metabolism , Gene Targeting/methods , Nucleosomes/metabolism , RNA, Guide, Kinetoplastida/metabolism , Animals , CRISPR-Associated Protein 9 , Chromosome Mapping , Humans , Mice
5.
Cell Stem Cell ; 18(4): 541-53, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-26971820

ABSTRACT

Developing technologies for efficient and scalable disruption of gene expression will provide powerful tools for studying gene function, developmental pathways, and disease mechanisms. Here, we develop clustered regularly interspaced short palindromic repeat interference (CRISPRi) to repress gene expression in human induced pluripotent stem cells (iPSCs). CRISPRi, in which a doxycycline-inducible deactivated Cas9 is fused to a KRAB repression domain, can specifically and reversibly inhibit gene expression in iPSCs and iPSC-derived cardiac progenitors, cardiomyocytes, and T lymphocytes. This gene repression system is tunable and has the potential to silence single alleles. Compared with CRISPR nuclease (CRISPRn), CRISPRi gene repression is more efficient and homogenous across cell populations. The CRISPRi system in iPSCs provides a powerful platform to perform genome-scale screens in a wide range of iPSC-derived cell types, dissect developmental pathways, and model disease.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Gene Silencing , Induced Pluripotent Stem Cells/metabolism , Humans
6.
Elife ; 52016 03 17.
Article in English | MEDLINE | ID: mdl-26987018

ABSTRACT

The prokaryotic CRISPR (clustered regularly interspaced palindromic repeats)-associated protein, Cas9, has been widely adopted as a tool for editing, imaging, and regulating eukaryotic genomes. However, our understanding of how to select single-guide RNAs (sgRNAs) that mediate efficient Cas9 activity is incomplete, as we lack insight into how chromatin impacts Cas9 targeting. To address this gap, we analyzed large-scale genetic screens performed in human cell lines using either nuclease-active or nuclease-dead Cas9 (dCas9). We observed that highly active sgRNAs for Cas9 and dCas9 were found almost exclusively in regions of low nucleosome occupancy. In vitro experiments demonstrated that nucleosomes in fact directly impede Cas9 binding and cleavage, while chromatin remodeling can restore Cas9 access. Our results reveal a critical role of eukaryotic chromatin in dictating the targeting specificity of this transplanted bacterial enzyme, and provide rules for selecting Cas9 target sites distinct from and complementary to those based on sequence properties.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , DNA/metabolism , Endonucleases/antagonists & inhibitors , Nucleosomes/metabolism , CRISPR-Associated Protein 9 , Cell Line , Gene Targeting/methods , Humans
7.
PLoS Genet ; 11(10): e1005592, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26485701

ABSTRACT

Early embryogenesis is a unique developmental stage where genetic control of development is handed off from mother to zygote. Yet the contribution of this transition to the evolution of gene expression is poorly understood. Here we study two aspects of gene expression specific to early embryogenesis in Drosophila: sex-biased gene expression prior to the onset of canonical X chromosomal dosage compensation, and the contribution of maternally supplied mRNAs. We sequenced mRNAs from individual unfertilized eggs and precisely staged and sexed blastoderm embryos, and compared levels between D. melanogaster, D. yakuba, D. pseudoobscura and D. virilis. First, we find that mRNA content is highly conserved for a given stage and that studies relying on pooled embryos likely systematically overstate the degree of gene expression divergence. Unlike studies done on larvae and adults where most species show a larger proportion of genes with male-biased expression, we find that transcripts in Drosophila embryos are largely female-biased in all species, likely due to incomplete dosage compensation prior to the activation of the canonical dosage compensation mechanism. The divergence of sex-biased gene expression across species is observed to be often due to lineage-specific decrease of expression; the most drastic example of which is the overall reduction of male expression from the neo-X chromosome in D. pseudoobscura, leading to a pervasive female-bias on this chromosome. We see no evidence for a faster evolution of expression on the X chromosome in embryos (no "faster-X" effect), unlike in adults, and contrary to a previous study on pooled non-sexed embryos. Finally, we find that most genes are conserved in regard to their maternal or zygotic origin of transcription, and present evidence that differences in maternal contribution to the blastoderm transcript pool may be due to species-specific divergence of transcript degradation rates.


Subject(s)
Blastoderm/growth & development , Dosage Compensation, Genetic , Embryonic Development/genetics , Evolution, Molecular , RNA, Messenger/genetics , Animals , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Embryo, Nonmammalian , Female , Gene Expression Regulation, Developmental , Male , RNA, Messenger/biosynthesis , Sex Ratio , Species Specificity , X Chromosome/genetics
8.
Genome Res ; 25(11): 1715-26, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26335634

ABSTRACT

The transition from a specified germ cell to a population of pluripotent cells occurs rapidly following fertilization. During this developmental transition, the zygotic genome is largely transcriptionally quiescent and undergoes significant chromatin remodeling. In Drosophila, the DNA-binding protein Zelda (also known as Vielfaltig) is required for this transition and for transcriptional activation of the zygotic genome. Open chromatin is associated with Zelda-bound loci, as well as more generally with regions of active transcription. Nonetheless, the extent to which Zelda influences chromatin accessibility across the genome is largely unknown. Here we used formaldehyde-assisted isolation of regulatory elements to determine the role of Zelda in regulating regions of open chromatin in the early embryo. We demonstrate that Zelda is essential for hundreds of regions of open chromatin. This Zelda-mediated chromatin accessibility facilitates transcription-factor recruitment and early gene expression. Thus, Zelda possesses some key characteristics of a pioneer factor. Unexpectedly, chromatin at a large subset of Zelda-bound regions remains open even in the absence of Zelda. The GAGA factor-binding motif and embryonic GAGA factor binding are specifically enriched in these regions. We propose that both Zelda and GAGA factor function to specify sites of open chromatin and together facilitate the remodeling of the early embryonic genome.


Subject(s)
Chromatin/genetics , DNA-Binding Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Transcription Factors/metabolism , Animals , Chromatin/metabolism , DNA-Binding Proteins/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Female , Genetic Association Studies , Genetic Loci , Male , Nuclear Proteins , Promoter Regions, Genetic , Regulatory Sequences, Nucleic Acid , Sequence Analysis, DNA , Transcription Factors/genetics , Transcriptional Activation
9.
Proc Natl Acad Sci U S A ; 112(26): E3384-91, 2015 Jun 30.
Article in English | MEDLINE | ID: mdl-26080438

ABSTRACT

Genetic screening based on loss-of-function phenotypes is a powerful discovery tool in biology. Although the recent development of clustered regularly interspaced short palindromic repeats (CRISPR)-based screening approaches in mammalian cell culture has enormous potential, RNA interference (RNAi)-based screening remains the method of choice in several biological contexts. We previously demonstrated that ultracomplex pooled short-hairpin RNA (shRNA) libraries can largely overcome the problem of RNAi off-target effects in genome-wide screens. Here, we systematically optimize several aspects of our shRNA library, including the promoter and microRNA context for shRNA expression, selection of guide strands, and features relevant for postscreen sample preparation for deep sequencing. We present next-generation high-complexity libraries targeting human and mouse protein-coding genes, which we grouped into 12 sublibraries based on biological function. A pilot screen suggests that our next-generation RNAi library performs comparably to current CRISPR interference (CRISPRi)-based approaches and can yield complementary results with high sensitivity and high specificity.


Subject(s)
Genome , RNA Interference , Animals , Artificial Intelligence , Humans , Mice , RNA, Small Interfering/genetics
10.
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
11.
Elife ; 32014 Oct 14.
Article in English | MEDLINE | ID: mdl-25313869

ABSTRACT

We describe the genome-wide distributions and temporal dynamics of nucleosomes and post-translational histone modifications throughout the maternal-to-zygotic transition in embryos of Drosophila melanogaster. At mitotic cycle 8, when few zygotic genes are being transcribed, embryonic chromatin is in a relatively simple state: there are few nucleosome free regions, undetectable levels of the histone methylation marks characteristic of mature chromatin, and low levels of histone acetylation at a relatively small number of loci. Histone acetylation increases by cycle 12, but it is not until cycle 14 that nucleosome free regions and domains of histone methylation become widespread. Early histone acetylation is strongly associated with regions that we have previously shown to be bound in early embryos by the maternally deposited transcription factor Zelda, suggesting that Zelda triggers a cascade of events, including the accumulation of specific histone modifications, that plays a role in the subsequent activation of these sequences.


Subject(s)
Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Genome, Insect/genetics , Zygote/metabolism , Animals , Binding Sites , Blastoderm/metabolism , Drosophila Proteins/genetics , Embryo, Nonmammalian/metabolism , Enhancer Elements, Genetic/genetics , Female , Gene Expression Regulation, Developmental , Histones/metabolism , Male , Models, Genetic , Nucleosomes/metabolism , Protein Processing, Post-Translational , Transcription Factors/metabolism , Transcription, Genetic
12.
PLoS Genet ; 10(2): e1004159, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24550743

ABSTRACT

Sex chromosome dosage differences between females and males are a significant form of natural genetic variation in many species. Like many species with chromosomal sex determination, Drosophila females have two X chromosomes, while males have one X and one Y. Fusions of sex chromosomes with autosomes have occurred along the lineage leading to D. pseudoobscura and D. miranda. The resulting neo-sex chromosomes are gradually evolving the properties of sex chromosomes, and neo-X chromosomes are becoming targets for the molecular mechanisms that compensate for differences in X chromosome dose between sexes. We have previously shown that D. melanogaster possess at least two dosage compensation mechanisms: the well- characterized MSL-mediated dosage compensation active in most somatic tissues, and another system active during early embryogenesis prior to the onset of MSL-mediated dosage compensation. To better understand the developmental constraints on sex chromosome gene expression and evolution, we sequenced mRNA from individual male and female embryos of D. pseudoobscura and D. miranda, from ∼0.5 to 8 hours of development. Autosomal expression levels are highly conserved between these species. But, unlike D. melanogaster, we observe a general lack of dosage compensation in D. pseudoobscura and D. miranda prior to the onset of MSL-mediated dosage compensation. Thus, either there has been a lineage-specific gain or loss in early dosage compensation mechanism(s) or increasing X chromosome dose may strain dosage compensation systems and make them less effective. The extent of female bias on the X chromosomes decreases through developmental time with the establishment of MSL-mediated dosage compensation, but may do so more slowly in D. miranda than D. pseudoobscura. These results also prompt a number of questions about whether species with more sex-linked genes have more sex-specific phenotypes, and how much transcript level variance is tolerable during critical stages of development.


Subject(s)
Evolution, Molecular , Sex Characteristics , Sex Chromosomes/genetics , Sex Determination Processes , Animals , Dosage Compensation, Genetic , Drosophila melanogaster/genetics , Female , Gene Expression Regulation, Developmental , Humans , Male , Species Specificity
13.
PLoS Genet ; 9(9): e1003748, 2013.
Article in English | MEDLINE | ID: mdl-24068946

ABSTRACT

To better characterize how variation in regulatory sequences drives divergence in gene expression, we undertook a systematic study of transcription factor binding and gene expression in blastoderm embryos of four species, which sample much of the diversity in the 40 million-year old genus Drosophila: D. melanogaster, D. yakuba, D. pseudoobscura and D. virilis. We compared gene expression, measured by mRNA-seq, to the genome-wide binding, measured by ChIP-seq, of four transcription factors involved in early anterior-posterior patterning. We found that mRNA levels are much better conserved than individual transcription factor binding events, and that changes in a gene's expression were poorly explained by changes in adjacent transcription factor binding. However, highly bound sites, sites in regions bound by multiple factors and sites near genes are conserved more frequently than other binding, suggesting that a considerable amount of transcription factor binding is weakly or non-functional and not subject to purifying selection.


Subject(s)
Drosophila melanogaster/embryology , Gene Expression Regulation, Developmental , Genetic Variation , Transcription Factors/genetics , Animals , Base Sequence , Binding Sites , Blastoderm/cytology , Blastoderm/growth & development , Blastoderm/metabolism , Conserved Sequence/genetics , Embryo, Nonmammalian , Enhancer Elements, Genetic , Protein Binding
14.
PLoS Biol ; 9(2): e1000590, 2011 Feb 08.
Article in English | MEDLINE | ID: mdl-21346796

ABSTRACT

When Drosophila melanogaster embryos initiate zygotic transcription around mitotic cycle 10, the dose-sensitive expression of specialized genes on the X chromosome triggers a sex-determination cascade that, among other things, compensates for differences in sex chromosome dose by hypertranscribing the single X chromosome in males. However, there is an approximately 1 hour delay between the onset of zygotic transcription and the establishment of canonical dosage compensation near the end of mitotic cycle 14. During this time, zygotic transcription drives segmentation, cellularization, and other important developmental events. Since many of the genes involved in these processes are on the X chromosome, we wondered whether they are transcribed at higher levels in females and whether this might lead to sex-specific early embryonic patterning. To investigate this possibility, we developed methods to precisely stage, sex, and characterize the transcriptomes of individual embryos. We measured genome-wide mRNA abundance in male and female embryos at eight timepoints, spanning mitotic cycle 10 through late cycle 14, using polymorphisms between parental lines to distinguish maternal and zygotic transcription. We found limited sex-specific zygotic transcription, with a weak tendency for genes on the X to be expressed at higher levels in females. However, transcripts derived from the single X chromosome in males were more abundant that those derived from either X chromosome in females, demonstrating that there is widespread dosage compensation prior to the activation of the canonical MSL-mediated dosage compensation system. Crucially, this new system of early zygotic dosage compensation results in nearly identical transcript levels for key X-linked developmental regulators, including giant (gt), brinker (brk), buttonhead (btd), and short gastrulation (sog), in male and female embryos.


Subject(s)
Body Patterning/genetics , Dosage Compensation, Genetic , Drosophila melanogaster/embryology , Embryonic Development/genetics , Gene Expression Regulation, Developmental , X Chromosome , Animals , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Female , Gene Expression Profiling , Male , Polymorphism, Genetic , RNA, Messenger/metabolism , Repressor Proteins/genetics , Sex Characteristics , Sex Factors , Time Factors , Transcription Factors/genetics , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...