Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
1.
Cell ; 184(3): 689-708.e20, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33482083

ABSTRACT

The most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) is a GGGGCC repeat expansion in the C9orf72 gene. We developed a platform to interrogate the chromatin accessibility landscape and transcriptional program within neurons during degeneration. We provide evidence that neurons expressing the dipeptide repeat protein poly(proline-arginine), translated from the C9orf72 repeat expansion, activate a highly specific transcriptional program, exemplified by a single transcription factor, p53. Ablating p53 in mice completely rescued neurons from degeneration and markedly increased survival in a C9orf72 mouse model. p53 reduction also rescued axonal degeneration caused by poly(glycine-arginine), increased survival of C9orf72 ALS/FTD-patient-induced pluripotent stem cell (iPSC)-derived motor neurons, and mitigated neurodegeneration in a C9orf72 fly model. We show that p53 activates a downstream transcriptional program, including Puma, which drives neurodegeneration. These data demonstrate a neurodegenerative mechanism dynamically regulated through transcription-factor-binding events and provide a framework to apply chromatin accessibility and transcription program profiles to neurodegeneration.


Subject(s)
C9orf72 Protein/metabolism , DNA Repeat Expansion/genetics , Nerve Degeneration/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Axons/metabolism , C9orf72 Protein/genetics , Cell Death , Cells, Cultured , Cerebral Cortex/pathology , Chromatin/metabolism , DNA Damage , Disease Models, Animal , Drosophila , Mice, Inbred C57BL , Nerve Degeneration/pathology , Protein Stability , Transcription, Genetic , Tumor Suppressor Proteins/metabolism
2.
Cell ; 183(7): 2020-2035.e16, 2020 12 23.
Article in English | MEDLINE | ID: mdl-33326746

ABSTRACT

Thousands of proteins localize to the nucleus; however, it remains unclear which contain transcriptional effectors. Here, we develop HT-recruit, a pooled assay where protein libraries are recruited to a reporter, and their transcriptional effects are measured by sequencing. Using this approach, we measure gene silencing and activation for thousands of domains. We find a relationship between repressor function and evolutionary age for the KRAB domains, discover that Homeodomain repressor strength is collinear with Hox genetic organization, and identify activities for several domains of unknown function. Deep mutational scanning of the CRISPRi KRAB maps the co-repressor binding surface and identifies substitutions that improve stability/silencing. By tiling 238 proteins, we find repressors as short as ten amino acids. Finally, we report new activator domains, including a divergent KRAB. These results provide a resource of 600 human proteins containing effectors and demonstrate a scalable strategy for assigning functions to protein domains.


Subject(s)
High-Throughput Screening Assays , Transcription Factors/metabolism , Amino Acid Sequence , CRISPR-Cas Systems/genetics , Female , Gene Silencing , Genes, Reporter , HEK293 Cells , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , K562 Cells , Lentivirus/physiology , Molecular Sequence Annotation , Mutation/genetics , Nuclear Proteins/metabolism , Promoter Regions, Genetic/genetics , Protein Domains , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Reproducibility of Results , Transcription, Genetic , Zinc Fingers
3.
Mol Cell ; 84(11): 2104-2118.e6, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38761795

ABSTRACT

Circular RNAs (circRNAs) are stable RNAs present in cell-free RNA, which may comprise cellular debris and pathogen genomes. Here, we investigate the phenomenon and mechanism of cellular uptake and intracellular fate of exogenous circRNAs. Human myeloid cells and B cells selectively internalize extracellular circRNAs. Macrophage uptake of circRNA is rapid, energy dependent, and saturable. CircRNA uptake can lead to translation of encoded sequences and antigen presentation. The route of internalization influences immune activation after circRNA uptake, with distinct gene expression programs depending on the route of RNA delivery. Genome-scale CRISPR screens and chemical inhibitor studies nominate macrophage scavenger receptor MSR1, Toll-like receptors, and mTOR signaling as key regulators of receptor-mediated phagocytosis of circRNAs, a dominant pathway to internalize circRNAs in parallel to macropinocytosis. These results suggest that cell-free circRNA serves as an "eat me" signal and danger-associated molecular pattern, indicating orderly pathways of recognition and disposal.


Subject(s)
Macrophages , Phagocytosis , RNA, Circular , Signal Transduction , RNA, Circular/genetics , RNA, Circular/metabolism , Humans , Macrophages/metabolism , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/genetics , Animals , Toll-Like Receptors/metabolism , Toll-Like Receptors/genetics , B-Lymphocytes/metabolism , B-Lymphocytes/immunology , Scavenger Receptors, Class A/metabolism , Scavenger Receptors, Class A/genetics , Antigen Presentation , Pinocytosis , Mice
4.
Mol Cell ; 83(10): 1623-1639.e8, 2023 05 18.
Article in English | MEDLINE | ID: mdl-37164018

ABSTRACT

The HUSH complex recognizes and silences foreign DNA such as viruses, transposons, and transgenes without prior exposure to its targets. Here, we show that endogenous targets of the HUSH complex fall into two distinct classes based on the presence or absence of H3K9me3. These classes are further distinguished by their transposon content and differential response to the loss of HUSH. A de novo genomic rearrangement at the Sox2 locus induces a switch from H3K9me3-independent to H3K9me3-associated HUSH targeting, resulting in silencing. We further demonstrate that HUSH interacts with the termination factor WDR82 and-via its component MPP8-with nascent RNA. HUSH accumulates at sites of high RNAPII occupancy including long exons and transcription termination sites in a manner dependent on WDR82 and CPSF. Together, our results uncover the functional diversity of HUSH targets and show that this vertebrate-specific complex exploits evolutionarily ancient transcription termination machinery for co-transcriptional chromatin targeting and genome surveillance.


Subject(s)
Gene Silencing , Transcription Factors , Transcription Factors/metabolism , Transcription, Genetic , Genome/genetics , RNA
5.
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
6.
Nature ; 616(7956): 365-372, 2023 04.
Article in English | MEDLINE | ID: mdl-37020022

ABSTRACT

Human gene expression is regulated by more than 2,000 transcription factors and chromatin regulators1,2. Effector domains within these proteins can activate or repress transcription. However, for many of these regulators we do not know what type of effector domains they contain, their location in the protein, their activation and repression strengths, and the sequences that are necessary for their functions. Here, we systematically measure the effector activity of more than 100,000 protein fragments tiling across most chromatin regulators and transcription factors in human cells (2,047 proteins). By testing the effect they have when recruited at reporter genes, we annotate 374 activation domains and 715 repression domains, roughly 80% of which are new and have not been previously annotated3-5. Rational mutagenesis and deletion scans across all the effector domains reveal aromatic and/or leucine residues interspersed with acidic, proline, serine and/or glutamine residues are necessary for activation domain activity. Furthermore, most repression domain sequences contain sites for small ubiquitin-like modifier (SUMO)ylation, short interaction motifs for recruiting corepressors or are structured binding domains for recruiting other repressive proteins. We discover bifunctional domains that can both activate and repress, some of which dynamically split a cell population into high- and low-expression subpopulations. Our systematic annotation and characterization of effector domains provide a rich resource for understanding the function of human transcription factors and chromatin regulators, engineering compact tools for controlling gene expression and refining predictive models of effector domain function.


Subject(s)
Gene Expression Regulation , Mutagenesis , Protein Domains , Transcription Factors , Transcription, Genetic , Humans , Chromatin/genetics , Chromatin/metabolism , Genes, Reporter/genetics , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism , Protein Domains/genetics , Repressor Proteins/chemistry , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sumoylation
7.
Nature ; 622(7982): 359-366, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37758944

ABSTRACT

The assembly of cortical circuits involves the generation and migration of interneurons from the ventral to the dorsal forebrain1-3, which has been challenging to study at inaccessible stages of late gestation and early postnatal human development4. Autism spectrum disorder and other neurodevelopmental disorders (NDDs) have been associated with abnormal cortical interneuron development5, but which of these NDD genes affect interneuron generation and migration, and how they mediate these effects remains unknown. We previously developed a platform to study interneuron development and migration in subpallial organoids and forebrain assembloids6. Here we integrate assembloids with CRISPR screening to investigate the involvement of 425 NDD genes in human interneuron development. The first screen aimed at interneuron generation revealed 13 candidate genes, including CSDE1 and SMAD4. We subsequently conducted an interneuron migration screen in more than 1,000 forebrain assembloids that identified 33 candidate genes, including cytoskeleton-related genes and the endoplasmic reticulum-related gene LNPK. We discovered that, during interneuron migration, the endoplasmic reticulum is displaced along the leading neuronal branch before nuclear translocation. LNPK deletion interfered with this endoplasmic reticulum displacement and resulted in abnormal migration. These results highlight the power of this CRISPR-assembloid platform to systematically map NDD genes onto human development and reveal disease mechanisms.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Neurodevelopmental Disorders , Female , Humans , Infant, Newborn , Pregnancy , Cell Movement/genetics , CRISPR-Cas Systems/genetics , Interneurons/cytology , Interneurons/metabolism , Interneurons/pathology , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/pathology , Organoids/cytology , Organoids/embryology , Organoids/growth & development , Organoids/metabolism , Organoids/pathology , Endoplasmic Reticulum/metabolism , Prosencephalon/cytology , Prosencephalon/embryology , Prosencephalon/growth & development , Prosencephalon/metabolism , Prosencephalon/pathology , Active Transport, Cell Nucleus
8.
Cell ; 152(4): 909-22, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-23394947

ABSTRACT

Genetic interaction (GI) maps, comprising pairwise measures of how strongly the function of one gene depends on the presence of a second, have enabled the systematic exploration of gene function in microorganisms. Here, we present a two-stage strategy to construct high-density GI maps in mammalian cells. First, we use ultracomplex pooled shRNA libraries (25 shRNAs/gene) to identify high-confidence hit genes for a given phenotype and effective shRNAs. We then construct double-shRNA libraries from these to systematically measure GIs between hits. A GI map focused on ricin susceptibility broadly recapitulates known pathways and provides many unexpected insights. These include a noncanonical role for COPI, a previously uncharacterized protein complex affecting toxin clearance, a specialized role for the ribosomal protein RPS25, and functionally distinct mammalian TRAPP complexes. The ability to rapidly generate mammalian GI maps provides a potentially transformative tool for defining gene function and designing combination therapies based on synergistic pairs.


Subject(s)
Biological Transport , Epistasis, Genetic , Ricin/toxicity , Atorvastatin , Carrier Proteins/metabolism , Cell Line, Tumor , Coat Protein Complex I/metabolism , Endoplasmic Reticulum/metabolism , Heptanoic Acids/pharmacology , Humans , Membrane Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Pyrroles/pharmacology , RNA, Small Interfering , Ribosomal Proteins/metabolism , Vesicular Transport Proteins/metabolism
9.
Mol Cell ; 80(4): 578-591.e5, 2020 11 19.
Article in English | MEDLINE | ID: mdl-33171122

ABSTRACT

Extracellular 2'3'-cyclic-GMP-AMP (cGAMP) is an immunotransmitter exported by diseased cells and imported into host cells to activate the innate immune STING pathway. We previously identified SLC19A1 as a cGAMP importer, but its use across human cell lines is limited. Here, we identify LRRC8A heteromeric channels, better known as volume-regulated anion channels (VRAC), as widely expressed cGAMP transporters. LRRC8A forms complexes with LRRC8C and/or LRRC8E, depending on their expression levels, to transport cGAMP and other 2'3'-cyclic dinucleotides. In contrast, LRRC8D inhibits cGAMP transport. We demonstrate that cGAMP is effluxed or influxed via LRRC8 channels, as dictated by the cGAMP electrochemical gradient. Activation of LRRC8A channels, which can occur under diverse stresses, strongly potentiates cGAMP transport. We identify activator sphingosine 1-phosphate and inhibitor DCPIB as chemical tools to manipulate channel-mediated cGAMP transport. Finally, LRRC8A channels are key cGAMP transporters in resting primary human vasculature cells and universal human cGAMP transporters when activated.


Subject(s)
CRISPR-Cas Systems , Membrane Proteins/metabolism , Nucleotides, Cyclic/metabolism , Biological Transport , Cyclopentanes/pharmacology , Humans , Indans/pharmacology , Lysophospholipids/pharmacology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , U937 Cells
10.
Mol Cell ; 80(3): 452-469.e9, 2020 11 05.
Article in English | MEDLINE | ID: mdl-33157015

ABSTRACT

Although TP53 is the most commonly mutated gene in human cancers, the p53-dependent transcriptional programs mediating tumor suppression remain incompletely understood. Here, to uncover critical components downstream of p53 in tumor suppression, we perform unbiased RNAi and CRISPR-Cas9-based genetic screens in vivo. These screens converge upon the p53-inducible gene Zmat3, encoding an RNA-binding protein, and we demonstrate that ZMAT3 is an important tumor suppressor downstream of p53 in mouse KrasG12D-driven lung and liver cancers and human carcinomas. Integrative analysis of the ZMAT3 RNA-binding landscape and transcriptomic profiling reveals that ZMAT3 directly modulates exon inclusion in transcripts encoding proteins of diverse functions, including the p53 inhibitors MDM4 and MDM2, splicing regulators, and components of varied cellular processes. Interestingly, these exons are enriched in NMD signals, and, accordingly, ZMAT3 broadly affects target transcript stability. Collectively, these studies reveal ZMAT3 as a novel RNA-splicing and homeostasis regulator and a key component of p53-mediated tumor suppression.


Subject(s)
RNA-Binding Proteins/genetics , Tumor Suppressor Protein p53/genetics , Adenocarcinoma/genetics , Alternative Splicing , Animals , Cell Cycle Proteins/metabolism , Exons , Gene Expression Profiling/methods , Genes, Tumor Suppressor , Humans , Liver Neoplasms/genetics , Male , Mice , Mice, Inbred ICR , Mice, SCID , RNA Interference , RNA Splicing , RNA-Binding Proteins/metabolism , Tumor Suppressor Protein p53/metabolism
11.
Nat Methods ; 21(4): 723-734, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38504114

ABSTRACT

The ENCODE Consortium's efforts to annotate noncoding cis-regulatory elements (CREs) have advanced our understanding of gene regulatory landscapes. Pooled, noncoding CRISPR screens offer a systematic approach to investigate cis-regulatory mechanisms. The ENCODE4 Functional Characterization Centers conducted 108 screens in human cell lines, comprising >540,000 perturbations across 24.85 megabases of the genome. Using 332 functionally confirmed CRE-gene links in K562 cells, we established guidelines for screening endogenous noncoding elements with CRISPR interference (CRISPRi), including accurate detection of CREs that exhibit variable, often low, transcriptional effects. Benchmarking five screen analysis tools, we find that CASA produces the most conservative CRE calls and is robust to artifacts of low-specificity single guide RNAs. We uncover a subtle DNA strand bias for CRISPRi in transcribed regions with implications for screen design and analysis. Together, we provide an accessible data resource, predesigned single guide RNAs for targeting 3,275,697 ENCODE SCREEN candidate CREs with CRISPRi and screening guidelines to accelerate functional characterization of the noncoding genome.


Subject(s)
CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Humans , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , CRISPR-Cas Systems/genetics , Genome , K562 Cells , RNA, Guide, CRISPR-Cas Systems
12.
Nature ; 597(7877): 549-554, 2021 09.
Article in English | MEDLINE | ID: mdl-34497417

ABSTRACT

Monoclonal antibody therapies targeting tumour antigens drive cancer cell elimination in large part by triggering macrophage phagocytosis of cancer cells1-7. However, cancer cells evade phagocytosis using mechanisms that are incompletely understood. Here we develop a platform for unbiased identification of factors that impede antibody-dependent cellular phagocytosis (ADCP) using complementary genome-wide CRISPR knockout and overexpression screens in both cancer cells and macrophages. In cancer cells, beyond known factors such as CD47, we identify many regulators of susceptibility to ADCP, including the poorly characterized enzyme adipocyte plasma membrane-associated protein (APMAP). We find that loss of APMAP synergizes with tumour antigen-targeting monoclonal antibodies and/or CD47-blocking monoclonal antibodies to drive markedly increased phagocytosis across a wide range of cancer cell types, including those that are otherwise resistant to ADCP. Additionally, we show that APMAP loss synergizes with several different tumour-targeting monoclonal antibodies to inhibit tumour growth in mice. Using genome-wide counterscreens in macrophages, we find that the G-protein-coupled receptor GPR84 mediates enhanced phagocytosis of APMAP-deficient cancer cells. This work reveals a cancer-intrinsic regulator of susceptibility to antibody-driven phagocytosis and, more broadly, expands our knowledge of the mechanisms governing cancer resistance to macrophage phagocytosis.


Subject(s)
Antibody-Dependent Cell Cytotoxicity/genetics , CRISPR-Cas Systems , Cytophagocytosis/genetics , Macrophages/immunology , Neoplasms/immunology , Neoplasms/pathology , Animals , Antibodies, Monoclonal/immunology , Antibody-Dependent Cell Cytotoxicity/immunology , Antigens, Neoplasm/immunology , CD47 Antigen/antagonists & inhibitors , Cell Line, Tumor , Cells, Cultured , Female , Gene Editing , Gene Knockout Techniques , Humans , Lymphoma, T-Cell/immunology , Lymphoma, T-Cell/pathology , Macrophages/cytology , Macrophages/metabolism , Male , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Receptors, G-Protein-Coupled/metabolism
13.
Nature ; 592(7856): 794-798, 2021 04.
Article in English | MEDLINE | ID: mdl-33854239

ABSTRACT

The initiation of cell division integrates a large number of intra- and extracellular inputs. D-type cyclins (hereafter, cyclin D) couple these inputs to the initiation of DNA replication1. Increased levels of cyclin D promote cell division by activating cyclin-dependent kinases 4 and 6 (hereafter, CDK4/6), which in turn phosphorylate and inactivate the retinoblastoma tumour suppressor. Accordingly, increased levels and activity of cyclin D-CDK4/6 complexes are strongly linked to unchecked cell proliferation and cancer2,3. However, the mechanisms that regulate levels of cyclin D are incompletely understood4,5. Here we show that autophagy and beclin 1 regulator 1 (AMBRA1) is the main regulator of the degradation of cyclin D. We identified AMBRA1 in a genome-wide screen to investigate the genetic basis of  the response to CDK4/6 inhibition. Loss of AMBRA1 results in high levels of cyclin D in cells and in mice, which promotes proliferation and decreases sensitivity to CDK4/6 inhibition. Mechanistically, AMBRA1 mediates ubiquitylation and proteasomal degradation of cyclin D as a substrate receptor for the cullin 4 E3 ligase complex. Loss of AMBRA1 enhances the growth of lung adenocarcinoma in a mouse model, and low levels of AMBRA1 correlate with worse survival in patients with lung adenocarcinoma. Thus, AMBRA1 regulates cellular levels of cyclin D, and contributes to cancer development and the response of cancer cells to CDK4/6 inhibitors.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cyclin D/metabolism , Adenocarcinoma of Lung/genetics , Animals , Cell Division , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclin-Dependent Kinase 6/metabolism , Genes, Tumor Suppressor , Humans , Lung Neoplasms/genetics , Mice , Piperazines/pharmacology , Pyridines/pharmacology , U937 Cells , Ubiquitination
14.
Mol Cell ; 75(2): 372-381.e5, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31126740

ABSTRACT

2'3'-cyclic-GMP-AMP (cGAMP) is a second messenger that activates the antiviral stimulator of interferon genes (STING) pathway. We recently identified a novel role for cGAMP as a soluble, extracellular immunotransmitter that is produced and secreted by cancer cells. Secreted cGAMP is then sensed by host cells, eliciting an antitumoral immune response. Due to the antitumoral effects of cGAMP, other CDN-based STING agonists are currently under investigation in clinical trials for metastatic solid tumors. However, it is unknown how cGAMP and other CDNs cross the cell membrane to activate intracellular STING. Using a genome-wide CRISPR screen, we identified SLC19A1 as the first known importer of cGAMP and other CDNs, including the investigational new drug 2'3'-bisphosphosphothioate-cyclic-di-AMP (2'3'-CDAS). These discoveries will provide insight into cGAMP's role as an immunotransmitter and aid in the development of more targeted CDN-based cancer therapeutics.


Subject(s)
Immunity, Innate/genetics , Neoplasms/genetics , Nucleotides, Cyclic/genetics , Reduced Folate Carrier Protein/genetics , CRISPR-Cas Systems/genetics , Cell Line, Tumor , Cell Membrane/genetics , Genome, Human/genetics , Humans , Membrane Proteins/genetics , Neoplasms/immunology , Nucleotides, Cyclic/immunology , Signal Transduction/genetics
15.
Mol Cell ; 73(2): 377-389.e11, 2019 01 17.
Article in English | MEDLINE | ID: mdl-30581143

ABSTRACT

The ubiquitin proteasome system (UPS) maintains the integrity of the proteome by selectively degrading misfolded or mis-assembled proteins, but the rules that govern how conformationally defective proteins in the secretory pathway are selected from the structurally and topologically diverse constellation of correctly folded membrane and secretory proteins for efficient degradation by cytosolic proteasomes is not well understood. Here, we combine parallel pooled genome-wide CRISPR-Cas9 forward genetic screening with a highly quantitative and sensitive protein turnover assay to discover a previously undescribed collaboration between membrane-embedded cytoplasmic ubiquitin E3 ligases to conjugate heterotypic branched or mixed ubiquitin (Ub) chains on substrates of endoplasmic-reticulum-associated degradation (ERAD). These findings demonstrate that parallel CRISPR analysis can be used to deconvolve highly complex cell biological processes and identify new biochemical pathways in protein quality control.


Subject(s)
CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Endoplasmic Reticulum-Associated Degradation , Genome-Wide Association Study/methods , Proteasome Endopeptidase Complex/metabolism , Proteostasis , CRISPR-Associated Protein 9/metabolism , Endoplasmic Reticulum-Associated Degradation/drug effects , Endoplasmic Reticulum-Associated Degradation/genetics , HEK293 Cells , Humans , K562 Cells , Kinetics , Proteasome Endopeptidase Complex/genetics , Protein Folding , Proteolysis , Proteostasis/drug effects , Proteostasis/genetics , Ricin/pharmacology , Substrate Specificity , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
16.
Nature ; 580(7801): 136-141, 2020 04.
Article in English | MEDLINE | ID: mdl-32238925

ABSTRACT

Cancer genomics studies have identified thousands of putative cancer driver genes1. Development of high-throughput and accurate models to define the functions of these genes is a major challenge. Here we devised a scalable cancer-spheroid model and performed genome-wide CRISPR screens in 2D monolayers and 3D lung-cancer spheroids. CRISPR phenotypes in 3D more accurately recapitulated those of in vivo tumours, and genes with differential sensitivities between 2D and 3D conditions were highly enriched for genes that are mutated in lung cancers. These analyses also revealed drivers that are essential for cancer growth in 3D and in vivo, but not in 2D. Notably, we found that carboxypeptidase D is responsible for removal of a C-terminal RKRR motif2 from the α-chain of the insulin-like growth factor 1 receptor that is critical for receptor activity. Carboxypeptidase D expression correlates with patient outcomes in patients with lung cancer, and loss of carboxypeptidase D reduced tumour growth. Our results reveal key differences between 2D and 3D cancer models, and establish a generalizable strategy for performing CRISPR screens in spheroids to reveal cancer vulnerabilities.


Subject(s)
CRISPR-Cas Systems/genetics , Cell Culture Techniques/methods , Cell Proliferation/genetics , Genome, Human/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Spheroids, Cellular/pathology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Amino Acid Motifs , Animals , Carboxypeptidases/antagonists & inhibitors , Carboxypeptidases/deficiency , Carboxypeptidases/genetics , Carboxypeptidases/metabolism , Female , Humans , Lung Neoplasms/metabolism , Mice , Molecular Targeted Therapy , Mutation , Phenotype , Receptor, IGF Type 1/chemistry , Receptor, IGF Type 1/metabolism , Signal Transduction , Spheroids, Cellular/metabolism , Xenograft Model Antitumor Assays
17.
Nature ; 575(7784): 688-692, 2019 11.
Article in English | MEDLINE | ID: mdl-31634900

ABSTRACT

Ferroptosis is a form of regulated cell death that is caused by the iron-dependent peroxidation of lipids1,2. The glutathione-dependent lipid hydroperoxidase glutathione peroxidase 4 (GPX4) prevents ferroptosis by converting lipid hydroperoxides into non-toxic lipid alcohols3,4. Ferroptosis has previously been implicated in the cell death that underlies several degenerative conditions2, and induction of ferroptosis by the inhibition of GPX4 has emerged as a therapeutic strategy to trigger cancer cell death5. However, sensitivity to GPX4 inhibitors varies greatly across cancer cell lines6, which suggests that additional factors govern resistance to ferroptosis. Here, using a synthetic lethal CRISPR-Cas9 screen, we identify ferroptosis suppressor protein 1 (FSP1) (previously known as apoptosis-inducing factor mitochondrial 2 (AIFM2)) as a potent ferroptosis-resistance factor. Our data indicate that myristoylation recruits FSP1 to the plasma membrane where it functions as an oxidoreductase that reduces coenzyme Q10 (CoQ) (also known as ubiquinone-10), which acts as a lipophilic radical-trapping antioxidant that halts the propagation of lipid peroxides. We further find that FSP1 expression positively correlates with ferroptosis resistance across hundreds of cancer cell lines, and that FSP1 mediates resistance to ferroptosis in lung cancer cells in culture and in mouse tumour xenografts. Thus, our data identify FSP1 as a key component of a non-mitochondrial CoQ antioxidant system that acts in parallel to the canonical glutathione-based GPX4 pathway. These findings define a ferroptosis suppression pathway and indicate that pharmacological inhibition of FSP1 may provide an effective strategy to sensitize cancer cells to ferroptosis-inducing chemotherapeutic agents.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Ferroptosis/genetics , Mitochondrial Proteins/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Ubiquinone/analogs & derivatives , Animals , Apoptosis Regulatory Proteins/genetics , Cell Line, Tumor , Cell Membrane/metabolism , Gene Expression Regulation, Enzymologic , Heterografts , Humans , Lipid Peroxides/metabolism , Male , Mice , Mice, SCID , Mitochondrial Proteins/genetics , Ubiquinone/metabolism
18.
Nature ; 568(7751): 187-192, 2019 04.
Article in English | MEDLINE | ID: mdl-30944478

ABSTRACT

Microglia maintain homeostasis in the central nervous system through phagocytic clearance of protein aggregates and cellular debris. This function deteriorates during ageing and neurodegenerative disease, concomitant with cognitive decline. However, the mechanisms of impaired microglial homeostatic function and the cognitive effects of restoring this function remain unknown. We combined CRISPR-Cas9 knockout screens with RNA sequencing analysis to discover age-related genetic modifiers of microglial phagocytosis. These screens identified CD22, a canonical B cell receptor, as a negative regulator of phagocytosis that is upregulated on aged microglia. CD22 mediates the anti-phagocytic effect of α2,6-linked sialic acid, and inhibition of CD22 promotes the clearance of myelin debris, amyloid-ß oligomers and α-synuclein fibrils in vivo. Long-term central nervous system delivery of an antibody that blocks CD22 function reprograms microglia towards a homeostatic transcriptional state and improves cognitive function in aged mice. These findings elucidate a mechanism of age-related microglial impairment and a strategy to restore homeostasis in the ageing brain.


Subject(s)
Aging/physiology , Brain/cytology , Homeostasis/drug effects , Microglia/drug effects , N-Acetylneuraminic Acid/pharmacology , Phagocytosis/drug effects , Sialic Acid Binding Ig-like Lectin 2/antagonists & inhibitors , Aging/drug effects , Aging/genetics , Animals , Brain/drug effects , Brain/physiology , CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems/genetics , Cognition/drug effects , Cognition/physiology , Female , Homeostasis/genetics , Male , Mice , Mice, Inbred C57BL , Microglia/cytology , N-Acetylneuraminic Acid/chemistry , Phagocytosis/genetics , Sequence Analysis, RNA , Sialic Acid Binding Ig-like Lectin 2/genetics , Sialic Acid Binding Ig-like Lectin 2/metabolism
19.
Mol Cell ; 68(1): 26-43, 2017 Oct 05.
Article in English | MEDLINE | ID: mdl-28985508

ABSTRACT

The past several years have seen an explosion in development of applications for the CRISPR-Cas9 system, from efficient genome editing, to high-throughput screening, to recruitment of a range of DNA and chromatin-modifying enzymes. While homology-directed repair (HDR) coupled with Cas9 nuclease cleavage has been used with great success to repair and re-write genomes, recently developed base-editing systems present a useful orthogonal strategy to engineer nucleotide substitutions. Base editing relies on recruitment of cytidine deaminases to introduce changes (rather than double-stranded breaks and donor templates) and offers potential improvements in efficiency while limiting damage and simplifying the delivery of editing machinery. At the same time, these systems enable novel mutagenesis strategies to introduce sequence diversity for engineering and discovery. Here, we review the different base-editing platforms, including their deaminase recruitment strategies and editing outcomes, and compare them to other CRISPR genome-editing technologies. Additionally, we discuss how these systems have been applied in therapeutic, engineering, and research settings. Lastly, we explore future directions of this emerging technology.


Subject(s)
CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , DNA Repair , Endonucleases/genetics , Gene Editing/methods , Genome , Animals , Directed Molecular Evolution , Endonucleases/metabolism , Eukaryotic Cells/cytology , Eukaryotic Cells/metabolism , Genetic Engineering , Humans , Mutagenesis , Nucleotides/genetics , Nucleotides/metabolism , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism
20.
Mol Cell ; 67(6): 1037-1048.e6, 2017 Sep 21.
Article in English | MEDLINE | ID: mdl-28890333

ABSTRACT

The three-dimensional arrangement of the human genome comprises a complex network of structural and regulatory chromatin loops important for coordinating changes in transcription during human development. To better understand the mechanisms underlying context-specific 3D chromatin structure and transcription during cellular differentiation, we generated comprehensive in situ Hi-C maps of DNA loops in human monocytes and differentiated macrophages. We demonstrate that dynamic looping events are regulatory rather than structural in nature and uncover widespread coordination of dynamic enhancer activity at preformed and acquired DNA loops. Enhancer-bound loop formation and enhancer activation of preformed loops together form multi-loop activation hubs at key macrophage genes. Activation hubs connect 3.4 enhancers per promoter and exhibit a strong enrichment for activator protein 1 (AP-1)-binding events, suggesting that multi-loop activation hubs involving cell-type-specific transcription factors represent an important class of regulatory chromatin structures for the spatiotemporal control of transcription.


Subject(s)
Cell Differentiation , Chromatin Assembly and Disassembly , Chromatin/metabolism , DNA/metabolism , Macrophages/metabolism , Transcription Factor AP-1/metabolism , Transcription, Genetic , Binding Sites , Cell Line, Tumor , Chromatin/chemistry , Chromatin/genetics , DNA/chemistry , DNA/genetics , Enhancer Elements, Genetic , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Humans , Nucleic Acid Conformation , Phenotype , Protein Binding , Time Factors , Transcription Factor AP-1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL