Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
Add more filters

Publication year range
1.
Cell ; 165(6): 1389-1400, 2016 Jun 02.
Article in English | MEDLINE | ID: mdl-27212235

ABSTRACT

Bivalent promoters in embryonic stem cells (ESCs) carry methylation marks on two lysine residues, K4 and K27, in histone3 (H3). K4me2/3 is generally considered to promote transcription, and Polycomb Repressive Complex 2 (PRC2) places K27me3, which is erased at lineage-restricted genes when ESCs differentiate in culture. Molecular defects in various PRC2 null adult tissues lack a unifying explanation. We found that epigenomes in adult mouse intestine and other self-renewing tissues show fewer and distinct bivalent promoters compared to ESCs. Groups of tissue-specific genes that carry bivalent marks are repressed, despite the presence of promoter H3K4me2/3. These are the predominant genes de-repressed in PRC2-deficient adult cells, where aberrant expression is proportional to the H3K4me2/3 levels observed at their promoters in wild-type cells. Thus, in adult animals, PRC2 specifically represses genes with acquired, tissue-restricted promoter bivalency. These findings provide new insights into specificity in chromatin-based gene regulation.


Subject(s)
Embryonic Stem Cells/metabolism , Polycomb Repressive Complex 2/genetics , Promoter Regions, Genetic , Animals , Cell Differentiation/genetics , DNA Methylation , Gene Expression Regulation , Histones/metabolism , Intestinal Mucosa/metabolism , Intestines/cytology , Lysine/metabolism , Mice , Mice, Inbred C57BL , Polycomb Repressive Complex 2/metabolism
2.
Nat Immunol ; 18(2): 184-195, 2017 02.
Article in English | MEDLINE | ID: mdl-27992400

ABSTRACT

Invariant natural killer T cells (iNKT cells) are innate-like lymphocytes that protect against infection, autoimmune disease and cancer. However, little is known about the epigenetic regulation of iNKT cell development. Here we found that the H3K27me3 histone demethylase UTX was an essential cell-intrinsic factor that controlled an iNKT-cell lineage-specific gene-expression program and epigenetic landscape in a demethylase-activity-dependent manner. UTX-deficient iNKT cells exhibited impaired expression of iNKT cell signature genes due to a decrease in activation-associated H3K4me3 marks and an increase in repressive H3K27me3 marks within the promoters occupied by UTX. We found that JunB regulated iNKT cell development and that the expression of genes that were targets of both JunB and the iNKT cell master transcription factor PLZF was UTX dependent. We identified iNKT cell super-enhancers and demonstrated that UTX-mediated regulation of super-enhancer accessibility was a key mechanism for commitment to the iNKT cell lineage. Our findings reveal how UTX regulates the development of iNKT cells through multiple epigenetic mechanisms.


Subject(s)
Cell Differentiation , Epigenesis, Genetic , Gene Expression Regulation , Histone Demethylases/metabolism , Natural Killer T-Cells/physiology , Animals , Cell Lineage , Cells, Cultured , Enhancer Elements, Genetic/genetics , Histone Demethylases/genetics , Immunity, Innate/genetics , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Inbred C57BL , Organ Specificity , Promoter Regions, Genetic/genetics , Promyelocytic Leukemia Zinc Finger Protein , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Mol Cell ; 79(1): 11-29, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32619467

ABSTRACT

The CRISPR-Cas system offers a programmable platform for eukaryotic genome and epigenome editing. The ability to perform targeted genetic and epigenetic perturbations enables researchers to perform a variety of tasks, ranging from investigating questions in basic biology to potentially developing novel therapeutics for the treatment of disease. While CRISPR systems have been engineered to target DNA and RNA with increased precision, efficiency, and flexibility, assays to identify off-target editing are becoming more comprehensive and sensitive. Furthermore, techniques to perform high-throughput genome and epigenome editing can be paired with a variety of readouts and are uncovering important cellular functions and mechanisms. These technological advances drive and are driven by accompanying computational approaches. Here, we briefly present available CRISPR technologies and review key computational advances and considerations for various CRISPR applications. In particular, we focus on the analysis of on- and off-target editing and CRISPR pooled screen data.


Subject(s)
CRISPR-Cas Systems , Computational Biology/methods , Epigenomics , Gene Editing , Genome, Human , Humans
4.
Mol Cell ; 74(6): 1148-1163.e7, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31005419

ABSTRACT

Self-renewal and pluripotency of the embryonic stem cell (ESC) state are established and maintained by multiple regulatory networks that comprise transcription factors and epigenetic regulators. While much has been learned regarding transcription factors, the function of epigenetic regulators in these networks is less well defined. We conducted a CRISPR-Cas9-mediated loss-of-function genetic screen that identified two epigenetic regulators, TAF5L and TAF6L, components or co-activators of the GNAT-HAT complexes for the mouse ESC (mESC) state. Detailed molecular studies demonstrate that TAF5L/TAF6L transcriptionally activate c-Myc and Oct4 and their corresponding MYC and CORE regulatory networks. Besides, TAF5L/TAF6L predominantly regulate their target genes through H3K9ac deposition and c-MYC recruitment that eventually activate the MYC regulatory network for self-renewal of mESCs. Thus, our findings uncover a role of TAF5L/TAF6L in directing the MYC regulatory network that orchestrates gene expression programs to control self-renewal for the maintenance of mESC state.


Subject(s)
Embryonic Stem Cells/metabolism , Gene Regulatory Networks , Induced Pluripotent Stem Cells/metabolism , Proto-Oncogene Proteins c-myc/genetics , TATA-Binding Protein Associated Factors/genetics , Animals , CRISPR-Cas Systems , Cell Cycle/genetics , Cell Proliferation , Cellular Reprogramming , Embryo, Mammalian , Embryonic Stem Cells/cytology , Epigenesis, Genetic , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Editing , Gene Expression Regulation , HEK293 Cells , Histones/genetics , Histones/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Mice , Primary Cell Culture , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , TATA-Binding Protein Associated Factors/metabolism
5.
Nat Methods ; 20(9): 1368-1378, 2023 09.
Article in English | MEDLINE | ID: mdl-37537351

ABSTRACT

Gene regulatory networks (GRNs) are key determinants of cell function and identity and are dynamically rewired during development and disease. Despite decades of advancement, challenges remain in GRN inference, including dynamic rewiring, causal inference, feedback loop modeling and context specificity. To address these challenges, we develop Dictys, a dynamic GRN inference and analysis method that leverages multiomic single-cell assays of chromatin accessibility and gene expression, context-specific transcription factor footprinting, stochastic process network and efficient probabilistic modeling of single-cell RNA-sequencing read counts. Dictys improves GRN reconstruction accuracy and reproducibility and enables the inference and comparative analysis of context-specific and dynamic GRNs across developmental contexts. Dictys' network analyses recover unique insights in human blood and mouse skin development with cell-type-specific and dynamic GRNs. Its dynamic network visualizations enable time-resolved discovery and investigation of developmental driver transcription factors and their regulated targets. Dictys is available as a free, open-source and user-friendly Python package.


Subject(s)
Gene Regulatory Networks , Multiomics , Animals , Mice , Humans , Reproducibility of Results , Transcription Factors/genetics , Algorithms
6.
Nat Methods ; 2023 May 29.
Article in English | MEDLINE | ID: mdl-37248389

ABSTRACT

Most current single-cell analysis pipelines are limited to cell embeddings and rely heavily on clustering, while lacking the ability to explicitly model interactions between different feature types. Furthermore, these methods are tailored to specific tasks, as distinct single-cell problems are formulated differently. To address these shortcomings, here we present SIMBA, a graph embedding method that jointly embeds single cells and their defining features, such as genes, chromatin-accessible regions and DNA sequences, into a common latent space. By leveraging the co-embedding of cells and features, SIMBA allows for the study of cellular heterogeneity, clustering-free marker discovery, gene regulation inference, batch effect removal and omics data integration. We show that SIMBA provides a single framework that allows diverse single-cell problems to be formulated in a unified way and thus simplifies the development of new analyses and extension to new single-cell modalities. SIMBA is implemented as a comprehensive Python library ( https://simba-bio.readthedocs.io ).

7.
Brief Bioinform ; 24(3)2023 05 19.
Article in English | MEDLINE | ID: mdl-37099664

ABSTRACT

Transcription factors (TFs) are key regulatory proteins that control the transcriptional rate of cells by binding short DNA sequences called transcription factor binding sites (TFBS) or motifs. Identifying and characterizing TFBS is fundamental to understanding the regulatory mechanisms governing the transcriptional state of cells. During the last decades, several experimental methods have been developed to recover DNA sequences containing TFBS. In parallel, computational methods have been proposed to discover and identify TFBS motifs based on these DNA sequences. This is one of the most widely investigated problems in bioinformatics and is referred to as the motif discovery problem. In this manuscript, we review classical and novel experimental and computational methods developed to discover and characterize TFBS motifs in DNA sequences, highlighting their advantages and drawbacks. We also discuss open challenges and future perspectives that could fill the remaining gaps in the field.


Subject(s)
Algorithms , Transcription Factors , Protein Binding , Transcription Factors/metabolism , Binding Sites , Base Sequence , Computational Biology
8.
Nat Methods ; 18(9): 1075-1081, 2021 09.
Article in English | MEDLINE | ID: mdl-34354266

ABSTRACT

Epigenetic editing is an emerging technology that uses artificial transcription factors (aTFs) to regulate expression of a target gene. Although human genes can be robustly upregulated by targeting aTFs to promoters, the activation induced by directing aTFs to distal transcriptional enhancers is substantially less robust and consistent. Here we show that long-range activation using CRISPR-based aTFs in human cells can be made more efficient and reliable by concurrently targeting an aTF to the target gene promoter. We used this strategy to direct target gene choice for enhancers capable of regulating more than one promoter and to achieve allele-selective activation of human genes by targeting aTFs to single-nucleotide polymorphisms embedded in distally located sequences. Our results broaden the potential applications of the epigenetic editing toolbox for research and therapeutics.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Gene Targeting/methods , Promoter Regions, Genetic , Transcription Factors/genetics , Alleles , Apolipoprotein C-III/genetics , Apolipoproteins A/genetics , Cell Line , Enhancer Elements, Genetic , Humans , Interleukin-2 Receptor alpha Subunit/genetics , MyoD Protein/genetics , Polymorphism, Single Nucleotide , Transcriptional Activation , beta-Globins/genetics
9.
Nature ; 561(7723): 416-419, 2018 09.
Article in English | MEDLINE | ID: mdl-30209390

ABSTRACT

CRISPR-Cas genome-editing nucleases hold substantial promise for developing human therapeutic applications1-6 but identifying unwanted off-target mutations is important for clinical translation7. A well-validated method that can reliably identify off-targets in vivo has not been described to date, which means it is currently unclear whether and how frequently these mutations occur. Here we describe 'verification of in vivo off-targets' (VIVO), a highly sensitive strategy that can robustly identify the genome-wide off-target effects of CRISPR-Cas nucleases in vivo. We use VIVO and a guide RNA deliberately designed to be promiscuous to show that CRISPR-Cas nucleases can induce substantial off-target mutations in mouse livers in vivo. More importantly, we also use VIVO to show that appropriately designed guide RNAs can direct efficient in vivo editing in mouse livers with no detectable off-target mutations. VIVO provides a general strategy for defining and quantifying the off-target effects of gene-editing nucleases in whole organisms, thereby providing a blueprint to foster the development of therapeutic strategies that use in vivo gene editing.


Subject(s)
CRISPR-Associated Proteins/metabolism , CRISPR-Cas Systems/genetics , Gene Editing/methods , Gene Editing/standards , Genome/genetics , Mutation , Substrate Specificity/genetics , Animals , CRISPR-Associated Proteins/genetics , Female , Humans , INDEL Mutation , Male , Mice , Mice, Inbred C57BL , Proprotein Convertase 9/genetics , Transgenes/genetics
10.
Nature ; 560(7717): E26, 2018 08.
Article in English | MEDLINE | ID: mdl-29849139

ABSTRACT

In Fig. 4e of this Article, the labels for 'Control' and 'HFD' were reversed ('Control' should have been labelled blue rather than purple, and 'HFD' should have been labelled purple rather than blue). Similarly, in Fig. 4f of this Article, the labels for 'V' and 'GW' were reversed ('V' should have been labelled blue rather than purple, and 'GW' should have been labelled purple instead of blue). The original figure has been corrected online.

11.
Mol Cell ; 57(2): 304-316, 2015 Jan 22.
Article in English | MEDLINE | ID: mdl-25578878

ABSTRACT

Polycomb repressive complex 2 (PRC2) plays crucial roles in transcriptional regulation and stem cell development. However, the context-specific functions associated with alternative subunits remain largely unexplored. Here we show that the related enzymatic subunits EZH1 and EZH2 undergo an expression switch during blood cell development. An erythroid-specific enhancer mediates transcriptional activation of EZH1, and a switch from GATA2 to GATA1 controls the developmental EZH1/2 switch by differential association with EZH1 enhancers. We further examine the in vivo stoichiometry of the PRC2 complexes by quantitative proteomics and reveal the existence of an EZH1-SUZ12 subcomplex lacking EED. EZH1 together with SUZ12 form a non-canonical PRC2 complex, occupy active chromatin, and positively regulate gene expression. Loss of EZH2 expression leads to repositioning of EZH1 to EZH2 targets. Thus, the lineage- and developmental stage-specific regulation of PRC2 subunit composition leads to a switch from canonical silencing to non-canonical functions during blood stem cell specification.


Subject(s)
GATA Transcription Factors/physiology , Polycomb Repressive Complex 2/metabolism , Base Sequence , Carcinogenesis , Enhancer of Zeste Homolog 2 Protein , Epigenesis, Genetic , Erythroid Cells/metabolism , Hematopoiesis , Hematopoietic Stem Cells , Histones/metabolism , Humans , K562 Cells , Methylation , Promoter Regions, Genetic , Protein Processing, Post-Translational , Protein Subunits
12.
Mol Cell ; 57(4): 636-647, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25699710

ABSTRACT

The mechanisms contributing to transcription-associated genomic instability are both complex and incompletely understood. Although R-loops are normal transcriptional intermediates, they are also associated with genomic instability. Here, we show that BRCA1 is recruited to R-loops that form normally over a subset of transcription termination regions. There it mediates the recruitment of a specific, physiological binding partner, senataxin (SETX). Disruption of this complex led to R-loop-driven DNA damage at those loci as reflected by adjacent γ-H2AX accumulation and ssDNA breaks within the untranscribed strand of relevant R-loop structures. Genome-wide analysis revealed widespread BRCA1 binding enrichment at R-loop-rich termination regions (TRs) of actively transcribed genes. Strikingly, within some of these genes in BRCA1 null breast tumors, there are specific insertion/deletion mutations located close to R-loop-mediated BRCA1 binding sites within TRs. Thus, BRCA1/SETX complexes support a DNA repair mechanism that addresses R-loop-based DNA damage at transcriptional pause sites.


Subject(s)
BRCA1 Protein/physiology , DNA Repair , Models, Genetic , RNA Helicases/physiology , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , DNA Damage , DNA Helicases , HeLa Cells , Humans , Multifunctional Enzymes , RNA Helicases/genetics , RNA Helicases/metabolism , Transcription Termination, Genetic , Transcription, Genetic
13.
Bioinformatics ; 37(15): 2103-2111, 2021 Aug 09.
Article in English | MEDLINE | ID: mdl-33532840

ABSTRACT

MOTIVATION: Genome-wide association studies (GWASs) have identified thousands of common trait-associated genetic variants but interpretation of their function remains challenging. These genetic variants can overlap the binding sites of transcription factors (TFs) and therefore could alter gene expression. However, we currently lack a systematic understanding on how this mechanism contributes to phenotype. RESULTS: We present Motif-Raptor, a TF-centric computational tool that integrates sequence-based predictive models, chromatin accessibility, gene expression datasets and GWAS summary statistics to systematically investigate how TF function is affected by genetic variants. Given trait-associated non-coding variants, Motif-Raptor can recover relevant cell types and critical TFs to drive hypotheses regarding their mechanism of action. We tested Motif-Raptor on complex traits such as rheumatoid arthritis and red blood cell count and demonstrated its ability to prioritize relevant cell types, potential regulatory TFs and non-coding SNPs which have been previously characterized and validated. AVAILABILITY AND IMPLEMENTATION: Motif-Raptor is freely available as a Python package at: https://github.com/pinellolab/MotifRaptor. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.

14.
PLoS Comput Biol ; 17(9): e1009444, 2021 09.
Article in English | MEDLINE | ID: mdl-34570769

ABSTRACT

Transcription factors (TFs) are proteins that promote or reduce the expression of genes by binding short genomic DNA sequences known as transcription factor binding sites (TFBS). While several tools have been developed to scan for potential occurrences of TFBS in linear DNA sequences or reference genomes, no tool exists to find them in pangenome variation graphs (VGs). VGs are sequence-labelled graphs that can efficiently encode collections of genomes and their variants in a single, compact data structure. Because VGs can losslessly compress large pangenomes, TFBS scanning in VGs can efficiently capture how genomic variation affects the potential binding landscape of TFs in a population of individuals. Here we present GRAFIMO (GRAph-based Finding of Individual Motif Occurrences), a command-line tool for the scanning of known TF DNA motifs represented as Position Weight Matrices (PWMs) in VGs. GRAFIMO extends the standard PWM scanning procedure by considering variations and alternative haplotypes encoded in a VG. Using GRAFIMO on a VG based on individuals from the 1000 Genomes project we recover several potential binding sites that are enhanced, weakened or missed when scanning only the reference genome, and which could constitute individual-specific binding events. GRAFIMO is available as an open-source tool, under the MIT license, at https://github.com/pinellolab/GRAFIMO and https://github.com/InfOmics/GRAFIMO.


Subject(s)
Genetic Variation , Nucleotide Motifs , Software , Transcription Factors/metabolism , Base Sequence , Binding Sites/genetics , Computational Biology , Computer Graphics , Genome, Human , Genomics , Haplotypes , Humans , Protein Binding/genetics
15.
Nature ; 531(7592): 53-8, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26935695

ABSTRACT

Little is known about how pro-obesity diets regulate tissue stem and progenitor cell function. Here we show that high-fat diet (HFD)-induced obesity augments the numbers and function of Lgr5(+) intestinal stem cells of the mammalian intestine. Mechanistically, a HFD induces a robust peroxisome proliferator-activated receptor delta (PPAR-δ) signature in intestinal stem cells and progenitor cells (non-intestinal stem cells), and pharmacological activation of PPAR-δ recapitulates the effects of a HFD on these cells. Like a HFD, ex vivo treatment of intestinal organoid cultures with fatty acid constituents of the HFD enhances the self-renewal potential of these organoid bodies in a PPAR-δ-dependent manner. Notably, HFD- and agonist-activated PPAR-δ signalling endow organoid-initiating capacity to progenitors, and enforced PPAR-δ signalling permits these progenitors to form in vivo tumours after loss of the tumour suppressor Apc. These findings highlight how diet-modulated PPAR-δ activation alters not only the function of intestinal stem and progenitor cells, but also their capacity to initiate tumours.


Subject(s)
Cell Transformation, Neoplastic/drug effects , Colonic Neoplasms/pathology , Diet, High-Fat/adverse effects , Intestines/pathology , Stem Cells/drug effects , Stem Cells/pathology , Animals , Cell Count , Cell Self Renewal/drug effects , Female , Genes, APC , Humans , Male , Mice , Obesity/chemically induced , Obesity/pathology , Organoids/drug effects , Organoids/metabolism , Organoids/pathology , PPAR delta/metabolism , Signal Transduction/drug effects , Stem Cell Niche/drug effects , Stem Cells/metabolism , beta Catenin/metabolism
16.
Mol Cell ; 53(1): 32-48, 2014 01 09.
Article in English | MEDLINE | ID: mdl-24361252

ABSTRACT

Self-renewal and pluripotency of embryonic stem cells (ESCs) are established by multiple regulatory pathways operating at several levels. The roles of histone demethylases (HDMs) in these programs are incompletely defined. We conducted a functional RNAi screen for HDMs and identified five potential HDMs essential for mouse ESC identity. In-depth analyses demonstrate that the closely related HDMs Jmjd2b and Jmjd2c are necessary for self-renewal of ESCs and induced pluripotent stem cell generation. Genome-wide occupancy studies reveal that Jmjd2b unique, Jmjd2c unique, and Jmjd2b-Jmjd2c common target sites belong to functionally separable Core, Polycomb repressive complex (PRC), and Myc regulatory modules, respectively. Jmjd2b and Nanog act through an interconnected regulatory loop, whereas Jmjd2c assists PRC2 in transcriptional repression. Thus, two HDMs of the same subclass exhibit distinct and combinatorial functions in control of the ESC state. Such complexity of HDM function reveals an aspect of multilayered transcriptional control.


Subject(s)
Embryonic Stem Cells/enzymology , Jumonji Domain-Containing Histone Demethylases/metabolism , Pluripotent Stem Cells/enzymology , Transcription, Genetic/physiology , Animals , Cell Line , Embryonic Stem Cells/cytology , Genome-Wide Association Study , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Mice , Nanog Homeobox Protein , Pluripotent Stem Cells/cytology , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism
17.
J Biol Chem ; 295(47): 15797-15809, 2020 11 20.
Article in English | MEDLINE | ID: mdl-32994224

ABSTRACT

Regulatory elements (REs) consist of enhancers and promoters that occupy a significant portion of the noncoding genome and control gene expression programs either in cis or in trans Putative REs have been identified largely based on their regulatory features (co-occupancy of ESC-specific transcription factors, enhancer histone marks, and DNase hypersensitivity) in mouse embryonic stem cells (mESCs). However, less has been established regarding their regulatory functions in their native context. We deployed cis- and trans-regulatory elements scanning through saturating mutagenesis and sequencing (ctSCAN-SMS) to target elements within the ∼12-kb cis-region (cis-REs; CREs) of the Oct4 gene locus, as well as genome-wide 2,613 high-confidence trans-REs (TREs), in mESCs. ctSCAN-SMS identified 10 CREs and 12 TREs as novel candidate REs of the Oct4 gene in mESCs. Furthermore, deletions of these candidate REs confirmed that the majority of the REs are functionally active, and CREs are more active than TREs in controlling Oct4 gene expression. A subset of active CREs and TREs physically interact with the Oct4 promoter to varying degrees; specifically, a greater number of active CREs, compared with active TREs, physically interact with the Oct4 promoter. Moreover, comparative genomics analysis reveals that a greater number of active CREs than active TREs are evolutionarily conserved between mice and primates, including humans. Taken together, our study demonstrates the reliability and robustness of ctSCAN-SMS screening to identify critical REs and investigate their roles in the regulation of transcriptional output of a target gene (in this case Oct4) in their native context.


Subject(s)
Genetic Loci , Mouse Embryonic Stem Cells/metabolism , Octamer Transcription Factor-3/metabolism , Regulatory Elements, Transcriptional , Animals , CRISPR-Cas Systems , Cell Line , Genome-Wide Association Study , Humans , Mice , Mouse Embryonic Stem Cells/cytology , Octamer Transcription Factor-3/genetics
18.
Bioinformatics ; 36(7): 2001-2008, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31764961

ABSTRACT

MOTIVATION: Clustered regularly interspaced short palindromic repeats (CRISPR) technologies allow for facile genomic modification in a site-specific manner. A key step in this process is the in silico design of single guide RNAs to efficiently and specifically target a site of interest. To this end, it is necessary to enumerate all potential off-target sites within a given genome that could be inadvertently altered by nuclease-mediated cleavage. Currently available software for this task is limited by computational efficiency, variant support or annotation, and assessment of the functional impact of potential off-target effects. RESULTS: To overcome these limitations, we have developed CRISPRitz, a suite of software tools to support the design and analysis of CRISPR/CRISPR-associated (Cas) experiments. Using efficient data structures combined with parallel computation, we offer a rapid, reliable, and exhaustive search mechanism to enumerate a comprehensive list of putative off-target sites. As proof-of-principle, we performed a head-to-head comparison with other available tools on several datasets. This analysis highlighted the unique features and superior computational performance of CRISPRitz including support for genomic searching with DNA/RNA bulges and mismatches of arbitrary size as specified by the user as well as consideration of genetic variants (variant-aware). In addition, graphical reports are offered for coding and non-coding regions that annotate the potential impact of putative off-target sites that lie within regions of functional genomic annotation (e.g. insulator and chromatin accessible sites from the ENCyclopedia Of DNA Elements [ENCODE] project). AVAILABILITY AND IMPLEMENTATION: The software is freely available at: https://github.com/pinellolab/CRISPRitzhttps://github.com/InfOmics/CRISPRitz. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Gene Editing , CRISPR-Cas Systems , RNA, Guide, Kinetoplastida , Software
19.
Blood ; 133(21): 2255-2262, 2019 05 23.
Article in English | MEDLINE | ID: mdl-30704988

ABSTRACT

The thalassemias are compelling targets for therapeutic genome editing in part because monoallelic correction of a subset of hematopoietic stem cells (HSCs) would be sufficient for enduring disease amelioration. A primary challenge is the development of efficient repair strategies that are effective in HSCs. Here, we demonstrate that allelic disruption of aberrant splice sites, one of the major classes of thalassemia mutations, is a robust approach to restore gene function. We target the IVS1-110G>A mutation using Cas9 ribonucleoprotein (RNP) and the IVS2-654C>T mutation by Cas12a/Cpf1 RNP in primary CD34+ hematopoietic stem and progenitor cells (HSPCs) from ß-thalassemia patients. Each of these nuclease complexes achieves high efficiency and penetrance of therapeutic edits. Erythroid progeny of edited patient HSPCs show reversal of aberrant splicing and restoration of ß-globin expression. This strategy could enable correction of a substantial fraction of transfusion-dependent ß-thalassemia genotypes with currently available gene-editing technology.


Subject(s)
Gene Editing , Gene Expression Regulation , Hematopoietic Stem Cells , RNA Splice Sites , RNA Splicing , beta-Globins , beta-Thalassemia , CRISPR-Cas Systems , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Point Mutation , beta-Globins/biosynthesis , beta-Globins/genetics , beta-Thalassemia/genetics , beta-Thalassemia/metabolism , beta-Thalassemia/therapy
20.
Nat Chem Biol ; 15(5): 529-539, 2019 05.
Article in English | MEDLINE | ID: mdl-30992567

ABSTRACT

Understanding the mechanism of small molecules is a critical challenge in chemical biology and drug discovery. Medicinal chemistry is essential for elucidating drug mechanism, enabling variation of small molecule structure to gain structure-activity relationships (SARs). However, the development of complementary approaches that systematically vary target protein structure could provide equally informative SARs for investigating drug mechanism and protein function. Here we explore the ability of CRISPR-Cas9 mutagenesis to profile the interactions between lysine-specific histone demethylase 1 (LSD1) and chemical inhibitors in the context of acute myeloid leukemia (AML). Through this approach, termed CRISPR-suppressor scanning, we elucidate drug mechanism of action by showing that LSD1 enzyme activity is not required for AML survival and that LSD1 inhibitors instead function by disrupting interactions between LSD1 and the transcription factor GFI1B on chromatin. Our studies clarify how LSD1 inhibitors mechanistically operate in AML and demonstrate how CRISPR-suppressor scanning can uncover novel aspects of target biology.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Histone Demethylases/genetics , Histone Demethylases/metabolism , Leukemia, Myeloid, Acute/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Histone Demethylases/antagonists & inhibitors , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Models, Molecular , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Repressor Proteins/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL