Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Mol Cell ; 84(3): 463-475.e5, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38242128

ABSTRACT

Type I CRISPR-Cas systems utilize the RNA-guided Cascade complex to identify matching DNA targets and the nuclease-helicase Cas3 to degrade them. Among the seven subtypes, type I-C is compact in size and highly active in creating large-sized genome deletions in human cells. Here, we use four cryoelectron microscopy snapshots to define its RNA-guided DNA binding and cleavage mechanisms in high resolution. The non-target DNA strand (NTS) is accommodated by I-C Cascade in a continuous binding groove along the juxtaposed Cas11 subunits. Binding of Cas3 further traps a flexible bulge in NTS, enabling NTS nicking. We identified two anti-CRISPR proteins AcrIC8 and AcrIC9 that strongly inhibit Neisseria lactamica I-C function. Structural analysis showed that AcrIC8 inhibits PAM recognition through allosteric inhibition, whereas AcrIC9 achieves so through direct competition. Both Acrs potently inhibit I-C-mediated genome editing and transcriptional modulation in human cells, providing the first off-switches for type I CRISPR eukaryotic genome engineering.


Subject(s)
CRISPR-Associated Proteins , Gene Editing , Humans , CRISPR-Cas Systems , Cryoelectron Microscopy , CRISPR-Associated Proteins/metabolism , DNA/metabolism , RNA
2.
Mol Cell ; 82(4): 852-867.e5, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35051351

ABSTRACT

Leading CRISPR-Cas technologies employ Cas9 and Cas12 enzymes that generate RNA-guided dsDNA breaks. Yet, the most abundant microbial adaptive immune systems, Type I CRISPRs, are under-exploited for eukaryotic applications. Here, we report the adoption of a minimal CRISPR-Cas3 from Neisseria lactamica (Nla) type I-C system to create targeted large deletions in the human genome. RNP delivery of its processive Cas3 nuclease and target recognition complex Cascade can confer ∼95% editing efficiency. Unexpectedly, NlaCascade assembly in bacteria requires internal translation of a hidden component Cas11 from within the cas8 gene. Furthermore, expressing a separately encoded NlaCas11 is the key to enable plasmid- and mRNA-based editing in human cells. Finally, we demonstrate that supplying cas11 is a universal strategy to systematically implement divergent I-C, I-D, and I-B CRISPR-Cas3 editors with compact sizes, distinct PAM preferences, and guide orthogonality. These findings greatly expand our ability to engineer long-range genome edits.


Subject(s)
Bacterial Proteins/genetics , CRISPR-Associated Proteins/genetics , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Deletion , Gene Editing , Genome, Human , Neisseria lactamica/genetics , Bacterial Proteins/metabolism , CRISPR-Associated Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Neisseria lactamica/enzymology , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism
3.
Cell ; 153(5): 1134-48, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23664764

ABSTRACT

Epigenetic mechanisms have been proposed to play crucial roles in mammalian development, but their precise functions are only partially understood. To investigate epigenetic regulation of embryonic development, we differentiated human embryonic stem cells into mesendoderm, neural progenitor cells, trophoblast-like cells, and mesenchymal stem cells and systematically characterized DNA methylation, chromatin modifications, and the transcriptome in each lineage. We found that promoters that are active in early developmental stages tend to be CG rich and mainly engage H3K27me3 upon silencing in nonexpressing lineages. By contrast, promoters for genes expressed preferentially at later stages are often CG poor and primarily employ DNA methylation upon repression. Interestingly, the early developmental regulatory genes are often located in large genomic domains that are generally devoid of DNA methylation in most lineages, which we termed DNA methylation valleys (DMVs). Our results suggest that distinct epigenetic mechanisms regulate early and late stages of ES cell differentiation.


Subject(s)
DNA Methylation , Embryonic Stem Cells/metabolism , Epigenomics , Gene Expression Regulation, Developmental , Animals , Cell Differentiation , Chromatin/metabolism , CpG Islands , Embryonic Stem Cells/cytology , Histones/metabolism , Humans , Methylation , Neoplasms/genetics , Promoter Regions, Genetic , Zebrafish/embryology
4.
Trends Biochem Sci ; 48(1): 9-10, 2023 01.
Article in English | MEDLINE | ID: mdl-36100522

ABSTRACT

High-resolution structures solved by Schuler et al. shed light on how Cas9's evolutionary ancestor IscB operates as an RNA-guided nuclease. With only two-fifths the size of Cas9, IscB holds great promise for alleviating the cargo size constraint of in vivo CRISPR delivery.


Subject(s)
CRISPR-Cas Systems , RNA , Endonucleases/genetics
5.
Mol Cell ; 74(5): 936-950.e5, 2019 06 06.
Article in English | MEDLINE | ID: mdl-30975459

ABSTRACT

CRISPR-Cas systems enable microbial adaptive immunity and provide eukaryotic genome editing tools. These tools employ a single effector enzyme of type II or V CRISPR to generate RNA-guided, precise genome breaks. Here we demonstrate the feasibility of using type I CRISPR-Cas to effectively introduce a spectrum of long-range chromosomal deletions with a single RNA guide in human embryonic stem cells and HAP1 cells. Type I CRISPR systems rely on the multi-subunit ribonucleoprotein (RNP) complex Cascade to identify DNA targets and on the helicase-nuclease enzyme Cas3 to degrade DNA processively. With RNP delivery of T. fusca Cascade and Cas3, we obtained 13%-60% editing efficiency. Long-range PCR-based and high-throughput-sequencing-based lesion analyses reveal that a variety of deletions, ranging from a few hundred base pairs to 100 kilobases, are created upstream of the target site. These results highlight the potential utility of type I CRISPR-Cas for long-range genome manipulations and deletion screens in eukaryotes.


Subject(s)
CRISPR-Cas Systems/genetics , Human Embryonic Stem Cells , RNA, Guide, Kinetoplastida/genetics , Sequence Deletion/genetics , Endonucleases/chemistry , Endonucleases/genetics , Escherichia coli/genetics , Gene Editing/methods , Genome, Human/genetics , Genomics , Humans , Ribonucleoproteins/genetics
6.
Mol Cell ; 70(5): 757-758, 2018 06 07.
Article in English | MEDLINE | ID: mdl-29883600

ABSTRACT

Cas4 nucleases are conserved factors in many CRISPR systems, yet their molecular role has remained enigmatic. In this issue of Molecular Cell, Shiimori et al. (2018) report that two Cas4 nucleases together determine the size, orientation, and PAM for foreign DNA snippets acquired by CRISPR loci as immunological memory.


Subject(s)
CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Adaptation, Physiological , DNA , Endonucleases
7.
Mol Cell ; 69(5): 906-914.e4, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29456189

ABSTRACT

The microbial CRISPR systems enable adaptive defense against mobile elements and also provide formidable tools for genome engineering. The Cas9 proteins are type II CRISPR-associated, RNA-guided DNA endonucleases that identify double-stranded DNA targets by sequence complementarity and protospacer adjacent motif (PAM) recognition. Here we report that the type II-C CRISPR-Cas9 from Neisseria meningitidis (Nme) is capable of programmable, RNA-guided, site-specific cleavage and recognition of single-stranded RNA targets and that this ribonuclease activity is independent of the PAM sequence. We define the mechanistic feature and specificity constraint for RNA cleavage by NmeCas9 and also show that nuclease null dNmeCas9 binds to RNA target complementary to CRISPR RNA. Finally, we demonstrate that NmeCas9-catalyzed RNA cleavage can be blocked by three families of type II-C anti-CRISPR proteins. These results fundamentally expand the targeting capacities of CRISPR-Cas9 and highlight the potential utility of NmeCas9 as a single platform to target both RNA and DNA.


Subject(s)
CRISPR-Cas Systems/physiology , Neisseria meningitidis/metabolism , RNA Stability/physiology , RNA, Bacterial/metabolism , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/metabolism , Neisseria meningitidis/genetics , RNA, Bacterial/genetics
8.
Mol Cell ; 70(2): 211-227.e8, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29656925

ABSTRACT

Flux through kinase and ubiquitin-driven signaling systems depends on the modification kinetics, stoichiometry, primary site specificity, and target abundance within the pathway, yet we rarely understand these parameters and their spatial organization within cells. Here we develop temporal digital snapshots of ubiquitin signaling on the mitochondrial outer membrane in embryonic stem cell-derived neurons, and we model HeLa cell systems upon activation of the PINK1 kinase and PARKIN ubiquitin ligase by proteomic counting of ubiquitylation and phosphorylation events. We define the kinetics and site specificity of PARKIN-dependent target ubiquitylation, and we demonstrate the power of this approach to quantify pathway modulators and to mechanistically define the role of PARKIN UBL phosphorylation in pathway activation in induced neurons. Finally, through modulation of pS65-Ub on mitochondria, we demonstrate that Ub hyper-phosphorylation is inhibitory to mitophagy receptor recruitment, indicating that pS65-Ub stoichiometry in vivo is optimized to coordinate PARKIN recruitment via pS65-Ub and mitophagy receptors via unphosphorylated chains.


Subject(s)
Human Embryonic Stem Cells/enzymology , Mitochondrial Membranes/enzymology , Neural Stem Cells/enzymology , Neurogenesis , Neurons/enzymology , Proteomics/methods , Ubiquitin-Protein Ligases/metabolism , Enzyme Activation , HeLa Cells , Humans , Kinetics , Mitophagy , Phenotype , Phosphorylation , Protein Kinases/genetics , Protein Kinases/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics , Ubiquitination , Voltage-Dependent Anion Channel 1/genetics , Voltage-Dependent Anion Channel 1/metabolism
9.
Methods ; 172: 32-41, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31228550

ABSTRACT

The majority of bacteria and archaea rely on CRISPR-Cas systems for RNA-guided, adaptive immunity against mobile genetic elements. The Cas9 family of type II CRISPR-associated DNA endonucleases generates programmable double strand breaks in the CRISPR-complementary DNA targets flanked by the PAM motif. Nowadays, CRISPR-Cas9 provides a set of powerful tools for precise genome manipulation in eukaryotes and prokaryotes. Recently, a few Cas9 orthologs have been reported to possess intrinsic CRISPR-guided, sequence-specific ribonuclease activities. These discoveries fundamentally expanded the targeting capability of CRISPR-Cas9 systems, and promise to provide new CRISPR tools to manipulate specific cellular RNA transcripts. Here we present a detailed method for the biochemical characterization of Cas9's RNA-targeting potential.


Subject(s)
CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems/genetics , Enzyme Assays/methods , RNA, Guide, Kinetoplastida/metabolism , Bacteriophages/metabolism , CRISPR-Associated Protein 9/antagonists & inhibitors , Enzyme Assays/instrumentation , Neisseria meningitidis/enzymology , Neisseria meningitidis/genetics , Neisseria meningitidis/virology , RNA, Guide, Kinetoplastida/genetics , Viral Proteins/metabolism
10.
Proc Natl Acad Sci U S A ; 114(30): E6072-E6078, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28696312

ABSTRACT

Here, we report the derivation of arterial endothelial cells from human pluripotent stem cells that exhibit arterial-specific functions in vitro and in vivo. We combine single-cell RNA sequencing of embryonic mouse endothelial cells with an EFNB2-tdTomato/EPHB4-EGFP dual reporter human embryonic stem cell line to identify factors that regulate arterial endothelial cell specification. The resulting xeno-free protocol produces cells with gene expression profiles, oxygen consumption rates, nitric oxide production levels, shear stress responses, and TNFα-induced leukocyte adhesion rates characteristic of arterial endothelial cells. Arterial endothelial cells were robustly generated from multiple human embryonic and induced pluripotent stem cell lines and have potential applications for both disease modeling and regenerative medicine.


Subject(s)
Arteries/cytology , Endothelial Cells/transplantation , Neovascularization, Physiologic , Pluripotent Stem Cells/physiology , Tissue Engineering/methods , Animals , CRISPR-Cas Systems , Cell Line , Endothelial Cells/cytology , Humans , Mice , Myocardial Infarction/therapy , Sequence Analysis, RNA
11.
Proc Natl Acad Sci U S A ; 112(40): 12516-21, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26392547

ABSTRACT

Human pluripotent stem cell-based in vitro models that reflect human physiology have the potential to reduce the number of drug failures in clinical trials and offer a cost-effective approach for assessing chemical safety. Here, human embryonic stem (ES) cell-derived neural progenitor cells, endothelial cells, mesenchymal stem cells, and microglia/macrophage precursors were combined on chemically defined polyethylene glycol hydrogels and cultured in serum-free medium to model cellular interactions within the developing brain. The precursors self-assembled into 3D neural constructs with diverse neuronal and glial populations, interconnected vascular networks, and ramified microglia. Replicate constructs were reproducible by RNA sequencing (RNA-Seq) and expressed neurogenesis, vasculature development, and microglia genes. Linear support vector machines were used to construct a predictive model from RNA-Seq data for 240 neural constructs treated with 34 toxic and 26 nontoxic chemicals. The predictive model was evaluated using two standard hold-out testing methods: a nearly unbiased leave-one-out cross-validation for the 60 training compounds and an unbiased blinded trial using a single hold-out set of 10 additional chemicals. The linear support vector produced an estimate for future data of 0.91 in the cross-validation experiment and correctly classified 9 of 10 chemicals in the blinded trial.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Neural Stem Cells/cytology , Pluripotent Stem Cells/cytology , Brain/cytology , Brain/growth & development , Brain/metabolism , Cell Communication/drug effects , Cell Communication/genetics , Cells, Cultured , Culture Media, Serum-Free/pharmacology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Expression Regulation, Developmental , Gene Ontology , Humans , Hydrogels/pharmacology , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Microglia/cytology , Microglia/drug effects , Microglia/metabolism , Models, Biological , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurogenesis/drug effects , Neurogenesis/genetics , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Polyethylene Glycols/pharmacology , Support Vector Machine , Tissue Engineering/methods , Xenobiotics/classification , Xenobiotics/pharmacology
12.
Proc Natl Acad Sci U S A ; 110(39): 15644-9, 2013 Sep 24.
Article in English | MEDLINE | ID: mdl-23940360

ABSTRACT

Genome engineering in human pluripotent stem cells (hPSCs) holds great promise for biomedical research and regenerative medicine. Recently, an RNA-guided, DNA-cleaving interference pathway from bacteria [the type II clustered, regularly interspaced, short palindromic repeats (CRISPR)-CRISPR-associated (Cas) pathway] has been adapted for use in eukaryotic cells, greatly facilitating genome editing. Only two CRISPR-Cas systems (from Streptococcus pyogenes and Streptococcus thermophilus), each with their own distinct targeting requirements and limitations, have been developed for genome editing thus far. Furthermore, limited information exists about homology-directed repair (HDR)-mediated gene targeting using long donor DNA templates in hPSCs with these systems. Here, using a distinct CRISPR-Cas system from Neisseria meningitidis, we demonstrate efficient targeting of an endogenous gene in three hPSC lines using HDR. The Cas9 RNA-guided endonuclease from N. meningitidis (NmCas9) recognizes a 5'-NNNNGATT-3' protospacer adjacent motif (PAM) different from those recognized by Cas9 proteins from S. pyogenes and S. thermophilus (SpCas9 and StCas9, respectively). Similar to SpCas9, NmCas9 is able to use a single-guide RNA (sgRNA) to direct its activity. Because of its distinct protospacer adjacent motif, the N. meningitidis CRISPR-Cas machinery increases the sequence contexts amenable to RNA-directed genome editing.


Subject(s)
Bacterial Proteins/metabolism , Genetic Engineering/methods , Genome, Human/genetics , Neisseria meningitidis/metabolism , Pluripotent Stem Cells/metabolism , Animals , Base Sequence , Cell Line , Gene Deletion , Gene Targeting , Humans , Mammals , Molecular Sequence Data , RNA/metabolism , RNA Editing/genetics , RNA, Small Untranslated
13.
Bioinformatics ; 30(18): 2665-7, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24872422

ABSTRACT

UNLABELLED: Aptamers are 'synthetic antibodies' that can bind to target molecules with high affinity and specificity. Aptamers are chemically synthesized and their discovery can be performed completely in vitro, rather than relying on in vivo biological processes, making them well-suited for high-throughput discovery. However, a large fraction of the most enriched aptamers in Systematic Evolution of Ligands by EXponential enrichment (SELEX) rounds display poor binding activity. Here, we present MPBind, a Meta-motif-based statistical framework and pipeline to Predict the BIND: ing potential of SELEX-derived aptamers. Using human embryonic stem cell SELEX-Seq data, MPBind achieved high prediction accuracy for binding potential. Further analysis showed that MPBind is robust to both polymerase chain reaction amplification bias and incomplete sequencing of aptamer pools. These two biases usually confound aptamer analysis. AVAILABILITY AND IMPLEMENTATION: MPBind software and documents are available at http://www.morgridge.net/MPBind.html. The human embryonic stem cells whole-cell SELEX-Seq data are available at http://www.morgridge.net/Aptamer/.


Subject(s)
Aptamers, Nucleotide/metabolism , Computational Biology/methods , SELEX Aptamer Technique , Software , Embryonic Stem Cells/metabolism , Humans , Ligands , Oligonucleotides/metabolism , Sequence Analysis , Substrate Specificity
14.
Proc Natl Acad Sci U S A ; 109(19): 7162-8, 2012 May 08.
Article in English | MEDLINE | ID: mdl-22474382

ABSTRACT

The transcription factor OCT4 is fundamental to maintaining pluripotency and self-renewal. To better understand protein-level regulation of OCT4, we applied liquid chromatography-MS to identify 14 localized sites of phosphorylation, 11 of which were previously unknown. Functional analysis of two sites, T234 and S235, suggested that phosphorylation within the homeobox region of OCT4 negatively regulates its activity by interrupting sequence-specific DNA binding. Mutating T234 and S235 to mimic constitutive phosphorylation at these sites reduces transcriptional activation from an OCT4-responsive reporter and decreases reprogramming efficiency. We also cataloged 144 unique phosphopeptides on known OCT4 interacting partners, including SOX2 and SALL4, that copurified during immunoprecipitation. These proteins were enriched for phosphorylation at motifs associated with ERK signaling. Likewise, OCT4 harbored several putative ERK phosphorylation sites. Kinase assays confirmed that ERK2 phosphorylated these sites in vitro, providing a direct link between ERK signaling and the transcriptional machinery that governs pluripotency.


Subject(s)
Embryonic Stem Cells/metabolism , Octamer Transcription Factor-3/metabolism , Serine/metabolism , Threonine/metabolism , Amino Acid Sequence , Binding Sites/genetics , Blotting, Western , Cells, Cultured , HEK293 Cells , Humans , Immunoprecipitation , Mitogen-Activated Protein Kinase 1/metabolism , Models, Molecular , Molecular Sequence Data , Mutation , Octamer Transcription Factor-3/chemistry , Octamer Transcription Factor-3/genetics , Phosphorylation , Protein Binding , Protein Structure, Tertiary , SOXB1 Transcription Factors/metabolism , Sequence Homology, Amino Acid , Serine/chemistry , Serine/genetics , Threonine/chemistry , Threonine/genetics , Transcription Factors/metabolism , Transcriptional Activation
15.
Nat Methods ; 8(5): 424-9, 2011 May.
Article in English | MEDLINE | ID: mdl-21478862

ABSTRACT

We re-examine the individual components for human embryonic stem cell (ESC) and induced pluripotent stem cell (iPSC) culture and formulate a cell culture system in which all protein reagents for liquid media, attachment surfaces and splitting are chemically defined. A major improvement is the lack of a serum albumin component, as variations in either animal- or human-sourced albumin batches have previously plagued human ESC and iPSC culture with inconsistencies. Using this new medium (E8) and vitronectin-coated surfaces, we demonstrate improved derivation efficiencies of vector-free human iPSCs with an episomal approach. This simplified E8 medium should facilitate both the research use and clinical applications of human ESCs and iPSCs and their derivatives, and should be applicable to other reprogramming methods.


Subject(s)
Cell Culture Techniques/methods , Culture Media/chemistry , Induced Pluripotent Stem Cells/cytology , Animals , Biopsy , Cattle , Cell Proliferation , Cell Survival , Coated Materials, Biocompatible , Culture Media, Serum-Free/chemistry , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Fibroblasts/cytology , Gene Expression , Growth Substances , Humans , Induced Pluripotent Stem Cells/metabolism , Karyotyping , Serum Albumin, Bovine , Skin/cytology , Vitronectin
16.
PLoS Comput Biol ; 9(10): e1003252, 2013.
Article in English | MEDLINE | ID: mdl-24146602

ABSTRACT

Regulatory networks that control gene expression are important in diverse biological contexts including stress response and development. Each gene's regulatory program is determined by module-level regulation (e.g. co-regulation via the same signaling system), as well as gene-specific determinants that can fine-tune expression. We present a novel approach, Modular regulatory network learning with per gene information (MERLIN), that infers regulatory programs for individual genes while probabilistically constraining these programs to reveal module-level organization of regulatory networks. Using edge-, regulator- and module-based comparisons of simulated networks of known ground truth, we find MERLIN reconstructs regulatory programs of individual genes as well or better than existing approaches of network reconstruction, while additionally identifying modular organization of the regulatory networks. We use MERLIN to dissect global transcriptional behavior in two biological contexts: yeast stress response and human embryonic stem cell differentiation. Regulatory modules inferred by MERLIN capture co-regulatory relationships between signaling proteins and downstream transcription factors thereby revealing the upstream signaling systems controlling transcriptional responses. The inferred networks are enriched for regulators with genetic or physical interactions, supporting the inference, and identify modules of functionally related genes bound by the same transcriptional regulators. Our method combines the strengths of per-gene and per-module methods to reveal new insights into transcriptional regulation in stress and development.


Subject(s)
Computational Biology/methods , Gene Regulatory Networks/genetics , Models, Genetic , Signal Transduction/genetics , Cluster Analysis , Gene Regulatory Networks/physiology , Humans , Saccharomyces cerevisiae , Signal Transduction/physiology , Software , Transcription Factors/genetics , Transcription Factors/metabolism
17.
Stem Cells ; 30(4): 623-30, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22213113

ABSTRACT

Fibroblast growth factor (FGF), transforming growth factor (TGF)/Nodal, and Insulin/insulin-like growth factor (IGF) signaling pathways are sufficient to maintain human embryonic stem cells (ESCs) and induced pluripotent stem cells in a proliferative, undifferentiated state. Here, we show that only a few FGF family members (FGF2, FGF4, FGF6, and FGF9) are able to sustain strong extracellular-signal-regulated kinase (ERK) phosphorylation and NANOG expression levels in human ESCs. Surprisingly, FGF1, which is reported to target the same set of receptors as FGF2, fails to sustain ERK phosphorylation and NANOG expression under standard culture conditions. We find that the failure of FGF1 to sustain ES is due to thermal instability of the wild-type protein, not receptor specificity, and that a mutated thermal-stable FGF1 sustains human ESCs and supports both differentiation and reprogramming protocols.


Subject(s)
Cell Differentiation/drug effects , Cellular Reprogramming/drug effects , Fibroblast Growth Factors/pharmacology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Temperature , Cell Proliferation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblast Growth Factors/metabolism , Heparin/metabolism , Humans , Mutant Proteins/metabolism , Mutation/genetics , Phosphorylation/drug effects , Pluripotent Stem Cells/enzymology , Protein Stability/drug effects , Receptors, Fibroblast Growth Factor/metabolism
18.
bioRxiv ; 2023 Aug 06.
Article in English | MEDLINE | ID: mdl-37577534

ABSTRACT

Type I CRISPR-Cas systems utilize the RNA-guided Cascade complex to identify matching DNA targets, and the nuclease-helicase Cas3 to degrade them. Among seven subtypes, Type I-C is compact in size and highly active in creating large-sized genome deletions in human cells. Here we use four cryo-electron microscopy snapshots to define its RNA-guided DNA binding and cleavage mechanisms in high resolution. The non-target DNA strand (NTS) is accommodated by I-C Cascade in a continuous binding groove along the juxtaposed Cas11 subunits. Binding of Cas3 further traps a flexible bulge in NTS, enabling efficient NTS nicking. We identified two anti-CRISPR proteins AcrIC8 and AcrIC9, that strongly inhibit N. lactamica I-C function. Structural analysis showed that AcrIC8 inhibits PAM recognition through direct competition, whereas AcrIC9 achieves so through allosteric inhibition. Both Acrs potently inhibit I-C-mediated genome editing and transcriptional modulation in human cells, providing the first off-switches for controllable Type I CRISPR genome engineering.

19.
PLoS Genet ; 5(12): e1000755, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19997491

ABSTRACT

The origin recognition complex (ORC) marks chromosomal sites as replication origins and is essential for replication initiation. In yeast, ORC also binds to DNA elements called silencers, where its primary function is to recruit silent information regulator (SIR) proteins to establish transcriptional silencing. Indeed, silencers function poorly as chromosomal origins. Several genetic, molecular, and biochemical studies of HMR-E have led to a model proposing that when ORC becomes limiting in the cell (such as in the orc2-1 mutant) only sites that bind ORC tightly (such as HMR-E) remain fully occupied by ORC, while lower affinity sites, including many origins, lose ORC occupancy. Since HMR-E possessed a unique non-replication function, we reasoned that other tight sites might reveal novel functions for ORC on chromosomes. Therefore, we comprehensively determined ORC "affinity" genome-wide by performing an ORC ChIP-on-chip in ORC2 and orc2-1 strains. Here we describe a novel group of orc2-1-resistant ORC-interacting chromosomal sites (ORF-ORC sites) that did not function as replication origins or silencers. Instead, ORF-ORC sites were comprised of protein-coding regions of highly transcribed metabolic genes. In contrast to the ORC-silencer paradigm, transcriptional activation promoted ORC association with these genes. Remarkably, ORF-ORC genes were enriched in proximity to origins of replication and, in several instances, were transcriptionally regulated by these origins. Taken together, these results suggest a surprising connection among ORC, replication origins, and cellular metabolism.


Subject(s)
Metabolic Networks and Pathways/genetics , Origin Recognition Complex/metabolism , Replication Origin/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Binding Sites , Chromatin Immunoprecipitation , Chromosomes, Fungal/genetics , Gene Expression Regulation, Fungal , Open Reading Frames/genetics , Protein Binding , Reproducibility of Results , Saccharomyces cerevisiae Proteins/metabolism , Sequence Deletion , Silencer Elements, Transcriptional/genetics , Transcription, Genetic
20.
Curr Protoc ; 2(2): e361, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35129865

ABSTRACT

CRISPR-Cas systems provide researchers with eukaryotic genome editing tools and therapeutic platforms that make it possible to target disease mutations in somatic organs. Most of these tools employ Type II (e.g., Cas9) or Type V (e.g., Cas12a) CRISPR enzymes to create RNA-guided precise double-strand breaks in the genome. However, such technologies are limited in their capacity to make targeted large deletions. Recently, the Type I CRISPR system, which is prevalent in microbes and displays unique enzymatic features, has been harnessed to effectively create large chromosomal deletions in human cells. Type I CRISPR first uses a multisubunit ribonucleoprotein (RNP) complex called Cascade to find its guide-complementary target site, and then recruits a helicase-nuclease enzyme, Cas3, to travel along and shred the target DNA over a long distance with high processivity. When introduced into human cells as purified RNPs, the CRISPR-Cas3 complex can efficiently induce large genomic deletions of varying lengths (1-100 kb) from the CRISPR-targeted site. Because of this unique editing outcome, CRISPR-Cas3 holds great promise for tasks such as the removal of integrated viral genomes and the interrogation of structural variants affecting gene function and human disease. Here, we provide detailed protocols for introducing large deletions using CRISPR-Cas3. We describe step-by-step procedures for purifying the Type I-E CRISPR proteins Cascade and Cas3 from Thermobifida fusca, electroporating RNPs into human cells, and characterizing DNA deletions using PCR and sequencing. We focus here on human pluripotent stem cells due to their clinical potential, but these protocols will be broadly useful for other cell lines and model organisms for applications including large genomic deletion, full-gene or -chromosome removal, and CRISPR screening for noncoding elements, among others. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Expression and purification of Tfu Cascade RNP Support Protocol 1: Expression and purification of TfuCas3 protein Support Protocol 2: Culture of human pluripotent stem cells Basic Protocol 2: Introduction of Tfu Cascade RNP and Cas3 protein into hPSCs via electroporation Basic Protocol 3: Characterization of genomic DNA lesions using long-range PCR, TOPO cloning, and Sanger sequencing Alternate Protocol: Comprehensive analysis of genomic lesions by Tn5-based next-generation sequencing Support Protocol 3: Single-cell clonal isolation.


Subject(s)
CRISPR-Associated Proteins , Pluripotent Stem Cells , CRISPR-Associated Proteins/genetics , CRISPR-Cas Systems/genetics , Gene Editing , Genomics , Humans , Pluripotent Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL