Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Cell Rep ; 43(6): 114313, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38838224

ABSTRACT

Personalized cancer therapeutics bring directed treatment options to patients based on their tumor's genetic signature. Unfortunately, tumor genomes are remarkably adaptable, and acquired resistance through gene mutation frequently occurs. Identifying mutations that promote resistance within drug-treated patient populations can be cost, resource, and time intensive. Accordingly, base editing, enabled by Cas9-deaminase domain fusions, has emerged as a promising approach for rapid, large-scale gene variant screening in situ. Here, we adapt and optimize a conditional activation-induced cytidine deaminase (AID)-dead Cas9 (dCas9) system, which demonstrates greater heterogeneity of edits with an expanded footprint compared to the most commonly utilized cytosine base editor, BE4. In combination with a custom single guide RNA (sgRNA) library, we identify individual and compound variants in epidermal growth factor receptor (EGFR) and v-raf murine sarcoma viral oncogene homolog B1 (BRAF) that confer resistance to established EGFR inhibitors. This system and analytical pipeline provide a simple, highly scalable platform for cis or trans drug-modifying variant discovery and for uncovering valuable insights into protein structure-function relationships.


Subject(s)
Drug Resistance, Neoplasm , ErbB Receptors , Humans , Drug Resistance, Neoplasm/genetics , ErbB Receptors/metabolism , ErbB Receptors/genetics , ErbB Receptors/antagonists & inhibitors , Cell Line, Tumor , Gene Editing/methods , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , CRISPR-Cas Systems/genetics , Mutation/genetics , Mutagenesis
2.
Nat Commun ; 14(1): 902, 2023 02 17.
Article in English | MEDLINE | ID: mdl-36804928

ABSTRACT

CRISPR-mediated transcriptional activation (CRISPRa) is a powerful technology for inducing gene expression from endogenous loci with exciting applications in high throughput gain-of-function genomic screens and the engineering of cell-based models. However, current strategies for generating potent, stable, CRISPRa-competent cell lines present limitations for the broad utility of this approach. Here, we provide a high-efficiency, self-selecting CRISPRa enrichment strategy, which combined with piggyBac transposon technology enables rapid production of CRISPRa-ready cell populations compatible with a variety of downstream assays. We complement this with an optimized guide RNA scaffold that significantly enhances CRISPRa functionality. Finally, we describe a synthetic guide RNA tool set that enables transient, population-wide gene activation when used with the self-selecting CRISPRa system. Taken together, this versatile platform greatly enhances the potential for CRISPRa across a wide variety of cellular contexts.


Subject(s)
Genome , RNA , Transcriptional Activation , Cell Line , CRISPR-Cas Systems/genetics
3.
Nat Commun ; 12(1): 5771, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34599190

ABSTRACT

Germline specification in mammals occurs through an inductive process whereby competent cells in the post-implantation epiblast differentiate into primordial germ cells (PGC). The intrinsic factors that endow epiblast cells with the competence to respond to germline inductive signals remain unknown. Single-cell RNA sequencing across multiple stages of an in vitro PGC-like cells (PGCLC) differentiation system shows that PGCLC genes initially expressed in the naïve pluripotent stage become homogeneously dismantled in germline competent epiblast like-cells (EpiLC). In contrast, the decommissioning of enhancers associated with these germline genes is incomplete. Namely, a subset of these enhancers partly retain H3K4me1, accumulate less heterochromatic marks and remain accessible and responsive to transcriptional activators. Subsequently, as in vitro germline competence is lost, these enhancers get further decommissioned and lose their responsiveness to transcriptional activators. Importantly, using H3K4me1-deficient cells, we show that the loss of this histone modification reduces the germline competence of EpiLC and decreases PGCLC differentiation efficiency. Our work suggests that, although H3K4me1 might not be essential for enhancer function, it can facilitate the (re)activation of enhancers and the establishment of gene expression programs during specific developmental transitions.


Subject(s)
Enhancer Elements, Genetic , Germ Cells/metabolism , Histones/metabolism , Lysine/metabolism , Animals , Cell Differentiation , Chromatin/metabolism , Embryo, Mammalian/cytology , Gene Expression Regulation , Germ Cells/cytology , Germ Layers/cytology , Male , Methylation , Mice , Mice, Transgenic , Mouse Embryonic Stem Cells/cytology , Mutation/genetics , Otx Transcription Factors/genetics , Otx Transcription Factors/metabolism , RNA-Seq , Single-Cell Analysis , Transcription Initiation Site , Transcription, Genetic
5.
Cell Res ; 28(2): 204-220, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29313530

ABSTRACT

Long-range chromatin interactions between enhancers and promoters are essential for transcription of many developmentally controlled genes in mammals and other metazoans. Currently, the exact mechanisms that connect distal enhancers to their specific target promoters remain to be fully elucidated. Here, we show that the enhancer-specific histone H3 lysine 4 monomethylation (H3K4me1) and the histone methyltransferases MLL3 and MLL4 (MLL3/4) play an active role in this process. We demonstrate that in differentiating mouse embryonic stem cells, MLL3/4-dependent deposition of H3K4me1 at enhancers correlates with increased levels of chromatin interactions, whereas loss of this histone modification leads to reduced levels of chromatin interactions and defects in gene activation during differentiation. H3K4me1 facilitates recruitment of the Cohesin complex, a known regulator of chromatin organization, to chromatin in vitro and in vivo, providing a potential mechanism for MLL3/4 to promote chromatin interactions between enhancers and promoters. Taken together, our results support a role for MLL3/4-dependent H3K4me1 in orchestrating long-range chromatin interactions at enhancers in mammalian cells.


Subject(s)
Chromatin/metabolism , Enhancer Elements, Genetic/physiology , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Animals , Cell Cycle Proteins/metabolism , Cell Differentiation/physiology , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/metabolism , Gene Expression/physiology , In Situ Hybridization, Fluorescence , Methylation , Mice , Promoter Regions, Genetic/physiology , SOXB1 Transcription Factors/metabolism , Sequence Analysis, RNA , Cohesins
6.
Mol Cell ; 66(4): 568-576.e4, 2017 May 18.
Article in English | MEDLINE | ID: mdl-28483418

ABSTRACT

Monomethylation of histone H3 at lysine 4 (H3K4me1) and acetylation of histone H3 at lysine 27 (H3K27ac) are correlated with transcriptionally engaged enhancer elements, but the functional impact of these modifications on enhancer activity is not well understood. Here we used CRISPR/Cas9 genome editing to separate catalytic activity-dependent and independent functions of Mll3 (Kmt2c) and Mll4 (Kmt2d, Mll2), the major enhancer H3K4 monomethyltransferases. Loss of H3K4me1 from enhancers in Mll3/4 catalytically deficient cells causes partial reduction of H3K27ac, but has surprisingly minor effects on transcription from either enhancers or promoters. In contrast, loss of Mll3/4 proteins leads to strong depletion of enhancer Pol II occupancy and eRNA synthesis, concomitant with downregulation of target genes. Interestingly, downregulated genes exhibit reduced polymerase levels in gene bodies, but not at promoters, suggestive of pause-release defects. Altogether, our results suggest that enhancer H3K4me1 provides only a minor contribution to the long-range coactivator function of Mll3/4.


Subject(s)
Embryonic Stem Cells/enzymology , Enhancer Elements, Genetic , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Promoter Regions, Genetic , RNA/biosynthesis , Transcription, Genetic , Animals , CRISPR-Cas Systems , Cell Line , Gene Editing , Gene Expression Regulation, Developmental , Histone-Lysine N-Methyltransferase/genetics , Male , Methylation , Mice , Mutation , RNA/genetics , Time Factors , Transcriptional Activation , Transfection
7.
Development ; 140(20): 4182-92, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24004944

ABSTRACT

Members of the Polycomb group of repressors and trithorax group of activators maintain heritable states of transcription by modifying nucleosomal histones or remodeling chromatin. Although tremendous progress has been made toward defining the biochemical activities of Polycomb and trithorax group proteins, much remains to be learned about how they interact with each other and the general transcription machinery to maintain on or off states of gene expression. The trithorax group protein Kismet (KIS) is related to the SWI/SNF and CHD families of chromatin remodeling factors. KIS promotes transcription elongation, facilitates the binding of the trithorax group histone methyltransferases ASH1 and TRX to active genes, and counteracts repressive methylation of histone H3 on lysine 27 (H3K27) by Polycomb group proteins. Here, we sought to clarify the mechanism of action of KIS and how it interacts with ASH1 to antagonize H3K27 methylation in Drosophila. We present evidence that KIS promotes transcription elongation and counteracts Polycomb group repression via distinct mechanisms. A chemical inhibitor of transcription elongation, DRB, had no effect on ASH1 recruitment or H3K27 methylation. Conversely, loss of ASH1 function had no effect on transcription elongation. Mutations in kis cause a global reduction in the di- and tri-methylation of histone H3 on lysine 36 (H3K36) - modifications that antagonize H3K27 methylation in vitro. Furthermore, loss of ASH1 significantly decreases H3K36 dimethylation, providing further evidence that ASH1 is an H3K36 dimethylase in vivo. These and other findings suggest that KIS antagonizes Polycomb group repression by facilitating ASH1-dependent H3K36 dimethylation.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila/metabolism , Homeodomain Proteins/metabolism , Polycomb-Group Proteins/metabolism , Transcription Factors/metabolism , Animals , Chromatin Assembly and Disassembly , Dichlororibofuranosylbenzimidazole/pharmacology , Drosophila/genetics , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/metabolism , Histones , Methylation , Nucleic Acid Synthesis Inhibitors/pharmacology , Salivary Glands/metabolism , Transcription, Genetic/drug effects
8.
PLoS Genet ; 8(8): e1002878, 2012.
Article in English | MEDLINE | ID: mdl-22912596

ABSTRACT

dMi-2 is a highly conserved ATP-dependent chromatin-remodeling factor that regulates transcription and cell fates by altering the structure or positioning of nucleosomes. Here we report an unanticipated role for dMi-2 in the regulation of higher-order chromatin structure in Drosophila. Loss of dMi-2 function causes salivary gland polytene chromosomes to lose their characteristic banding pattern and appear more condensed than normal. Conversely, increased expression of dMi-2 triggers decondensation of polytene chromosomes accompanied by a significant increase in nuclear volume; this effect is relatively rapid and is dependent on the ATPase activity of dMi-2. Live analysis revealed that dMi-2 disrupts interactions between the aligned chromatids of salivary gland polytene chromosomes. dMi-2 and the cohesin complex are enriched at sites of active transcription; fluorescence-recovery after photobleaching (FRAP) assays showed that dMi-2 decreases stable association of cohesin with polytene chromosomes. These findings demonstrate that dMi-2 is an important regulator of both chromosome condensation and cohesin binding in interphase cells.


Subject(s)
Adenosine Triphosphatases/genetics , Autoantigens/genetics , Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Nucleosomes/genetics , Polytene Chromosomes/genetics , Adenosine Triphosphatases/metabolism , Animals , Autoantigens/metabolism , Cell Cycle Proteins/metabolism , Chromatids , Chromosomal Proteins, Non-Histone/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Fluorescence Recovery After Photobleaching , Interphase/genetics , Protein Binding , Salivary Glands/cytology , Salivary Glands/metabolism , Cohesins
9.
Genes Dev ; 24(23): 2693-704, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21123654

ABSTRACT

Splicing regulatory networks are essential components of eukaryotic gene expression programs, yet little is known about how they are integrated with transcriptional regulatory networks into coherent gene expression programs. Here we define the MER1 splicing regulatory network and examine its role in the gene expression program during meiosis in budding yeast. Mer1p splicing factor promotes splicing of just four pre-mRNAs. All four Mer1p-responsive genes also require Nam8p for splicing activation by Mer1p; however, other genes require Nam8p but not Mer1p, exposing an overlapping meiotic splicing network controlled by Nam8p. MER1 mRNA and three of the four Mer1p substrate pre-mRNAs are induced by the transcriptional regulator Ume6p. This unusual arrangement delays expression of Mer1p-responsive genes relative to other genes under Ume6p control. Products of Mer1p-responsive genes are required for initiating and completing recombination and for activation of Ndt80p, the activator of the transcriptional network required for subsequent steps in the program. Thus, the MER1 splicing regulatory network mediates the dependent relationship between the UME6 and NDT80 transcriptional regulatory networks in the meiotic gene expression program. This study reveals how splicing regulatory networks can be interlaced with transcriptional regulatory networks in eukaryotic gene expression programs.


Subject(s)
Gene Expression Regulation, Fungal , RNA Splicing , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Epistasis, Genetic/genetics , Gene Deletion , Genome, Fungal/genetics , Introns/genetics , Meiosis , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
10.
PLoS Genet ; 4(10): e1000217, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18846226

ABSTRACT

Polycomb and trithorax group proteins regulate cellular pluripotency and differentiation by maintaining hereditable states of transcription. Many Polycomb and trithorax group proteins have been implicated in the covalent modification or remodeling of chromatin, but how they interact with each other and the general transcription machinery to regulate transcription is not well understood. The trithorax group protein Kismet-L (KIS-L) is a member of the CHD subfamily of chromatin-remodeling factors that plays a global role in transcription by RNA polymerase II (Pol II). Mutations in CHD7, the human counterpart of kis, are associated with CHARGE syndrome, a developmental disorder affecting multiple tissues and organs. To clarify how KIS-L activates gene expression and counteracts Polycomb group silencing, we characterized defects resulting from the loss of KIS-L function in Drosophila. These studies revealed that KIS-L acts downstream of P-TEFb recruitment to stimulate elongation by Pol II. The presence of two chromodomains in KIS-L suggested that its recruitment or function might be regulated by the methylation of histone H3 lysine 4 by the trithorax group proteins ASH1 and TRX. Although we observed significant overlap between the distributions of KIS-L, ASH1, and TRX on polytene chromosomes, KIS-L did not bind methylated histone tails in vitro, and loss of TRX or ASH1 function did not alter the association of KIS-L with chromatin. By contrast, loss of kis function led to a dramatic reduction in the levels of TRX and ASH1 associated with chromatin and was accompanied by increased histone H3 lysine 27 methylation-a modification required for Polycomb group repression. A similar increase in H3 lysine 27 methylation was observed in ash1 and trx mutant larvae. Our findings suggest that KIS-L promotes early elongation and counteracts Polycomb group repression by recruiting the ASH1 and TRX histone methyltransferases to chromatin.


Subject(s)
DNA Helicases/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Epigenesis, Genetic , Histones/metabolism , Homeodomain Proteins/metabolism , RNA Polymerase II/metabolism , Animals , Chromatin Assembly and Disassembly/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA Helicases/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Genes, Insect , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/chemistry , Histones/genetics , Homeodomain Proteins/genetics , Lysine/chemistry , Methylation , Protein Processing, Post-Translational , Transcription Factors/genetics , Transcription Factors/metabolism
11.
Hum Immunol ; 68(5): 309-23, 2007 May.
Article in English | MEDLINE | ID: mdl-17462498

ABSTRACT

The influence of donor and recipient killer immunoglobulin-like receptor (KIR) genotype on the outcome of hematopoietic cell transplantation between human leukocyte antigen (HLA)-matched siblings was investigated. Transplants were divided into four groups according to the combination of group A and B KIR haplotypes in the transplant donor and recipient. Overall survival of myeloid patients varied with KIR genotype combination. Best survival was associated with the donor lacking and the recipient having group B KIR haplotypes; poorest survival was associated with the donor having and the recipient lacking group B KIR haplotypes. The latter combination was also associated with increased relapse and acute graft-versus-host disease (GVHD). However, its detrimental effects were seen only for transplants where the recipient and donor were homozygous for the C1 KIR ligand and therefore lacked the C2 ligand. Presence of the Bw4 ligand was also associated with increased acute GVHD. In contrast presence of both KIR3DL1 and its cognate Bw4 ligand was associated with decreased nonrelapse mortality. Analysis of the KIR genes individually revealed KIR2DS3 as a protective factor for chronic GVHD. The results suggest how simple assessments of KIR genotype might inform the selection of donors for hematopoietic cell transplantation.


Subject(s)
Hematopoietic Stem Cell Transplantation , Histocompatibility Antigens Class I/genetics , Leukemia/surgery , Receptors, Immunologic/genetics , Adolescent , Adult , Aged , Disease-Free Survival , Gene Frequency , Genotype , Graft vs Host Disease/genetics , HLA-A Antigens/genetics , HLA-B Antigens/genetics , HLA-C Antigens/genetics , Haplotypes , Histocompatibility Testing/statistics & numerical data , Humans , Leukemia/genetics , Leukemia/immunology , Middle Aged , Proportional Hazards Models , Receptors, KIR , Receptors, KIR3DL1 , Risk Factors , Siblings , Survival Analysis , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...