Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Cell ; 173(3): 624-633.e8, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29656892

ABSTRACT

CTLA-4 immune checkpoint blockade is clinically effective in a subset of patients with metastatic melanoma. We identify a subcluster of MAGE-A cancer-germline antigens, located within a narrow 75 kb region of chromosome Xq28, that predicts resistance uniquely to blockade of CTLA-4, but not PD-1. We validate this gene expression signature in an independent anti-CTLA-4-treated cohort and show its specificity to the CTLA-4 pathway with two independent anti-PD-1-treated cohorts. Autophagy, a process critical for optimal anti-cancer immunity, has previously been shown to be suppressed by the MAGE-TRIM28 ubiquitin ligase in vitro. We now show that the expression of the key autophagosome component LC3B and other activators of autophagy are negatively associated with MAGE-A protein levels in human melanomas, including samples from patients with resistance to CTLA-4 blockade. Our findings implicate autophagy suppression in resistance to CTLA-4 blockade in melanoma, suggesting exploitation of autophagy induction for potential therapeutic synergy with CTLA-4 inhibitors.


Subject(s)
CTLA-4 Antigen/genetics , CTLA-4 Antigen/immunology , Epigenesis, Genetic , Germ-Line Mutation , Neoplasms/genetics , Neoplasms/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Autophagy , Cell Line, Tumor , DNA Methylation , Female , Gene Expression Profiling , Humans , Immunotherapy , Ipilimumab/pharmacology , Male , Melanoma/genetics , Melanoma/immunology , Melanoma-Specific Antigens/genetics , Melanoma-Specific Antigens/immunology , Mice , Mice, Transgenic , Skin Neoplasms/genetics , Skin Neoplasms/immunology
2.
Mol Cell ; 46(5): 705-13, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22681891

ABSTRACT

Extensive changes in posttranslational histone modifications accompany the rewiring of the transcriptional program during stem cell differentiation. However, the mechanisms controlling the changes in specific chromatin modifications and their function during differentiation remain only poorly understood. We show that histone H2B monoubiquitination (H2Bub1) significantly increases during differentiation of human mesenchymal stem cells (hMSCs) and various lineage-committed precursor cells and in diverse organisms. Furthermore, the H2B ubiquitin ligase RNF40 is required for the induction of differentiation markers and transcriptional reprogramming of hMSCs. This function is dependent upon CDK9 and the WAC adaptor protein, which are required for H2B monoubiquitination. Finally, we show that RNF40 is required for the resolution of the H3K4me3/H3K27me3 bivalent poised state on lineage-specific genes during the transition from an inactive to an active chromatin conformation. Thus, these data indicate that H2Bub1 is required for maintaining multipotency of hMSCs and plays a central role in controlling stem cell differentiation.


Subject(s)
Cell Differentiation/genetics , Histones/metabolism , Mesenchymal Stem Cells/cytology , Multipotent Stem Cells/cytology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/physiology , Cell Line , Chromatin Assembly and Disassembly , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/physiology , Humans , Mesenchymal Stem Cells/metabolism , Multipotent Stem Cells/metabolism , Protein Processing, Post-Translational , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/physiology , Ubiquitination
3.
Cancer Discov ; 12(1): 220-235, 2022 01.
Article in English | MEDLINE | ID: mdl-34429321

ABSTRACT

Clonal hematopoiesis is a prevalent age-related condition associated with a greatly increased risk of hematologic disease; mutations in DNA methyltransferase 3A (DNMT3A) are the most common driver of this state. DNMT3A variants occur across the gene with some particularly associated with malignancy, but the functional relevance and mechanisms of pathogenesis of the majority of mutations are unknown. Here, we systematically investigated the methyltransferase activity and protein stability of 253 disease-associated DNMT3A mutations, and found that 74% were loss-of-function mutations. Half of these variants exhibited reduced protein stability and, as a class, correlated with greater clonal expansion and acute myeloid leukemia development. We investigated the mechanisms underlying the instability using a CRISPR screen and uncovered regulated destruction of DNMT3A mediated by the DCAF8 E3 ubiquitin ligase adaptor. We establish a new paradigm to classify novel variants that has prognostic and potential therapeutic significance for patients with hematologic disease. SIGNIFICANCE: DNMT3A has emerged as the most important epigenetic regulator and tumor suppressor in the hematopoietic system. Our study represents a systematic and high-throughput method to characterize the molecular impact of DNMT3A missense mutations and the discovery of a regulated destruction mechanism of DNMT3A offering new prognostic and future therapeutic avenues.See related commentary by Ma and Will, p. 23.This article is highlighted in the In This Issue feature, p. 1.


Subject(s)
DNA Methyltransferase 3A/genetics , Leukemia, Myeloid, Acute/genetics , Ubiquitin-Protein Ligases/genetics , Animals , HEK293 Cells , Humans , Leukocytes, Mononuclear , Mice , Mutation, Missense
4.
Nat Struct Mol Biol ; 28(7): 594-603, 2021 07.
Article in English | MEDLINE | ID: mdl-34140676

ABSTRACT

DNA methylation plays a critical role during development, particularly in repressing retrotransposons. The mammalian methylation landscape is dependent on the combined activities of the canonical maintenance enzyme Dnmt1 and the de novo Dnmts, 3a and 3b. Here, we demonstrate that Dnmt1 displays de novo methylation activity in vitro and in vivo with specific retrotransposon targeting. We used whole-genome bisulfite and long-read Nanopore sequencing in genetically engineered methylation-depleted mouse embryonic stem cells to provide an in-depth assessment and quantification of this activity. Utilizing additional knockout lines and molecular characterization, we show that the de novo methylation activity of Dnmt1 depends on Uhrf1, and its genomic recruitment overlaps with regions that enrich for Uhrf1, Trim28 and H3K9 trimethylation. Our data demonstrate that Dnmt1 can catalyze DNA methylation in both a de novo and maintenance context, especially at retrotransposons, where this mechanism may provide additional stability for long-term repression and epigenetic propagation throughout development.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Methylation/genetics , DNA Transposable Elements/genetics , Embryonic Development/genetics , Animals , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cells, Cultured , Chromatin/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , Gene Knockout Techniques , Genome/genetics , Histones/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Tripartite Motif-Containing Protein 28/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Whole Genome Sequencing , DNA Methyltransferase 3B
5.
Nat Commun ; 12(1): 4219, 2021 07 09.
Article in English | MEDLINE | ID: mdl-34244505

ABSTRACT

Streptococcus pyogenes (Spy) Cas9 has potential as a component of gene therapeutics for incurable diseases. One of its limitations is its large size, which impedes its formulation and delivery in therapeutic applications. Smaller Cas9s are an alternative, but lack robust activity or specificity and frequently recognize longer PAMs. Here, we investigated four uncharacterized, smaller Cas9s and found three employing a "GG" dinucleotide PAM similar to SpyCas9. Protein engineering generated synthetic RNA-guided nucleases (sRGNs) with editing efficiencies and specificities exceeding even SpyCas9 in vitro and in human cell lines on disease-relevant targets. sRGN mRNA lipid nanoparticles displayed manufacturing advantages and high in vivo editing efficiency in the mouse liver. Finally, sRGNs, but not SpyCas9, could be packaged into all-in-one AAV particles with a gRNA and effected robust in vivo editing of non-human primate (NHP) retina photoreceptors. Human gene therapy efforts are expected to benefit from these improved alternatives to existing CRISPR nucleases.


Subject(s)
CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems/genetics , Gene Editing/methods , Staphylococcus/enzymology , Animals , CRISPR-Associated Protein 9/isolation & purification , Cell Line, Tumor , Dependovirus , Disease Models, Animal , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , HEK293 Cells , Humans , Macaca fascicularis , Male , Mice , Parvovirinae/genetics , Protein Engineering , Ribonucleases , Staphylococcus/genetics , Substrate Specificity , Usher Syndromes/genetics , Usher Syndromes/therapy , RNA, Guide, CRISPR-Cas Systems
6.
Nat Cell Biol ; 22(4): 389-400, 2020 04.
Article in English | MEDLINE | ID: mdl-32231305

ABSTRACT

In mouse embryonic stem cells (mESCs), chemical blockade of Gsk3α/ß and Mek1/2 (2i) instructs a self-renewing ground state whose endogenous inducers are unknown. Here we show that the axon guidance cue Netrin-1 promotes naive pluripotency by triggering profound signalling, transcriptomic and epigenetic changes in mESCs. Furthermore, we demonstrate that Netrin-1 can substitute for blockade of Gsk3α/ß and Mek1/2 to sustain self-renewal of mESCs in combination with leukaemia inhibitory factor and regulates the formation of the mouse pluripotent blastocyst. Mechanistically, we reveal how Netrin-1 and the balance of its receptors Neo1 and Unc5B co-regulate Wnt and MAPK pathways in both mouse and human ESCs. Netrin-1 induces Fak kinase to inactivate Gsk3α/ß and stabilize ß-catenin while increasing the phosphatase activity of a Ppp2r2c-containing Pp2a complex to reduce Erk1/2 activity. Collectively, this work identifies Netrin-1 as a regulator of pluripotency and reveals that it mediates different effects in mESCs depending on its receptor dosage, opening perspectives for balancing self-renewal and lineage commitment.


Subject(s)
Gene Expression Regulation, Developmental , Nerve Tissue Proteins/genetics , Netrin Receptors/genetics , Netrin-1/genetics , Receptors, Cell Surface/genetics , Wnt Signaling Pathway/genetics , Animals , Cell Line , Embryo, Mammalian , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Leukemia Inhibitory Factor/genetics , Leukemia Inhibitory Factor/metabolism , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Kinase 2/genetics , MAP Kinase Kinase 2/metabolism , Male , Mice , Mice, Knockout , Mice, SCID , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Nerve Tissue Proteins/metabolism , Netrin Receptors/metabolism , Netrin-1/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Receptors, Cell Surface/metabolism , beta Catenin/genetics , beta Catenin/metabolism
7.
Nat Biotechnol ; 37(12): 1478-1481, 2019 12.
Article in English | MEDLINE | ID: mdl-31740840

ABSTRACT

Expansions of short tandem repeats are genetic variants that have been implicated in several neuropsychiatric and other disorders, but their assessment remains challenging with current polymerase-based methods1-4. Here we introduce a CRISPR-Cas-based enrichment strategy for nanopore sequencing combined with an algorithm for raw signal analysis. Our method, termed STRique for short tandem repeat identification, quantification and evaluation, integrates conventional sequence mapping of nanopore reads with raw signal alignment for the localization of repeat boundaries and a hidden Markov model-based repeat counting mechanism. We demonstrate the precise quantification of repeat numbers in conjunction with the determination of CpG methylation states in the repeat expansion and in adjacent regions at the single-molecule level without amplification. Our method enables the study of previously inaccessible genomic regions and their epigenetic marks.


Subject(s)
DNA Methylation/genetics , Genomics/methods , Microsatellite Repeats/genetics , Nanopore Sequencing/methods , Algorithms , Amyotrophic Lateral Sclerosis/genetics , C9orf72 Protein/genetics , CRISPR-Cas Systems/genetics , Cells, Cultured , Humans , Nanopores
8.
Nat Commun ; 9(1): 597, 2018 02 09.
Article in English | MEDLINE | ID: mdl-29426832

ABSTRACT

In normal mammalian development cytosine methylation is essential and is directed to specific regions of the genome. Despite notable advances through mapping its genome-wide distribution, studying the direct contribution of DNA methylation to gene and genome regulation has been limited by the lack of tools for its precise manipulation. Thus, combining the targeting capability of the CRISPR-Cas9 system with an epigenetic modifier has attracted interest in the scientific community. In contrast to profiling the genome-wide cleavage of a nuclease competent Cas9, tracing the global activity of a dead Cas9 (dCas9) methyltransferase fusion protein is challenging within a highly methylated genome. Here, we report the generation and use of an engineered, methylation depleted but maintenance competent mouse ES cell line and find surprisingly ubiquitous nuclear activity of dCas9-methyltransferases. Subsequent experiments in human somatic cells refine these observations and point to an important difference between genetic and epigenetic editing tools that require unique experimental considerations.


Subject(s)
Cell Line , DNA (Cytosine-5-)-Methyltransferases/metabolism , Embryonic Stem Cells/enzymology , Animals , Bacterial Proteins , CRISPR-Associated Protein 9 , Endonucleases , Gene Editing , Humans , Mice
9.
Cell Stem Cell ; 22(4): 559-574.e9, 2018 04 05.
Article in English | MEDLINE | ID: mdl-29551301

ABSTRACT

The somatic DNA methylation (DNAme) landscape is established early in development but remains highly dynamic within focal regions that overlap with gene regulatory elements. The significance of these dynamic changes, particularly in the central nervous system, remains unresolved. Here, we utilize a powerful human embryonic stem cell differentiation model for the generation of motor neurons (MNs) in combination with genetic mutations in the de novo DNAme machinery. We quantitatively dissect the role of DNAme in directing somatic cell fate with high-resolution genome-wide bisulfite-, bulk-, and single-cell-RNA sequencing. We find defects in neuralization and MN differentiation in DNMT3A knockouts (KO) that can be rescued by the targeting of DNAme to key developmental loci using catalytically inactive dCas9. We also find decreased dendritic arborization and altered electrophysiological properties in DNMT3A KO MNs. Our work provides a list of DNMT3A-regulated targets and a mechanistic link between de novo DNAme, cellular differentiation, and human MN function.


Subject(s)
Cell Differentiation , DNA Methylation , Motor Neurons/cytology , Motor Neurons/metabolism , Biocatalysis , Cell Differentiation/genetics , DNA (Cytosine-5-)-Methyltransferases/deficiency , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/genetics , DNA Methyltransferase 3A , Humans
11.
Cell Stem Cell ; 17(4): 462-70, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26235340

ABSTRACT

Mouse embryonic stem cells (mESCs) cultured under serum/LIF conditions exhibit heterogeneous expression of pluripotency-associated factors that can be overcome by two inhibitors (2i) of the MEK and GSK3 pathways. Several studies have shown that the "ground state" induced by 2i is characterized by global hypomethylation and specific transcriptional profiles, but little is known about the contributing effectors. Here we show that 2i conditions rapidly alter the global binding landscape of OCT4, SOX2, and NANOG. The dynamic binding influences enhancer activity and shows enrichment for regulators linked to Wnt and Erk signaling. Epigenomic characterization provided limited insights to the immediate transcriptional dynamics, suggesting that these are likely more secondary effects. Likewise, loss of the PRC2 component EED to prevent H3K27me3 deposition had minimal effect on the transcriptome, implying that it is largely dispensable for continued repression of bivalent genes and de novo silencing in 2i.


Subject(s)
Epigenesis, Genetic , Mouse Embryonic Stem Cells/physiology , Animals , Cells, Cultured , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/genetics , Mice , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Polycomb Repressive Complex 2/genetics , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Transcriptome
12.
Nat Genet ; 47(5): 469-78, 2015 May.
Article in English | MEDLINE | ID: mdl-25822089

ABSTRACT

DNA methylation is a key epigenetic modification involved in regulating gene expression and maintaining genomic integrity. Here we inactivated all three catalytically active DNA methyltransferases (DNMTs) in human embryonic stem cells (ESCs) using CRISPR/Cas9 genome editing to further investigate the roles and genomic targets of these enzymes. Disruption of DNMT3A or DNMT3B individually as well as of both enzymes in tandem results in viable, pluripotent cell lines with distinct effects on the DNA methylation landscape, as assessed by whole-genome bisulfite sequencing. Surprisingly, in contrast to findings in mouse, deletion of DNMT1 resulted in rapid cell death in human ESCs. To overcome this immediate lethality, we generated a doxycycline-responsive tTA-DNMT1* rescue line and readily obtained homozygous DNMT1-mutant lines. However, doxycycline-mediated repression of exogenous DNMT1* initiates rapid, global loss of DNA methylation, followed by extensive cell death. Our data provide a comprehensive characterization of DNMT-mutant ESCs, including single-base genome-wide maps of the targets of these enzymes.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation , Embryonic Stem Cells/enzymology , Animals , Apoptosis , Base Sequence , Catalytic Domain , Cell Differentiation , Cell Proliferation , Cell Survival , Cells, Cultured , Coculture Techniques , CpG Islands , DNA (Cytosine-5-)-Methyltransferase 1 , DNA Methyltransferase 3A , Embryonic Stem Cells/physiology , Epigenesis, Genetic , Gene Expression , Gene Knockout Techniques , Humans , Mice , DNA Methyltransferase 3B
13.
Stem Cell Reports ; 2(3): 245-52, 2014 Mar 11.
Article in English | MEDLINE | ID: mdl-24672748

ABSTRACT

Pluripotent stem cells retain the ability to differentiate into the three germ layers and germline. As a result, there is a major interest in characterizing regulators that establish and maintain pluripotency. The network of transcription factors continues to expand in complexity, and one factor, undifferentiated embryonic cell transcription factor 1 (UTF1), has recently moved more into the limelight. To facilitate the study of UTF1, we report the generation and characterization of two reporter lines that enable efficient tracking, mapping, and purification of endogenous UTF1. In particular, we include a built-in biotinylation system in our targeted locus that allows efficient and reliable pulldown. We also use this reporter to show the dynamic regulation of Utf1 in distinct stem cell conditions and demonstrate its utility for reprogramming studies. The multipurpose design of the reporter lines enables many directions of future study and should lead to a better understanding of UTF1's diverse roles.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Animals , Biomarkers , Cell Differentiation , Cell Line , Cellular Reprogramming/genetics , DNA/genetics , DNA/metabolism , Embryo, Mammalian , Embryonic Stem Cells , Gene Expression , Gene Expression Regulation, Developmental , Gene Order , Genes, Reporter , Genetic Vectors/genetics , Mice , Pluripotent Stem Cells , Protein Binding , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL