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










Publication year range
1.
Blood Cancer Discov ; 2024 Apr 07.
Article in English | MEDLINE | ID: mdl-38583144
2.
Proc Natl Acad Sci U S A ; 118(30)2021 07 27.
Article in English | MEDLINE | ID: mdl-34285077

ABSTRACT

Dysfunction in T cells limits the efficacy of cancer immunotherapy. We profiled the epigenome, transcriptome, and enhancer connectome of exhaustion-prone GD2-targeting HA-28z chimeric antigen receptor (CAR) T cells and control CD19-targeting CAR T cells, which present less exhaustion-inducing tonic signaling, at multiple points during their ex vivo expansion. We found widespread, dynamic changes in chromatin accessibility and three-dimensional (3D) chromosome conformation preceding changes in gene expression, notably at loci proximal to exhaustion-associated genes such as PDCD1, CTLA4, and HAVCR2, and increased DNA motif access for AP-1 family transcription factors, which are known to promote exhaustion. Although T cell exhaustion has been studied in detail in mice, we find that the regulatory networks of T cell exhaustion differ between species and involve distinct loci of accessible chromatin and cis-regulated target genes in human CAR T cell exhaustion. Deletion of exhaustion-specific candidate enhancers of PDCD1 suppress the expression of PD-1 in an in vitro model of T cell dysfunction and in HA-28z CAR T cells, suggesting enhancer editing as a path forward in improving cancer immunotherapy.


Subject(s)
Chromatin/metabolism , Neoplasms/therapy , Programmed Cell Death 1 Receptor/metabolism , Receptors, Chimeric Antigen , T-Lymphocytes/physiology , Animals , Antigens, CD19 , Cell Line , Chromatin/genetics , Gene Expression Regulation, Neoplastic , Humans , Mice , Programmed Cell Death 1 Receptor/genetics
3.
Nature ; 576(7786): 293-300, 2019 12.
Article in English | MEDLINE | ID: mdl-31802004

ABSTRACT

Chimeric antigen receptor (CAR) T cells mediate anti-tumour effects in a small subset of patients with cancer1-3, but dysfunction due to T cell exhaustion is an important barrier to progress4-6. To investigate the biology of exhaustion in human T cells expressing CAR receptors, we used a model system with a tonically signaling CAR, which induces hallmark features of exhaustion6. Exhaustion was associated with a profound defect in the production of IL-2, along with increased chromatin accessibility of AP-1 transcription factor motifs and overexpression of the bZIP and IRF transcription factors that have been implicated in mediating dysfunction in exhausted T cells7-10. Here we show that CAR T cells engineered to overexpress the canonical AP-1 factor c-Jun have enhanced expansion potential, increased functional capacity, diminished terminal differentiation and improved anti-tumour potency in five different mouse tumour models in vivo. We conclude that a functional deficiency in c-Jun mediates dysfunction in exhausted human T cells, and that engineering CAR T cells to overexpress c-Jun renders them resistant to exhaustion, thereby addressing a major barrier to progress for this emerging class of therapeutic agents.


Subject(s)
Proto-Oncogene Proteins c-jun/metabolism , Receptors, Antigen, T-Cell/immunology , Animals , Cell Line, Tumor , Epigenesis, Genetic , Gene Expression Regulation , Humans , Mice , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , Proto-Oncogene Proteins c-jun/genetics , Receptors, Antigen, T-Cell/genetics , Transcription Factor AP-1/genetics , Transcription Factor AP-1/immunology , Transcription, Genetic
4.
Nat Methods ; 16(6): 489-492, 2019 06.
Article in English | MEDLINE | ID: mdl-31133759

ABSTRACT

Modular domains of long non-coding RNAs can serve as scaffolds to bring distant regions of the linear genome into spatial proximity. Here, we present HiChIRP, a method leveraging bio-orthogonal chemistry and optimized chromosome conformation capture conditions, which enables interrogation of chromatin architecture focused around a specific RNA of interest down to approximately ten copies per cell. HiChIRP of three nuclear RNAs reveals insights into promoter interactions (7SK), telomere biology (telomerase RNA component) and inflammatory gene regulation (lincRNA-EPS).


Subject(s)
Chromatin/chemistry , Chromatin/genetics , Embryonic Stem Cells/metabolism , Gene Expression Regulation , RNA, Long Noncoding/genetics , RNA/chemistry , Telomerase/chemistry , Animals , Cells, Cultured , Chromosomes , Embryonic Stem Cells/cytology , Genome , Mice , Promoter Regions, Genetic , RNA/genetics , Telomerase/genetics
5.
Nat Med ; 24(5): 580-590, 2018 05.
Article in English | MEDLINE | ID: mdl-29686426

ABSTRACT

T cells create vast amounts of diversity in the genes that encode their T cell receptors (TCRs), which enables individual clones to recognize specific peptide-major histocompatibility complex (MHC) ligands. Here we combined sequencing of the TCR-encoding genes with assay for transposase-accessible chromatin with sequencing (ATAC-seq) analysis at the single-cell level to provide information on the TCR specificity and epigenomic state of individual T cells. By using this approach, termed transcript-indexed ATAC-seq (T-ATAC-seq), we identified epigenomic signatures in immortalized leukemic T cells, primary human T cells from healthy volunteers and primary leukemic T cells from patient samples. In peripheral blood CD4+ T cells from healthy individuals, we identified cis and trans regulators of naive and memory T cell states and found substantial heterogeneity in surface-marker-defined T cell populations. In patients with a leukemic form of cutaneous T cell lymphoma, T-ATAC-seq enabled identification of leukemic and nonleukemic regulatory pathways in T cells from the same individual by allowing separation of the signals that arose from the malignant clone from the background T cell noise. Thus, T-ATAC-seq is a new tool that enables analysis of epigenomic landscapes in clonal T cells and should be valuable for studies of T cell malignancy, immunity and immunotherapy.


Subject(s)
Chromatin/metabolism , High-Throughput Nucleotide Sequencing/methods , Transposases/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cell Line, Transformed , Clone Cells , Epigenomics , Humans , Immunity , Jurkat Cells , Leukemia/immunology , Leukemia/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Antigen, T-Cell/metabolism , Single-Cell Analysis
7.
Cell ; 166(6): 1500-1511.e9, 2016 Sep 08.
Article in English | MEDLINE | ID: mdl-27610572

ABSTRACT

Reversing the dysfunctional T cell state that arises in cancer and chronic viral infections is the focus of therapeutic interventions; however, current therapies are effective in only some patients and some tumor types. To gain a deeper molecular understanding of the dysfunctional T cell state, we analyzed population and single-cell RNA profiles of CD8(+) tumor-infiltrating lymphocytes (TILs) and used genetic perturbations to identify a distinct gene module for T cell dysfunction that can be uncoupled from T cell activation. This distinct dysfunction module is downstream of intracellular metallothioneins that regulate zinc metabolism and can be identified at single-cell resolution. We further identify Gata-3, a zinc-finger transcription factor in the dysfunctional module, as a regulator of dysfunction, and we use CRISPR-Cas9 genome editing to show that it drives a dysfunctional phenotype in CD8(+) TILs. Our results open novel avenues for targeting dysfunctional T cell states while leaving activation programs intact.


Subject(s)
CD8-Positive T-Lymphocytes/pathology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Animals , CD8-Positive T-Lymphocytes/immunology , CRISPR-Cas Systems , Carcinogenesis/genetics , Carcinogenesis/immunology , Female , GATA3 Transcription Factor/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Humans , Melanoma/immunology , Melanoma/physiopathology , Metallothionein/deficiency , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
8.
Nat Biotechnol ; 33(5): 495-502, 2015 May.
Article in English | MEDLINE | ID: mdl-25867923

ABSTRACT

Spatial localization is a key determinant of cellular fate and behavior, but methods for spatially resolved, transcriptome-wide gene expression profiling across complex tissues are lacking. RNA staining methods assay only a small number of transcripts, whereas single-cell RNA-seq, which measures global gene expression, separates cells from their native spatial context. Here we present Seurat, a computational strategy to infer cellular localization by integrating single-cell RNA-seq data with in situ RNA patterns. We applied Seurat to spatially map 851 single cells from dissociated zebrafish (Danio rerio) embryos and generated a transcriptome-wide map of spatial patterning. We confirmed Seurat's accuracy using several experimental approaches, then used the strategy to identify a set of archetypal expression patterns and spatial markers. Seurat correctly localizes rare subpopulations, accurately mapping both spatially restricted and scattered groups. Seurat will be applicable to mapping cellular localization within complex patterned tissues in diverse systems.


Subject(s)
In Situ Hybridization, Fluorescence/methods , Single-Cell Analysis/methods , Zebrafish/growth & development , Animals , Gene Expression Regulation, Developmental , High-Throughput Nucleotide Sequencing , Image Processing, Computer-Assisted , Transcriptome/genetics , Zebrafish/genetics
9.
Nature ; 516(7529): 56-61, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25471879

ABSTRACT

Pluripotent stem cells (PSCs) are capable of dynamic interconversion between distinct substates; however, the regulatory circuits specifying these states and enabling transitions between them are not well understood. Here we set out to characterize transcriptional heterogeneity in mouse PSCs by single-cell expression profiling under different chemical and genetic perturbations. Signalling factors and developmental regulators show highly variable expression, with expression states for some variable genes heritable through multiple cell divisions. Expression variability and population heterogeneity can be influenced by perturbation of signalling pathways and chromatin regulators. Notably, either removal of mature microRNAs or pharmacological blockage of signalling pathways drives PSCs into a low-noise ground state characterized by a reconfigured pluripotency network, enhanced self-renewal and a distinct chromatin state, an effect mediated by opposing microRNA families acting on the Myc/Lin28/let-7 axis. These data provide insight into the nature of transcriptional heterogeneity in PSCs.


Subject(s)
Gene Expression Regulation, Developmental , Pluripotent Stem Cells/physiology , Animals , Cell Death , Cell Division , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Gene Expression Profiling , Mice , MicroRNAs/metabolism , Pluripotent Stem Cells/cytology , Signal Transduction
10.
Curr Protoc Mol Biol ; 107: 4.22.1-4.22.17, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24984854

ABSTRACT

For the past several decades, due to technical limitations, the field of transcriptomics has focused on population-level measurements that can mask significant differences between individual cells. With the advent of single-cell RNA-Seq, it is now possible to profile the responses of individual cells at unprecedented depth and thereby uncover, transcriptome-wide, the heterogeneity that exists within these populations. This unit describes a method that merges several important technologies to produce, in high-throughput, single-cell RNA-Seq libraries. Complementary DNA (cDNA) is made from full-length mRNA transcripts using a reverse transcriptase that has terminal transferase activity. This, when combined with a second "template-switch" primer, allows for cDNAs to be constructed that have two universal priming sequences. Following preamplification from these common sequences, Nextera XT is used to prepare a pool of 96 uniquely indexed samples ready for Illumina sequencing.


Subject(s)
Gene Library , High-Throughput Nucleotide Sequencing/methods , RNA, Messenger , Sequence Analysis, RNA/methods , Animals , DNA, Complementary/chemistry , DNA, Complementary/genetics , Humans , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/isolation & purification
11.
Nature ; 496(7446): 461-8, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23467089

ABSTRACT

Despite their importance, the molecular circuits that control the differentiation of naive T cells remain largely unknown. Recent studies that reconstructed regulatory networks in mammalian cells have focused on short-term responses and relied on perturbation-based approaches that cannot be readily applied to primary T cells. Here we combine transcriptional profiling at high temporal resolution, novel computational algorithms, and innovative nanowire-based perturbation tools to systematically derive and experimentally validate a model of the dynamic regulatory network that controls the differentiation of mouse TH17 cells, a proinflammatory T-cell subset that has been implicated in the pathogenesis of multiple autoimmune diseases. The TH17 transcriptional network consists of two self-reinforcing, but mutually antagonistic, modules, with 12 novel regulators, the coupled action of which may be essential for maintaining the balance between TH17 and other CD4(+) T-cell subsets. Our study identifies and validates 39 regulatory factors, embeds them within a comprehensive temporal network and reveals its organizational principles; it also highlights novel drug targets for controlling TH17 cell differentiation.


Subject(s)
Cell Differentiation/genetics , Gene Regulatory Networks/genetics , Th17 Cells/cytology , Th17 Cells/metabolism , Animals , Cells, Cultured , DNA/genetics , DNA/metabolism , Forkhead Transcription Factors/metabolism , Gene Knockdown Techniques , Genome/genetics , Interferon-gamma/biosynthesis , Interleukin-2/genetics , Mice , Mice, Inbred C57BL , Nanowires , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , Silicon , Th17 Cells/immunology , Time Factors , Trans-Activators/metabolism , Transcription Factors/metabolism , Transcription, Genetic/genetics , fas Receptor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...