Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 122
Filter
Add more filters

Publication year range
1.
Cell ; 185(7): 1223-1239.e20, 2022 03 31.
Article in English | MEDLINE | ID: mdl-35290801

ABSTRACT

While CRISPR screens are helping uncover genes regulating many cell-intrinsic processes, existing approaches are suboptimal for identifying extracellular gene functions, particularly in the tissue context. Here, we developed an approach for spatial functional genomics called Perturb-map. We applied Perturb-map to knock out dozens of genes in parallel in a mouse model of lung cancer and simultaneously assessed how each knockout influenced tumor growth, histopathology, and immune composition. Moreover, we paired Perturb-map and spatial transcriptomics for unbiased analysis of CRISPR-edited tumors. We found that in Tgfbr2 knockout tumors, the tumor microenvironment (TME) was converted to a fibro-mucinous state, and T cells excluded, concomitant with upregulated TGFß and TGFß-mediated fibroblast activation, indicating that TGFß-receptor loss on cancer cells increased TGFß bioavailability and its immunosuppressive effects on the TME. These studies establish Perturb-map for functional genomics within the tissue at single-cell resolution with spatial architecture preserved and provide insight into how TGFß responsiveness of cancer cells can affect the TME.


Subject(s)
Neoplasms , Tumor Microenvironment , Animals , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Genomics , Mice , Neoplasms/genetics , Transforming Growth Factor beta/genetics
2.
Nat Immunol ; 24(6): 1020-1035, 2023 06.
Article in English | MEDLINE | ID: mdl-37127830

ABSTRACT

While regulatory T (Treg) cells are traditionally viewed as professional suppressors of antigen presenting cells and effector T cells in both autoimmunity and cancer, recent findings of distinct Treg cell functions in tissue maintenance suggest that their regulatory purview extends to a wider range of cells and is broader than previously assumed. To elucidate tumoral Treg cell 'connectivity' to diverse tumor-supporting accessory cell types, we explored immediate early changes in their single-cell transcriptomes upon punctual Treg cell depletion in experimental lung cancer and injury-induced inflammation. Before any notable T cell activation and inflammation, fibroblasts, endothelial and myeloid cells exhibited pronounced changes in their gene expression in both cancer and injury settings. Factor analysis revealed shared Treg cell-dependent gene programs, foremost, prominent upregulation of VEGF and CCR2 signaling-related genes upon Treg cell deprivation in either setting, as well as in Treg cell-poor versus Treg cell-rich human lung adenocarcinomas. Accordingly, punctual Treg cell depletion combined with short-term VEGF blockade showed markedly improved control of PD-1 blockade-resistant lung adenocarcinoma progression in mice compared to the corresponding monotherapies, highlighting a promising factor-based querying approach to elucidating new rational combination treatments of solid organ cancers.


Subject(s)
Neoplasms , T-Lymphocytes, Regulatory , Animals , Mice , Humans , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Tumor Microenvironment , Neoplasms/metabolism
3.
Cell ; 183(3): 702-716.e14, 2020 10 29.
Article in English | MEDLINE | ID: mdl-33125890

ABSTRACT

The cellular complexity and scale of the early liver have constrained analyses examining its emergence during organogenesis. To circumvent these issues, we analyzed 45,334 single-cell transcriptomes from embryonic day (E)7.5, when endoderm progenitors are specified, to E10.5 liver, when liver parenchymal and non-parenchymal cell lineages emerge. Our data detail divergence of vascular and sinusoidal endothelia, including a distinct transcriptional profile for sinusoidal endothelial specification by E8.75. We characterize two distinct mesothelial cell types as well as early hepatic stellate cells and reveal distinct spatiotemporal distributions for these populations. We capture transcriptional profiles for hepatoblast specification and migration, including the emergence of a hepatomesenchymal cell type and evidence for hepatoblast collective cell migration. Further, we identify cell-cell interactions during the organization of the primitive sinusoid. This study provides a comprehensive atlas of liver lineage establishment from the endoderm and mesoderm through to the organization of the primitive sinusoid at single-cell resolution.


Subject(s)
Cell Lineage/genetics , Liver/cytology , Liver/metabolism , Single-Cell Analysis , Transcriptome/genetics , Animals , Cell Movement , Embryo, Mammalian/cytology , Endothelium/cytology , Mesoderm/cytology , Mice , Signal Transduction , Stem Cells/cytology
4.
Cell ; 181(2): 236-249, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32302568

ABSTRACT

Crucial transitions in cancer-including tumor initiation, local expansion, metastasis, and therapeutic resistance-involve complex interactions between cells within the dynamic tumor ecosystem. Transformative single-cell genomics technologies and spatial multiplex in situ methods now provide an opportunity to interrogate this complexity at unprecedented resolution. The Human Tumor Atlas Network (HTAN), part of the National Cancer Institute (NCI) Cancer Moonshot Initiative, will establish a clinical, experimental, computational, and organizational framework to generate informative and accessible three-dimensional atlases of cancer transitions for a diverse set of tumor types. This effort complements both ongoing efforts to map healthy organs and previous large-scale cancer genomics approaches focused on bulk sequencing at a single point in time. Generating single-cell, multiparametric, longitudinal atlases and integrating them with clinical outcomes should help identify novel predictive biomarkers and features as well as therapeutically relevant cell types, cell states, and cellular interactions across transitions. The resulting tumor atlases should have a profound impact on our understanding of cancer biology and have the potential to improve cancer detection, prevention, and therapeutic discovery for better precision-medicine treatments of cancer patients and those at risk for cancer.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Neoplasms/metabolism , Tumor Microenvironment/physiology , Atlases as Topic , Cell Transformation, Neoplastic/pathology , Genomics/methods , Humans , Precision Medicine/methods , Single-Cell Analysis/methods
5.
Cell ; 179(4): 846-863.e24, 2019 10 31.
Article in English | MEDLINE | ID: mdl-31668803

ABSTRACT

Dendritic cells (DCs) play a critical role in orchestrating adaptive immune responses due to their unique ability to initiate T cell responses and direct their differentiation into effector lineages. Classical DCs have been divided into two subsets, cDC1 and cDC2, based on phenotypic markers and their distinct abilities to prime CD8 and CD4 T cells. While the transcriptional regulation of the cDC1 subset has been well characterized, cDC2 development and function remain poorly understood. By combining transcriptional and chromatin analyses with genetic reporter expression, we identified two principal cDC2 lineages defined by distinct developmental pathways and transcriptional regulators, including T-bet and RORγt, two key transcription factors known to define innate and adaptive lymphocyte subsets. These novel cDC2 lineages were characterized by distinct metabolic and functional programs. Extending our findings to humans revealed conserved DC heterogeneity and the presence of the newly defined cDC2 subsets in human cancer.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/genetics , Genetic Heterogeneity , Neoplasms/immunology , Adaptive Immunity/genetics , Animals , Cell Differentiation/immunology , Chromatin/genetics , Dendritic Cells/immunology , Gene Expression Regulation, Developmental , Humans , Immunity, Innate/genetics , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Mice , Neoplasms/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transcription, Genetic/immunology
6.
Cell ; 174(3): 716-729.e27, 2018 07 26.
Article in English | MEDLINE | ID: mdl-29961576

ABSTRACT

Single-cell RNA sequencing technologies suffer from many sources of technical noise, including under-sampling of mRNA molecules, often termed "dropout," which can severely obscure important gene-gene relationships. To address this, we developed MAGIC (Markov affinity-based graph imputation of cells), a method that shares information across similar cells, via data diffusion, to denoise the cell count matrix and fill in missing transcripts. We validate MAGIC on several biological systems and find it effective at recovering gene-gene relationships and additional structures. Applied to the epithilial to mesenchymal transition, MAGIC reveals a phenotypic continuum, with the majority of cells residing in intermediate states that display stem-like signatures, and infers known and previously uncharacterized regulatory interactions, demonstrating that our approach can successfully uncover regulatory relations without perturbations.


Subject(s)
Gene Expression Profiling/methods , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Algorithms , Cell Line , Epistasis, Genetic/genetics , Gene Regulatory Networks/genetics , Humans , Markov Chains , MicroRNAs/genetics , RNA, Messenger/genetics , Software
7.
Cell ; 174(5): 1293-1308.e36, 2018 08 23.
Article in English | MEDLINE | ID: mdl-29961579

ABSTRACT

Knowledge of immune cell phenotypes in the tumor microenvironment is essential for understanding mechanisms of cancer progression and immunotherapy response. We profiled 45,000 immune cells from eight breast carcinomas, as well as matched normal breast tissue, blood, and lymph nodes, using single-cell RNA-seq. We developed a preprocessing pipeline, SEQC, and a Bayesian clustering and normalization method, Biscuit, to address computational challenges inherent to single-cell data. Despite significant similarity between normal and tumor tissue-resident immune cells, we observed continuous phenotypic expansions specific to the tumor microenvironment. Analysis of paired single-cell RNA and T cell receptor (TCR) sequencing data from 27,000 additional T cells revealed the combinatorial impact of TCR utilization on phenotypic diversity. Our results support a model of continuous activation in T cells and do not comport with the macrophage polarization model in cancer. Our results have important implications for characterizing tumor-infiltrating immune cells.


Subject(s)
Breast Neoplasms/immunology , Gene Expression Regulation, Neoplastic , Receptors, Antigen, T-Cell/metabolism , Sequence Analysis, RNA , Single-Cell Analysis , Tumor Microenvironment/immunology , Bayes Theorem , Breast Neoplasms/pathology , Cluster Analysis , Computational Biology , Female , Gene Expression Profiling , Humans , Immune System , Immunotherapy/methods , Lymph Nodes , Lymphocytes, Tumor-Infiltrating , Macrophages/metabolism , Phenotype , Transcriptome
8.
Cell ; 170(6): 1120-1133.e17, 2017 Sep 07.
Article in English | MEDLINE | ID: mdl-28803728

ABSTRACT

Immune-checkpoint blockade is able to achieve durable responses in a subset of patients; however, we lack a satisfying comprehension of the underlying mechanisms of anti-CTLA-4- and anti-PD-1-induced tumor rejection. To address these issues, we utilized mass cytometry to comprehensively profile the effects of checkpoint blockade on tumor immune infiltrates in human melanoma and murine tumor models. These analyses reveal a spectrum of tumor-infiltrating T cell populations that are highly similar between tumor models and indicate that checkpoint blockade targets only specific subsets of tumor-infiltrating T cell populations. Anti-PD-1 predominantly induces the expansion of specific tumor-infiltrating exhausted-like CD8 T cell subsets. In contrast, anti-CTLA-4 induces the expansion of an ICOS+ Th1-like CD4 effector population in addition to engaging specific subsets of exhausted-like CD8 T cells. Thus, our findings indicate that anti-CTLA-4 and anti-PD-1 checkpoint-blockade-induced immune responses are driven by distinct cellular mechanisms.


Subject(s)
CTLA-4 Antigen/antagonists & inhibitors , Melanoma/immunology , Melanoma/therapy , Neoplasm Metastasis/immunology , Neoplasm Metastasis/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocyte Subsets/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Female , Flow Cytometry , Gene Expression Regulation , Humans , Immunotherapy , Melanoma/pathology , Mice , Mice, Inbred C57BL , Neoplasm Metastasis/pathology , Single-Cell Analysis , Transcription, Genetic
9.
Cell ; 169(4): 736-749.e18, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28475899

ABSTRACT

Immune cells in the tumor microenvironment modulate cancer progression and are attractive therapeutic targets. Macrophages and T cells are key components of the microenvironment, yet their phenotypes and relationships in this ecosystem and to clinical outcomes are ill defined. We used mass cytometry with extensive antibody panels to perform in-depth immune profiling of samples from 73 clear cell renal cell carcinoma (ccRCC) patients and five healthy controls. In 3.5 million measured cells, we identified 17 tumor-associated macrophage phenotypes, 22 T cell phenotypes, and a distinct immune composition correlated with progression-free survival, thereby presenting an in-depth human atlas of the immune tumor microenvironment in this disease. This study revealed potential biomarkers and targets for immunotherapy development and validated tools that can be used for immune profiling of other tumor types.


Subject(s)
Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Tumor Microenvironment , Humans , Image Cytometry , Immune Tolerance , Kidney/cytology , Macrophages/immunology , Macrophages/pathology , Single-Cell Analysis , T-Lymphocytes/immunology , T-Lymphocytes/pathology
10.
Cell ; 169(4): 750-765.e17, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28475900

ABSTRACT

To guide the design of immunotherapy strategies for patients with early stage lung tumors, we developed a multiscale immune profiling strategy to map the immune landscape of early lung adenocarcinoma lesions to search for tumor-driven immune changes. Utilizing a barcoding method that allows a simultaneous single-cell analysis of the tumor, non-involved lung, and blood cells, we provide a detailed immune cell atlas of early lung tumors. We show that stage I lung adenocarcinoma lesions already harbor significantly altered T cell and NK cell compartments. Moreover, we identified changes in tumor-infiltrating myeloid cell (TIM) subsets that likely compromise anti-tumor T cell immunity. Paired single-cell analyses thus offer valuable knowledge of tumor-driven immune changes, providing a powerful tool for the rational design of immune therapies. VIDEO ABSTRACT.


Subject(s)
Adenocarcinoma/immunology , Adenocarcinoma/pathology , Immunity, Innate , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Single-Cell Analysis/methods , Adenocarcinoma of Lung , Dendritic Cells/pathology , Humans , Killer Cells, Natural/pathology , Macrophages/pathology , T-Lymphocytes/pathology , Tumor Microenvironment
11.
Cell ; 164(1-2): 293-309, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26771497

ABSTRACT

Large-scale genomic studies have identified multiple somatic aberrations in breast cancer, including copy number alterations and point mutations. Still, identifying causal variants and emergent vulnerabilities that arise as a consequence of genetic alterations remain major challenges. We performed whole-genome small hairpin RNA (shRNA) "dropout screens" on 77 breast cancer cell lines. Using a hierarchical linear regression algorithm to score our screen results and integrate them with accompanying detailed genetic and proteomic information, we identify vulnerabilities in breast cancer, including candidate "drivers," and reveal general functional genomic properties of cancer cells. Comparisons of gene essentiality with drug sensitivity data suggest potential resistance mechanisms, effects of existing anti-cancer drugs, and opportunities for combination therapy. Finally, we demonstrate the utility of this large dataset by identifying BRD4 as a potential target in luminal breast cancer and PIK3CA mutations as a resistance determinant for BET-inhibitors.


Subject(s)
Algorithms , Breast Neoplasms/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Cycle Proteins , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases , Cluster Analysis , Drug Resistance, Neoplasm , Gene Dosage , Gene Expression Profiling , Genome-Wide Association Study , Humans , Linear Models , Nuclear Proteins/genetics , Phosphatidylinositol 3-Kinases , Transcription Factors/genetics
12.
Cell ; 162(1): 184-97, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-26095251

ABSTRACT

Acute myeloid leukemia (AML) manifests as phenotypically and functionally diverse cells, often within the same patient. Intratumor phenotypic and functional heterogeneity have been linked primarily by physical sorting experiments, which assume that functionally distinct subpopulations can be prospectively isolated by surface phenotypes. This assumption has proven problematic, and we therefore developed a data-driven approach. Using mass cytometry, we profiled surface and intracellular signaling proteins simultaneously in millions of healthy and leukemic cells. We developed PhenoGraph, which algorithmically defines phenotypes in high-dimensional single-cell data. PhenoGraph revealed that the surface phenotypes of leukemic blasts do not necessarily reflect their intracellular state. Using hematopoietic progenitors, we defined a signaling-based measure of cellular phenotype, which led to isolation of a gene expression signature that was predictive of survival in independent cohorts. This study presents new methods for large-scale analysis of single-cell heterogeneity and demonstrates their utility, yielding insights into AML pathophysiology.


Subject(s)
Computational Biology/methods , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/physiopathology , Single-Cell Analysis/methods , Bone Marrow/pathology , Child , Cohort Studies , Genetic Heterogeneity , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/pathology , Neoplastic Stem Cells/pathology , Transcriptome
13.
Cell ; 157(3): 714-25, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24766814

ABSTRACT

Tissue regeneration is an orchestrated progression of cells from an immature state to a mature one, conventionally represented as distinctive cell subsets. A continuum of transitional cell states exists between these discrete stages. We combine the depth of single-cell mass cytometry and an algorithm developed to leverage this continuum by aligning single cells of a given lineage onto a unified trajectory that accurately predicts the developmental path de novo. Applied to human B cell lymphopoiesis, the algorithm (termed Wanderlust) constructed trajectories spanning from hematopoietic stem cells through to naive B cells. This trajectory revealed nascent fractions of B cell progenitors and aligned them with developmentally cued regulatory signaling including IL-7/STAT5 and cellular events such as immunoglobulin rearrangement, highlighting checkpoints across which regulatory signals are rewired paralleling changes in cellular state. This study provides a comprehensive analysis of human B lymphopoiesis, laying a foundation to apply this approach to other tissues and "corrupted" developmental processes including cancer.


Subject(s)
Algorithms , B-Lymphocytes/cytology , Lymphopoiesis , Humans , Interleukin-7/metabolism , Precursor Cells, B-Lymphoid/cytology , STAT5 Transcription Factor/metabolism , V(D)J Recombination
14.
Cell ; 159(6): 1461-75, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25433701

ABSTRACT

Identifying driver genes in cancer remains a crucial bottleneck in therapeutic development and basic understanding of the disease. We developed Helios, an algorithm that integrates genomic data from primary tumors with data from functional RNAi screens to pinpoint driver genes within large recurrently amplified regions of DNA. Applying Helios to breast cancer data identified a set of candidate drivers highly enriched with known drivers (p < 10(-14)). Nine of ten top-scoring Helios genes are known drivers of breast cancer, and in vitro validation of 12 candidates predicted by Helios found ten conferred enhanced anchorage-independent growth, demonstrating Helios's exquisite sensitivity and specificity. We extensively characterized RSF-1, a driver identified by Helios whose amplification correlates with poor prognosis, and found increased tumorigenesis and metastasis in mouse models. We have demonstrated a powerful approach for identifying driver genes and how it can yield important insights into cancer.


Subject(s)
Algorithms , Breast Neoplasms/genetics , Animals , Bayes Theorem , Breast Neoplasms/pathology , Cell Line, Tumor , DNA Copy Number Variations , Female , Genome-Wide Association Study , Humans , Mice, Inbred NOD , Mice, SCID , RNA Interference
15.
Nature ; 616(7958): 806-813, 2023 04.
Article in English | MEDLINE | ID: mdl-36991128

ABSTRACT

Metastasis frequently develops from disseminated cancer cells that remain dormant after the apparently successful treatment of a primary tumour. These cells fluctuate between an immune-evasive quiescent state and a proliferative state liable to immune-mediated elimination1-6. Little is known about the clearing of reawakened metastatic cells and how this process could be therapeutically activated to eliminate residual disease in patients. Here we use models of indolent lung adenocarcinoma metastasis to identify cancer cell-intrinsic determinants of immune reactivity during exit from dormancy. Genetic screens of tumour-intrinsic immune regulators identified the stimulator of interferon genes (STING) pathway as a suppressor of metastatic outbreak. STING activity increases in metastatic progenitors that re-enter the cell cycle and is dampened by hypermethylation of the STING promoter and enhancer in breakthrough metastases or by chromatin repression in cells re-entering dormancy in response to TGFß. STING expression in cancer cells derived from spontaneous metastases suppresses their outgrowth. Systemic treatment of mice with STING agonists eliminates dormant metastasis and prevents spontaneous outbreaks in a T cell- and natural killer cell-dependent manner-these effects require cancer cell STING function. Thus, STING provides a checkpoint against the progression of dormant metastasis and a therapeutically actionable strategy for the prevention of disease relapse.


Subject(s)
Lung Neoplasms , Neoplasm Metastasis , Animals , Mice , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/immunology , Adenocarcinoma of Lung/pathology , Cell Cycle , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/genetics , Neoplasm Metastasis/immunology , Neoplasm Metastasis/pathology , Neoplasm Recurrence, Local/drug therapy , T-Lymphocytes/immunology , Transforming Growth Factor beta , Killer Cells, Natural/immunology
16.
Mol Cell ; 81(11): 2477-2493.e10, 2021 06 03.
Article in English | MEDLINE | ID: mdl-33891860

ABSTRACT

CD8 T cells play an essential role in defense against viral and bacterial infections and in tumor immunity. Deciphering T cell loss of functionality is complicated by the conspicuous heterogeneity of CD8 T cell states described across experimental and clinical settings. By carrying out a unified analysis of over 300 assay for transposase-accessible chromatin sequencing (ATAC-seq) and RNA sequencing (RNA-seq) experiments from 12 studies of CD8 T cells in cancer and infection, we defined a shared differentiation trajectory toward dysfunction and its underlying transcriptional drivers and revealed a universal early bifurcation of functional and dysfunctional T cell states across models. Experimental dissection of acute and chronic viral infection using single-cell ATAC (scATAC)-seq and allele-specific single-cell RNA (scRNA)-seq identified state-specific drivers and captured the emergence of similar TCF1+ progenitor-like populations at an early branch point, at which functional and dysfunctional T cells diverge. Our atlas of CD8 T cell states will facilitate mechanistic studies of T cell immunity and translational efforts.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epigenesis, Genetic/immunology , Immunity, Cellular , Lymphocytic Choriomeningitis/genetics , Neoplasms/genetics , Transcription Factors/genetics , Acute Disease , Atlases as Topic , CD8-Positive T-Lymphocytes/classification , CD8-Positive T-Lymphocytes/pathology , Chromatin/chemistry , Chromatin/immunology , Chronic Disease , Gene Expression Profiling , Gene Regulatory Networks , High-Throughput Nucleotide Sequencing/methods , Humans , Lymphocyte Activation , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/pathology , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/pathogenicity , Neoplasms/immunology , Neoplasms/pathology , Principal Component Analysis , Single-Cell Analysis , Transcription Factors/classification , Transcription Factors/immunology , Transcription, Genetic , Transposases/genetics , Transposases/metabolism
17.
Immunity ; 50(5): 1202-1217.e7, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31027997

ABSTRACT

Stable changes in chromatin states and gene expression in cells of the immune system form the basis for memory of infections and other challenges. Here, we used naturally occurring cis-regulatory variation in wild-derived inbred mouse strains to explore the mechanisms underlying long-lasting versus transient gene regulation in CD8 T cells responding to acute viral infection. Stably responsive DNA elements were characterized by dramatic and congruent chromatin remodeling events affecting multiple neighboring sites and required distinct transcription factor (TF) binding motifs for their accessibility. Specifically, we found that cooperative recruitment of T-box and Runx family transcription factors to shared targets mediated stable chromatin remodeling upon T cell activation. Our observations provide insights into the molecular mechanisms driving virus-specific CD8 T cell responses and suggest a general mechanism for the formation of transcriptional and epigenetic memory applicable to other immune and non-immune cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Chromatin Assembly and Disassembly/genetics , Core Binding Factor Alpha 2 Subunit/genetics , Gene Expression Regulation/genetics , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , T-Box Domain Proteins/genetics , Animals , CD8-Positive T-Lymphocytes/virology , Cell Line , Chromatin/genetics , Epigenesis, Genetic/genetics , Female , Gene Expression/genetics , Gene Expression Regulation/immunology , Genetic Variation , Immunologic Memory/genetics , Immunologic Memory/immunology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphocytic Choriomeningitis/virology , Male , Mice , Mice, Inbred C57BL , Transcription, Genetic/genetics
18.
Immunity ; 50(4): 1084-1098.e10, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30926234

ABSTRACT

Co-stimulation regulates T cell activation, but it remains unclear whether co-stimulatory pathways also control T cell differentiation. We used mass cytometry to profile T cells generated in the genetic absence of the negative co-stimulatory molecules CTLA-4 and PD-1. Our data indicate that negative co-stimulation constrains the possible cell states that peripheral T cells can acquire. CTLA-4 imposes major boundaries on CD4+ T cell phenotypes, whereas PD-1 subtly limits CD8+ T cell phenotypes. By computationally reconstructing T cell differentiation paths, we identified protein expression changes that underlied the abnormal phenotypic expansion and pinpointed when lineage choice events occurred during differentiation. Similar alterations in T cell phenotypes were observed after anti-CTLA-4 and anti-PD-1 antibody blockade. These findings implicate negative co-stimulation as a key regulator and determinant of T cell differentiation and suggest that checkpoint blockade might work in part by altering the limits of T cell phenotypes.


Subject(s)
CTLA-4 Antigen/immunology , Lymphocyte Activation , Lymphopoiesis , Programmed Cell Death 1 Receptor/immunology , T-Lymphocyte Subsets/cytology , Animals , CD4-Positive T-Lymphocytes/classification , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/deficiency , CTLA-4 Antigen/genetics , Cell Lineage , Immunophenotyping , Lymph Nodes/cytology , Mice, Knockout , Thymus Gland/cytology
19.
Nat Methods ; 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38871986

ABSTRACT

Single-cell RNA sequencing allows us to model cellular state dynamics and fate decisions using expression similarity or RNA velocity to reconstruct state-change trajectories; however, trajectory inference does not incorporate valuable time point information or utilize additional modalities, whereas methods that address these different data views cannot be combined or do not scale. Here we present CellRank 2, a versatile and scalable framework to study cellular fate using multiview single-cell data of up to millions of cells in a unified fashion. CellRank 2 consistently recovers terminal states and fate probabilities across data modalities in human hematopoiesis and endodermal development. Our framework also allows combining transitions within and across experimental time points, a feature we use to recover genes promoting medullary thymic epithelial cell formation during pharyngeal endoderm development. Moreover, we enable estimating cell-specific transcription and degradation rates from metabolic-labeling data, which we apply to an intestinal organoid system to delineate differentiation trajectories and pinpoint regulatory strategies.

20.
Immunity ; 48(5): 1029-1045.e5, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29768164

ABSTRACT

Exhausted CD8 T (Tex) cells are immunotherapy targets in chronic infection and cancer, but a comprehensive assessment of Tex cell diversity in human disease is lacking. Here, we developed a transcriptomic- and epigenetic-guided mass cytometry approach to define core exhaustion-specific genes and disease-induced changes in Tex cells in HIV and human cancer. Single-cell proteomic profiling identified 9 distinct Tex cell clusters using phenotypic, functional, transcription factor, and inhibitory receptor co-expression patterns. An exhaustion severity metric was developed and integrated with high-dimensional phenotypes to define Tex cell clusters that were present in healthy subjects, common across chronic infection and cancer or enriched in either disease, linked to disease severity, and changed with HIV therapy. Combinatorial patterns of immunotherapy targets on different Tex cell clusters were also defined. This approach and associated datasets present a resource for investigating human Tex cell biology, with implications for immune monitoring and immunomodulation in chronic infections, autoimmunity, and cancer.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epigenomics/methods , Flow Cytometry/methods , Gene Expression Profiling/methods , HIV Infections/immunology , Lung Neoplasms/immunology , CD8-Positive T-Lymphocytes/metabolism , HIV Infections/genetics , HIV Infections/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Proteomics/methods , Transcription Factors/genetics , Transcription Factors/immunology , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL