Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 239
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Annu Rev Immunol ; 36: 813-842, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29677477

ABSTRACT

Given the many cell types and molecular components of the human immune system, along with vast variations across individuals, how should we go about developing causal and predictive explanations of immunity? A central strategy in human studies is to leverage natural variation to find relationships among variables, including DNA variants, epigenetic states, immune phenotypes, clinical descriptors, and others. Here, we focus on how natural variation is used to find patterns, infer principles, and develop predictive models for two areas: (a) immune cell activation-how single-cell profiling boosts our ability to discover immune cell types and states-and (b) antigen presentation and recognition-how models can be generated to predict presentation of antigens on MHC molecules and their detection by T cell receptors. These are two examples of a shift in how we find the drivers and targets of immunity, especially in the human system in the context of health and disease.


Subject(s)
Immune System , Immunity , Animals , Antigen Presentation/immunology , Biomarkers , Disease Susceptibility/immunology , Disease Susceptibility/metabolism , Epitopes/immunology , Genomics/methods , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immune System/cytology , Immune System/physiology , Ligands , Major Histocompatibility Complex/genetics , Major Histocompatibility Complex/immunology , Peptides/immunology , Protein Transport , Proteolysis , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
2.
Cell ; 186(7): 1417-1431.e20, 2023 03 30.
Article in English | MEDLINE | ID: mdl-37001502

ABSTRACT

Senescent cell accumulation has been implicated in the pathogenesis of aging-associated diseases, including cancer. The mechanism that prevents the accumulation of senescent cells in aging human organs is unclear. Here, we demonstrate that a virus-immune axis controls the senescent fibroblast accumulation in the human skin. Senescent fibroblasts increased in old skin compared with young skin. However, they did not increase with advancing age in the elderly. Increased CXCL9 and cytotoxic CD4+ T cells (CD4 CTLs) recruitment were significantly associated with reduced senescent fibroblasts in the old skin. Senescent fibroblasts expressed human leukocyte antigen class II (HLA-II) and human cytomegalovirus glycoprotein B (HCMV-gB), becoming direct CD4 CTL targets. Skin-resident CD4 CTLs eliminated HCMV-gB+ senescent fibroblasts in an HLA-II-dependent manner, and HCMV-gB activated CD4 CTLs from the human skin. Collectively, our findings demonstrate HCMV reactivation in senescent cells, which CD4 CTLs can directly eliminate through the recognition of the HCMV-gB antigen.


Subject(s)
Antineoplastic Agents , Cytomegalovirus Infections , Humans , Aged , Cytomegalovirus , T-Lymphocytes, Cytotoxic , HLA Antigens , CD4-Positive T-Lymphocytes , Cellular Senescence
3.
Nat Immunol ; 25(6): 1033-1045, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38745085

ABSTRACT

The etiology and effect of age-related immune dysfunction in cancer is not completely understood. Here we show that limited priming of CD8+ T cells in the aged tumor microenvironment (TME) outweighs cell-intrinsic defects in limiting tumor control. Increased tumor growth in aging is associated with reduced CD8+ T cell infiltration and function. Transfer of T cells from young mice does not restore tumor control in aged mice owing to rapid induction of T cell dysfunction. Cell-extrinsic signals in the aged TME drive a tumor-infiltrating age-associated dysfunctional (TTAD) cell state that is functionally, transcriptionally and epigenetically distinct from canonical T cell exhaustion. Altered natural killer cell-dendritic cell-CD8+ T cell cross-talk in aged tumors impairs T cell priming by conventional type 1 dendritic cells and promotes TTAD cell formation. Aged mice are thereby unable to benefit from therapeutic tumor vaccination. Critically, myeloid-targeted therapy to reinvigorate conventional type 1 dendritic cells can improve tumor control and restore CD8+ T cell immunity in aging.


Subject(s)
Aging , CD8-Positive T-Lymphocytes , Dendritic Cells , Tumor Microenvironment , Animals , Tumor Microenvironment/immunology , CD8-Positive T-Lymphocytes/immunology , Mice , Dendritic Cells/immunology , Aging/immunology , Mice, Inbred C57BL , Killer Cells, Natural/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Humans , Neoplasms/immunology , Cell Line, Tumor , Female , Lymphocyte Activation/immunology
4.
Nat Immunol ; 25(4): 644-658, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38503922

ABSTRACT

The organization of immune cells in human tumors is not well understood. Immunogenic tumors harbor spatially localized multicellular 'immunity hubs' defined by expression of the T cell-attracting chemokines CXCL10/CXCL11 and abundant T cells. Here, we examined immunity hubs in human pre-immunotherapy lung cancer specimens and found an association with beneficial response to PD-1 blockade. Critically, we discovered the stem-immunity hub, a subtype of immunity hub strongly associated with favorable PD-1-blockade outcome. This hub is distinct from mature tertiary lymphoid structures and is enriched for stem-like TCF7+PD-1+CD8+ T cells, activated CCR7+LAMP3+ dendritic cells and CCL19+ fibroblasts as well as chemokines that organize these cells. Within the stem-immunity hub, we find preferential interactions between CXCL10+ macrophages and TCF7-CD8+ T cells as well as between mature regulatory dendritic cells and TCF7+CD4+ and regulatory T cells. These results provide a picture of the spatial organization of the human intratumoral immune response and its relevance to patient immunotherapy outcomes.


Subject(s)
Lung Neoplasms , Humans , CD8-Positive T-Lymphocytes , Programmed Cell Death 1 Receptor , Chemokines/metabolism , Immunotherapy/methods , Tumor Microenvironment
5.
Cell ; 184(15): 3962-3980.e17, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34171305

ABSTRACT

T cell-mediated immunity plays an important role in controlling SARS-CoV-2 infection, but the repertoire of naturally processed and presented viral epitopes on class I human leukocyte antigen (HLA-I) remains uncharacterized. Here, we report the first HLA-I immunopeptidome of SARS-CoV-2 in two cell lines at different times post infection using mass spectrometry. We found HLA-I peptides derived not only from canonical open reading frames (ORFs) but also from internal out-of-frame ORFs in spike and nucleocapsid not captured by current vaccines. Some peptides from out-of-frame ORFs elicited T cell responses in a humanized mouse model and individuals with COVID-19 that exceeded responses to canonical peptides, including some of the strongest epitopes reported to date. Whole-proteome analysis of infected cells revealed that early expressed viral proteins contribute more to HLA-I presentation and immunogenicity. These biological insights, as well as the discovery of out-of-frame ORF epitopes, will facilitate selection of peptides for immune monitoring and vaccine development.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Histocompatibility Antigens Class I/immunology , Open Reading Frames/genetics , Peptides/immunology , Proteome/immunology , SARS-CoV-2/immunology , A549 Cells , Alleles , Amino Acid Sequence , Animals , Antigen Presentation/immunology , COVID-19/immunology , COVID-19/virology , Female , HEK293 Cells , Humans , Kinetics , Male , Mice , Peptides/chemistry , T-Lymphocytes/immunology
6.
Cell ; 184(26): 6262-6280.e26, 2021 12 22.
Article in English | MEDLINE | ID: mdl-34910928

ABSTRACT

Colorectal cancers (CRCs) arise from precursor polyps whose cellular origins, molecular heterogeneity, and immunogenic potential may reveal diagnostic and therapeutic insights when analyzed at high resolution. We present a single-cell transcriptomic and imaging atlas of the two most common human colorectal polyps, conventional adenomas and serrated polyps, and their resulting CRC counterparts. Integrative analysis of 128 datasets from 62 participants reveals adenomas arise from WNT-driven expansion of stem cells, while serrated polyps derive from differentiated cells through gastric metaplasia. Metaplasia-associated damage is coupled to a cytotoxic immune microenvironment preceding hypermutation, driven partly by antigen-presentation differences associated with tumor cell-differentiation status. Microsatellite unstable CRCs contain distinct non-metaplastic regions where tumor cells acquire stem cell properties and cytotoxic immune cells are depleted. Our multi-omic atlas provides insights into malignant progression of colorectal polyps and their microenvironment, serving as a framework for precision surveillance and prevention of CRC.


Subject(s)
Colonic Polyps/pathology , Colorectal Neoplasms/pathology , Tumor Microenvironment , Adaptive Immunity , Adenoma/genetics , Adenoma/pathology , Adult , Aged , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Death , Cell Differentiation , Colonic Polyps/genetics , Colonic Polyps/immunology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Genetic Heterogeneity , Humans , Male , Mice , Middle Aged , Mutation/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , RNA-Seq , Reproducibility of Results , Single-Cell Analysis , Tumor Microenvironment/immunology
7.
Nat Immunol ; 24(8): 1382-1390, 2023 08.
Article in English | MEDLINE | ID: mdl-37500887

ABSTRACT

Microglia, the macrophages of the brain parenchyma, are key players in neurodegenerative diseases such as Alzheimer's disease. These cells adopt distinct transcriptional subtypes known as states. Understanding state function, especially in human microglia, has been elusive owing to a lack of tools to model and manipulate these cells. Here, we developed a platform for modeling human microglia transcriptional states in vitro. We found that exposure of human stem-cell-differentiated microglia to synaptosomes, myelin debris, apoptotic neurons or synthetic amyloid-beta fibrils generated transcriptional diversity that mapped to gene signatures identified in human brain microglia, including disease-associated microglia, a state enriched in neurodegenerative diseases. Using a new lentiviral approach, we demonstrated that the transcription factor MITF drives a disease-associated transcriptional signature and a highly phagocytic state. Together, these tools enable the manipulation and functional interrogation of human microglial states in both homeostatic and disease-relevant contexts.


Subject(s)
Alzheimer Disease , Induced Pluripotent Stem Cells , Neurodegenerative Diseases , Humans , Microglia , Alzheimer Disease/genetics , Brain
8.
Cell ; 182(6): 1474-1489.e23, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32841603

ABSTRACT

Widespread changes to DNA methylation and chromatin are well documented in cancer, but the fate of higher-order chromosomal structure remains obscure. Here we integrated topological maps for colon tumors and normal colons with epigenetic, transcriptional, and imaging data to characterize alterations to chromatin loops, topologically associated domains, and large-scale compartments. We found that spatial partitioning of the open and closed genome compartments is profoundly compromised in tumors. This reorganization is accompanied by compartment-specific hypomethylation and chromatin changes. Additionally, we identify a compartment at the interface between the canonical A and B compartments that is reorganized in tumors. Remarkably, similar shifts were evident in non-malignant cells that have accumulated excess divisions. Our analyses suggest that these topological changes repress stemness and invasion programs while inducing anti-tumor immunity genes and may therefore restrain malignant progression. Our findings call into question the conventional view that tumor-associated epigenomic alterations are primarily oncogenic.


Subject(s)
Chromatin/metabolism , Chromosomes/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic/genetics , Cell Division , Cellular Senescence/genetics , Chromatin Immunoprecipitation Sequencing , Chromosomes/genetics , Cohort Studies , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Computational Biology , DNA Methylation/genetics , Epigenomics , HCT116 Cells , Humans , In Situ Hybridization, Fluorescence , Microscopy, Electron, Transmission , Molecular Dynamics Simulation , RNA-Seq , Spatial Analysis , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
9.
Cell ; 183(3): 818-834.e13, 2020 10 29.
Article in English | MEDLINE | ID: mdl-33038342

ABSTRACT

Many approaches to identify therapeutically relevant neoantigens couple tumor sequencing with bioinformatic algorithms and inferred rules of tumor epitope immunogenicity. However, there are no reference data to compare these approaches, and the parameters governing tumor epitope immunogenicity remain unclear. Here, we assembled a global consortium wherein each participant predicted immunogenic epitopes from shared tumor sequencing data. 608 epitopes were subsequently assessed for T cell binding in patient-matched samples. By integrating peptide features associated with presentation and recognition, we developed a model of tumor epitope immunogenicity that filtered out 98% of non-immunogenic peptides with a precision above 0.70. Pipelines prioritizing model features had superior performance, and pipeline alterations leveraging them improved prediction performance. These findings were validated in an independent cohort of 310 epitopes prioritized from tumor sequencing data and assessed for T cell binding. This data resource enables identification of parameters underlying effective anti-tumor immunity and is available to the research community.


Subject(s)
Antigens, Neoplasm/immunology , Epitopes/immunology , Neoplasms/immunology , Alleles , Antigen Presentation/immunology , Cohort Studies , Humans , Peptides/immunology , Programmed Cell Death 1 Receptor , Reproducibility of Results
10.
Nat Immunol ; 23(10): 1495-1506, 2022 10.
Article in English | MEDLINE | ID: mdl-36151395

ABSTRACT

The immune system can eliminate tumors, but checkpoints enable immune escape. Here, we identify immune evasion mechanisms using genome-scale in vivo CRISPR screens across cancer models treated with immune checkpoint blockade (ICB). We identify immune evasion genes and important immune inhibitory checkpoints conserved across cancers, including the non-classical major histocompatibility complex class I (MHC class I) molecule Qa-1b/HLA-E. Surprisingly, loss of tumor interferon-γ (IFNγ) signaling sensitizes many models to immunity. The immune inhibitory effects of tumor IFN sensing are mediated through two mechanisms. First, tumor upregulation of classical MHC class I inhibits natural killer cells. Second, IFN-induced expression of Qa-1b inhibits CD8+ T cells via the NKG2A/CD94 receptor, which is induced by ICB. Finally, we show that strong IFN signatures are associated with poor response to ICB in individuals with renal cell carcinoma or melanoma. This study reveals that IFN-mediated upregulation of classical and non-classical MHC class I inhibitory checkpoints can facilitate immune escape.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Histocompatibility Antigens Class I/metabolism , Humans , Immune Checkpoint Inhibitors , Immune Evasion , Interferon-gamma/genetics , Interferon-gamma/metabolism , NK Cell Lectin-Like Receptor Subfamily C
11.
Cell ; 176(6): 1325-1339.e22, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30827679

ABSTRACT

Lineage tracing provides key insights into the fate of individual cells in complex organisms. Although effective genetic labeling approaches are available in model systems, in humans, most approaches require detection of nuclear somatic mutations, which have high error rates, limited scale, and do not capture cell state information. Here, we show that somatic mutations in mtDNA can be tracked by single-cell RNA or assay for transposase accessible chromatin (ATAC) sequencing. We leverage somatic mtDNA mutations as natural genetic barcodes and demonstrate their utility as highly accurate clonal markers to infer cellular relationships. We track native human cells both in vitro and in vivo and relate clonal dynamics to gene expression and chromatin accessibility. Our approach should allow clonal tracking at a 1,000-fold greater scale than with nuclear genome sequencing, with simultaneous information on cell state, opening the way to chart cellular dynamics in human health and disease.


Subject(s)
DNA, Mitochondrial/genetics , Mitochondria/genetics , Base Sequence , Cell Lineage , Chromatin , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Genomics/methods , HEK293 Cells , Hematopoietic Stem Cells/physiology , High-Throughput Nucleotide Sequencing/methods , Humans , Mutation , Single-Cell Analysis , Transposases
12.
Nat Immunol ; 22(8): 1030-1041, 2021 08.
Article in English | MEDLINE | ID: mdl-34312544

ABSTRACT

T cell exhaustion is associated with failure to clear chronic infections and malignant cells. Defining the molecular mechanisms of T cell exhaustion and reinvigoration is essential to improving immunotherapeutic modalities. Here we confirmed pervasive phenotypic, functional and transcriptional differences between memory and exhausted antigen-specific CD8+ T cells in human hepatitis C virus (HCV) infection before and after treatment. After viral cure, phenotypic changes in clonally stable exhausted T cell populations suggested differentiation toward a memory-like profile. However, functionally, the cells showed little improvement, and critical transcriptional regulators remained in the exhaustion state. Notably, T cells from chronic HCV infection that were exposed to antigen for less time because of viral escape mutations were functionally and transcriptionally more similar to memory T cells from spontaneously resolved HCV infection. Thus, the duration of T cell stimulation impacts exhaustion recovery, with antigen removal after long-term exhaustion being insufficient for the development of functional T cell memory.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Hepacivirus/immunology , Hepatitis C, Chronic/immunology , Immunologic Memory/immunology , Antiviral Agents/therapeutic use , Cell Differentiation/immunology , Epitopes/genetics , Hepatitis C, Chronic/drug therapy , Humans , Phenotype
13.
Cell ; 175(4): 998-1013.e20, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30388456

ABSTRACT

Treatment of cancer has been revolutionized by immune checkpoint blockade therapies. Despite the high rate of response in advanced melanoma, the majority of patients succumb to disease. To identify factors associated with success or failure of checkpoint therapy, we profiled transcriptomes of 16,291 individual immune cells from 48 tumor samples of melanoma patients treated with checkpoint inhibitors. Two distinct states of CD8+ T cells were defined by clustering and associated with patient tumor regression or progression. A single transcription factor, TCF7, was visualized within CD8+ T cells in fixed tumor samples and predicted positive clinical outcome in an independent cohort of checkpoint-treated patients. We delineated the epigenetic landscape and clonality of these T cell states and demonstrated enhanced antitumor immunity by targeting novel combinations of factors in exhausted cells. Our study of immune cell transcriptomes from tumors demonstrates a strategy for identifying predictors, mechanisms, and targets for enhancing checkpoint immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy/methods , Melanoma/immunology , Transcriptome , Animals , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Antigens, CD/immunology , Antineoplastic Agents, Immunological/immunology , Antineoplastic Agents, Immunological/pharmacology , Apyrase/antagonists & inhibitors , Apyrase/immunology , Cell Line, Tumor , Humans , Leukocyte Common Antigens/antagonists & inhibitors , Leukocyte Common Antigens/immunology , Melanoma/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T Cell Transcription Factor 1/metabolism
14.
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
15.
Cell ; 169(4): 567-569, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28475889

ABSTRACT

A deep understanding of the immune landscape in human cancer is essential for guiding the development of immunotherapy to benefit more patients with long-lasting efficacy. Now, two studies from Lavin et al. and Chevrier et al. employ mass cytometry to study immune infiltrates in lung adenocarcinoma and clear cell renal cell carcinoma, respectively.


Subject(s)
Carcinoma, Renal Cell/immunology , Immunotherapy , Humans , Metals, Heavy
17.
Nat Immunol ; 20(9): 1231-1243, 2019 09.
Article in English | MEDLINE | ID: mdl-31358999

ABSTRACT

Understanding resistance to antibody to programmed cell death protein 1 (PD-1; anti-PD-1) is crucial for the development of reversal strategies. In anti-PD-1-resistant models, simultaneous anti-PD-1 and vaccine therapy reversed resistance, while PD-1 blockade before antigen priming abolished therapeutic outcomes. This was due to induction of dysfunctional PD-1+CD38hi CD8+ cells by PD-1 blockade in suboptimally primed CD8 cell conditions induced by tumors. This results in erroneous T cell receptor signaling and unresponsiveness to antigenic restimulation. On the other hand, PD-1 blockade of optimally primed CD8 cells prevented the induction of dysfunctional CD8 cells, reversing resistance. Depleting PD-1+CD38hi CD8+ cells enhanced therapeutic outcomes. Furthermore, non-responding patients showed more PD-1+CD38+CD8+ cells in tumor and blood than responders. In conclusion, the status of CD8+ T cell priming is a major contributor to anti-PD-1 therapeutic resistance. PD-1 blockade in unprimed or suboptimally primed CD8 cells induces resistance through the induction of PD-1+CD38hi CD8+ cells that is reversed by optimal priming. PD-1+CD38hi CD8+ cells serve as a predictive and therapeutic biomarker for anti-PD-1 treatment. Sequencing of anti-PD-1 and vaccine is crucial for successful therapy.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , CD8-Positive T-Lymphocytes/immunology , Drug Resistance, Neoplasm/immunology , Membrane Glycoproteins/metabolism , Neoplasms/immunology , Programmed Cell Death 1 Receptor/immunology , ADP-ribosyl Cyclase 1/genetics , Animals , Antibodies/immunology , CD8-Positive T-Lymphocytes/pathology , Cancer Vaccines/immunology , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Female , Humans , Immunotherapy/methods , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Tumor Microenvironment/immunology
19.
Nat Immunol ; 20(7): 928-942, 2019 07.
Article in English | MEDLINE | ID: mdl-31061532

ABSTRACT

To define the cell populations that drive joint inflammation in rheumatoid arthritis (RA), we applied single-cell RNA sequencing (scRNA-seq), mass cytometry, bulk RNA sequencing (RNA-seq) and flow cytometry to T cells, B cells, monocytes, and fibroblasts from 51 samples of synovial tissue from patients with RA or osteoarthritis (OA). Utilizing an integrated strategy based on canonical correlation analysis of 5,265 scRNA-seq profiles, we identified 18 unique cell populations. Combining mass cytometry and transcriptomics revealed cell states expanded in RA synovia: THY1(CD90)+HLA-DRAhi sublining fibroblasts, IL1B+ pro-inflammatory monocytes, ITGAX+TBX21+ autoimmune-associated B cells and PDCD1+ peripheral helper T (TPH) cells and follicular helper T (TFH) cells. We defined distinct subsets of CD8+ T cells characterized by GZMK+, GZMB+, and GNLY+ phenotypes. We mapped inflammatory mediators to their source cell populations; for example, we attributed IL6 expression to THY1+HLA-DRAhi fibroblasts and IL1B production to pro-inflammatory monocytes. These populations are potentially key mediators of RA pathogenesis.


Subject(s)
Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/metabolism , Gene Expression Profiling , Synovial Membrane/metabolism , Transcriptome , Arthritis, Rheumatoid/pathology , Autoimmunity/genetics , Biomarkers , Computational Biology/methods , Cross-Sectional Studies , Cytokines/metabolism , Fibroblasts/metabolism , Flow Cytometry , Gene Expression , Gene Expression Profiling/methods , High-Throughput Nucleotide Sequencing , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Humans , Leukocytes/immunology , Leukocytes/metabolism , Monocytes/immunology , Monocytes/metabolism , Signal Transduction , Single-Cell Analysis/methods , Synovial Membrane/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Workflow
20.
Nat Immunol ; 20(7): 902-914, 2019 07.
Article in English | MEDLINE | ID: mdl-31209404

ABSTRACT

Lupus nephritis is a potentially fatal autoimmune disease for which the current treatment is ineffective and often toxic. To develop mechanistic hypotheses of disease, we analyzed kidney samples from patients with lupus nephritis and from healthy control subjects using single-cell RNA sequencing. Our analysis revealed 21 subsets of leukocytes active in disease, including multiple populations of myeloid cells, T cells, natural killer cells and B cells that demonstrated both pro-inflammatory responses and inflammation-resolving responses. We found evidence of local activation of B cells correlated with an age-associated B-cell signature and evidence of progressive stages of monocyte differentiation within the kidney. A clear interferon response was observed in most cells. Two chemokine receptors, CXCR4 and CX3CR1, were broadly expressed, implying a potentially central role in cell trafficking. Gene expression of immune cells in urine and kidney was highly correlated, which would suggest that urine might serve as a surrogate for kidney biopsies.


Subject(s)
Kidney/immunology , Lupus Nephritis/immunology , Biomarkers , Biopsy , Cluster Analysis , Computational Biology/methods , Epithelial Cells/metabolism , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation , Humans , Immunophenotyping , Interferons/metabolism , Kidney/metabolism , Kidney/pathology , Leukocytes/immunology , Leukocytes/metabolism , Lupus Nephritis/genetics , Lupus Nephritis/metabolism , Lupus Nephritis/pathology , Lymphocytes/immunology , Lymphocytes/metabolism , Molecular Sequence Annotation , Myeloid Cells/immunology , Myeloid Cells/metabolism , Single-Cell Analysis , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL