Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Bioinformatics ; 39(12)2023 12 01.
Article in English | MEDLINE | ID: mdl-38113416

ABSTRACT

SUMMARY: Single-cell RNA sequencing (scRNA-seq) has revolutionized the study of gene expression at the individual cell level, unraveling unprecedented insights into cellular heterogeneity. However, the analysis of scRNA-seq data remains a challenging and time-consuming task, often demanding advanced computational expertise, rendering it impractical for high-volume environments and applications. We present CellBridge, an automated workflow designed to simplify the standard procedures entailed in scRNA-seq data analysis, eliminating the need for specialized computational expertise. CellBridge utilizes state-of-the-art computational methods, integrating a range of advanced functionalities, covering the entire process from raw unaligned sequencing reads to cell type annotation. Hence, CellBridge accelerates the pace of discovery by seamlessly enabling insights into vast volumes of scRNA-seq data, without compromising workflow control and reproducibility. AVAILABILITY AND IMPLEMENTATION: The source code, detailed documentation, and materials required to reproduce the results are available on GitHub and archived in Zenodo. For the CellBridge pre-processing step (v1.0.0), access the GitHub repository at https://github.com/Sanofi-Public/PMCB-ToBridge and the Zenodo archive at https://zenodo.org/records/10246161. For the CellBridge processing step (v1.0.0), visit the GitHub repository at https://github.com/Sanofi-Public/PMCB-CellBridge and the Zenodo archive at https://zenodo.org/records/10246046.


Subject(s)
Gene Expression Profiling , Single-Cell Gene Expression Analysis , Gene Expression Profiling/methods , Sequence Analysis, RNA/methods , Workflow , Reproducibility of Results , Single-Cell Analysis , Software
2.
MethodsX ; 10: 102196, 2023.
Article in English | MEDLINE | ID: mdl-37424758

ABSTRACT

Single-cell RNA sequencing (scRNA-seq) experiments provide opportunities to peer into complex tissues at single-cell resolution. However, insightful biological interpretation of scRNA-seq data relies upon precise identification of cell types. The ability to identify the origin of a cell quickly and accurately will greatly improve downstream analyses. We present Sargent, a transformation-free, cluster-free, single-cell annotation algorithm for rapidly identifying the cell types of origin based on cell type-specific markers. We demonstrate Sargent's high accuracy by annotating simulated datasets. Further, we compare Sargent performance against expert-annotated scRNA-seq data from human organs including PBMC, heart, kidney, and lung. We demonstrate that Sargent retains both the flexibility and biological interpretability of cluster-based manual annotation. Additionally, the automation eliminates the labor intensive and potentially biased user annotation, producing robust, reproducible, and scalable outputs.•Sargent is a transformation-free, cluster-free, single-cell annotation algorithm for rapidly identifying the cell types of origin based on cell type-specific markers.•Sargent retains both the flexibility and biological interpretability of cluster-based manual annotation.•Automation eliminates the labor intensive and potentially biased user annotation, producing robust, reproducible, and scalable outputs.

3.
Am J Respir Cell Mol Biol ; 69(4): 404-421, 2023 10.
Article in English | MEDLINE | ID: mdl-37369139

ABSTRACT

Systemic sclerosis (SSc) is a progressive, multiorgan disease with limited treatment options. Although a recent proof-of-concept study using romilkimab or SAR156597, a bispecific IL-4/IL-13 antibody, suggests a direct role of these cytokines in the pathophysiology of SSc, their contributions to the balance between inflammation and fibrosis are unclear. Here, we determine the roles of type 2 inflammation in fibrogenesis using FRA2-Tg (Fos-related antigen 2-overexpressing transgenic) mice, which develop spontaneous, age-dependent progressive lung fibrosis. We defined the molecular signatures of inflammation and fibrosis at three key stages in disease progression, corresponding to preonset, inflammatory dominant, and fibrosis dominant biology, and revealed an early increase in cytokine-cytokine receptor interactions and antigen-processing and presentation pathways followed by enhanced Th2- and M2 macrophage-driven type 2 responses. This type 2 inflammation progressed to extensive fibrotic pathology by 14-18 weeks of age, with these gene signatures overlapping significantly with those seen in the lungs of patients with SSc with interstitial lung disease (ILD). These changes were also evident in the histopathology, which showed perivascular and peribronchiolar inflammation with prominent eosinophilia and accumulation of profibrotic M2-like macrophages followed by rapid progression to fibrosis with thickened alveolar walls with multifocal fibrotic bands and signs of interstitial pneumonia. Critically, treatment with a bispecific antibody targeting IL-4 and IL-13 during the inflammatory phase abrogated the Th2 and M2 responses and led to near-complete abrogation of lung fibrosis. These data recapitulate important features of fibrotic progression in the lungs of patients with SSc-ILD and enhance our understanding of the progressive pathobiology of SSc. This study also further establishes FRA2-Tg mice as a valuable tool for testing future therapeutic agents in SSc-ILD.


Subject(s)
Lung Diseases, Interstitial , Pulmonary Fibrosis , Scleroderma, Systemic , Humans , Mice , Animals , Pulmonary Fibrosis/pathology , Interleukin-13 , Interleukin-4 , Lung Diseases, Interstitial/pathology , Fibrosis , Lung/pathology , Inflammation/pathology , Mice, Transgenic , Cytokines
4.
NAR Genom Bioinform ; 5(1): lqad030, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36968431

ABSTRACT

Most cell-cell interactions and crosstalks are mediated by ligand-receptor interactions. The advent of single-cell RNA-sequencing (scRNA-seq) techniques has enabled characterizing tissue heterogeneity at single-cell level. In the past few years, several methods have been developed to study ligand-receptor interactions at cell type level using scRNA-seq data. However, there is still no easy way to query the activity of a specific user-defined signaling pathway in a targeted way or to map the interactions of the same subunit with different ligands as part of different receptor complexes. Here, we present DiSiR, a fast and easy-to-use permutation-based software framework to investigate how individual cells are interacting with each other by analyzing signaling pathways of multi-subunit ligand-activated receptors from scRNA-seq data, not only for available curated databases of ligand-receptor interactions, but also for interactions that are not listed in these databases. We show that, when utilized to infer ligand-receptor interactions from both simulated and real datasets, DiSiR outperforms other well-known permutation-based methods, e.g. CellPhoneDB and ICELLNET. Finally, to demonstrate DiSiR's utility in exploring data and generating biologically relevant hypotheses, we apply it to COVID lung and rheumatoid arthritis (RA) synovium scRNA-seq datasets and highlight potential differences between inflammatory pathways at cell type level for control versus disease samples.

5.
Sci Transl Med ; 15(681): eabq4419, 2023 02.
Article in English | MEDLINE | ID: mdl-36724239

ABSTRACT

Rheumatoid arthritis (RA) is one of the most common autoimmune diseases affecting primarily the joints. Despite successful therapies including antibodies against tumor necrosis factor (TNF) and interleukin-6 (IL-6) receptor, only 20 to 30% of patients experience remission. We studied whether inhibiting both TNF and IL-6 would result in improved efficacy. Using backtranslation from single-cell RNA sequencing (scRNA-seq) data from individuals with RA, we hypothesized that TNF and IL-6 act synergistically on fibroblast-like synoviocytes (FLS) and T cells. Coculture of FLS from individuals with RA and T cells supported this hypothesis, revealing effects on both disease-driving pathways and biomarkers. Combining anti-TNF and anti-IL-6 antibodies in collagen-induced arthritis (CIA) mouse models resulted in sustained long-term remission, improved histology, and effects on bone remodeling pathways. These promising data initiated the development of an anti-TNF/IL-6 bispecific nanobody compound 1, with similar potencies against TNF and IL-6. We observed additive efficacy of compound 1 in a FLS/T cell coculture affecting arthritis and T helper 17 (TH17) pathways. This nanobody compound transcript signature inversely overlapped with described RA endotypes, indicating a potential efficacy in a broader patient population. In summary, we showed superiority of a bispecific anti-TNF/IL-6 nanobody compound or combination treatment over monospecific treatments in both in vitro and in vivo models. We anticipate improved efficacy in upcoming clinical studies.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Synoviocytes , Animals , Humans , Mice , Arthritis, Experimental/drug therapy , Cells, Cultured , Fibroblasts/pathology , Synovial Membrane/pathology , Synoviocytes/metabolism , Synoviocytes/pathology , Tumor Necrosis Factor Inhibitors/metabolism , Tumor Necrosis Factor Inhibitors/pharmacology , Tumor Necrosis Factor Inhibitors/therapeutic use , Tumor Necrosis Factor-alpha/metabolism , Interleukin-6/immunology
6.
Nat Neurosci ; 26(1): 12-26, 2023 01.
Article in English | MEDLINE | ID: mdl-36536241

ABSTRACT

Iron dysregulation has been implicated in multiple neurodegenerative diseases, including Parkinson's disease (PD). Iron-loaded microglia are frequently found in affected brain regions, but how iron accumulation influences microglia physiology and contributes to neurodegeneration is poorly understood. Here we show that human induced pluripotent stem cell-derived microglia grown in a tri-culture system are highly responsive to iron and susceptible to ferroptosis, an iron-dependent form of cell death. Furthermore, iron overload causes a marked shift in the microglial transcriptional state that overlaps with a transcriptomic signature found in PD postmortem brain microglia. Our data also show that this microglial response contributes to neurodegeneration, as removal of microglia from the tri-culture system substantially delayed iron-induced neurotoxicity. To elucidate the mechanisms regulating iron response in microglia, we performed a genome-wide CRISPR screen and identified novel regulators of ferroptosis, including the vesicle trafficking gene SEC24B. These data suggest a critical role for microglia iron overload and ferroptosis in neurodegeneration.


Subject(s)
Ferroptosis , Induced Pluripotent Stem Cells , Iron Overload , Parkinson Disease , Humans , Induced Pluripotent Stem Cells/metabolism , Iron/metabolism , Iron Overload/metabolism , Microglia/metabolism , Parkinson Disease/genetics
7.
Cancer Immunol Res ; 10(1): 40-55, 2022 01.
Article in English | MEDLINE | ID: mdl-34795032

ABSTRACT

Macrophages often abound within tumors, express colony-stimulating factor 1 receptor (CSF1R), and are linked to adverse patient survival. Drugs blocking CSF1R signaling have been used to suppress tumor-promoting macrophage responses; however, their mechanisms of action remain incompletely understood. Here, we assessed the lung tumor immune microenvironment in mice treated with BLZ945, a prototypical small-molecule CSF1R inhibitor, using single-cell RNA sequencing and mechanistic validation approaches. We showed that tumor control was not caused by CSF1R+ cell depletion; instead, CSF1R targeting reshaped the CSF1R+ cell landscape, which unlocked cross-talk between antitumoral CSF1R- cells. These cells included IFNγ-producing natural killer and T cells, and an IL12-producing dendritic cell subset, denoted as DC3, which were all necessary for CSF1R inhibitor-mediated lung tumor control. These data indicate that CSF1R targeting can activate a cardinal cross-talk between cells that are not macrophages and that are essential to mediate the effects of T cell-targeted immunotherapies and promote antitumor immunity.See related Spotlight by Burrello and de Visser, p. 4.


Subject(s)
Dendritic Cells/immunology , Immunotherapy/methods , Interferon-gamma/metabolism , Interleukin-12/metabolism , Lung Neoplasms/therapy , Animals , Benzothiazoles/pharmacology , Cell Line, Tumor , Female , Lung Neoplasms/immunology , Mice , Mice, Inbred C57BL , Picolinic Acids/pharmacology , Tumor Microenvironment/drug effects , Tumor-Associated Macrophages/drug effects , Tumor-Associated Macrophages/metabolism , Xenograft Model Antitumor Assays
8.
Commun Biol ; 4(1): 370, 2021 04 14.
Article in English | MEDLINE | ID: mdl-33854168

ABSTRACT

Lung cancer is the leading cause of cancer deaths. Tumor heterogeneity, which hampers development of targeted therapies, was herein deconvoluted via single cell RNA sequencing in aggressive human adenocarcinomas (carrying Kras-mutations) and comparable murine model. We identified a tumor-specific, mutant-KRAS-associated subpopulation which is conserved in both human and murine lung cancer. We previously reported a key role for the oncogene BMI-1 in adenocarcinomas. We therefore investigated the effects of in vivo PTC596 treatment, which affects BMI-1 activity, in our murine model. Post-treatment, MRI analysis showed decreased tumor size, while single cell transcriptomics concomitantly detected near complete ablation of the mutant-KRAS-associated subpopulation, signifying the presence of a pharmacologically targetable, tumor-associated subpopulation. Our findings therefore hold promise for the development of a targeted therapy for KRAS-mutant adenocarcinomas.


Subject(s)
Benzimidazoles/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Epithelial Cells/drug effects , Lung Neoplasms/drug therapy , Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Pyrazines/pharmacology , A549 Cells , Animals , Antineoplastic Agents , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Molecular Targeted Therapy , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA-Seq , Single-Cell Analysis , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
9.
Sci Rep ; 10(1): 6321, 2020 Apr 14.
Article in English | MEDLINE | ID: mdl-32286331

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

10.
Sci Rep ; 10(1): 2219, 2020 02 10.
Article in English | MEDLINE | ID: mdl-32042039

ABSTRACT

Establishing clinically relevant single-cell (SC) transcriptomic workflows from cryopreserved tissue is essential to move this emerging immune monitoring technology from the bench to the bedside. Improper sample preparation leads to detrimental cascades, resulting in loss of precious time, money and finally compromised data. There is an urgent need to establish protocols specifically designed to overcome the inevitable variations in sample quality resulting from uncontrollable factors in a clinical setting. Here, we explore sample preparation techniques relevant to a range of clinically relevant scenarios, where SC gene expression and repertoire analysis are applied to a cryopreserved sample derived from a small amount of blood, with unknown or partially known preservation history. We compare a total of ten cell-counting, viability-improvement, and lymphocyte-enrichment methods to highlight a number of unexpected findings. Trypan blue-based automated counters, typically recommended for single-cell sample quantitation, consistently overestimate viability. Advanced sample clean-up procedures significantly impact total cell yield, while only modestly increasing viability. Finally, while pre-enrichment of B cells from whole peripheral blood mononuclear cells (PBMCs) results in the most reliable BCR repertoire data, comparable T-cell enrichment strategies distort the ratio of CD4+ and CD8+ cells. Furthermore, we provide high-resolution analysis of gene expression and clonotype repertoire of different B cell subtypes. Together these observations provide both qualitative and quantitative sample preparation guidelines that increase the chances of obtaining high-quality single-cell transcriptomic and repertoire data from human PBMCs in a variety of clinical settings.


Subject(s)
Gene Expression Profiling/methods , Leukocytes, Mononuclear/metabolism , Single-Cell Analysis/methods , Workflow , Cryopreservation , Humans , Leukocyte Count/methods , Transcriptome
11.
Immunity ; 50(5): 1317-1334.e10, 2019 05 21.
Article in English | MEDLINE | ID: mdl-30979687

ABSTRACT

Tumor-infiltrating myeloid cells (TIMs) comprise monocytes, macrophages, dendritic cells, and neutrophils, and have emerged as key regulators of cancer growth. These cells can diversify into a spectrum of states, which might promote or limit tumor outgrowth but remain poorly understood. Here, we used single-cell RNA sequencing (scRNA-seq) to map TIMs in non-small-cell lung cancer patients. We uncovered 25 TIM states, most of which were reproducibly found across patients. To facilitate translational research of these populations, we also profiled TIMs in mice. In comparing TIMs across species, we identified a near-complete congruence of population structures among dendritic cells and monocytes; conserved neutrophil subsets; and species differences among macrophages. By contrast, myeloid cell population structures in patients' blood showed limited overlap with those of TIMs. This study determines the lung TIM landscape and sets the stage for future investigations into the potential of TIMs as immunotherapy targets.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Dendritic Cells/immunology , Lung Neoplasms/immunology , Macrophages/immunology , Monocytes/immunology , Neutrophils/immunology , Animals , Base Sequence , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Gene Expression Profiling , Humans , Lung/immunology , Lung/pathology , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Sequence Analysis, RNA
13.
Nature ; 560(7718): 377-381, 2018 08.
Article in English | MEDLINE | ID: mdl-30069046

ABSTRACT

The functions of epithelial tissues are dictated by the types, abundance and distribution of the differentiated cells they contain. Attempts to restore tissue function after damage require knowledge of how physiological tasks are distributed among cell types, and how cell states vary between homeostasis, injury-repair and disease. In the conducting airway, a heterogeneous basal cell population gives rise to specialized luminal cells that perform mucociliary clearance1. Here we perform single-cell profiling of human bronchial epithelial cells and mouse tracheal epithelial cells to obtain a comprehensive census of cell types in the conducting airway and their behaviour in homeostasis and regeneration. Our analysis reveals cell states that represent known and novel cell populations, delineates their heterogeneity and identifies distinct differentiation trajectories during homeostasis and tissue repair. Finally, we identified a novel, rare cell type that we call the 'pulmonary ionocyte', which co-expresses FOXI1, multiple subunits of the vacuolar-type H+-ATPase (V-ATPase) and CFTR, the gene that is mutated in cystic fibrosis. Using immunofluorescence, modulation of signalling pathways and electrophysiology, we show that Notch signalling is necessary and FOXI1 expression is sufficient to drive the production of the pulmonary ionocyte, and that the pulmonary ionocyte is a major source of CFTR activity in the conducting airway epithelium.


Subject(s)
Bronchi/cytology , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Profiling , Single-Cell Analysis , Trachea/cytology , Adolescent , Adult , Animals , Cell Differentiation/genetics , Cells, Cultured , Child , Child, Preschool , Cystic Fibrosis/genetics , Female , Fluorescent Antibody Technique , Forkhead Transcription Factors/metabolism , Homeostasis/genetics , Humans , Male , Mice , Organ Specificity , Receptors, Notch/metabolism , Regeneration/genetics , Sequence Analysis, RNA , Signal Transduction/genetics , Vacuolar Proton-Translocating ATPases/metabolism , Young Adult
14.
Science ; 358(6367)2017 12 01.
Article in English | MEDLINE | ID: mdl-29191879

ABSTRACT

Bone marrow-derived myeloid cells can accumulate within tumors and foster cancer outgrowth. Local immune-neoplastic interactions have been intensively investigated, but the contribution of the systemic host environment to tumor growth remains poorly understood. Here, we show in mice and cancer patients (n = 70) that lung adenocarcinomas increase bone stromal activity in the absence of bone metastasis. Animal studies reveal that the cancer-induced bone phenotype involves bone-resident osteocalcin-expressing (Ocn+) osteoblastic cells. These cells promote cancer by remotely supplying a distinct subset of tumor-infiltrating SiglecFhigh neutrophils, which exhibit cancer-promoting properties. Experimentally reducing Ocn+ cell numbers suppresses the neutrophil response and lung tumor outgrowth. These observations posit osteoblasts as remote regulators of lung cancer and identify SiglecFhigh neutrophils as myeloid cell effectors of the osteoblast-driven protumoral response.


Subject(s)
Adenocarcinoma/pathology , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Bone and Bones/pathology , Lectins/metabolism , Lung Neoplasms/pathology , Neutrophil Infiltration , Neutrophils/metabolism , Neutrophils/pathology , Osteoblasts/pathology , Adenocarcinoma of Lung , Animals , Bone Density , Bone Marrow Cells/pathology , Bone and Bones/metabolism , Cell Line, Tumor , Humans , Mice , Mice, Inbred C57BL , Myeloid Cells/pathology , Neoplasms, Experimental/pathology , Osteocalcin/metabolism , Receptor for Advanced Glycation End Products/metabolism
15.
Dev Biol ; 424(2): 181-188, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28283406

ABSTRACT

We characterize the genetic diversity of Xenopus laevis strains using RNA-seq data and allele-specific analysis. This data provides a catalogue of coding variation, which can be used for improving the genomic sequence, as well as for better sequence alignment, probe design, and proteomic analysis. In addition, we paint a broad picture of the genetic landscape of the species by functionally annotating different classes of mutations with a well-established prediction tool (PolyPhen-2). Further, we specifically compare the variation in the progeny of four crosses: inbred genomic (J)-strain, outbred albino (B)-strain, and two hybrid crosses of J and B strains. We identify a subset of mutations specific to the B strain, which allows us to investigate the selection pressures affecting duplicated genes in this allotetraploid. From these crosses we find the ratio of non-synonymous to synonymous mutations is lower in duplicated genes, which suggests that they are under greater purifying selection. Surprisingly, we also find that function-altering ("damaging") mutations constitute a greater fraction of the non-synonymous variants in this group, which suggests a role for subfunctionalization in coding variation affecting duplicated genes.


Subject(s)
Genetic Variation , Open Reading Frames/genetics , Transcriptome/genetics , Xenopus laevis/genetics , Animals , Base Sequence , Crosses, Genetic , Gene Duplication , Genome , Hybridization, Genetic , Inbreeding , Mass Spectrometry , Mutation, Missense/genetics , Polymorphism, Single Nucleotide/genetics , Reproducibility of Results , Sequence Analysis, RNA , Xenopus Proteins/chemistry , Xenopus Proteins/genetics , Xenopus Proteins/metabolism
16.
Nat Protoc ; 12(1): 44-73, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27929523

ABSTRACT

Single-cell RNA sequencing has recently emerged as a powerful tool for mapping cellular heterogeneity in diseased and healthy tissues, yet high-throughput methods are needed for capturing the unbiased diversity of cells. Droplet microfluidics is among the most promising candidates for capturing and processing thousands of individual cells for whole-transcriptome or genomic analysis in a massively parallel manner with minimal reagent use. We recently established a method called inDrops, which has the capability to index >15,000 cells in an hour. A suspension of cells is first encapsulated into nanoliter droplets with hydrogel beads (HBs) bearing barcoding DNA primers. Cells are then lysed and mRNA is barcoded (indexed) by a reverse transcription (RT) reaction. Here we provide details for (i) establishing an inDrops platform (1 d); (ii) performing hydrogel bead synthesis (4 d); (iii) encapsulating and barcoding cells (1 d); and (iv) RNA-seq library preparation (2 d). inDrops is a robust and scalable platform, and it is unique in its ability to capture and profile >75% of cells in even very small samples, on a scale of thousands or tens of thousands of cells.


Subject(s)
High-Throughput Nucleotide Sequencing/instrumentation , Lab-On-A-Chip Devices , Sequence Analysis, RNA/instrumentation , Single-Cell Analysis/instrumentation , Equipment Design
17.
Nat Genet ; 49(1): 10-16, 2017 01.
Article in English | MEDLINE | ID: mdl-27869828

ABSTRACT

There is a striking and unexplained male predominance across many cancer types. A subset of X-chromosome genes can escape X-inactivation, which would protect females from complete functional loss by a single mutation. To identify putative 'escape from X-inactivation tumor-suppressor' (EXITS) genes, we examined somatic alterations from >4,100 cancers across 21 tumor types for sex bias. Six of 783 non-pseudoautosomal region (PAR) X-chromosome genes (ATRX, CNKSR2, DDX3X, KDM5C, KDM6A, and MAGEC3) harbored loss-of-function mutations more frequently in males (based on a false discovery rate < 0.1), in comparison to zero of 18,055 autosomal and PAR genes (Fisher's exact P < 0.0001). Male-biased mutations in genes that escape X-inactivation were observed in combined analysis across many cancers and in several individual tumor types, suggesting a generalized phenomenon. We conclude that biallelic expression of EXITS genes in females explains a portion of the reduced cancer incidence in females as compared to males across a variety of tumor types.


Subject(s)
Chromosomes, Human, X/genetics , Genes, Tumor Suppressor , Genes, X-Linked/genetics , Mutation/genetics , Neoplasms/genetics , Sexism/statistics & numerical data , X Chromosome Inactivation/genetics , Female , Humans , Male
18.
Nat Genet ; 48(3): 231-237, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26808112

ABSTRACT

An unexpectedly large number of human autosomal genes are subject to monoallelic expression (MAE). Our analysis of 4,227 such genes uncovers surprisingly high genetic variation across human populations. This increased diversity is unlikely to reflect relaxed purifying selection. Remarkably, MAE genes exhibit an elevated recombination rate and an increased density of hypermutable sequence contexts. However, these factors do not fully account for the increased diversity. We find that the elevated nucleotide diversity of MAE genes is also associated with greater allelic age: variants in these genes tend to be older and are enriched in polymorphisms shared by Neanderthals and chimpanzees. Both synonymous and nonsynonymous alleles of MAE genes have elevated average population frequencies. We also observed strong enrichment of the MAE signature among genes reported to evolve under balancing selection. We propose that an important biological function of widespread MAE might be the generation of cell-to-cell heterogeneity; the increased genetic variation contributes to this heterogeneity.


Subject(s)
Gene Expression Regulation , Genetic Variation , Alleles , Animals , Genetics, Population , Humans , Neanderthals/genetics , Pan troglodytes/genetics
19.
Nucleic Acids Res ; 44(D1): D753-6, 2016 Jan 04.
Article in English | MEDLINE | ID: mdl-26503248

ABSTRACT

Recently, data on 'random' autosomal monoallelic expression has become available for the entire genome in multiple human and mouse tissues and cell types, creating a need for better access and dissemination. The database of autosomal monoallelic expression (dbMAE; https://mae.hms.harvard.edu) incorporates data from multiple recent reports of genome-wide analyses. These include transcriptome-wide analyses of allelic imbalance in clonal cell populations based on sequence polymorphisms, as well as indirect identification, based on a specific chromatin signature present in MAE gene bodies. Currently, dbMAE contains transcriptome-wide chromatin identification calls for 8 human and 21 mouse tissues, and describes over 16 000 murine and ∼ 700 human cases of directly measured biased expression, compiled from allele-specific RNA-seq and genotyping array data. All data are manually curated. To ensure cross-publication uniformity, we performed re-analysis of transcriptome-wide RNA-seq data using the same pipeline. Data are accessed through an interface that allows for basic and advanced searches; all source references, including raw data, are clearly described and hyperlinked. This ensures the utility of the resource as an initial screening tool for those interested in investigating the role of monoallelic expression in their specific genes and tissues of interest.


Subject(s)
Alleles , Databases, Genetic , Gene Expression Profiling , Animals , Chromatin/metabolism , Humans , Mice , Transcriptome
20.
Cell Rep ; 11(10): 1549-63, 2015 Jun 16.
Article in English | MEDLINE | ID: mdl-26051943

ABSTRACT

Basal-like and luminal breast tumors have distinct clinical behavior and molecular profiles, yet the underlying mechanisms are poorly defined. To interrogate processes that determine these distinct phenotypes and their inheritance pattern, we generated somatic cell fusions and performed integrated genetic and epigenetic (DNA methylation and chromatin) profiling. We found that the basal-like trait is generally dominant and is largely defined by epigenetic repression of luminal transcription factors. Definition of super-enhancers highlighted a core program common in luminal cells but a high degree of heterogeneity in basal-like breast cancers that correlates with clinical outcome. We also found that protein extracts of basal-like cells are sufficient to induce a luminal-to-basal phenotypic switch, implying a trigger of basal-like autoregulatory circuits. We determined that KDM6A might be required for luminal-basal fusions, and we identified EN1, TBX18, and TCF4 as candidate transcriptional regulators of the luminal-to-basal switch. Our findings highlight the remarkable epigenetic plasticity of breast cancer cells.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Cell Fusion/methods , Cell Line, Tumor , Epigenomics , Female , Gene Expression , Gene Expression Profiling , Hepatocyte Nuclear Factor 3-alpha/genetics , Humans , MCF-7 Cells , Promoter Regions, Genetic , Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...