13 research outputs found

    Identification of the stress granule transcriptome via RNA-editing in single cells and in vivo

    Get PDF
    Stress granules are phase-separated assemblies formed around RNAs. So far, the techniques available to identify these RNAs are not suitable for single cells and small tissues displaying cell heterogeneity. Here, we used TRIBE (target of RNA-binding proteins identified by editing) to profile stress granule RNAs. We used an RNA-binding protein (FMR1) fused to the catalytic domain of an RNA-editing enzyme (ADAR), which coalesces into stress granules upon oxidative stress. RNAs colocalized with this fusion are edited, producing mutations that are detectable by VASA sequencing. Using single-molecule FISH, we validated that this purification-free method can reliably identify stress granule RNAs in bulk and single S2 cells and in Drosophila neurons. Similar to mammalian cells, we find that stress granule mRNAs encode ATP binding, cell cycle, and transcription factors. This method opens the possibility to identify stress granule RNAs and other RNA-based assemblies in other single cells and tissues

    Spatially resolved and single cell transcriptomics

    No full text
    In recent years, massive parallel sequencing has revolutionized the field of biology and has provided us with a vast number of new discoveries in fields such as neurology, developmental biology and cancer research. A significant area is deciphering gene expression patterns, as well as other aspects of transcriptome information, such as the impact of splice variants and mutations on biological functions and disease development. By applying RNA-sequencing, one can extract this type of information in a large-scale manner. The most recent approaches include high-resolution techniques such as single cell sequencing and in situ methods in order to circumvent the problems with gene expression averaging in homogenized samples, and loss of spatial information. The research in this thesis is focused on the development of a novel genome-wide spatial transcriptomics method. The technique is used for analysis of intact tissue sections as well as single cells from solution, with the aim to combine gene expression and morphological information. In Paper I, the method is described in detail, and it is shown that the method is able to generate spatial high quality data from mouse olfactory bulb tissue sections (a part of the forebrain) as well as from tissue sections from breast cancer samples. In Paper III, we adapt the library preparation method in order to be able to execute it on a robotic workstation, thus increasing the reproducibility and the throughput, and decreasing the hands-on time. In Paper IV, we generate 3D-data from breast cancer samples by serial sectioning. We show that the gene expression can be highly variable along all three axes of a tumor, and we track pathways with specific spatial activity, as well as perform subtype classification with three-dimensional resolution. In Paper II, we present a high-throughput method for single cell transcriptomics of cells in solution. The method is based on the same type of solid surface capture as the tissue protocol described in Papers I, III and IV. Again, we show that we can generate high-quality gene expression data, and connect this to morphological characteristics of the analyzed single cells; both using cultured cells and samples from patients with leukemia.QC 20170125</p

    Gene expression profiling of periodontitis-affected gingival tissue by spatial transcriptomics

    No full text
    Periodontitis is a highly prevalent chronic inflammatory disease of the periodontium, leading ultimately to tooth loss. In order to characterize the gene expression of periodontitis-affected gingival tissue, we have here simultaneously quantified and localized gene expression in periodontal tissue using spatial transcriptomics, combining RNA sequencing with histological analysis. Our analyses revealed distinct clusters of gene expression, which were identified to correspond to epithelium, inflamed areas of connective tissue, and non-inflamed areas of connective tissue. Moreover, 92 genes were identified as significantly up-regulated in inflamed areas of the gingival connective tissue compared to non-inflamed tissue. Among these, immunoglobulin lambda-like polypeptide 5 (IGLL5), signal sequence receptor subunit 4 (SSR4), marginal zone B and B1 cell specific protein (MZB1), and X-box binding protein 1 (XBP1) were the four most highly up-regulated genes. These genes were also verified as significantly higher expressed in gingival tissue of patients with periodontitis compared to healthy controls, using reverse transcription quantitative polymerase chain reaction. Moreover, the protein expressions of up-regulated genes were verified in gingival biopsies by immunohistochemistry. In summary, in this study, we report distinct gene expression signatures within periodontitis-affected gingival tissue, as well as specific genes that are up-regulated in inflamed areas compared to non-inflamed areas of gingival tissue. The results obtained from this study may add novel information on the genes and cell types contributing to pathogenesis of the chronic inflammatory disease periodontitis

    An automated approach to prepare tissue-derived spatially barcoded RNA-sequencing libraries.

    No full text
    Sequencing the nucleic acid content of individual cells or specific biological samples is becoming increasingly common. This drives the need for robust, scalable and automated library preparation protocols. Furthermore, an increased understanding of tissue heterogeneity has lead to the development of several unique sequencing protocols that aim to retain or infer spatial context. In this study, a protocol for retaining spatial information of transcripts has been adapted to run on a robotic workstation. The method spatial transcriptomics is evaluated in terms of robustness and variability through the preparation of reference RNA, as well as through preparation and sequencing of six replicate sections of a gingival tissue biopsy from a patient with periodontitis. The results are reduced technical variability between replicates and a higher throughput, processing four times more samples with less than a third of the hands on time, compared to the standard protocol.QC 20161124</p

    Spatial Deconvolution of HER2-positive Breast Tumors Reveals Novel Intercellular Relationships

    No full text
    In the past decades, transcriptomic studies have revolutionized cancer treatment and diagnosis. However, tumor sequencing strategies typically result in loss of spatial information, critical to understand cell interactions and their functional relevance. To address this, we investigate spatial gene expression in HER2-positive breast tumors using Spatial Transcriptomics technology. We show that expression-based clustering enables data-driven tumor annotation and assessment of intra-and interpatient heterogeneity; from which we discover shared gene signatures for immune and tumor processes. We integrate and spatially map tumor-associated types from single cell data to find: segregated epithelial cells, interactions between B and T-cells and myeloid cells, co-localization of macrophage and T-cell subsets. A model is constructed to infer presence of tertiary lymphoid structures, applicable across tissue types and technical platforms. Taken together, we combine different data modalities to define novel interactions between tumor-infiltrating cells in breast cancer and provide tools generalizing across tissues and diseases

    Spatial deconvolution of HER2-positive breast cancer delineates tumor-associated cell type interactions

    No full text
    In the past decades, transcriptomic studies have revolutionized cancer treatment and diagnosis. However, tumor sequencing strategies typically result in loss of spatial information, critical to understand cell interactions and their functional relevance. To address this, we investigate spatial gene expression in HER2-positive breast tumors using Spatial Transcriptomics technology. We show that expression-based clustering enables data-driven tumor annotation and assessment of intra- and interpatient heterogeneity; from which we discover shared gene signatures for immune and tumor processes. By integration with single cell data, we spatially map tumor-associated cell types to find tertiary lymphoid-like structures, and a type I interferon response overlapping with regions of T-cell and macrophage subset colocalization. We construct a predictive model to infer presence of tertiary lymphoid-like structures, applicable across tissue types and technical platforms. Taken together, we combine different data modalities to define a high resolution map of cellular interactions in tumors and provide tools generalizing across tissues and diseases

    Spatial detection of fetal marker genes expressed at low level in adult human heart tissue

    No full text
    Abstract Heart failure is a major health problem linked to poor quality of life and high mortality rates. Hence, novel biomarkers, such as fetal marker genes with low expression levels, could potentially differentiate disease states in order to improve therapy. In many studies on heart failure, cardiac biopsies have been analyzed as uniform pieces of tissue with bulk techniques, but this homogenization approach can mask medically relevant phenotypes occurring only in isolated parts of the tissue. This study examines such spatial variations within and between regions of cardiac biopsies. In contrast to standard RNA sequencing, this approach provides a spatially resolved transcriptome- and tissue-wide perspective of the adult human heart, and enables detection of fetal marker genes expressed by minor subpopulations of cells within the tissue. Analysis of patients with heart failure, with preserved ejection fraction, demonstrated spatially divergent expression of fetal genes in cardiac biopsies
    corecore