Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Cell ; 185(15): 2840-2840.e1, 2022 07 21.
Article in English | MEDLINE | ID: mdl-35868280

ABSTRACT

Spatially resolved transcriptomics methodologies using RNA sequencing principles have and will continue to contribute to decode the molecular landscape of tissues. Linking quantitative sequencing data with tissue morphology empowers profiling of cellular morphology and transcription over time and space in health and disease. To view this SnapShot, open or download the PDF.


Subject(s)
Transcriptome , Animals , Humans , Sequence Analysis, RNA , Spatial Analysis
2.
Nature ; 608(7922): 360-367, 2022 08.
Article in English | MEDLINE | ID: mdl-35948708

ABSTRACT

Defining the transition from benign to malignant tissue is fundamental to improving early diagnosis of cancer1. Here we use a systematic approach to study spatial genome integrity in situ and describe previously unidentified clonal relationships. We used spatially resolved transcriptomics2 to infer spatial copy number variations in >120,000 regions across multiple organs, in benign and malignant tissues. We demonstrate that genome-wide copy number variation reveals distinct clonal patterns within tumours and in nearby benign tissue using an organ-wide approach focused on the prostate. Our results suggest a model for how genomic instability arises in histologically benign tissue that may represent early events in cancer evolution. We highlight the power of capturing the molecular and spatial continuums in a tissue context and challenge the rationale for treatment paradigms, including focal therapy.


Subject(s)
Clone Cells , DNA Copy Number Variations , Genomic Instability , Neoplasms , Spatial Analysis , Clone Cells/metabolism , Clone Cells/pathology , DNA Copy Number Variations/genetics , Early Detection of Cancer , Genome, Human , Genomic Instability/genetics , Genomics , Humans , Male , Models, Biological , Neoplasms/genetics , Neoplasms/pathology , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Transcriptome/genetics
3.
Nat Methods ; 21(4): 673-679, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38438615

ABSTRACT

Spatial landmarks are crucial in describing histological features between samples or sites, tracking regions of interest in microscopy, and registering tissue samples within a common coordinate framework. Although other studies have explored unsupervised landmark detection, existing methods are not well-suited for histological image data as they often require a large number of images to converge, are unable to handle nonlinear deformations between tissue sections and are ineffective for z-stack alignment, other modalities beyond image data or multimodal data. We address these challenges by introducing effortless landmark detection, a new unsupervised landmark detection and registration method using neural-network-guided thin-plate splines. Our proposed method is evaluated on a diverse range of datasets including histology and spatially resolved transcriptomics, demonstrating superior performance in both accuracy and stability compared to existing approaches.


Subject(s)
Deep Learning , Image Processing, Computer-Assisted/methods
4.
Nat Methods ; 16(10): 987-990, 2019 10.
Article in English | MEDLINE | ID: mdl-31501547

ABSTRACT

Spatial and molecular characteristics determine tissue function, yet high-resolution methods to capture both concurrently are lacking. Here, we developed high-definition spatial transcriptomics, which captures RNA from histological tissue sections on a dense, spatially barcoded bead array. Each experiment recovers several hundred thousand transcript-coupled spatial barcodes at 2-µm resolution, as demonstrated in mouse brain and primary breast cancer. This opens the way to high-resolution spatial analysis of cells and tissues.


Subject(s)
Gene Expression Profiling , Transcriptome , Animals , Breast Neoplasms/pathology , Female , Humans , Mice , Olfactory Bulb/cytology , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Tissue Array Analysis
5.
BMC Bioinformatics ; 21(1): 161, 2020 Apr 29.
Article in English | MEDLINE | ID: mdl-32349652

ABSTRACT

BACKGROUND: Technological developments in the emerging field of spatial transcriptomics have opened up an unexplored landscape where transcript information is put in a spatial context. Clustering commonly constitutes a central component in analyzing this type of data. However, deciding on the number of clusters to use and interpreting their relationships can be difficult. RESULTS: We introduce SpatialCPie, an R package designed to facilitate cluster evaluation for spatial transcriptomics data. SpatialCPie clusters the data at multiple resolutions. The results are visualized with pie charts that indicate the similarity between spatial regions and clusters and a cluster graph that shows the relationships between clusters at different resolutions. We demonstrate SpatialCPie on several publicly available datasets. CONCLUSIONS: SpatialCPie provides intuitive visualizations of cluster relationships when dealing with Spatial Transcriptomics data.


Subject(s)
Software , Transcriptome/genetics , Cluster Analysis , Gene Expression Regulation, Developmental , Heart/embryology , Humans
6.
BMC Genomics ; 21(1): 298, 2020 Apr 15.
Article in English | MEDLINE | ID: mdl-32293264

ABSTRACT

BACKGROUND: Interest in studying the spatial distribution of gene expression in tissues is rapidly increasing. Spatial Transcriptomics is a novel sequencing-based technology that generates high-throughput information on the distribution, heterogeneity and co-expression of cells in tissues. Unfortunately, manual preparation of high-quality sequencing libraries is time-consuming and subject to technical variability due to human error during manual pipetting, which results in sample swapping and the accidental introduction of batch effects. All these factors complicate the production and interpretation of biological datasets. RESULTS: We have integrated an Agilent Bravo Automated Liquid Handling Platform into the Spatial Transcriptomics workflow. Compared to the previously reported Magnatrix 8000+ automated protocol, this approach increases the number of samples processed per run, reduces sample preparation time by 35%, and minimizes batch effects between samples. The new approach is also shown to be highly accurate and almost completely free from technical variability between prepared samples. CONCLUSIONS: The new automated Spatial Transcriptomics protocol using the Agilent Bravo Automated Liquid Handling Platform rapidly generates high-quality Spatial Transcriptomics libraries. Given the wide use of the Agilent Bravo Automated Liquid Handling Platform in research laboratories and facilities, this will allow many researchers to quickly create robust Spatial Transcriptomics libraries.


Subject(s)
Gene Expression Regulation/genetics , High-Throughput Nucleotide Sequencing/methods , Transcriptome , Animals , Automation , Computational Biology , Gene Library , High-Throughput Nucleotide Sequencing/instrumentation , Mice , Mice, Inbred C57BL , Olfactory Bulb/metabolism , Robotics
7.
Bioinformatics ; 34(11): 1966-1968, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29360929

ABSTRACT

Motiviation: Spatial Transcriptomics (ST) is a method which combines high resolution tissue imaging with high troughput transcriptome sequencing data. This data must be aligned with the images for correct visualization, a process that involves several manual steps. Results: Here we present ST Spot Detector, a web tool that automates and facilitates this alignment through a user friendly interface. Contact: jose.fernandez.navarro@scilifelab.se. Supplementary information: Supplementary data are available at Bioinformatics online.


Subject(s)
Gene Expression Profiling/methods , Image Interpretation, Computer-Assisted/methods , Software , Animals , Humans , Internet , Plants , Sequence Analysis, RNA/methods , Spatial Analysis
8.
Nat Commun ; 13(1): 5475, 2022 09 17.
Article in English | MEDLINE | ID: mdl-36115838

ABSTRACT

The molecular mechanisms underlying lethal castration-resistant prostate cancer remain poorly understood, with intratumoral heterogeneity a likely contributing factor. To examine the temporal aspects of resistance, we analyze tumor heterogeneity in needle biopsies collected before and after treatment with androgen deprivation therapy. By doing so, we are able to couple clinical responsiveness and morphological information such as Gleason score to transcriptome-wide data. Our data-driven analysis of transcriptomes identifies several distinct intratumoral cell populations, characterized by their unique gene expression profiles. Certain cell populations present before treatment exhibit gene expression profiles that match those of resistant tumor cell clusters, present after treatment. We confirm that these clusters are resistant by the localization of active androgen receptors to the nuclei in cancer cells post-treatment. Our data also demonstrates that most stromal cells adjacent to resistant clusters do not express the androgen receptor, and we identify differentially expressed genes for these cells. Altogether, this study shows the potential to increase the power in predicting resistant tumors.


Subject(s)
Prostatic Neoplasms , Receptors, Androgen , Androgen Antagonists/pharmacology , Androgen Antagonists/therapeutic use , Androgens/metabolism , Clone Cells/metabolism , Humans , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Spatio-Temporal Analysis
9.
Nat Biotechnol ; 40(4): 476-479, 2022 04.
Article in English | MEDLINE | ID: mdl-34845373

ABSTRACT

Current methods for spatial transcriptomics are limited by low spatial resolution. Here we introduce a method that integrates spatial gene expression data with histological image data from the same tissue section to infer higher-resolution expression maps. Using a deep generative model, our method characterizes the transcriptome of micrometer-scale anatomical features and can predict spatial gene expression from histology images alone.


Subject(s)
Transcriptome , Transcriptome/genetics
10.
Nat Med ; 27(3): 546-559, 2021 03.
Article in English | MEDLINE | ID: mdl-33654293

ABSTRACT

Angiotensin-converting enzyme 2 (ACE2) and accessory proteases (TMPRSS2 and CTSL) are needed for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) cellular entry, and their expression may shed light on viral tropism and impact across the body. We assessed the cell-type-specific expression of ACE2, TMPRSS2 and CTSL across 107 single-cell RNA-sequencing studies from different tissues. ACE2, TMPRSS2 and CTSL are coexpressed in specific subsets of respiratory epithelial cells in the nasal passages, airways and alveoli, and in cells from other organs associated with coronavirus disease 2019 (COVID-19) transmission or pathology. We performed a meta-analysis of 31 lung single-cell RNA-sequencing studies with 1,320,896 cells from 377 nasal, airway and lung parenchyma samples from 228 individuals. This revealed cell-type-specific associations of age, sex and smoking with expression levels of ACE2, TMPRSS2 and CTSL. Expression of entry factors increased with age and in males, including in airway secretory cells and alveolar type 2 cells. Expression programs shared by ACE2+TMPRSS2+ cells in nasal, lung and gut tissues included genes that may mediate viral entry, key immune functions and epithelial-macrophage cross-talk, such as genes involved in the interleukin-6, interleukin-1, tumor necrosis factor and complement pathways. Cell-type-specific expression patterns may contribute to the pathogenesis of COVID-19, and our work highlights putative molecular pathways for therapeutic intervention.


Subject(s)
COVID-19/epidemiology , COVID-19/genetics , Host-Pathogen Interactions/genetics , SARS-CoV-2/physiology , Sequence Analysis, RNA/statistics & numerical data , Single-Cell Analysis/statistics & numerical data , Virus Internalization , Adult , Aged , Aged, 80 and over , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/virology , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism , COVID-19/pathology , COVID-19/virology , Cathepsin L/genetics , Cathepsin L/metabolism , Datasets as Topic/statistics & numerical data , Demography , Female , Gene Expression Profiling/statistics & numerical data , Humans , Lung/metabolism , Lung/virology , Male , Middle Aged , Organ Specificity/genetics , Respiratory System/metabolism , Respiratory System/virology , Sequence Analysis, RNA/methods , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Single-Cell Analysis/methods
11.
Commun Biol ; 3(1): 565, 2020 10 09.
Article in English | MEDLINE | ID: mdl-33037292

ABSTRACT

The field of spatial transcriptomics is rapidly expanding, and with it the repertoire of available technologies. However, several of the transcriptome-wide spatial assays do not operate on a single cell level, but rather produce data comprised of contributions from a - potentially heterogeneous - mixture of cells. Still, these techniques are attractive to use when examining complex tissue specimens with diverse cell populations, where complete expression profiles are required to properly capture their richness. Motivated by an interest to put gene expression into context and delineate the spatial arrangement of cell types within a tissue, we here present a model-based probabilistic method that uses single cell data to deconvolve the cell mixtures in spatial data. To illustrate the capacity of our method, we use data from different experimental platforms and spatially map cell types from the mouse brain and developmental heart, which arrange as expected.


Subject(s)
Computational Biology , Gene Expression Profiling , Single-Cell Analysis , Transcriptome , Animals , Computational Biology/methods , Computational Biology/standards , Gene Expression Profiling/methods , Humans , Mice , Organ Specificity , Organogenesis/genetics , Single-Cell Analysis/methods , Single-Cell Analysis/standards
12.
Nat Biomed Eng ; 4(8): 827-834, 2020 08.
Article in English | MEDLINE | ID: mdl-32572199

ABSTRACT

Spatial transcriptomics allows for the measurement of RNA abundance at a high spatial resolution, making it possible to systematically link the morphology of cellular neighbourhoods and spatially localized gene expression. Here, we report the development of a deep learning algorithm for the prediction of local gene expression from haematoxylin-and-eosin-stained histopathology images using a new dataset of 30,612 spatially resolved gene expression data matched to histopathology images from 23 patients with breast cancer. We identified over 100 genes, including known breast cancer biomarkers of intratumoral heterogeneity and the co-localization of tumour growth and immune activation, the expression of which can be predicted from the histopathology images at a resolution of 100 µm. We also show that the algorithm generalizes well to The Cancer Genome Atlas and to other breast cancer gene expression datasets without the need for re-training. Predicting the spatially resolved transcriptome of a tissue directly from tissue images may enable image-based screening for molecular biomarkers with spatial variation.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Deep Learning , Algorithms , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Female , Gene Expression Profiling/methods , Humans , Image Processing, Computer-Assisted , Reproducibility of Results , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL