Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 43
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Nat Genet ; 56(8): 1725-1736, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38951642

RÉSUMÉ

Idiopathic pulmonary fibrosis (IPF) is a progressive lung disease with poor prognosis and limited treatment options. Efforts to identify effective treatments are thwarted by limited understanding of IPF pathogenesis and poor translatability of available preclinical models. Here we generated spatially resolved transcriptome maps of human IPF (n = 4) and bleomycin-induced mouse pulmonary fibrosis (n = 6) to address these limitations. We uncovered distinct fibrotic niches in the IPF lung, characterized by aberrant alveolar epithelial cells in a microenvironment dominated by transforming growth factor beta signaling alongside predicted regulators, such as TP53 and APOE. We also identified a clear divergence between the arrested alveolar regeneration in the IPF fibrotic niches and the active tissue repair in the acutely fibrotic mouse lung. Our study offers in-depth insights into the IPF transcriptional landscape and proposes alveolar regeneration as a promising therapeutic strategy for IPF.


Sujet(s)
Bléomycine , Fibrose pulmonaire idiopathique , Transcriptome , Animaux , Humains , Souris , Fibrose pulmonaire idiopathique/génétique , Fibrose pulmonaire idiopathique/anatomopathologie , Bléomycine/toxicité , Modèles animaux de maladie humaine , Facteur de croissance transformant bêta/métabolisme , Facteur de croissance transformant bêta/génétique , Poumon/métabolisme , Poumon/anatomopathologie , Souris de lignée C57BL , Transduction du signal/génétique , Mâle , Analyse de profil d'expression de gènes , Pneumocytes/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Régénération/génétique , Apolipoprotéines E/génétique
3.
BMC Bioinformatics ; 24(1): 461, 2023 Dec 07.
Article de Anglais | MEDLINE | ID: mdl-38062356

RÉSUMÉ

BACKGROUND: Basecalling long DNA sequences is a crucial step in nanopore-based DNA sequencing protocols. In recent years, the CTC-RNN model has become the leading basecalling model, supplanting preceding hidden Markov models (HMMs) that relied on pre-segmenting ion current measurements. However, the CTC-RNN model operates independently of prior biological and physical insights. RESULTS: We present a novel basecaller named Lokatt: explicit duration Markov model and residual-LSTM network. It leverages an explicit duration HMM (EDHMM) designed to model the nanopore sequencing processes. Trained on a newly generated library with methylation-free Ecoli samples and MinION R9.4.1 chemistry, the Lokatt basecaller achieves basecalling performances with a median single read identity score of 0.930, a genome coverage ratio of 99.750%, on par with existing state-of-the-art structure when trained on the same datasets. CONCLUSION: Our research underlines the potential of incorporating prior knowledge into the basecalling processes, particularly through integrating HMMs and recurrent neural networks. The Lokatt basecaller showcases the efficacy of a hybrid approach, emphasizing its capacity to achieve high-quality basecalling performance while accommodating the nuances of nanopore sequencing. These outcomes pave the way for advanced basecalling methodologies, with potential implications for enhancing the accuracy and efficiency of nanopore-based DNA sequencing protocols.


Sujet(s)
Nanopores , ADN/génétique , Analyse de séquence d'ADN/méthodes , , Séquence nucléotidique , Séquençage nucléotidique à haut débit/méthodes
4.
Bioinformatics ; 39(10)2023 10 03.
Article de Anglais | MEDLINE | ID: mdl-37846051

RÉSUMÉ

SUMMARY: Spatially resolved transcriptomics technologies generate gene expression data with retained positional information from a tissue section, often accompanied by a corresponding histological image. Computational tools should make it effortless to incorporate spatial information into data analyses and present analysis results in their histological context. Here, we present semla, an R package for processing, analysis, and visualization of spatially resolved transcriptomics data generated by the Visium platform, that includes interactive web applications for data exploration and tissue annotation. AVAILABILITY AND IMPLEMENTATION: The R package semla is available on GitHub (https://github.com/ludvigla/semla), under the MIT License, and deposited on Zenodo (https://doi.org/10.5281/zenodo.8321645). Documentation and tutorials with detailed descriptions of usage can be found at https://ludvigla.github.io/semla/.


Sujet(s)
Biologie informatique , Transcriptome , Biologie informatique/méthodes , Logiciel , Analyse de profil d'expression de gènes , Documentation
5.
Placenta ; 139: 213-216, 2023 08.
Article de Anglais | MEDLINE | ID: mdl-37481829

RÉSUMÉ

Spatial transcriptomics (ST) maps RNA level patterns within a tissue. This technology has not been previously applied to human placental tissue. We demonstrate analysis of human placental samples with ST. Unsupervised clustering revealed that distinct RNA patterns were found corresponding to different morphological structures. Additionally, when focusing upon terminal villi and hemoglobin associated structures, RNA levels differed between placentas from full term healthy pregnancies and those complicated by preeclampsia. The results from this study can provide a benchmark for future ST studies in placenta.


Sujet(s)
Placenta , Pré-éclampsie , Grossesse , Humains , Femelle , ARN , Transcriptome , Pré-éclampsie/génétique , Analyse de profil d'expression de gènes
6.
Nat Commun ; 14(1): 1438, 2023 03 15.
Article de Anglais | MEDLINE | ID: mdl-36922516

RÉSUMÉ

To date, single-cell studies of human white adipose tissue (WAT) have been based on small cohort sizes and no cellular consensus nomenclature exists. Herein, we performed a comprehensive meta-analysis of publicly available and newly generated single-cell, single-nucleus, and spatial transcriptomic results from human subcutaneous, omental, and perivascular WAT. Our high-resolution map is built on data from ten studies and allowed us to robustly identify >60 subpopulations of adipocytes, fibroblast and adipogenic progenitors, vascular, and immune cells. Using these results, we deconvolved spatial and bulk transcriptomic data from nine additional cohorts to provide spatial and clinical dimensions to the map. This identified cell-cell interactions as well as relationships between specific cell subtypes and insulin resistance, dyslipidemia, adipocyte volume, and lipolysis upon long-term weight changes. Altogether, our meta-map provides a rich resource defining the cellular and microarchitectural landscape of human WAT and describes the associations between specific cell types and metabolic states.


Sujet(s)
Tissu adipeux blanc , Transcriptome , Humains , Transcriptome/génétique , Tissu adipeux blanc/métabolisme , Adipocytes/métabolisme , Analyse de profil d'expression de gènes , Adipogenèse/génétique , Tissu adipeux
7.
Nat Biotechnol ; 41(8): 1085-1088, 2023 08.
Article de Anglais | MEDLINE | ID: mdl-36604544

RÉSUMÉ

Current methods for epigenomic profiling are limited in their ability to obtain genome-wide information with spatial resolution. We introduce spatial ATAC, a method that integrates transposase-accessible chromatin profiling in tissue sections with barcoded solid-phase capture to perform spatially resolved epigenomics. We show that spatial ATAC enables the discovery of the regulatory programs underlying spatial gene expression during mouse organogenesis, lineage differentiation and in human pathology.


Sujet(s)
Chromatine , Transposases , Animaux , Humains , Souris , Chromatine/génétique , Transposases/génétique , Transposases/métabolisme , Épigénomique/méthodes , Séquençage nucléotidique à haut débit/méthodes , Analyse de séquence d'ADN/méthodes
8.
Sci Rep ; 12(1): 11876, 2022 07 13.
Article de Anglais | MEDLINE | ID: mdl-35831338

RÉSUMÉ

B cells play a significant role in established Rheumatoid Arthritis (RA). However, it is unclear to what extent differentiated B cells are present in joint tissue already at the onset of disease. Here, we studied synovial biopsies (n = 8) captured from untreated patients at time of diagnosis. 3414 index-sorted B cells underwent RNA sequencing and paired tissue pieces were subjected to spatial transcriptomics (n = 4). We performed extensive bioinformatics analyses to dissect the local B cell composition. Select plasma cell immunoglobulin sequences were expressed as monoclonal antibodies and tested by ELISA. Memory and plasma cells were found irrespective of autoantibody status of the patients. Double negative memory B cells were prominent, but did not display a distinct transcriptional profile. The tissue architecture implicate both local B cell maturation via T cell help and plasma cell survival niches with a strong CXCL12-CXCR4 axis. The immunoglobulin sequence analyses revealed clonality between the memory B and plasma cell pools further supporting local maturation. One of the plasma cell-derived antibodies displayed citrulline autoreactivity, demonstrating local autoreactive plasma cell differentiation in joint biopsies captured from untreated early RA. Hence, plasma cell niches are not a consequence of chronic inflammation, but are already present at the time of diagnosis.


Sujet(s)
Polyarthrite rhumatoïde , Membrane synoviale , Autoanticorps , Différenciation cellulaire , Humains , Membrane synoviale/anatomopathologie , Transcriptome
9.
Commun Biol ; 5(1): 129, 2022 02 11.
Article de Anglais | MEDLINE | ID: mdl-35149753

RÉSUMÉ

The inflamed rheumatic joint is a highly heterogeneous and complex tissue with dynamic recruitment and expansion of multiple cell types that interact in multifaceted ways within a localized area. Rheumatoid arthritis synovium has primarily been studied either by immunostaining or by molecular profiling after tissue homogenization. Here, we use Spatial Transcriptomics, where tissue-resident RNA is spatially labeled in situ with barcodes in a transcriptome-wide fashion, to study local tissue interactions at the site of chronic synovial inflammation. We report comprehensive spatial RNA-Seq data coupled to cell type-specific localization patterns at and around organized structures of infiltrating leukocyte cells in the synovium. Combining morphological features and high-throughput spatially resolved transcriptomics may be able to provide higher statistical power and more insights into monitoring disease severity and treatment-specific responses in seropositive and seronegative rheumatoid arthritis.


Sujet(s)
Polyarthrite rhumatoïde , Transcriptome , Polyarthrite rhumatoïde/génétique , Polyarthrite rhumatoïde/métabolisme , Humains , Membrane synoviale/métabolisme
11.
Cell Metab ; 33(9): 1869-1882.e6, 2021 09 07.
Article de Anglais | MEDLINE | ID: mdl-34380013

RÉSUMÉ

The contribution of cellular heterogeneity and architecture to white adipose tissue (WAT) function is poorly understood. Herein, we combined spatially resolved transcriptional profiling with single-cell RNA sequencing and image analyses to map human WAT composition and structure. This identified 18 cell classes with unique propensities to form spatially organized homo- and heterotypic clusters. Of these, three constituted mature adipocytes that were similar in size, but distinct in their spatial arrangements and transcriptional profiles. Based on marker genes, we termed these AdipoLEP, AdipoPLIN, and AdipoSAA. We confirmed, in independent datasets, that their respective gene profiles associated differently with both adipocyte and whole-body insulin sensitivity. Corroborating our observations, insulin stimulation in vivo by hyperinsulinemic-euglycemic clamp showed that only AdipoPLIN displayed a transcriptional response to insulin. Altogether, by mining this multimodal resource we identify that human WAT is composed of three classes of mature adipocytes, only one of which is insulin responsive.


Sujet(s)
Insulinorésistance , Insuline , Adipocytes , Tissu adipeux , Tissu adipeux blanc , Humains , Insuline/pharmacologie
13.
Cell Rep ; 35(8): 109174, 2021 05 25.
Article de Anglais | MEDLINE | ID: mdl-34038736

RÉSUMÉ

The CD8+ T cell response to an antigen is composed of many T cell clones with unique T cell receptors, together forming a heterogeneous repertoire of effector and memory cells. How individual T cell clones contribute to this heterogeneity throughout immune responses remains largely unknown. In this study, we longitudinally track human CD8+ T cell clones expanding in response to yellow fever virus (YFV) vaccination at the single-cell level. We observed a drop in clonal diversity in blood from the acute to memory phase, suggesting that clonal selection shapes the circulating memory repertoire. Clones in the memory phase display biased differentiation trajectories along a gradient from stem cell to terminally differentiated effector memory fates. In secondary responses, YFV- and influenza-specific CD8+ T cell clones are poised to recapitulate skewed differentiation trajectories. Collectively, we show that the sum of distinct clonal phenotypes results in the multifaceted human T cell response to acute viral infections.


Sujet(s)
Lymphocytes T CD8+/immunologie , Maladies virales/virologie , Fièvre jaune/virologie , Maladie aigüe , Différenciation cellulaire , Cellules cultivées , Humains
14.
Breast Cancer Res ; 22(1): 6, 2020 01 13.
Article de Anglais | MEDLINE | ID: mdl-31931856

RÉSUMÉ

BACKGROUND: Distinguishing ductal carcinoma in situ (DCIS) from invasive ductal carcinoma (IDC) regions in clinical biopsies constitutes a diagnostic challenge. Spatial transcriptomics (ST) is an in situ capturing method, which allows quantification and visualization of transcriptomes in individual tissue sections. In the past, studies have shown that breast cancer samples can be used to study their transcriptomes with spatial resolution in individual tissue sections. Previously, supervised machine learning methods were used in clinical studies to predict the clinical outcomes for cancer types. METHODS: We used four publicly available ST breast cancer datasets from breast tissue sections annotated by pathologists as non-malignant, DCIS, or IDC. We trained and tested a machine learning method (support vector machine) based on the expert annotation as well as based on automatic selection of cell types by their transcriptome profiles. RESULTS: We identified expression signatures for expert annotated regions (non-malignant, DCIS, and IDC) and build machine learning models. Classification results for 798 expression signature transcripts showed high coincidence with the expert pathologist annotation for DCIS (100%) and IDC (96%). Extending our analysis to include all 25,179 expressed transcripts resulted in an accuracy of 99% for DCIS and 98% for IDC. Further, classification based on an automatically identified expression signature covering all ST spots of tissue sections resulted in prediction accuracy of 95% for DCIS and 91% for IDC. CONCLUSIONS: This concept study suggest that the ST signatures learned from expert selected breast cancer tissue sections can be used to identify breast cancer regions in whole tissue sections including regions not trained on. Furthermore, the identified expression signatures can classify cancer regions in tissue sections not used for training with high accuracy. Expert-generated but even automatically generated cancer signatures from ST data might be able to classify breast cancer regions and provide clinical decision support for pathologists in the future.


Sujet(s)
Marqueurs biologiques tumoraux/génétique , Tumeurs du sein/diagnostic , Carcinome canalaire du sein/diagnostic , Carcinome intracanalaire non infiltrant/diagnostic , Apprentissage machine , Typage moléculaire/méthodes , Transcriptome , Tumeurs du sein/classification , Tumeurs du sein/génétique , Carcinome canalaire du sein/génétique , Carcinome intracanalaire non infiltrant/génétique , Femelle , Humains , Courbe ROC , Analyse spatiale
15.
Sci Rep ; 9(1): 18975, 2019 12 12.
Article de Anglais | MEDLINE | ID: mdl-31831833

RÉSUMÉ

Lately it has become possible to analyze transcriptomic profiles in tissue sections with retained cellular context. We aimed to explore synovial biopsies from rheumatoid arthritis (RA) and spondyloarthritis (SpA) patients, using Spatial Transcriptomics (ST) as a proof of principle approach for unbiased mRNA studies at the site of inflammation in these chronic inflammatory diseases. Synovial tissue biopsies from affected joints were studied with ST. The transcriptome data was subjected to differential gene expression analysis (DEA), pathway analysis, immune cell type identification using Xcell analysis and validation with immunohistochemistry (IHC). The ST technology allows selective analyses on areas of interest, thus we analyzed morphologically distinct areas of mononuclear cell infiltrates. The top differentially expressed genes revealed an adaptive immune response profile and T-B cell interactions in RA, while in SpA, the profiles implicate functions associated with tissue repair. With spatially resolved gene expression data, overlaid on high-resolution histological images, we digitally portrayed pre-selected cell types in silico. The RA displayed an overrepresentation of central memory T cells, while in SpA effector memory T cells were most prominent. Consequently, ST allows for deeper understanding of cellular mechanisms and diversity in tissues from chronic inflammatory diseases.


Sujet(s)
Polyarthrite rhumatoïde/immunologie , Spondylarthrite/immunologie , Membrane synoviale/immunologie , Transcriptome/immunologie , Adulte , Polyarthrite rhumatoïde/anatomopathologie , Biopsie , Femelle , Analyse de profil d'expression de gènes , Humains , Inflammation/immunologie , Inflammation/anatomopathologie , Mâle , Adulte d'âge moyen , Séquençage par oligonucléotides en batterie , Spondylarthrite/anatomopathologie , Membrane synoviale/anatomopathologie
16.
Cell ; 179(7): 1647-1660.e19, 2019 Dec 12.
Article de Anglais | MEDLINE | ID: mdl-31835037

RÉSUMÉ

The process of cardiac morphogenesis in humans is incompletely understood. Its full characterization requires a deep exploration of the organ-wide orchestration of gene expression with a single-cell spatial resolution. Here, we present a molecular approach that reveals the comprehensive transcriptional landscape of cell types populating the embryonic heart at three developmental stages and that maps cell-type-specific gene expression to specific anatomical domains. Spatial transcriptomics identified unique gene profiles that correspond to distinct anatomical regions in each developmental stage. Human embryonic cardiac cell types identified by single-cell RNA sequencing confirmed and enriched the spatial annotation of embryonic cardiac gene expression. In situ sequencing was then used to refine these results and create a spatial subcellular map for the three developmental phases. Finally, we generated a publicly available web resource of the human developing heart to facilitate future studies on human cardiogenesis.


Sujet(s)
Régulation de l'expression des gènes au cours du développement , Coeur/embryologie , Myocytes cardiaques/métabolisme , Analyse sur cellule unique , Transcriptome , Femelle , Humains , Mâle , Morphogenèse , Myocytes cardiaques/cytologie , RNA-Seq
17.
Nat Methods ; 16(10): 987-990, 2019 10.
Article de Anglais | MEDLINE | ID: mdl-31501547

RÉSUMÉ

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.


Sujet(s)
Analyse de profil d'expression de gènes , Transcriptome , Animaux , Tumeurs du sein/anatomopathologie , Femelle , Humains , Souris , Bulbe olfactif/cytologie , Analyse de séquence d'ARN/méthodes , Analyse sur cellule unique/méthodes , Analyse sur puce à tissus
18.
Genome Biol ; 20(1): 68, 2019 04 01.
Article de Anglais | MEDLINE | ID: mdl-30935387

RÉSUMÉ

Accurate variant calling and genotyping represent major limiting factors for downstream applications of single-cell genomics. Here, we report Conbase for the identification of somatic mutations in single-cell DNA sequencing data. Conbase leverages phased read data from multiple samples in a dataset to achieve increased confidence in somatic variant calls and genotype predictions. Comparing the performance of Conbase to three other methods, we find that Conbase performs best in terms of false discovery rate and specificity and provides superior robustness on simulated data, in vitro expanded fibroblasts and clonal lymphocyte populations isolated directly from a healthy human donor.


Sujet(s)
Mutation , Analyse sur cellule unique , Logiciel , Lymphocytes T CD8+ , Fibroblastes , Humains , Polymorphisme de nucléotide simple , Analyse de séquence d'ADN
19.
Bioinformatics ; 35(6): 1058-1060, 2019 03 15.
Article de Anglais | MEDLINE | ID: mdl-30875427

RÉSUMÉ

MOTIVATION: Spatial Transcriptomics (ST) is a technique that combines high-resolution imaging with spatially resolved transcriptome-wide sequencing. This novel type of data opens up many possibilities for analysis and visualization, most of which are either not available with standard tools or too complex for normal users. RESULTS: Here, we present a tool, ST Viewer, which allows real-time interaction, analysis and visualization of Spatial Transcriptomics datasets through a seamless and smooth user interface. AVAILABILITY AND IMPLEMENTATION: The ST Viewer is open source under a MIT license and it is available at https://github.com/SpatialTranscriptomicsResearch/st_viewer. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Sujet(s)
Biologie informatique , Logiciel , Transcriptome
20.
Nat Protoc ; 13(11): 2501-2534, 2018 11.
Article de Anglais | MEDLINE | ID: mdl-30353172

RÉSUMÉ

Spatial resolution of gene expression enables gene expression events to be pinpointed to a specific location in biological tissue. Spatially resolved gene expression in tissue sections is traditionally analyzed using immunohistochemistry (IHC) or in situ hybridization (ISH). These technologies are invaluable tools for pathologists and molecular biologists; however, their throughput is limited to the analysis of only a few genes at a time. Recent advances in RNA sequencing (RNA-seq) have made it possible to obtain unbiased high-throughput gene expression data in bulk. Spatial Transcriptomics combines the benefits of traditional spatially resolved technologies with the massive throughput of RNA-seq. Here, we present a protocol describing how to apply the Spatial Transcriptomics technology to mammalian tissue. This protocol combines histological staining and spatially resolved RNA-seq data from intact tissue sections. Once suitable tissue-specific conditions have been established, library construction and sequencing can be completed in ~5-6 d. Data processing takes a few hours, with the exact timing dependent on the sequencing depth. Our method requires no special instruments and can be performed in any laboratory with access to a cryostat, microscope and next-generation sequencing.


Sujet(s)
Codage à barres de l'ADN pour la taxonomie/méthodes , Bulbe olfactif/métabolisme , ARN/génétique , Analyse sur puce à tissus/méthodes , Transcriptome , Animaux , Codage à barres de l'ADN pour la taxonomie/instrumentation , Banque de gènes , Séquençage nucléotidique à haut débit , Souris , Microtomie , Bulbe olfactif/ultrastructure , ARN/métabolisme , RT-PCR , Microextraction en phase solide/méthodes , Coloration et marquage/méthodes , Analyse sur puce à tissus/instrumentation , Fixation tissulaire/méthodes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE