100+ datasets found
  1. f

    Data_Sheet_1_CBA: Cluster-Guided Batch Alignment for Single Cell RNA-seq.PDF...

    • frontiersin.figshare.com
    pdf
    Updated Jun 1, 2023
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Wenbo Yu; Ahmed Mahfouz; Marcel J. T. Reinders (2023). Data_Sheet_1_CBA: Cluster-Guided Batch Alignment for Single Cell RNA-seq.PDF [Dataset]. http://doi.org/10.3389/fgene.2021.644211.s001
    Explore at:
    pdfAvailable download formats
    Dataset updated
    Jun 1, 2023
    Dataset provided by
    Frontiers
    Authors
    Wenbo Yu; Ahmed Mahfouz; Marcel J. T. Reinders
    License

    Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
    License information was derived automatically

    Description

    The power of single-cell RNA sequencing (scRNA-seq) in detecting cell heterogeneity or developmental process is becoming more and more evident every day. The granularity of this knowledge is further propelled when combining two batches of scRNA-seq into a single large dataset. This strategy is however hampered by technical differences between these batches. Typically, these batch effects are resolved by matching similar cells across the different batches. Current approaches, however, do not take into account that we can constrain this matching further as cells can also be matched on their cell type identity. We use an auto-encoder to embed two batches in the same space such that cells are matched. To accomplish this, we use a loss function that preserves: (1) cell-cell distances within each of the two batches, as well as (2) cell-cell distances between two batches when the cells are of the same cell-type. The cell-type guidance is unsupervised, i.e., a cell-type is defined as a cluster in the original batch. We evaluated the performance of our cluster-guided batch alignment (CBA) using pancreas and mouse cell atlas datasets, against six state-of-the-art single cell alignment methods: Seurat v3, BBKNN, Scanorama, Harmony, LIGER, and BERMUDA. Compared to other approaches, CBA preserves the cluster separation in the original datasets while still being able to align the two datasets. We confirm that this separation is biologically meaningful by identifying relevant differential expression of genes for these preserved clusters.

  2. d

    Data from: Large-scale integration of single-cell transcriptomic data...

    • dataone.org
    • data.niaid.nih.gov
    • +1more
    Updated May 2, 2025
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    David McKellar; Iwijn De Vlaminck; Benjamin Cosgrove (2025). Large-scale integration of single-cell transcriptomic data captures transitional progenitor states in mouse skeletal muscle regeneration [Dataset]. http://doi.org/10.5061/dryad.t4b8gtj34
    Explore at:
    Dataset updated
    May 2, 2025
    Dataset provided by
    Dryad Digital Repository
    Authors
    David McKellar; Iwijn De Vlaminck; Benjamin Cosgrove
    Time period covered
    Oct 22, 2021
    Description

    Skeletal muscle repair is driven by the coordinated self-renewal and fusion of myogenic stem and progenitor cells. Single-cell gene expression analyses of myogenesis have been hampered by the poor sampling of rare and transient cell states that are critical for muscle repair, and do not inform the spatial context that is important for myogenic differentiation. Here, we demonstrate how large-scale integration of single-cell and spatial transcriptomic data can overcome these limitations. We created a single-cell transcriptomic dataset of mouse skeletal muscle by integration, consensus annotation, and analysis of 23 newly collected scRNAseq datasets and 88 publicly available single-cell (scRNAseq) and single-nucleus (snRNAseq) RNA-sequencing datasets. The resulting dataset includes more than 365,000 cells and spans a wide range of ages, injury, and repair conditions. Together, these data enabled identification of the predominant cell types in skeletal muscle, and resolved cell subtypes, in...

  3. f

    Table1_Influence of single-cell RNA sequencing data integration on the...

    • frontiersin.figshare.com
    docx
    Updated Jun 13, 2023
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Tomasz Kujawa; Michał Marczyk; Joanna Polanska (2023). Table1_Influence of single-cell RNA sequencing data integration on the performance of differential gene expression analysis.docx [Dataset]. http://doi.org/10.3389/fgene.2022.1009316.s002
    Explore at:
    docxAvailable download formats
    Dataset updated
    Jun 13, 2023
    Dataset provided by
    Frontiers
    Authors
    Tomasz Kujawa; Michał Marczyk; Joanna Polanska
    License

    Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
    License information was derived automatically

    Description

    Large-scale comprehensive single-cell experiments are often resource-intensive and require the involvement of many laboratories and/or taking measurements at various times. This inevitably leads to batch effects, and systematic variations in the data that might occur due to different technology platforms, reagent lots, or handling personnel. Such technical differences confound biological variations of interest and need to be corrected during the data integration process. Data integration is a challenging task due to the overlapping of biological and technical factors, which makes it difficult to distinguish their individual contribution to the overall observed effect. Moreover, the choice of integration method may impact the downstream analyses, including searching for differentially expressed genes. From the existing data integration methods, we selected only those that return the full expression matrix. We evaluated six methods in terms of their influence on the performance of differential gene expression analysis in two single-cell datasets with the same biological study design that differ only in the way the measurement was done: one dataset manifests strong batch effects due to the measurements of each sample at a different time. Integrated data were visualized using the UMAP method. The evaluation was done both on individual gene level using parametric and non-parametric approaches for finding differentially expressed genes and on gene set level using gene set enrichment analysis. As an evaluation metric, we used two correlation coefficients, Pearson and Spearman, of the obtained test statistics between reference, test, and corrected studies. Visual comparison of UMAP plots highlighted ComBat-seq, limma, and MNN, which reduced batch effects and preserved differences between biological conditions. Most of the tested methods changed the data distribution after integration, which negatively impacts the use of parametric methods for the analysis. Two algorithms, MNN and Scanorama, gave very poor results in terms of differential analysis on gene and gene set levels. Finally, we highlight ComBat-seq as it led to the highest correlation of test statistics between reference and corrected dataset among others. Moreover, it does not distort the original distribution of gene expression data, so it can be used in all types of downstream analyses.

  4. Single-cell datasets for temporal gene expression integration

    • zenodo.org
    bin
    Updated Aug 12, 2022
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Jolene Ranek; Natalie Stanley; Jeremy Purvis; Jolene Ranek; Natalie Stanley; Jeremy Purvis (2022). Single-cell datasets for temporal gene expression integration [Dataset]. http://doi.org/10.5281/zenodo.6587903
    Explore at:
    binAvailable download formats
    Dataset updated
    Aug 12, 2022
    Dataset provided by
    Zenodohttp://zenodo.org/
    Authors
    Jolene Ranek; Natalie Stanley; Jeremy Purvis; Jolene Ranek; Natalie Stanley; Jeremy Purvis
    License

    MIT Licensehttps://opensource.org/licenses/MIT
    License information was derived automatically

    Description

    Contains loom files and preprocessed adata objects to compare methods for temporal gene expression integration. Loom files can be accessed using the 'read' function in Scvelo. Preprocessed adata objects can be accessed using the 'read_h5ad' function in Scanpy.

    The raw single-cell RNA sequencing datasets can be found under the following accession codes.

  5. Additional file 4 of scExtract: leveraging large language models for fully...

    • springernature.figshare.com
    xlsx
    Updated Jun 20, 2025
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Yuxuan Wu; Fuchou Tang (2025). Additional file 4 of scExtract: leveraging large language models for fully automated single-cell RNA-seq data annotation and prior-informed multi-dataset integration [Dataset]. http://doi.org/10.6084/m9.figshare.29368159.v1
    Explore at:
    xlsxAvailable download formats
    Dataset updated
    Jun 20, 2025
    Dataset provided by
    Figsharehttp://figshare.com/
    Authors
    Yuxuan Wu; Fuchou Tang
    License

    Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
    License information was derived automatically

    Description

    Additional file 4. Table S3: Skin scRNA-seq datasets selected for atlas construction.

  6. m

    Data from: CSS: cluster similarity spectrum integration of single-cell...

    • data.mendeley.com
    Updated Aug 15, 2020
    + more versions
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Zhisong He (2020). CSS: cluster similarity spectrum integration of single-cell genomics data [Dataset]. http://doi.org/10.17632/3kthhpw2pd.2
    Explore at:
    Dataset updated
    Aug 15, 2020
    Authors
    Zhisong He
    License

    Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
    License information was derived automatically

    Description

    It is a major challenge to integrate single-cell sequencing data across experiments, conditions, batches, timepoints and other technical considerations. New computational methods are required that can integrate samples while simultaneously preserving biological information. Here, we propose an unsupervised reference-free data representation, Cluster Similarity Spectrum (CSS), where each cell is represented by its similarities to clusters independently identified across samples. We show that CSS can be used to assess cellular heterogeneity and enable reconstruction of differentiation trajectories from cerebral organoid and other single-cell transcriptomic data, and to integrate data across experimental conditions and human individuals.

    The presented data set here includes 1) the seurat object of the published two-month-old human cerebral organoid scRNA-seq data (Kanton et al. 2019 Nature); 2) the single-cell RNA-seq data of cerebral organoid generated by inDrop; 3) the newly generated single-cell RNA-seq data of cerebral organoids with and without fixation conditions.

  7. Additional file 2 of scExtract: leveraging large language models for fully...

    • springernature.figshare.com
    xlsx
    Updated Jun 20, 2025
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Yuxuan Wu; Fuchou Tang (2025). Additional file 2 of scExtract: leveraging large language models for fully automated single-cell RNA-seq data annotation and prior-informed multi-dataset integration [Dataset]. http://doi.org/10.6084/m9.figshare.29368153.v1
    Explore at:
    xlsxAvailable download formats
    Dataset updated
    Jun 20, 2025
    Dataset provided by
    Figsharehttp://figshare.com/
    Authors
    Yuxuan Wu; Fuchou Tang
    License

    Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
    License information was derived automatically

    Description

    Additional file 2. Table S1: Cellxgene datasets used for annotation accuracy evaluation.

  8. q

    Single Cell Insights Into Cancer Transcriptomes: A Five-Part Single-Cell...

    • qubeshub.org
    Updated Nov 16, 2021
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Leigh Samsa*; Melissa Eslinger; Adam Kleinschmit; Amanda Solem; Carlos Goller* (2021). Single Cell Insights Into Cancer Transcriptomes: A Five-Part Single-Cell RNAseq Case Study Lesson [Dataset]. http://doi.org/10.24918/cs.2021.26
    Explore at:
    Dataset updated
    Nov 16, 2021
    Dataset provided by
    QUBES
    Authors
    Leigh Samsa*; Melissa Eslinger; Adam Kleinschmit; Amanda Solem; Carlos Goller*
    Description

    There is a growing need for integration of “Big Data” into undergraduate biology curricula. Transcriptomics is one venue to examine biology from an informatics perspective. RNA sequencing has largely replaced the use of microarrays for whole genome gene expression studies. Recently, single cell RNA sequencing (scRNAseq) has unmasked population heterogeneity, offering unprecedented views into the inner workings of individual cells. scRNAseq is transforming our understanding of development, cellular identity, cell function, and disease. As a ‘Big Data,’ scRNAseq can be intimidating for students to conceptualize and analyze, yet it plays an increasingly important role in modern biology. To address these challenges, we created an engaging case study that guides students through an exploration of scRNAseq technologies. Students work in groups to explore external resources, manipulate authentic data and experience how single cell RNA transcriptomics can be used for personalized cancer treatment. This five-part case study is intended for upper-level life science majors and graduate students in genetics, bioinformatics, molecular biology, cell biology, biochemistry, biology, and medical genomics courses. The case modules can be completed sequentially, or individual parts can be separately adapted. The first module can also be used as a stand-alone exercise in an introductory biology course. Students need an intermediate mastery of Microsoft Excel but do not need programming skills. Assessment includes both students’ self-assessment of their learning as answers to previous questions are used to progress through the case study and instructor assessment of final answers. This case provides a practical exercise in the use of high-throughput data analysis to explore the molecular basis of cancer at the level of single cells.

  9. Data from: Deep cross-omics cycle attention model for joint analysis of...

    • zenodo.org
    zip
    Updated Jun 17, 2022
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Chunman Zuo; Chunman Zuo (2022). Deep cross-omics cycle attention model for joint analysis of single-cell multi-omics data [Dataset]. http://doi.org/10.5281/zenodo.4762065
    Explore at:
    zipAvailable download formats
    Dataset updated
    Jun 17, 2022
    Dataset provided by
    Zenodohttp://zenodo.org/
    Authors
    Chunman Zuo; Chunman Zuo
    License

    Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
    License information was derived automatically

    Description

    We proposed DCCA for accurately dissecting the cellular heterogeneity on joint-profiling multi-omics data from the same individual cell by transferring representation between each other.

  10. Data from: Benchmarking deep learning methods for biologically conserved...

    • zenodo.org
    zip
    Updated Jan 12, 2025
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Chenxin Yi; Chenxin Yi (2025). Benchmarking deep learning methods for biologically conserved single-cell integration. [Dataset]. http://doi.org/10.5281/zenodo.14633468
    Explore at:
    zipAvailable download formats
    Dataset updated
    Jan 12, 2025
    Dataset provided by
    Zenodohttp://zenodo.org/
    Authors
    Chenxin Yi; Chenxin Yi
    License

    Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
    License information was derived automatically

    Description

    scIB-E is a comprehensive deep learning-based benchmarking framework for evaluating single-cell RNA sequencing (scRNA-seq) data integration methods.

    • Unified Benchmarking Framework:

      • Evaluates 16 deep-learning single-cell integration methods using a unified variational autoencoder (VAE) framework.
      • Incorporates batch information, cell-type labels, and combined strategies across three integration levels.
    • Refined Metrics for Intra-cell-type Variation:

      • Extends the single-cell integration benchmarking (scIB) metrics by adding new metrics to better capture intra-cell-type biological conservation.
    • Novel Loss Function:

      • Introduces Corr-MSE Loss, a correlation-based loss function designed to preserve global cellular relationships and enhance intra-cell-type biological variation.

    The preprocessed datasets are available at src/data.

  11. N

    Data from: Integrating multimodal data sets into a mathematical framework to...

    • data.niaid.nih.gov
    Updated May 10, 2021
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Johnson K; Howard GR; Morgan D; Brenner EA; Gardner AL; Durrett RE; Mo W; Al’Khafaji A; Sontag ED; Jarrett AM; Yankeelov TE; Brock A (2021). Integrating multimodal data sets into a mathematical framework to describe and predict therapeutic resistance in cancer [Dataset]. https://data.niaid.nih.gov/resources?id=gse154932
    Explore at:
    Dataset updated
    May 10, 2021
    Dataset provided by
    University of Texas at Austin
    Authors
    Johnson K; Howard GR; Morgan D; Brenner EA; Gardner AL; Durrett RE; Mo W; Al’Khafaji A; Sontag ED; Jarrett AM; Yankeelov TE; Brock A
    Description

    A significant challenge in the field of biomedicine is the development of methods to integrate the multitude of dispersed data sets into comprehensive frameworks to be used to generate optimal clinical decisions. Recent technological advances in single cell analysis allow for high-dimensional molecular characterization of cells and populations, but to date, few mathematical models have attempted to integrate measurements from the single cell scale with other data types. Here, we present a framework that actionizes static outputs from a machine learning model and leverages these as measurements of state variables in a dynamic mechanistic model of treatment response. We apply this framework to breast cancer cells to integrate single cell transcriptomic data with longitudinal population-size data. We demonstrate that the explicit inclusion of the transcriptomic information in the parameter estimation is critical for identification of the model parameters and enables accurate prediction of new treatment regimens. Inclusion of the transcriptomic data improves predictive accuracy in new treatment response dynamics with a concordance correlation coefficient (CCC) of 0.89 compared to a prediction accuracy of CCC = 0.79 without integration of the single cell RNA sequencing (scRNA-seq) data directly into the model calibration. To the best our knowledge, this is the first work that explicitly integrates single cell clonally-resolved transcriptome datasets with longitudinal treatment response data into a mechanistic mathematical model of drug resistance dynamics. We anticipate this approach to be a first step that demonstrates the feasibility of incorporating multimodal data sets into identifiable mathematical models to develop optimized treatment regimens from data. Single cell RNA-seq of MDA-MB-231 cell line with chemotherapy treatment

  12. pbmc single cell RNA-seq matrix

    • zenodo.org
    csv
    Updated May 4, 2021
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Samuel Buchet; Samuel Buchet; Francesco Carbone; Morgan Magnin; Morgan Magnin; Mickaël Ménager; Olivier Roux; Olivier Roux; Francesco Carbone; Mickaël Ménager (2021). pbmc single cell RNA-seq matrix [Dataset]. http://doi.org/10.5281/zenodo.4730807
    Explore at:
    csvAvailable download formats
    Dataset updated
    May 4, 2021
    Dataset provided by
    Zenodohttp://zenodo.org/
    Authors
    Samuel Buchet; Samuel Buchet; Francesco Carbone; Morgan Magnin; Morgan Magnin; Mickaël Ménager; Olivier Roux; Olivier Roux; Francesco Carbone; Mickaël Ménager
    License

    Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
    License information was derived automatically

    Description

    Single cell RNA-sequencing dataset of peripheral blood mononuclear cells (pbmc: T, B, NK and monocytes) extracted from two healthy donors.

    Cells labeled as C26 come from a 30 years old female and cells labeled as C27 come from a 53 years old male. Cells have been isolated from blood using ficoll. Samples were sequenced using standard 3' v3 chemistry protocols by 10x genomics. Cellranger v4.0.0 was used for the processing, and reads were aligned to the ensembl GRCg38 human genome (GRCg38_r98-ensembl_Sept2019). QC metrics were calculated on the count matrix generated by cellranger (filtered_feature_bc_matrix). Cells with less than 3 genes per cells, less than 500 reads per cell and more than 20% of mithocondrial genes were discarded.

    The processing steps was performed with the R package Seurat (https://satijalab.org/seurat/), including sample integration, data normalisation and scaling, dimensional reduction, and clustering. SCTransform method was adopted for the normalisation and scaling steps. The clustered cells were manually annotated using known cell type markers.

    Files content:

    - raw_dataset.csv: raw gene counts

    - normalized_dataset.csv: normalized gene counts (single cell matrix)

    - cell_types.csv: cell types identified from annotated cell clusters

    - cell_types_macro.csv: cell macro types

    - UMAP_coordinates.csv: 2d cell coordinates computed with UMAP algorithm in Seurat

  13. Additional file 3 of scRNASequest: an ecosystem of scRNA-seq analysis,...

    • springernature.figshare.com
    xlsx
    Updated Feb 13, 2024
    + more versions
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Kejie Li; Yu H. Sun; Zhengyu Ouyang; Soumya Negi; Zhen Gao; Jing Zhu; Wanli Wang; Yirui Chen; Sarbottam Piya; Wenxing Hu; Maria I. Zavodszky; Hima Yalamanchili; Shaolong Cao; Andrew Gehrke; Mark Sheehan; Dann Huh; Fergal Casey; Xinmin Zhang; Baohong Zhang (2024). Additional file 3 of scRNASequest: an ecosystem of scRNA-seq analysis, visualization, and publishing [Dataset]. http://doi.org/10.6084/m9.figshare.22735494.v1
    Explore at:
    xlsxAvailable download formats
    Dataset updated
    Feb 13, 2024
    Dataset provided by
    Figsharehttp://figshare.com/
    Authors
    Kejie Li; Yu H. Sun; Zhengyu Ouyang; Soumya Negi; Zhen Gao; Jing Zhu; Wanli Wang; Yirui Chen; Sarbottam Piya; Wenxing Hu; Maria I. Zavodszky; Hima Yalamanchili; Shaolong Cao; Andrew Gehrke; Mark Sheehan; Dann Huh; Fergal Casey; Xinmin Zhang; Baohong Zhang
    License

    Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
    License information was derived automatically

    Description

    Additional file 3: Supplementary Table S3. Detailed comparison of multiple single-cell RNA-seq data visualization software.

  14. o

    Test Data for Galaxy tutorial "Batch Correction and Integration" - Seurat...

    • ordo.open.ac.uk
    • zenodo.org
    bin
    Updated Apr 28, 2025
    + more versions
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Marisa Loach (2025). Test Data for Galaxy tutorial "Batch Correction and Integration" - Seurat version [Dataset]. http://doi.org/10.5281/zenodo.14713816
    Explore at:
    binAvailable download formats
    Dataset updated
    Apr 28, 2025
    Dataset provided by
    The Open University
    Authors
    Marisa Loach
    License

    Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
    License information was derived automatically

    Description

    This data is used for the Seurat version of the batch correction and integration tutorial on the Galaxy Training Network. The input data was provided by Seurat in the 'Integrative Analysis in Seurat v5' tutorial. The input dataset provided here has been filtered to include only cells for which nFeature_RNA > 1000. The other datasets were produced on Galaxy. The original dataset was published as: Ding, J., Adiconis, X., Simmons, S.K. et al. Systematic comparison of single-cell and single-nucleus RNA-sequencing methods. Nat Biotechnol 38, 737–746 (2020). https://doi.org/10.1038/s41587-020-0465-8.

  15. Data from: SIRV: Spatial inference of RNA velocity at the single-cell...

    • zenodo.org
    • data.niaid.nih.gov
    Updated Jul 6, 2022
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Tamim Abdelaal; Laurens Grossouw; Jeroen Pasterkamp; Boudewijn Lelieveldt; Marcel Reinders; Ahmed Mahfouz; Tamim Abdelaal; Laurens Grossouw; Jeroen Pasterkamp; Boudewijn Lelieveldt; Marcel Reinders; Ahmed Mahfouz (2022). SIRV: Spatial inference of RNA velocity at the single-cell resolution [Dataset]. http://doi.org/10.5281/zenodo.6798659
    Explore at:
    Dataset updated
    Jul 6, 2022
    Dataset provided by
    Zenodohttp://zenodo.org/
    Authors
    Tamim Abdelaal; Laurens Grossouw; Jeroen Pasterkamp; Boudewijn Lelieveldt; Marcel Reinders; Ahmed Mahfouz; Tamim Abdelaal; Laurens Grossouw; Jeroen Pasterkamp; Boudewijn Lelieveldt; Marcel Reinders; Ahmed Mahfouz
    License

    Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
    License information was derived automatically

    Description

    Spatial transcriptomics and scRNA-seq datasets used for integration and prediction of un/spliced expression for spatially measured genes using SIRV, used to infer the RNA velocity in the spatial context

  16. o

    Single-cell RNA-seq and TCR-seq datasets for the article: Intestinal...

    • explore.openaire.eu
    • zenodo.org
    Updated Apr 25, 2025
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Teruyuki Sano (2025). Single-cell RNA-seq and TCR-seq datasets for the article: Intestinal inflammation promotes gut commensal-specific CD4 T cell to initiate molecular mimicry-mediated neuroinflammation [Dataset]. http://doi.org/10.5281/zenodo.15276654
    Explore at:
    Dataset updated
    Apr 25, 2025
    Authors
    Teruyuki Sano
    Description

    This dataset accompanies the study titled "Intestinal inflammation promotes gut commensal-specific CD4 T cell to initiate molecular mimicry-mediated neuroinflammation" and contains: Processed bulk T cell receptor sequencing data, scripts for clonotype assembly and overlap analysis using MiXCR. Processed single-cell RNA-sequencing data, reference annotations, code for preprocessing, integration, and analysis, and associated table outputs.

  17. Data from: Cross-disease integration of single-cell RNA sequencing data from...

    • zenodo.org
    bin
    Updated May 16, 2025
    + more versions
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Catarina Pinto; Jakub Widawski; Sophie Zahalka; Barbara Thaler; Linda Schuster; Samuel Lukowski; Fidel Ramirez; Iñigo Tirapu; Catarina Pinto; Jakub Widawski; Sophie Zahalka; Barbara Thaler; Linda Schuster; Samuel Lukowski; Fidel Ramirez; Iñigo Tirapu (2025). Cross-disease integration of single-cell RNA sequencing data from lung myeloid cells reveals TAM signature in in vitro model [Dataset]. http://doi.org/10.5281/zenodo.15304326
    Explore at:
    binAvailable download formats
    Dataset updated
    May 16, 2025
    Dataset provided by
    Zenodohttp://zenodo.org/
    Authors
    Catarina Pinto; Jakub Widawski; Sophie Zahalka; Barbara Thaler; Linda Schuster; Samuel Lukowski; Fidel Ramirez; Iñigo Tirapu; Catarina Pinto; Jakub Widawski; Sophie Zahalka; Barbara Thaler; Linda Schuster; Samuel Lukowski; Fidel Ramirez; Iñigo Tirapu
    License

    Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
    License information was derived automatically

    Description

    Single cells from a 3D human cell-based model comprising tumor cell line-derived spheroids, cancer-associated fibroblasts and primary monocytes were dissociated and analyzed using scRNAseq. 4 monocyte donors were used in the 3D model, and 3 monocyte donors were used for 2D differentiation of macrophages.

  18. s

    Single-cell RNA-seq of the mouse and human lymph node lymphatic vasculature

    • purl.stanford.edu
    Updated Jan 2, 2020
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Menglan Xiang (2020). Single-cell RNA-seq of the mouse and human lymph node lymphatic vasculature [Dataset]. https://purl.stanford.edu/xr811qy1057
    Explore at:
    Dataset updated
    Jan 2, 2020
    Authors
    Menglan Xiang
    License

    Attribution 3.0 (CC BY 3.0)https://creativecommons.org/licenses/by/3.0/
    License information was derived automatically

    Description

    Single-cell transcriptomics promises to revolutionize our understanding of the vasculature. Emerging computational methods applied to high dimensional single cell data allow integration of results between samples and species, and illuminate the diversity and underlying developmental and architectural organization of cell populations. Here, we illustrate these methods in analysis of mouse lymph node (LN) lymphatic endothelial cells (LEC) at single cell resolution. Clustering identifies five well-delineated subsets, including two medullary sinus subsets not recognized previously as distinct. Nearest neighbor alignments in trajectory space position the major subsets in a sequence that recapitulates known and suggests novel features of LN lymphatic organization, providing a transcriptional map of the lymphatic endothelial niches and of the transitions between them. Differences in gene expression reveal specialized programs for (1) subcapsular ceiling endothelial interactions with the capsule connective tissue and cells, (2) subcapsular floor regulation of lymph borne cell entry into the LN parenchyma and antigen presentation, and (3) medullary subset specialization for pathogen interactions and LN remodeling. LEC of the subcapsular sinus floor and medulla, which represent major sites of cell entry and exit from the LN parenchyma respectively, respond robustly to oxazolone inflammation challenge with enriched signaling pathways that converge on both innate and adaptive immune responses. Integration of mouse and human single-cell profiles reveals a conserved cross-species pattern of lymphatic vascular niches and gene expression, as well as specialized human subsets and genes unique to each species. The examples provided demonstrate the power of single-cell analysis in elucidating endothelial cell heterogeneity, vascular organization and endothelial cell responses. We discuss the findings from the perspective of LEC functions in relation to niche formations in the unique stromal and highly immunological environment of the LN.

  19. S

    Single Cell RNA Sequencing Technology Report

    • datainsightsmarket.com
    doc, pdf, ppt
    Updated Jun 11, 2025
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Data Insights Market (2025). Single Cell RNA Sequencing Technology Report [Dataset]. https://www.datainsightsmarket.com/reports/single-cell-rna-sequencing-technology-1932041
    Explore at:
    ppt, pdf, docAvailable download formats
    Dataset updated
    Jun 11, 2025
    Dataset authored and provided by
    Data Insights Market
    License

    https://www.datainsightsmarket.com/privacy-policyhttps://www.datainsightsmarket.com/privacy-policy

    Time period covered
    2025 - 2033
    Area covered
    Global
    Variables measured
    Market Size
    Description

    The single-cell RNA sequencing (scRNA-seq) technology market is experiencing robust growth, projected to reach a significant market size driven by advancements in technology and increasing applications across diverse fields. The market's Compound Annual Growth Rate (CAGR) of 10.2% from 2019 to 2024, coupled with a 2025 market size of $144 million, indicates strong future potential. This growth is fueled by the technology's ability to provide unprecedented insights into cellular heterogeneity and gene expression at a single-cell level, revolutionizing biological research and clinical diagnostics. Key drivers include the rising adoption of scRNA-seq in oncology for identifying cancer subtypes and developing personalized therapies, immunology for understanding immune cell responses, and neuroscience for dissecting complex brain functions. Furthermore, ongoing technological advancements, such as the development of more efficient and cost-effective platforms, are expanding the accessibility and affordability of scRNA-seq, further fueling market expansion. The market's competitive landscape is characterized by a mix of established players like Illumina, Thermo Fisher Scientific, and 10x Genomics, along with emerging companies like Dolomite Bio and Pacific Biosciences, which are driving innovation and expanding applications. Looking ahead to 2033, the continued high CAGR suggests a substantial market expansion. The increasing demand for high-throughput scRNA-seq platforms, combined with the growing integration of bioinformatics and data analysis tools, will be crucial drivers. Challenges like data analysis complexity and the high cost of assays might somewhat restrain growth, but ongoing technological advancements are expected to mitigate these hurdles. The market segmentation, while not explicitly provided, is likely to be diverse, based on technology (e.g., microfluidic, plate-based), application (e.g., oncology, immunology, neuroscience), and end-user (e.g., academic research, pharmaceutical companies, clinical labs). Regional market share distribution will likely show a significant contribution from North America and Europe initially, followed by increasing adoption in Asia-Pacific and other emerging regions.

  20. 4

    Experiment Set - 4DNES8QRX3RH

    • data.4dnucleome.org
    Updated Mar 18, 2025
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Jesse Dixon, SALK (2025). Experiment Set - 4DNES8QRX3RH [Dataset]. https://data.4dnucleome.org/experiment-set-replicates/4DNES8QRX3RH/
    Explore at:
    Dataset updated
    Mar 18, 2025
    Dataset provided by
    4DN Data Coordination and Integration Center
    Authors
    Jesse Dixon, SALK
    Measurement technique
    single cell RNA-seq
    Description

    10X multiomic scRNA-seq on Peripheral blood monocytes from MCG016

Share
FacebookFacebook
TwitterTwitter
Email
Click to copy link
Link copied
Close
Cite
Wenbo Yu; Ahmed Mahfouz; Marcel J. T. Reinders (2023). Data_Sheet_1_CBA: Cluster-Guided Batch Alignment for Single Cell RNA-seq.PDF [Dataset]. http://doi.org/10.3389/fgene.2021.644211.s001

Data_Sheet_1_CBA: Cluster-Guided Batch Alignment for Single Cell RNA-seq.PDF

Related Article
Explore at:
pdfAvailable download formats
Dataset updated
Jun 1, 2023
Dataset provided by
Frontiers
Authors
Wenbo Yu; Ahmed Mahfouz; Marcel J. T. Reinders
License

Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically

Description

The power of single-cell RNA sequencing (scRNA-seq) in detecting cell heterogeneity or developmental process is becoming more and more evident every day. The granularity of this knowledge is further propelled when combining two batches of scRNA-seq into a single large dataset. This strategy is however hampered by technical differences between these batches. Typically, these batch effects are resolved by matching similar cells across the different batches. Current approaches, however, do not take into account that we can constrain this matching further as cells can also be matched on their cell type identity. We use an auto-encoder to embed two batches in the same space such that cells are matched. To accomplish this, we use a loss function that preserves: (1) cell-cell distances within each of the two batches, as well as (2) cell-cell distances between two batches when the cells are of the same cell-type. The cell-type guidance is unsupervised, i.e., a cell-type is defined as a cluster in the original batch. We evaluated the performance of our cluster-guided batch alignment (CBA) using pancreas and mouse cell atlas datasets, against six state-of-the-art single cell alignment methods: Seurat v3, BBKNN, Scanorama, Harmony, LIGER, and BERMUDA. Compared to other approaches, CBA preserves the cluster separation in the original datasets while still being able to align the two datasets. We confirm that this separation is biologically meaningful by identifying relevant differential expression of genes for these preserved clusters.

Search
Clear search
Close search
Google apps
Main menu