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...
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
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.
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
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.
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
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.
MIT Licensehttps://opensource.org/licenses/MIT
License information was derived automatically
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.
Mouse embryonic cell cycle dataset from Ref. (https://doi.org/10.1038/nbt.3102) was originally downloaded from ArrayExpress with the accession code E-MTAB-2805
Hematopoiesis differentiation dataset from Ref. (https://doi.org/10.1182/blood-2016-05-716480) was originally downloaded from the Gene Expression Omnibus with the accession code GSE81682
NKT cell differentiation dataset from Ref. (https://doi.org/10.1038/ni.3437) was originally downloaded from the Gene Expression Omnibus with the accession code GSE74596.
Hematopoiesis differentiation dataset from Ref. (https://doi.org/10.1038/nature19348) was originally downloaded from the Gene Expression Omnibus with the accession codes GSE70236, GSE70240, GSE70244
LPS stimulation dataset from Ref. (https://doi.org/10.1016/j.cels.2017.03.010) was originally downloaded from the Gene Expression Omnibus with the accession code GSE94383.
INF-gamma stimulation dataset from Ref. (https://doi.org/10.1038/s41587-020-00803-5) was originally downloaded from the Gene Expression Omnibus with the accession code GSE161465.
AML chemotherapy dataset from Ref. (https://doi.org/10.1038/s41591-018-0233-1) was originally downloaded from the Gene Expression Omnibus with the accession code GSE116481.
AML diagnosis/relapse dataset from Ref. (https://doi.org/10.1038/s41375-021-01338-7) was originally downloaded from the Gene Expression Omnibus with the accession code GSE126068.
MS case control PBMC and CSF datasets from Ref. (https://doi.org/10.1038/s41467-019-14118-w) was originally downloaded from the Gene Expression Omnibus with the accession code GSE138266.
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
Pancreas, Lung atlas, human immune cell, and human and mouse immune cell integration RNA integration tasks, and all ATAC mouse brain integration tasks from the manuscript "Benchmarking atlas-level data integration in single-cell genomics". These datasets were aggregated from public datasets, cell annotations were harmonized or reannotated, and the data was consistently preprocessed using scran pooling and log+1 transformation (for RNA tasks). In the immune cell datasets an erythrocyte development trajectory was also annotated. Details on dataset preprocessing can be found in the paper and in the accompanying Github at https://www.github.com/theislab/scib.Please cite the paper and the papers the individual datasets were aggregated from when using this data.
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
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.
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.
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
scIB-E is a comprehensive deep learning-based benchmarking framework for evaluating single-cell RNA sequencing (scRNA-seq) data integration methods.
Unified Benchmarking Framework:
Refined Metrics for Intra-cell-type Variation:
Novel Loss Function:
The preprocessed datasets are available at src/data.
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
Processed PBMC data for integration tutorial in https://github.com/rpmccordlab/SMILE.
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
seqFISH study of sagittal sections of mouse embryos at 8-10 somite stage. An additional round of hybridisation to capture cell membrane is performed to accurately segment cell boundaries.
Attribution 3.0 (CC BY 3.0)https://creativecommons.org/licenses/by/3.0/
License information was derived automatically
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.
Single Cell Analysis Market Size 2025-2029
The single cell analysis market size is forecast to increase by USD 4.63 billion at a CAGR of 18.2% between 2024 and 2029.
The market is experiencing significant growth due to the increasing prevalence of cancer and the rising incidence of chronic diseases and genetic disorders. This market is driven by the need for more precise and personalized diagnostic and therapeutic approaches, which single cell analysis provides. However, the high cost of single cell analysis products remains a major challenge for market expansion, limiting accessibility to this technology for many healthcare providers and research institutions. Despite this, the market's potential is vast, with opportunities in various end-user industries such as pharmaceuticals, biotechnology, and academia. This approach, which combines data from genomics, transcriptomics, proteomics, and metabolomics, among others, can provide valuable insights into cellular function and behavior.
Companies seeking to capitalize on this market's growth should focus on developing cost-effective solutions while maintaining the high-quality standards required for single cell analysis. Additionally, collaborations and partnerships with key opinion leaders and research institutions can help establish market presence and credibility. Overall, the market presents a compelling opportunity for companies to make a significant impact on the healthcare industry by enabling more accurate diagnoses and personalized treatments.
What will be the Size of the Single Cell Analysis Market during the forecast period?
Request Free Sample
Single-cell analysis, a cutting-edge technology, is revolutionizing the healthcare industry by enabling a more comprehensive knowledge of complex biological systems. This advanced approach allows for the examination of individual cells, providing insights into clinical trial design, tumor microenvironment, and patient stratification. Technologies such as single-cell spatial transcriptomics, microfluidic chips, and droplet microfluidics facilitate the analysis of cell diameter, morphology, immune cell infiltration, and cell cycle phase. Furthermore, single-cell lineage tracing, immune profiling, developmental trajectory analysis, and spatial proteomics offer valuable information on circulating tumor cells and tumor heterogeneity. Single-cell analysis software, genome-wide association studies, and epigenetic analysis contribute to the interpretation of vast amounts of data generated.
Drug response prediction, cell interactions, and biomarker validation are additional applications of this technology. Single-cell analysis services and consulting firms facilitate the implementation of this technology in research and clinical settings. Protein expression profiling, encapsulation, and cell-free DNA analysis through liquid biopsy further expand the scope of single-cell analysis. This technology's potential is vast, offering significant advancements in diagnostics, therapeutics, and fundamental research.
How is this Single Cell Analysis Industry segmented?
The single cell analysis industry research report provides comprehensive data (region-wise segment analysis), with forecasts and estimates in 'USD million' for the period 2025-2029, as well as historical data from 2019-2023 for the following segments.
Product
Consumables
Instrument
Type
Human cells
Animal cells
Technique
Flow cytometry
Next-generation sequencing (NGS)
Polymerase chain reaction (PCR)
Microscopy
Mass spectrometry
Application
Research
Medical
Geography
North America
US
Canada
Europe
France
Germany
Italy
UK
APAC
China
India
Japan
South Korea
By Product Insights
The consumables segment is estimated to witness significant growth during the forecast period. The market encompasses various technologies and applications, including cell stress analysis, omics data integration, cellular heterogeneity, cell engineering, single-cell immunophenotyping, single-cell DNA sequencing, cell proliferation assays, systems biology, precision medicine, cellular metabolism, single-cell proteomics, gene editing, imaging cytometry, academic research, mass cytometry, single-cell barcoding, single-cell spatial analysis, microarray analysis, single-cell sequencing, machine learning, biopharmaceutical industry, data visualization, next-generation sequencing, developmental biology, biotechnology industry, clinical diagnostics, cell cycle analysis, high-throughput screening, cell signaling, regenerative medicine, cell line development, cancer research, flow cytometry, drug discovery, stem cell research, cell culture, cell differentiation assays, biomarker discovery, personalized medicine, single-cell RNA sequencing, single-cell methylation analysis, single-cell data analysis, multiplexed analysi
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
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
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
This repository gathers the data and code used to generate hepatocellular carcinoma analyses in the paper presenting SeuratIntegrate. It contains the scripts to reproduce the figures presented in the article. Some figures are also available as pdf files.
To be able to fully reproduce the results from the paper, one shoud:
remotes::install_local("path/to/SeuratIntegrate_0.4.1.tar.gz")
conda env create --file SeuratIntegrate_bbknn_package-list.yml
conda env create --file SeuratIntegrate_scanorama_package-list.yml
conda env create --file SeuratIntegrate_scvi-tools_package-list.yml
conda env create --file SeuratIntegrate_trvae_package-list.yml
library(SeuratIntegrate)
UpdateEnvCache("bbknn", conda.env = "SeuratIntegrate_bbknn", conda.env.is.path = FALSE)
UpdateEnvCache("scanorama", conda.env = "SeuratIntegrate_scanorama", conda.env.is.path = FALSE)
UpdateEnvCache("scvi", conda.env = "SeuratIntegrate_scvi-tools", conda.env.is.path = FALSE)
UpdateEnvCache("trvae", conda.env = "SeuratIntegrate_trvae", conda.env.is.path = FALSE)
Once done, running the code in integrate.R should produce reproducible results. Note that lines 3 to 6 from integrate.R should be adapted to the user's setup.
integrate.R is subdivided into six main parts:
Intermediate SeuratObject
s have been saved between steps 3 and 4 and 5 and 6 (liver10k_integrated_object.RDS and liver10k_integrated_scored_object.RDS respectively). It is possible to start with these intermediate SeuratObject
s to avoid the preceding steps, given that the Preparation step is always run before.
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
Fig 2
Bone marrow (Fig 2B, D, E, F, H, Supplementary Fig 1A, 2,3)
1. Fig 2/BM/Reference/ Fig2_BM_prepare_data.R: Prepare bone marrow for CellFuse
2. Fig 2/BM/ BM_CellFuse_Integration.R: Run CellFuse
3. Fig 2/BM/BM_Running_Benchmark_Methods.R: Run benchmarking methods (Harmony, Seurat, FastMNN)
4. Fig 2/BM/BM_scIB_Benchmarking.ipynb: evaluate performance of CellFuse and other benchmarking methods using scIB framework proposed by Luecken et al.
5. Fig 2/BM/ BM_scIB_prepare_figures.R: Visualize results of scIB framework
6. Fig 2/BM/Sequential_Feature_drop/Prepare_data.R: Prepare data for evaluating sequential feature drop
7. Fig 2/BM/Sequential_Feature_drop/Run_methods.R: Run CellFuse, Harmony, Seurat and FastMNN for sequential feature drop
8. Fig 2/BM/Sequential_Feature_drop/Evaluate_results.R: Evaluate results features drop and visualize data.
PBMC (Fig 2G,I, Supplementary Fig 1B and 4)
1. Fig 2/PBMC/Reference/ Fig2_PBMC_prepare_data.R: Prepare PBMC data for CellFuse
2. Fig 2/ PBMC / PBMC_CellFuse_Integration.R: Run CellFuse
3. Fig 2/ PBMC /PBMC_Running_Benchmark_Methods.R: Run benchmarking methods (Harmony, Seurat, FastMNN)
4. Fig 2/ PBMC /PBMC_scIB_Benchmarking.ipynb: evaluate performace of CellFuse and other benchmarking methods using scIB framework proposed by Luecken et al., 2021
5. Fig 2/ PBMC /PBMC_scIB_prepare_figures.R: Visualize results of scIB framework
6. Fig 2/ PBMC/ RunTime_benchmark/Run_Benchmark.R: Prepare data, run benchmarking method and evaluate results.
Fig 3 and Supplementary Fig 5
1. Fig 3/Reference/ Fig3_CyTOF_prepare_data.R: Prepare CyTOF and CITE-Seq data for CellFuse
2. Fig 3/CellFuse_Integration_CyTOF.R: Run CellFuse to remove batch effect and integrate CyTOF data from day 7 post-infusion
3. Fig 3/CellFuse_Integration_CITESeq.R: Run CellFuse to integrate CyTOF and CITE-Seq data
4. Fig 3/CART_Data_visualisation.R: Visualize data
Fig 4
HuBMAP CODEX data (Fig. 4A, B, C, D and Supplementary Fig 6)
1. Fig 4/CODEX_colorectal/Reference/ CODEX_HuBMAP_prepare_data.R: Prepare CODEX data from annotated and unannotated donor
2. Fig 4/ CODEX_colorectal/ CODEX_HuBMAP_CellFuse_Predict.R: Run CellFuse on cells from from annotated and unannotated donor
3. Fig 4/ CODEX_colorectal/CODEX_HuBMAP_Data_visualisation.R: Visualize data and prepare figures.
4. Fig 4/ CODEX_colorectal/ CODEX_HuBMAP_Benchmark.R: Benchmarking CellFuse against CELESTA, SVM and Seurat using cells from annotated donors and prepare figures.
a. Astir is python package so run following python notebook: Fig 4/ CODEX_colorectal/ Benchmarking/Astir/Astrir.ipynb
5. Fig 4/ CODEX_colorectal/CODEX_HuBMAP_Suppl_figure_heatmap.R: F1score calculation per celltype per Benchmarking methods and heatmap comparing celltypes from annotated and unannotated donors (Supplementary Fig 6)
IMC Breast cancer data (Fig. 4E,F, G and Supplementary Fig 7)
1. Fig 4/ IMC_Breast_Cancer/ IMC_prepare_data.R: Prepare CODEX data from annotated and unannotated donor
2. Fig 4/ IMC_Breast_Cancer/ IMC_CellFuse_Predict.R: Run CellFuse to predict cell types
3. Fig 4/ IMC_Breast_Cancer/ IMC_dat_visualization.R: Visualize data and prepare figures.
Fig 5
1. Fig5/ Reference/ Fig5_CyTOF_Data_prep.R: Prepare CyTOF data from healthy PBMC and healthy colon single cells
2. Fig5/ MIBI_CellFuse_Predict.R: Run CellFuse to predicte cells from colon cancer patients
3. Fig5/ MIBI_PostPrediction.R: Visualize data and prepare figures
4. Fig5/ Predicted_Data/ mask_generation.ipynb: Post CellFuse prediction annotated cell types in segmented images. This will generate Fig5C and D
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
Data used for tutorial.
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
Additional file 3: Supplementary Table S3. Detailed comparison of multiple single-cell RNA-seq data visualization software.
https://www.datainsightsmarket.com/privacy-policyhttps://www.datainsightsmarket.com/privacy-policy
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.
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
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
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...