https://spdx.org/licenses/CC0-1.0.htmlhttps://spdx.org/licenses/CC0-1.0.html
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, including endothelial subtypes distinguished by vessel-type of origin, fibro/adipogenic progenitors defined by functional roles, and many distinct immune populations. The representation of different experimental conditions and the depth of transcriptome coverage enabled robust profiling of sparsely expressed genes. We built a densely sampled transcriptomic model of myogenesis, from stem cell quiescence to myofiber maturation and identified rare, transitional states of progenitor commitment and fusion that are poorly represented in individual datasets. We performed spatial RNA sequencing of mouse muscle at three time points after injury and used the integrated dataset as a reference to achieve a high-resolution, local deconvolution of cell subtypes. We also used the integrated dataset to explore ligand-receptor co-expression patterns and identify dynamic cell-cell interactions in muscle injury response. We provide a public web tool to enable interactive exploration and visualization of the data. Our work supports the utility of large-scale integration of single-cell transcriptomic data as a tool for biological discovery.
Methods Mice. The Cornell University Institutional Animal Care and Use Committee (IACUC) approved all animal protocols, and experiments were performed in compliance with its institutional guidelines. Adult C57BL/6J mice (mus musculus) were obtained from Jackson Laboratories (#000664; Bar Harbor, ME) and were used at 4-7 months of age. Aged C57BL/6J mice were obtained from the National Institute of Aging (NIA) Rodent Aging Colony and were used at 20 months of age. For new scRNAseq experiments, female mice were used in each experiment.
Mouse injuries and single-cell isolation. To induce muscle injury, both tibialis anterior (TA) muscles of old (20 months) C57BL/6J mice were injected with 10 µl of notexin (10 µg/ml; Latoxan; France). At 0, 1, 2, 3.5, 5, or 7 days post-injury (dpi), mice were sacrificed and TA muscles were collected and processed independently to generate single-cell suspensions. Muscles were digested with 8 mg/ml Collagenase D (Roche; Switzerland) and 10 U/ml Dispase II (Roche; Switzerland), followed by manual dissociation to generate cell suspensions. Cell suspensions were sequentially filtered through 100 and 40 μm filters (Corning Cellgro #431752 and #431750) to remove debris. Erythrocytes were removed through incubation in erythrocyte lysis buffer (IBI Scientific #89135-030).
Single-cell RNA-sequencing library preparation. After digestion, single-cell suspensions were washed and resuspended in 0.04% BSA in PBS at a concentration of 106 cells/ml. Cells were counted manually with a hemocytometer to determine their concentration. Single-cell RNA-sequencing libraries were prepared using the Chromium Single Cell 3’ reagent kit v3 (10x Genomics, PN-1000075; Pleasanton, CA) following the manufacturer’s protocol. Cells were diluted into the Chromium Single Cell A Chip to yield a recovery of 6,000 single-cell transcriptomes. After preparation, libraries were sequenced using on a NextSeq 500 (Illumina; San Diego, CA) using 75 cycle high output kits (Index 1 = 8, Read 1 = 26, and Read 2 = 58). Details on estimated sequencing saturation and the number of reads per sample are shown in Sup. Data 1.
Spatial RNA sequencing library preparation. Tibialis anterior muscles of adult (5 mo) C57BL6/J mice were injected with 10µl notexin (10 µg/ml) at 2, 5, and 7 days prior to collection. Upon collection, tibialis anterior muscles were isolated, embedded in OCT, and frozen fresh in liquid nitrogen. Spatially tagged cDNA libraries were built using the Visium Spatial Gene Expression 3’ Library Construction v1 Kit (10x Genomics, PN-1000187; Pleasanton, CA) (Fig. S7). Optimal tissue permeabilization time for 10 µm thick sections was found to be 15 minutes using the 10x Genomics Visium Tissue Optimization Kit (PN-1000193). H&E stained tissue sections were imaged using Zeiss PALM MicroBeam laser capture microdissection system and the images were stitched and processed using Fiji ImageJ software. cDNA libraries were sequenced on an Illumina NextSeq 500 using 150 cycle high output kits (Read 1=28bp, Read 2=120bp, Index 1=10bp, and Index 2=10bp). Frames around the capture area on the Visium slide were aligned manually and spots covering the tissue were selected using Loop Browser v4.0.0 software (10x Genomics). Sequencing data was then aligned to the mouse reference genome (mm10) using the spaceranger v1.0.0 pipeline to generate a feature-by-spot-barcode expression matrix (10x Genomics).
Download and alignment of single-cell RNA sequencing data. For all samples available via SRA, parallel-fastq-dump (github.com/rvalieris/parallel-fastq-dump) was used to download raw .fastq files. Samples which were only available as .bam files were converted to .fastq format using bamtofastq from 10x Genomics (github.com/10XGenomics/bamtofastq). Raw reads were aligned to the mm10 reference using cellranger (v3.1.0).
Preprocessing and batch correction of single-cell RNA sequencing datasets. First, ambient RNA signal was removed using the default SoupX (v1.4.5) workflow (autoEstCounts and adjustCounts; github.com/constantAmateur/SoupX). Samples were then preprocessed using the standard Seurat (v3.2.1) workflow (NormalizeData, ScaleData, FindVariableFeatures, RunPCA, FindNeighbors, FindClusters, and RunUMAP; github.com/satijalab/seurat). Cells with fewer than 750 features, fewer than 1000 transcripts, or more than 30% of unique transcripts derived from mitochondrial genes were removed. After preprocessing, DoubletFinder (v2.0) was used to identify putative doublets in each dataset, individually. BCmvn optimization was used for PK parameterization. Estimated doublet rates were computed by fitting the total number of cells after quality filtering to a linear regression of the expected doublet rates published in the 10x Chromium handbook. Estimated homotypic doublet rates were also accounted for using the modelHomotypic function. The default PN value (0.25) was used. Putative doublets were then removed from each individual dataset. After preprocessing and quality filtering, we merged the datasets and performed batch-correction with three tools, independently- Harmony (github.com/immunogenomics/harmony) (v1.0), Scanorama (github.com/brianhie/scanorama) (v1.3), and BBKNN (github.com/Teichlab/bbknn) (v1.3.12). We then used Seurat to process the integrated data. After initial integration, we removed the noisy cluster and re-integrated the data using each of the three batch-correction tools.
Cell type annotation. Cell types were determined for each integration method independently. For Harmony and Scanorama, dimensions accounting for 95% of the total variance were used to generate SNN graphs (Seurat::FindNeighbors). Louvain clustering was then performed on the output graphs (including the corrected graph output by BBKNN) using Seurat::FindClusters. A clustering resolution of 1.2 was used for Harmony (25 initial clusters), BBKNN (28 initial clusters), and Scanorama (38 initial clusters). Cell types were determined based on expression of canonical genes (Fig. S3). Clusters which had similar canonical marker gene expression patterns were merged.
Pseudotime workflow. Cells were subset based on the consensus cell types between all three integration methods. Harmony embedding values from the dimensions accounting for 95% of the total variance were used for further dimensional reduction with PHATE, using phateR (v1.0.4) (github.com/KrishnaswamyLab/phateR).
Deconvolution of spatial RNA sequencing spots. Spot deconvolution was performed using the deconvolution module in BayesPrism (previously known as “Tumor microEnvironment Deconvolution”, TED, v1.0; github.com/Danko-Lab/TED). First, myogenic cells were re-labeled, according to binning along the first PHATE dimension, as “Quiescent MuSCs” (bins 4-5), “Activated MuSCs” (bins 6-7), “Committed Myoblasts” (bins 8-10), and “Fusing Myoctes” (bins 11-18). Culture-associated muscle stem cells were ignored and myonuclei labels were retained as “Myonuclei (Type IIb)” and “Myonuclei (Type IIx)”. Next, highly and differentially expressed genes across the 25 groups of cells were identified with differential gene expression analysis using Seurat (FindAllMarkers, using Wilcoxon Rank Sum Test; results in Sup. Data 2). The resulting genes were filtered based on average log2-fold change (avg_logFC > 1) and the percentage of cells within the cluster which express each gene (pct.expressed > 0.5), yielding 1,069 genes. Mitochondrial and ribosomal protein genes were also removed from this list, in line with recommendations in the BayesPrism vignette. For each of the cell types, mean raw counts were calculated across the 1,069 genes to generate a gene expression profile for BayesPrism. Raw counts for each spot were then passed to the run.Ted function, using
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.
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
This integrated dataset represents the large-scale transcriptomics cell atlas of pons and medulla, compiled from 8 independent single-cell/single-nucleus RNA sequencing (sc/sn-RNA seq) datasets. This integration was performed using a standardized bioinformatic workflow, clustering, and marker-based annotation. This dataset compromised 317, 985 quality passed cells with 45 cell types. This dataset offers a valuable source for single-cell neuroscience to understand region-specific molecular insights and their cellular diversity. The "Final_Integrated_Pons_Medulla.rds" and "Pons_Medulla_Neurons_level_3.rds" contain the normalized expression data of all identified major cell types with their subtypes.
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
List of tumor microenvironment scRNA-seq datasets included in TMExplorer.
https://dataintelo.com/privacy-and-policyhttps://dataintelo.com/privacy-and-policy
As of 2023, the global single cell multi-omics market size is valued at approximately USD 2.5 billion, with a robust projected CAGR of 20.1% forecasted to propel the market to USD 9.8 billion by 2032. This remarkable growth is driven by several key factors, including technological advancements in single-cell analysis techniques, increased funding for omics research, and a growing emphasis on personalized medicine. The market is experiencing a surge in demand as researchers and healthcare providers seek more precise and comprehensive insights into cellular behavior, disease mechanisms, and therapeutic responses. The integration of multi-omics data at a single-cell level offers unparalleled resolution and depth, enabling a transformative understanding of complex biological systems.
One of the primary growth drivers of the single cell multi-omics market is the rapid advancement of technology, particularly in sequencing and analytical tools. Innovations in microfluidics, next-generation sequencing, and enhanced bioinformatics capabilities have significantly lowered the cost and increased the efficiency of single-cell analysis. These technological advancements allow researchers to dissect the heterogeneity of cellular populations with unprecedented precision, facilitating breakthroughs in understanding disease pathology and developing targeted therapeutics. Moreover, the continuous evolution of these technologies fosters their adoption across various fields, further expanding the market's scope and application.
Another significant factor contributing to market growth is the escalating demand for personalized medicine. As the healthcare industry shifts towards more individualized treatment approaches, the need for comprehensive insights at a cellular level becomes paramount. Single cell multi-omics provides a holistic view of cellular function by integrating genomic, transcriptomic, proteomic, and metabolomic data. This integrated approach not only enhances the understanding of disease mechanisms but also aids in the development of personalized therapeutic strategies, thereby driving the adoption of single cell multi-omics in clinical settings. The ability to tailor treatments based on unique cellular profiles is expected to significantly boost market demand over the forecast period.
Additionally, increasing funding and investments in life sciences research is acting as a catalyst for the growth of the single cell multi-omics market. Governments, academic institutions, and private entities are investing heavily in omics research to unlock new scientific insights and address pressing healthcare challenges. This influx of funding is facilitating the establishment of state-of-the-art research facilities and fostering collaborations between academic institutions and industry players. The enhanced research infrastructure and collaborative efforts are expected to accelerate scientific discoveries and propel the market's expansion, as researchers strive to unravel the complexities of biological systems at a single-cell level.
From a regional perspective, North America currently dominates the single cell multi-omics market, owing to its robust research infrastructure, presence of leading biotechnology firms, and substantial government funding for genomics and precision medicine initiatives. However, the Asia Pacific region is anticipated to exhibit the highest growth rate over the forecast period, driven by increasing investments in healthcare research, the rising prevalence of chronic diseases, and the burgeoning biotechnology sector. European countries are also witnessing a growing adoption of single cell multi-omics technologies, supported by collaborative research initiatives and favorable regulatory frameworks. These regional dynamics underscore the diverse growth opportunities within the global market, as stakeholders capitalize on regional strengths and address specific healthcare needs.
The technology segment within the single cell multi-omics market is predominantly categorized into single cell genomics, single cell transcriptomics, single cell proteomics, and single cell metabolomics. Each of these sub-segments plays a crucial role in providing comprehensive insights into cellular functions and interactions. Single cell genomics, which involves the analysis of DNA at a single-cell level, has become a cornerstone technology in this market. It enables researchers to investigate genetic variations, mutations, and chromosomal aberrations with unprecedented accuracy. This technology is pivotal in advancing our understanding of genetic predisposit
https://dataintelo.com/privacy-and-policyhttps://dataintelo.com/privacy-and-policy
The global single cell genome sequencing market is poised to expand significantly, with a projected market size of $1.5 billion in 2023 and an anticipated rise to $3.8 billion by 2032, reflecting a robust compound annual growth rate (CAGR) of 10.9%. This remarkable growth is primarily driven by the increasing demand for precision medicine and personalized therapies, advancements in sequencing technologies, and a growing focus on cancer research. As researchers and clinicians aim to uncover the complexities of cellular heterogeneity, single cell genome sequencing has emerged as a pivotal tool, catalyzing its widespread adoption across various scientific disciplines and healthcare sectors.
One of the key growth factors propelling the single cell genome sequencing market is the continuous advancement in sequencing technologies, such as Next Generation Sequencing (NGS). The transition from traditional bulk sequencing methods to single cell analysis allows for a more detailed and accurate representation of cellular architecture and genetic variation. This shift has been facilitated by technological innovations that have significantly reduced the cost and time required for sequencing, making these powerful tools more accessible to a broader range of researchers and institutions. Additionally, the integration of artificial intelligence and machine learning algorithms in data analysis enhances the interpretation of complex genomic data, further accelerating the market's growth trajectory.
Another critical driver is the increasing prevalence of cancer and other chronic diseases, which has intensified the need for personalized medicine approaches. Single cell genome sequencing enables the dissection of tumor heterogeneity and the identification of rare cell populations that may contribute to disease progression or treatment resistance. This capability is particularly valuable in oncology, where understanding the genomic landscape of individual tumor cells can inform tailored therapeutic strategies and improve patient outcomes. Moreover, the rising investment in cancer genomics research by both public and private entities underscores the importance of this technology in advancing precision oncology, thereby contributing to market expansion.
The burgeoning field of immunology also presents substantial opportunities for market growth. Single cell genome sequencing facilitates comprehensive profiling of immune cell populations, aiding in the discovery of novel immune cell subsets and their functional roles in health and disease. This has significant implications for the development of new immunotherapies and vaccines, especially in the context of infectious diseases and autoimmune disorders. Furthermore, the ongoing research initiatives focused on understanding the immune response to SARS-CoV-2 and other viral pathogens have underscored the value of single cell approaches, creating additional momentum for market growth.
From a regional perspective, North America remains a dominant force in the single cell genome sequencing market, driven by substantial R&D investments, a strong presence of key market players, and a supportive regulatory environment. The region's well-established healthcare infrastructure and robust academic and research institutions further bolster market growth. Meanwhile, Asia Pacific is emerging as a lucrative market, with countries like China and India investing heavily in biotechnology research and development. The growing prevalence of chronic diseases and increasing adoption of advanced sequencing technologies in this region are expected to fuel market expansion, making it a focal point for future growth.
The single cell genome sequencing market is segmented by technology into microfluidics, next-generation sequencing (NGS), polymerase chain reaction (PCR), quantitative PCR (qPCR), and other technologies. Each of these technologies plays a pivotal role in advancing the capabilities of single cell analysis. Microfluidics, for instance, has revolutionized the field by enabling the isolation and manipulation of individual cells in a highly controlled manner. This technology allows researchers to conduct high-throughput experiments with precision, which is essential for single cell sequencing applications. The miniaturization and integration of microfluidic devices have further enhanced the efficiency of cell sorting and preparation, thereby streamlining the workflow and reducing associated costs.
Next-generation sequencing (NGS) stands out as a cornerstone technology in the singl
https://dataintelo.com/privacy-and-policyhttps://dataintelo.com/privacy-and-policy
The global single cell genomics market size was valued at approximately USD 2.5 billion in 2023, and it is projected to reach around USD 8.3 billion by 2032, growing at a compound annual growth rate (CAGR) of about 14.1% during the forecast period. This growth can be primarily attributed to the increasing demand for precision medicine, advancements in technology, and the rising incidence of chronic diseases which necessitate detailed cellular level understanding. The burgeoning fields of personalized medicine and targeted therapies are also significant drivers, as they rely heavily on the insights provided by single cell genomic analyses to tailor interventions to individual genetic profiles.
The growth of the single cell genomics market is significantly bolstered by continuous technological advancements. Innovations such as next-generation sequencing (NGS) have made it feasible to conduct comprehensive analyses of individual cell genomes at a much faster rate and reduced cost. This technological progress enables researchers to explore cell heterogeneity, improve disease categorization, and discover novel therapeutic targets. Additionally, the development of novel bioinformatics tools and software has enhanced data interpretation capabilities, making it easier to manage the massive datasets generated from single cell analyses. These advancements not only optimize the research processes but also open new avenues for clinical applications, thereby driving market growth.
Another pivotal factor contributing to market expansion is the rising prevalence of complex genetic diseases, particularly cancer. Single cell genomics provides powerful tools to unravel the complexities of cancer biology by enabling the study of cancer at the genomic level, offering insights into tumor heterogeneity and the evolution of drug resistance. As cancer remains a leading health concern globally, the demand for effective diagnostic and therapeutic solutions is on the rise, thus propelling investments into single cell genomics research. Additionally, government funding and strategic collaborations between academic institutions and biopharmaceutical companies are fostering a conducive environment for research and development, further accelerating market growth.
The growing emphasis on personalized medicine also plays a crucial role in the expansion of the single cell genomics market. Personalized medicine aims to tailor healthcare solutions according to individual genetic makeup, and single cell genomics is instrumental in achieving this objective by providing insights into cellular heterogeneity. This paradigm shift in healthcare is supported by increasing regulatory approvals and the integration of genomic data into clinical practices. As more healthcare providers adopt these precision tools, the demand for single cell genomic technologies is expected to witness substantial growth, thereby driving the market forward.
Regionally, North America is expected to maintain its dominance in the single cell genomics market due to robust healthcare infrastructure, high adoption of advanced technologies, and substantial funding for genomic research. However, Asia Pacific is anticipated to exhibit the highest growth rate during the forecast period, fueled by increasing investments in biotechnology, rising awareness about precision medicine, and the growing number of genomics research centers in countries like China and India. The increasing prevalence of chronic and genetic diseases in these regions further amplifies the demand for single cell genomics solutions.
The technology segment in the single cell genomics market is diverse, comprising sequencing, quantitative polymerase chain reaction (qPCR), microarray, multiple displacement amplification (MDA), and other techniques. Sequencing technology, particularly single-cell RNA sequencing, is at the forefront of this segment, driving significant advancements in cellular research. This technique allows for in-depth analysis of gene expression profiles at a single-cell level, offering unparalleled insights into cellular functions and disease mechanisms. The rapid advancements in sequencing technologies, coupled with decreasing costs, are making this approach more accessible and scalable, thus fostering its widespread adoption in both research and clinical settings.
qPCR is another critical technology driving the single cell genomics market. Known for its high sensitivity, specificity, and speed, qPCR is widely used for gene expression studies, enabling researchers to quantify DNA sequences and monito
https://dataintelo.com/privacy-and-policyhttps://dataintelo.com/privacy-and-policy
The single cell omics market size is projected to grow from USD 2.5 billion in 2023 to USD 12 billion by 2032, demonstrating a remarkable CAGR of 18.9% over the forecast period. One of the primary growth factors contributing to this surge is the increasing demand for personalized medicine and advanced diagnostics. Growing investments in biomedical research and technological advancements in single cell analysis are driving the market expansion.
One of the key growth factors for the single cell omics market is the rapid development and integration of technologies such as next-generation sequencing (NGS) and mass spectrometry. These technologies have enabled researchers to delve deeper into the complexities of individual cells, providing insights that were previously unattainable. The ability to analyze the genome, transcriptome, proteome, and metabolome of a single cell has opened new frontiers in understanding cellular heterogeneity, disease mechanisms, and developmental biology. The continuous refinement and cost reduction of these technologies further bolster market growth.
Another significant growth driver is the increasing prevalence of chronic diseases, such as cancer and neurological disorders, which necessitate novel diagnostic and therapeutic approaches. Single cell omics offer unparalleled precision in identifying disease-associated biomarkers and understanding the molecular underpinnings of these conditions. This precision aids in the development of targeted therapies and personalized treatment plans, thereby improving patient outcomes. Additionally, government and private sector investments in healthcare infrastructure and research initiatives are facilitating the adoption of single cell omics technologies.
Moreover, the growing emphasis on personalized medicine is propelling the demand for single cell omics. Personalized medicine relies on the ability to tailor treatment strategies to individual patients based on their unique genetic, epigenetic, and phenotypic profiles. Single cell omics provide the necessary tools to achieve this level of precision, enabling healthcare providers to deliver more effective and less invasive treatments. The integration of single cell omics in clinical settings is expected to revolutionize patient care, driving market adoption.
Regionally, North America dominates the single cell omics market due to its well-established healthcare infrastructure, significant investment in research and development, and the presence of major market players. However, Asia Pacific is anticipated to witness the highest growth rate during the forecast period. Factors such as increasing government initiatives to promote biomedical research, rising healthcare expenditure, and the growing prevalence of chronic diseases contribute to this regional growth. Additionally, the expanding biotechnology and pharmaceutical industries in countries like China and India are expected to fuel market expansion in the Asia Pacific region.
In the realm of single cell omics, various technologies play a pivotal role in driving advancements and applications. The primary technologies include single cell genomics, single cell transcriptomics, single cell proteomics, and single cell metabolomics. Each of these technologies offers unique insights into cellular functions and heterogeneity, making them indispensable tools in biomedical research.
Single cell genomics focuses on the study of the genome at the individual cell level, allowing researchers to uncover genetic variations and mutations that contribute to disease. This technology is particularly beneficial in cancer research, where understanding the genetic makeup of cancerous cells can lead to the development of targeted therapies. The advancements in NGS have significantly enhanced the sensitivity and accuracy of single cell genomics, enabling the detection of rare genetic variants that were previously undetectable.
Single cell transcriptomics examines the transcriptome of individual cells, providing insights into gene expression patterns and regulatory mechanisms. This technology is essential for understanding cellular differentiation, development, and responses to environmental stimuli. The integration of high-throughput sequencing and bioinformatics tools has revolutionized single cell transcriptomics, allowing for the comprehensive analysis of thousands of cells simultaneously. This has led to significant discoveries in fields such as immunology, neurology, and stem cell research.
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
Datasets used in the paper 'Uncertainty-aware single-cell annotation with a hierarchical reject option'. This paper uses 5 open-source datasets:
1. The Allen Mouse Brain (AMB) dataset [1]: Filtered_mouse_allen_brain_labels.csv and Filtered_mouse_allen_brain_data.csv
2. The COVID dataset [2]: CocidBALLabel.csv and CovidBALCounts.csv
3. The Azimuth PBMC dataset [3]: pbmc.multimodal.h5ad
4. from the Flyatlas [4] the Flyhead dataset: Flyatlas_Fbbt_head.csv, Flyatlas_head_10x.loom and Flyatlas_Labels_head.csv
5. from the Flyatlas [4] the Flybody dataset: Flyatlas_Fbbt_body.csv, Flyatlas_body_10x.loom and Flyatlas_Labels_body.csv
(All the credits of these datasets go to the original creators of the datasets.)
References
[1] Tasic, B. et al. (2018). Shared and distinct transcriptomic cell types across neocortical areas. Nature, 563 (7729), 72–78. https://doi.org/10.1038/s41586-018-0654-5
[2] Chan Zuckerberg Initiative Single-Cell COVID-19 Consortia et al. (2020). Single cell profiling ofCOVID-19 patients: an international data resource from multiple tissues. Medrxiv preprint. https://doi.org/10.1101/2020.11.20.20227355
[3] Stuart, T. et al. (2019). Comprehensive Integration of Single-Cell Data. Cell, 177(7), 1888–1902.e21. https://doi.org/10.1016/j.cell.2019.05.031
[4] Li, H. et al. (2022). Fly Cell Atlas: A single-nucleus transcriptomic atlas of the adult fruit fly. Science, 375(6584), eabk2432. https://doi.org/10.1126/science.abk2432
https://dataintelo.com/privacy-and-policyhttps://dataintelo.com/privacy-and-policy
The global market size for Single Cell Analysis Systems was estimated to be approximately $2.5 billion in 2023, and it is projected to grow at a compound annual growth rate (CAGR) of 15% to reach around $7.5 billion by 2032. This growth can be attributed to several factors including technological advancements, increasing investments in research and development, and the rising prevalence of chronic diseases that require precise diagnostic tools.
One of the primary growth factors driving the Single Cell Analysis System market is the advancement in technology, particularly in the field of genomics and proteomics. Innovations such as next-generation sequencing (NGS) and high-throughput screening have significantly enhanced the ability to analyze individual cells at a molecular level. These technologies provide comprehensive data on cellular heterogeneity, which is crucial for understanding complex biological processes and developing targeted therapies. Additionally, the integration of artificial intelligence (AI) and machine learning (ML) algorithms into single-cell analysis tools has further revolutionized the field, enabling more accurate and faster data analysis.
Another significant growth factor is the increasing investment in biomedical research by both government and private sectors. Numerous countries are prioritizing healthcare advancements and are thus allocating substantial funds for research and development. For instance, the National Institutes of Health (NIH) in the United States and the European Research Council (ERC) in Europe are major contributors to biomedical research funding. This financial backing not only supports the development of new single-cell analysis technologies but also facilitates large-scale research projects that utilize these systems, thereby driving market growth.
The rising prevalence of chronic diseases such as cancer, neurological disorders, and immune-related conditions is also fueling the demand for single-cell analysis systems. These diseases often require precise diagnostic tools for early detection and personalized treatment plans. Single-cell analysis systems can provide detailed insights into the cellular makeup of tumors or infected tissues, enabling healthcare professionals to devise more effective treatment strategies. This capability is particularly crucial in oncology, where understanding tumor heterogeneity can significantly impact patient outcomes.
From a regional perspective, North America currently holds the largest share of the Single Cell Analysis System market, driven by a robust healthcare infrastructure, high research activity, and substantial funding. Europe follows closely, with significant contributions from countries like Germany, the UK, and France. The Asia Pacific region is also witnessing rapid growth, primarily due to increasing healthcare investments, rising awareness about advanced diagnostic tools, and the expansion of pharmaceutical and biotechnology industries. Latin America, the Middle East, and Africa are gradually catching up, with improvements in healthcare infrastructure and increasing adoption of advanced medical technologies.
The evolution of Cell Analysis Systems has been pivotal in advancing our understanding of cellular biology. These systems provide a comprehensive suite of tools that allow researchers to delve into the complexities of cellular functions and interactions. By enabling the detailed study of individual cells, these systems have opened new avenues for research in fields such as oncology, immunology, and neurology. The ability to analyze cellular responses at a granular level has significantly contributed to the development of targeted therapies and personalized medicine. As the demand for precise diagnostic tools continues to grow, the role of Cell Analysis Systems in biomedical research and clinical applications becomes increasingly critical.
The Single Cell Analysis System market is segmented into consumables, instruments, and software. Consumables are essential for performing single-cell analyses and include reagents, microplates, and assay kits. These products are in constant demand as they are required for each analysis, leading to a significant market share. Innovations in consumable products, such as the development of more efficient and sensitive reagents, are driving their adoption. Furthermore, the recurring nature of consumable purchases contributes to a steady revenue st
https://www.datainsightsmarket.com/privacy-policyhttps://www.datainsightsmarket.com/privacy-policy
The single-cell analysis software market is experiencing robust growth, driven by the increasing adoption of single-cell technologies in research and clinical settings. The market's expansion is fueled by several key factors, including the decreasing cost of single-cell sequencing technologies, the rising demand for personalized medicine, and the growing need for a deeper understanding of complex biological systems. Advancements in algorithms and computational power are enabling the analysis of increasingly larger and more complex datasets, leading to more accurate and insightful results. Furthermore, the development of user-friendly software interfaces is making single-cell analysis more accessible to a broader range of researchers, fostering wider adoption across diverse research areas such as oncology, immunology, and neuroscience. The competitive landscape is characterized by a mix of established players and emerging companies, each offering unique software features and capabilities. This competitive environment fosters innovation and drives the development of more sophisticated and comprehensive analysis tools. Looking ahead, the market is projected to maintain a healthy Compound Annual Growth Rate (CAGR) throughout the forecast period (2025-2033), exceeding 15% annually. This growth is expected to be driven by continued technological advancements, expanding applications in drug discovery and development, and increased funding for research initiatives focusing on single-cell technologies. The market segmentation will likely see continued growth across various research areas and therapeutic applications. While challenges such as data storage and management, and the need for specialized expertise, will remain, the overall outlook for the single-cell analysis software market is positive, indicating significant future opportunities for both established and emerging players in this rapidly evolving sector. The integration of artificial intelligence and machine learning within these software platforms will further enhance their analytical capabilities and accelerate market growth.
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
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
Cardiomyocytes, pivotal for heart contractility, are categorized into atrial (aCM) and ventricular (vCM) subtypes, each playing distinct roles in modulating blood flow, electrical signal conduction, pump function, and energy metabolism. Recent advancements in single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics have enhanced our understanding of cellular heterogeneity and intercellular communication within cardiac tissues. This study integrates scRNA-seq with spatial mapping to elucidate the spatial distribution and intercellular communication of aCM and vCM, focusing on their roles in energy metabolism, pump function, and regulatory functions. We performed scRNA-seq on isolated cardiac cells, followed by data normalization, PCA, and t-SNE clustering, identifying distinct cardiomyocyte subclusters. Ligand-receptor interaction analyses were conducted to explore cellular communication networks, and annotated single-cell data were projected onto heart tissue sections using spatial transcriptomics. Our results revealed distinct spatial distributions: vCM subclusters (vCM-1, vCM-2, vCM-3) predominantly occupied ventricular regions, while aCM subclusters (aCM-1, aCM-2) were primarily located in atrial regions with an increased presence of fibroblasts near atria. Igf2-Igf2r and Vegfb-Vegfr1 mediated communications were prominent in both regions, with extensive interactions between aCM-2 and vCM subclusters. This integration of scRNA-seq and spatial transcriptomics provides a comprehensive overview of cardiac tissue organization and intercellular communication, elucidating critical roles of vCM in energy metabolism and pump function, and aCM in regulating blood flow and electrical conduction. Understanding these interactions in anatomical context enhances our grasp of cardiac function complexity and identifies new therapeutic targets for cardiac diseases.
https://dataintelo.com/privacy-and-policyhttps://dataintelo.com/privacy-and-policy
The global cell analysis market size was valued at approximately USD 17 billion in 2023 and is projected to reach USD 32 billion by 2032, growing at a compound annual growth rate (CAGR) of 7.2% during the forecast period. This substantial growth is primarily driven by advancements in cell analysis technologies, increased research and development activities in the life sciences sector, and the rising prevalence of chronic diseases that necessitate novel therapeutic interventions. Furthermore, the growing focus on personalized medicine and the increasing demand for cell-based assays in drug discovery are significant factors contributing to the expansion of the cell analysis market.
One of the major growth drivers of the cell analysis market is the technological advancements in cell analysis instruments and methodologies. Innovations such as high-content screening, flow cytometry, and next-generation sequencing have revolutionized cell biology research, enabling more precise and comprehensive analyses of cellular functions. These advancements allow researchers to gain deeper insights into cellular mechanisms, leading to the development of targeted therapies and diagnostic tools. Moreover, the integration of artificial intelligence and machine learning in data analysis is enhancing the accuracy and efficiency of cell analysis, further propelling the market growth.
The increasing investment in research and development by pharmaceutical and biotechnology companies is another crucial factor fueling the growth of the cell analysis market. With the rising demand for targeted and effective therapeutics, these companies are actively exploring cell-based assays and technologies to accelerate drug discovery and development processes. Additionally, the growing importance of cell analysis in the development of biologics and biosimilars is driving the adoption of advanced cell analysis techniques. This trend is expected to continue as companies strive to meet the evolving needs of precision medicine and improve clinical outcomes.
The growing prevalence of chronic diseases such as cancer, cardiovascular diseases, and autoimmune disorders is significantly contributing to the expansion of the cell analysis market. These conditions require detailed cellular studies to understand disease mechanisms and identify potential therapeutic targets. Consequently, there is an increasing demand for cell analysis in clinical research and diagnostics. Furthermore, the emphasis on early detection and personalized treatment approaches is encouraging the adoption of cell analysis technologies in healthcare settings, thereby boosting market growth.
Single Cell Analysis is emerging as a transformative approach in the cell analysis market, offering unprecedented insights into cellular heterogeneity and function. By enabling the examination of individual cells rather than bulk populations, researchers can uncover unique cellular behaviors and interactions that were previously obscured. This level of detail is particularly valuable in understanding complex biological processes and disease mechanisms, such as cancer progression and immune responses. The integration of advanced technologies like microfluidics and next-generation sequencing with single cell analysis is enhancing its capabilities, making it an indispensable tool in both basic research and clinical applications. As the demand for precision medicine grows, single cell analysis is poised to play a crucial role in identifying novel therapeutic targets and developing personalized treatment strategies.
From a regional perspective, North America holds the largest share of the cell analysis market, driven by the presence of major pharmaceutical and biotechnology companies, advanced healthcare infrastructure, and increased funding for research activities. The Asia Pacific region is expected to witness the highest growth rate during the forecast period, attributed to the rising healthcare expenditure, growing biotechnology industry, and increasing focus on research and development activities in countries like China and India. Europe also presents significant growth opportunities owing to the strong presence of academic and research institutions and government support for scientific research.
The cell analysis market can be segmented by product into instruments, consumables, and software. Instruments form a crucial part of the cell analysis market, encompassing a wide range of equip
The cell analysis market size was valued at USD 17.7 billion in 2021 and is projected to reach USD 28.9 billion by 2030, expanding at a CAGR of around 10% during the forecast period, 2022 – 2030. The growth of the market is attributed to the increased cancer incidence and rising healthcare business.
The study of cells separated from tissues in multicellular and unicellular organisms is referred to as cell analysis. Cellular analysis plays an important role in protein identification epigenomics, gene identification, and other life science-related areas. The study of an organism's genetic and phonotypical traits is a natural process. Cellular analysis is used to assess and quantify cell number, cell condition, cell health & viability, proliferation, as well as chemical and cell-mediated toxicity.
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
Advances in single-cell RNA sequencing (scRNA-seq) technology have recently shed light on the molecular mechanisms of the spatial and temporal changes of thousands of cells simultaneously under homeostatic and ischemic conditions. The aim of this study is to investigate whether it is possible to integrate multiple similar scRNA-seq datasets for a more comprehensive understanding of diseases. In this study, we integrated three representative scRNA-seq datasets of 27,349 non-cardiomyocytes isolated at 3 and 7 days after myocardial infarction or sham surgery. In total, seven lineages, including macrophages, fibroblasts, endothelia, and lymphocytes, were identified in this analysis with distinct dynamic and functional properties in healthy and nonhealthy hearts. Myofibroblasts and endothelia were recognized as the central hubs of cellular communication via ligand-receptor interactions. Additionally, we showed that macrophages from different origins exhibited divergent transcriptional signatures, pathways, developmental trajectories, and transcriptional regulons. It was found that myofibroblasts predominantly expand at 7 days after myocardial infarction with pro-reparative characteristics. We identified signature genes of myofibroblasts, such as Postn, Cthrc1, and Ddah1, among which Ddah1 was exclusively expressed on activated fibroblasts and exhibited concordant upregulation in bulk RNA sequencing data and in vivo and in vitro experiments. Collectively, this compendium of scRNA-seq data provides a valuable entry point for understanding the transcriptional and dynamic changes of non-cardiomyocytes in healthy and nonhealthy hearts by integrating multiple datasets.
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
The metadata table contains information such as GEO accession, author, journal, year, PMID, sequencing technology, expression score type(s), source organism, type of cancer, number of patients, tumours, cells and genes, and the database that the data was obtained from. (XLSX)
Changes in cellular metabolism contribute to the development and progression of tumors, and can render tumors vulnerable to interventions. However, studies of human cancer metabolism remain limited due to technical challenges of detecting and quantifying small molecules, the highly interconnected nature of metabolic pathways, and the lack of designated tools to analyze and integrate metabolomics with other –omics data. Our study generates the largest comprehensive metabolomics dataset on a single cancer type, and provides a significant advance in integration of metabolomics with sequencing data. Our results highlight the massive re-organization of cellular metabolism as tumors progress and acquire more aggressive features. The results of our work are made available through an interactive public data portal for cancer research community. Overall design: 10 RNA samples from human ccRCC tumors analyzed from the high glutathione cluster
https://dataintelo.com/privacy-and-policyhttps://dataintelo.com/privacy-and-policy
The global omics based clinical trials market size was valued at approximately USD 3.5 billion in 2023 and is projected to grow to around USD 8.2 billion by 2032, exhibiting a Compound Annual Growth Rate (CAGR) of 9.8% during the forecast period. The growth of this market is driven by increasing demand for personalized medicine, advancements in omics technologies, and rising prevalence of chronic diseases worldwide.
One of the primary growth factors for the omics based clinical trials market is the rising demand for personalized medicine. Personalized medicine aims to tailor medical treatment to individual characteristics, such as genetic makeup, and is significantly facilitated by omics technologies like genomics, proteomics, and metabolomics. These technologies allow for a more precise understanding of disease mechanisms and patient responses to treatments, leading to more effective and targeted therapies. As the healthcare industry continues to shift towards personalized medicine, the demand for omics based clinical trials is expected to grow substantially.
Advancements in omics technologies represent another critical driver for market growth. Innovations in high-throughput sequencing, mass spectrometry, and bioinformatics have significantly enhanced the capabilities of omics technologies. These advancements have improved the accuracy, speed, and cost-effectiveness of omics based clinical trials. For instance, next-generation sequencing (NGS) technologies have revolutionized genomics by enabling rapid and comprehensive analysis of entire genomes, which is invaluable for identifying genetic variations associated with diseases. Such technological advancements are anticipated to propel the market forward.
The rising prevalence of chronic diseases, such as cancer, cardiovascular diseases, and neurological disorders, is also a significant factor fueling market expansion. Chronic diseases require long-term management and often entail complex treatment regimens that can benefit from the insights provided by omics technologies. For example, cancer treatment can be optimized through genomic profiling of tumors, allowing for the selection of the most effective therapies. As the global burden of chronic diseases continues to rise, the need for omics based clinical trials is expected to increase, driving market growth.
The integration of Human Single Cell Multi Omics is becoming increasingly pivotal in the landscape of clinical trials. This approach allows researchers to dissect the complex biological systems at a single-cell level, providing unprecedented insights into cellular heterogeneity and molecular dynamics. By analyzing individual cells, scientists can better understand the nuances of disease progression and treatment responses, which are often masked in bulk cell analyses. This granular level of detail is particularly beneficial in identifying rare cell populations that may play critical roles in disease mechanisms or therapeutic resistance. As the field of omics continues to evolve, Human Single Cell Multi Omics is expected to drive significant advancements in personalized medicine, offering more precise and effective treatment strategies tailored to individual patient profiles.
From a regional perspective, the North American market is poised to dominate the omics based clinical trials market. This dominance can be attributed to the region's well-established healthcare infrastructure, significant investment in research and development, and the presence of several leading pharmaceutical and biotechnology companies. Additionally, favorable regulatory policies and a strong focus on innovation further support market growth in this region. However, the Asia Pacific region is expected to experience the fastest growth during the forecast period, driven by increasing healthcare expenditure, growing awareness about personalized medicine, and rising investments in life sciences research.
The omics based clinical trials market is segmented by technology into genomics, proteomics, metabolomics, transcriptomics, and others. Genomics, which involves the study of an organism's complete set of DNA, is one of the most prominent segments. Advances in genomic technologies, particularly next-generation sequencing (NGS), have revolutionized the field by enabling rapid and comprehensive analysis of genetic information. This has led to significant improvements in understanding genetic p
https://spdx.org/licenses/CC0-1.0.htmlhttps://spdx.org/licenses/CC0-1.0.html
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, including endothelial subtypes distinguished by vessel-type of origin, fibro/adipogenic progenitors defined by functional roles, and many distinct immune populations. The representation of different experimental conditions and the depth of transcriptome coverage enabled robust profiling of sparsely expressed genes. We built a densely sampled transcriptomic model of myogenesis, from stem cell quiescence to myofiber maturation and identified rare, transitional states of progenitor commitment and fusion that are poorly represented in individual datasets. We performed spatial RNA sequencing of mouse muscle at three time points after injury and used the integrated dataset as a reference to achieve a high-resolution, local deconvolution of cell subtypes. We also used the integrated dataset to explore ligand-receptor co-expression patterns and identify dynamic cell-cell interactions in muscle injury response. We provide a public web tool to enable interactive exploration and visualization of the data. Our work supports the utility of large-scale integration of single-cell transcriptomic data as a tool for biological discovery.
Methods Mice. The Cornell University Institutional Animal Care and Use Committee (IACUC) approved all animal protocols, and experiments were performed in compliance with its institutional guidelines. Adult C57BL/6J mice (mus musculus) were obtained from Jackson Laboratories (#000664; Bar Harbor, ME) and were used at 4-7 months of age. Aged C57BL/6J mice were obtained from the National Institute of Aging (NIA) Rodent Aging Colony and were used at 20 months of age. For new scRNAseq experiments, female mice were used in each experiment.
Mouse injuries and single-cell isolation. To induce muscle injury, both tibialis anterior (TA) muscles of old (20 months) C57BL/6J mice were injected with 10 µl of notexin (10 µg/ml; Latoxan; France). At 0, 1, 2, 3.5, 5, or 7 days post-injury (dpi), mice were sacrificed and TA muscles were collected and processed independently to generate single-cell suspensions. Muscles were digested with 8 mg/ml Collagenase D (Roche; Switzerland) and 10 U/ml Dispase II (Roche; Switzerland), followed by manual dissociation to generate cell suspensions. Cell suspensions were sequentially filtered through 100 and 40 μm filters (Corning Cellgro #431752 and #431750) to remove debris. Erythrocytes were removed through incubation in erythrocyte lysis buffer (IBI Scientific #89135-030).
Single-cell RNA-sequencing library preparation. After digestion, single-cell suspensions were washed and resuspended in 0.04% BSA in PBS at a concentration of 106 cells/ml. Cells were counted manually with a hemocytometer to determine their concentration. Single-cell RNA-sequencing libraries were prepared using the Chromium Single Cell 3’ reagent kit v3 (10x Genomics, PN-1000075; Pleasanton, CA) following the manufacturer’s protocol. Cells were diluted into the Chromium Single Cell A Chip to yield a recovery of 6,000 single-cell transcriptomes. After preparation, libraries were sequenced using on a NextSeq 500 (Illumina; San Diego, CA) using 75 cycle high output kits (Index 1 = 8, Read 1 = 26, and Read 2 = 58). Details on estimated sequencing saturation and the number of reads per sample are shown in Sup. Data 1.
Spatial RNA sequencing library preparation. Tibialis anterior muscles of adult (5 mo) C57BL6/J mice were injected with 10µl notexin (10 µg/ml) at 2, 5, and 7 days prior to collection. Upon collection, tibialis anterior muscles were isolated, embedded in OCT, and frozen fresh in liquid nitrogen. Spatially tagged cDNA libraries were built using the Visium Spatial Gene Expression 3’ Library Construction v1 Kit (10x Genomics, PN-1000187; Pleasanton, CA) (Fig. S7). Optimal tissue permeabilization time for 10 µm thick sections was found to be 15 minutes using the 10x Genomics Visium Tissue Optimization Kit (PN-1000193). H&E stained tissue sections were imaged using Zeiss PALM MicroBeam laser capture microdissection system and the images were stitched and processed using Fiji ImageJ software. cDNA libraries were sequenced on an Illumina NextSeq 500 using 150 cycle high output kits (Read 1=28bp, Read 2=120bp, Index 1=10bp, and Index 2=10bp). Frames around the capture area on the Visium slide were aligned manually and spots covering the tissue were selected using Loop Browser v4.0.0 software (10x Genomics). Sequencing data was then aligned to the mouse reference genome (mm10) using the spaceranger v1.0.0 pipeline to generate a feature-by-spot-barcode expression matrix (10x Genomics).
Download and alignment of single-cell RNA sequencing data. For all samples available via SRA, parallel-fastq-dump (github.com/rvalieris/parallel-fastq-dump) was used to download raw .fastq files. Samples which were only available as .bam files were converted to .fastq format using bamtofastq from 10x Genomics (github.com/10XGenomics/bamtofastq). Raw reads were aligned to the mm10 reference using cellranger (v3.1.0).
Preprocessing and batch correction of single-cell RNA sequencing datasets. First, ambient RNA signal was removed using the default SoupX (v1.4.5) workflow (autoEstCounts and adjustCounts; github.com/constantAmateur/SoupX). Samples were then preprocessed using the standard Seurat (v3.2.1) workflow (NormalizeData, ScaleData, FindVariableFeatures, RunPCA, FindNeighbors, FindClusters, and RunUMAP; github.com/satijalab/seurat). Cells with fewer than 750 features, fewer than 1000 transcripts, or more than 30% of unique transcripts derived from mitochondrial genes were removed. After preprocessing, DoubletFinder (v2.0) was used to identify putative doublets in each dataset, individually. BCmvn optimization was used for PK parameterization. Estimated doublet rates were computed by fitting the total number of cells after quality filtering to a linear regression of the expected doublet rates published in the 10x Chromium handbook. Estimated homotypic doublet rates were also accounted for using the modelHomotypic function. The default PN value (0.25) was used. Putative doublets were then removed from each individual dataset. After preprocessing and quality filtering, we merged the datasets and performed batch-correction with three tools, independently- Harmony (github.com/immunogenomics/harmony) (v1.0), Scanorama (github.com/brianhie/scanorama) (v1.3), and BBKNN (github.com/Teichlab/bbknn) (v1.3.12). We then used Seurat to process the integrated data. After initial integration, we removed the noisy cluster and re-integrated the data using each of the three batch-correction tools.
Cell type annotation. Cell types were determined for each integration method independently. For Harmony and Scanorama, dimensions accounting for 95% of the total variance were used to generate SNN graphs (Seurat::FindNeighbors). Louvain clustering was then performed on the output graphs (including the corrected graph output by BBKNN) using Seurat::FindClusters. A clustering resolution of 1.2 was used for Harmony (25 initial clusters), BBKNN (28 initial clusters), and Scanorama (38 initial clusters). Cell types were determined based on expression of canonical genes (Fig. S3). Clusters which had similar canonical marker gene expression patterns were merged.
Pseudotime workflow. Cells were subset based on the consensus cell types between all three integration methods. Harmony embedding values from the dimensions accounting for 95% of the total variance were used for further dimensional reduction with PHATE, using phateR (v1.0.4) (github.com/KrishnaswamyLab/phateR).
Deconvolution of spatial RNA sequencing spots. Spot deconvolution was performed using the deconvolution module in BayesPrism (previously known as “Tumor microEnvironment Deconvolution”, TED, v1.0; github.com/Danko-Lab/TED). First, myogenic cells were re-labeled, according to binning along the first PHATE dimension, as “Quiescent MuSCs” (bins 4-5), “Activated MuSCs” (bins 6-7), “Committed Myoblasts” (bins 8-10), and “Fusing Myoctes” (bins 11-18). Culture-associated muscle stem cells were ignored and myonuclei labels were retained as “Myonuclei (Type IIb)” and “Myonuclei (Type IIx)”. Next, highly and differentially expressed genes across the 25 groups of cells were identified with differential gene expression analysis using Seurat (FindAllMarkers, using Wilcoxon Rank Sum Test; results in Sup. Data 2). The resulting genes were filtered based on average log2-fold change (avg_logFC > 1) and the percentage of cells within the cluster which express each gene (pct.expressed > 0.5), yielding 1,069 genes. Mitochondrial and ribosomal protein genes were also removed from this list, in line with recommendations in the BayesPrism vignette. For each of the cell types, mean raw counts were calculated across the 1,069 genes to generate a gene expression profile for BayesPrism. Raw counts for each spot were then passed to the run.Ted function, using