91 datasets found
  1. f

    Table_1_Read Mapping and Transcript Assembly: A Scalable and High-Throughput...

    • frontiersin.figshare.com
    xlsx
    Updated Jun 1, 2023
    + more versions
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Sateesh Peri; Sarah Roberts; Isabella R. Kreko; Lauren B. McHan; Alexandra Naron; Archana Ram; Rebecca L. Murphy; Eric Lyons; Brian D. Gregory; Upendra K. Devisetty; Andrew D. L. Nelson (2023). Table_1_Read Mapping and Transcript Assembly: A Scalable and High-Throughput Workflow for the Processing and Analysis of Ribonucleic Acid Sequencing Data.xlsx [Dataset]. http://doi.org/10.3389/fgene.2019.01361.s002
    Explore at:
    xlsxAvailable download formats
    Dataset updated
    Jun 1, 2023
    Dataset provided by
    Frontiers
    Authors
    Sateesh Peri; Sarah Roberts; Isabella R. Kreko; Lauren B. McHan; Alexandra Naron; Archana Ram; Rebecca L. Murphy; Eric Lyons; Brian D. Gregory; Upendra K. Devisetty; Andrew D. L. Nelson
    License

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

    Description

    Next-generation RNA-sequencing is an incredibly powerful means of generating a snapshot of the transcriptomic state within a cell, tissue, or whole organism. As the questions addressed by RNA-sequencing (RNA-seq) become both more complex and greater in number, there is a need to simplify RNA-seq processing workflows, make them more efficient and interoperable, and capable of handling both large and small datasets. This is especially important for researchers who need to process hundreds to tens of thousands of RNA-seq datasets. To address these needs, we have developed a scalable, user-friendly, and easily deployable analysis suite called RMTA (Read Mapping, Transcript Assembly). RMTA can easily process thousands of RNA-seq datasets with features that include automated read quality analysis, filters for lowly expressed transcripts, and read counting for differential expression analysis. RMTA is containerized using Docker for easy deployment within any compute environment [cloud, local, or high-performance computing (HPC)] and is available as two apps in CyVerse's Discovery Environment, one for normal use and one specifically designed for introducing undergraduates and high school to RNA-seq analysis. For extremely large datasets (tens of thousands of FASTq files) we developed a high-throughput, scalable, and parallelized version of RMTA optimized for launching on the Open Science Grid (OSG) from within the Discovery Environment. OSG-RMTA allows users to utilize the Discovery Environment for data management, parallelization, and submitting jobs to OSG, and finally, employ the OSG for distributed, high throughput computing. Alternatively, OSG-RMTA can be run directly on the OSG through the command line. RMTA is designed to be useful for data scientists, of any skill level, interested in rapidly and reproducibly analyzing their large RNA-seq data sets.

  2. CWL run of RNA-seq Analysis Workflow (CWLProv 0.5.0 Research Object)

    • zenodo.org
    • data.niaid.nih.gov
    • +2more
    bin, zip
    Updated Jan 24, 2020
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Farah Zaib Khan; Farah Zaib Khan; Stian Soiland-Reyes; Stian Soiland-Reyes (2020). CWL run of RNA-seq Analysis Workflow (CWLProv 0.5.0 Research Object) [Dataset]. http://doi.org/10.17632/xnwncxpw42.1
    Explore at:
    zip, binAvailable download formats
    Dataset updated
    Jan 24, 2020
    Dataset provided by
    Zenodohttp://zenodo.org/
    Authors
    Farah Zaib Khan; Farah Zaib Khan; Stian Soiland-Reyes; Stian Soiland-Reyes
    License

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

    Description

    This workflow adapts the approach and parameter settings of Trans-Omics for precision Medicine (TOPMed). The RNA-seq pipeline originated from the Broad Institute. There are in total five steps in the workflow starting from:

    1. Read alignment using STAR which produces aligned BAM files including the Genome BAM and Transcriptome BAM.
    2. The Genome BAM file is processed using Picard MarkDuplicates. producing an updated BAM file containing information on duplicate reads (such reads can indicate biased interpretation).
    3. SAMtools index is then employed to generate an index for the BAM file, in preparation for the next step.
    4. The indexed BAM file is processed further with RNA-SeQC which takes the BAM file, human genome reference sequence and Gene Transfer Format (GTF) file as inputs to generate transcriptome-level expression quantifications and standard quality control metrics.
    5. In parallel with transcript quantification, isoform expression levels are quantified by RSEM. This step depends only on the output of the STAR tool, and additional RSEM reference sequences.

    For testing and analysis, the workflow author provided example data created by down-sampling the read files of a TOPMed public access data. Chromosome 12 was extracted from the Homo Sapien Assembly 38 reference sequence and provided by the workflow authors. The required GTF and RSEM reference data files are also provided. The workflow is well-documented with a detailed set of instructions of the steps performed to down-sample the data are also provided for transparency. The availability of example input data, use of containerization for underlying software and detailed documentation are important factors in choosing this specific CWL workflow for CWLProv evaluation.

    This dataset folder is a CWLProv Research Object that captures the Common Workflow Language execution provenance, see https://w3id.org/cwl/prov/0.5.0 or use https://pypi.org/project/cwl

    Steps to reproduce

    To build the research object again, use Python 3 on macOS. Built with:

    • Processor 2.8GHz Intel Core i7
    • Memory: 16GB
    • OS: macOS High Sierra, Version 10.13.3
    • Storage: 250GB
    1. Install cwltool

      pip3 install cwltool==1.0.20180912090223
    2. Install git lfs
      The data download with the git repository requires the installation of Git lfs:
      https://www.atlassian.com/git/tutorials/git-lfs#installing-git-lfs

    3. Get the data and make the analysis environment ready:

      git clone https://github.com/FarahZKhan/cwl_workflows.git
      cd cwl_workflows/
      git checkout CWLProvTesting
      ./topmed-workflows/TOPMed_RNAseq_pipeline/input-examples/download_examples.sh
    4. Run the following commands to create the CWLProv Research Object:

      cwltool --provenance rnaseqwf_0.6.0_linux --tmp-outdir-prefix=/CWLProv_workflow_testing/intermediate_temp/temp --tmpdir-prefix=/CWLProv_workflow_testing/intermediate_temp/temp topmed-workflows/TOPMed_RNAseq_pipeline/rnaseq_pipeline_fastq.cwl topmed-workflows/TOPMed_RNAseq_pipeline/input-examples/Dockstore.json
      
      zip -r rnaseqwf_0.5.0_mac.zip rnaseqwf_0.5.0_mac
      sha256sum rnaseqwf_0.5.0_mac.zip > rnaseqwf_0.5.0_mac_mac.zip.sha256

    The https://github.com/FarahZKhan/cwl_workflows repository is a frozen snapshot from https://github.com/heliumdatacommons/TOPMed_RNAseq_CWL commit 027e8af41b906173aafdb791351fb29efc044120

  3. f

    Table_3_Read Mapping and Transcript Assembly: A Scalable and High-Throughput...

    • frontiersin.figshare.com
    • figshare.com
    docx
    Updated Jun 1, 2023
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Sateesh Peri; Sarah Roberts; Isabella R. Kreko; Lauren B. McHan; Alexandra Naron; Archana Ram; Rebecca L. Murphy; Eric Lyons; Brian D. Gregory; Upendra K. Devisetty; Andrew D. L. Nelson (2023). Table_3_Read Mapping and Transcript Assembly: A Scalable and High-Throughput Workflow for the Processing and Analysis of Ribonucleic Acid Sequencing Data.docx [Dataset]. http://doi.org/10.3389/fgene.2019.01361.s004
    Explore at:
    docxAvailable download formats
    Dataset updated
    Jun 1, 2023
    Dataset provided by
    Frontiers
    Authors
    Sateesh Peri; Sarah Roberts; Isabella R. Kreko; Lauren B. McHan; Alexandra Naron; Archana Ram; Rebecca L. Murphy; Eric Lyons; Brian D. Gregory; Upendra K. Devisetty; Andrew D. L. Nelson
    License

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

    Description

    Next-generation RNA-sequencing is an incredibly powerful means of generating a snapshot of the transcriptomic state within a cell, tissue, or whole organism. As the questions addressed by RNA-sequencing (RNA-seq) become both more complex and greater in number, there is a need to simplify RNA-seq processing workflows, make them more efficient and interoperable, and capable of handling both large and small datasets. This is especially important for researchers who need to process hundreds to tens of thousands of RNA-seq datasets. To address these needs, we have developed a scalable, user-friendly, and easily deployable analysis suite called RMTA (Read Mapping, Transcript Assembly). RMTA can easily process thousands of RNA-seq datasets with features that include automated read quality analysis, filters for lowly expressed transcripts, and read counting for differential expression analysis. RMTA is containerized using Docker for easy deployment within any compute environment [cloud, local, or high-performance computing (HPC)] and is available as two apps in CyVerse's Discovery Environment, one for normal use and one specifically designed for introducing undergraduates and high school to RNA-seq analysis. For extremely large datasets (tens of thousands of FASTq files) we developed a high-throughput, scalable, and parallelized version of RMTA optimized for launching on the Open Science Grid (OSG) from within the Discovery Environment. OSG-RMTA allows users to utilize the Discovery Environment for data management, parallelization, and submitting jobs to OSG, and finally, employ the OSG for distributed, high throughput computing. Alternatively, OSG-RMTA can be run directly on the OSG through the command line. RMTA is designed to be useful for data scientists, of any skill level, interested in rapidly and reproducibly analyzing their large RNA-seq data sets.

  4. Z

    Data from: The output and the log files from RNA-Seq workflow benchmark for...

    • data.niaid.nih.gov
    Updated Jan 24, 2020
    + more versions
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Ohta, Tazro (2020). The output and the log files from RNA-Seq workflow benchmark for CWL-metrics manuscript [Dataset]. https://data.niaid.nih.gov/resources?id=zenodo_2586546
    Explore at:
    Dataset updated
    Jan 24, 2020
    Dataset authored and provided by
    Ohta, Tazro
    License

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

    Description

    The output files and log files generated by the workflow executions for RNA-Seq workflow benchmark by CWL-metrics, from the manuscript "Accumulating computational resource usage of genomic data analysis workflow to optimize cloud computing instance selection" (https://doi.org/10.1101/456756).

  5. Ngs-Based Rna-Seq Market Analysis North America, Europe, Asia, Rest of World...

    • technavio.com
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Technavio, Ngs-Based Rna-Seq Market Analysis North America, Europe, Asia, Rest of World (ROW) - US, UK, Germany, Singapore, China - Size and Forecast 2024-2028 [Dataset]. https://www.technavio.com/report/ngs-based-rna-seq-market-analysis
    Explore at:
    Dataset provided by
    TechNavio
    Authors
    Technavio
    Time period covered
    2021 - 2025
    Area covered
    United States, Global
    Description

    Snapshot img

    NGS-Based Rna-Seq Market Size 2024-2028

    The NGS-based RNA-seq market size is forecast to increase by USD 6.66 billion, at a CAGR of 20.52% between 2023 and 2028.

    The market is witnessing significant growth, driven by the increased adoption of next-generation sequencing (NGS) methods for RNA-Seq analysis. The advanced capabilities of NGS techniques, such as high-throughput, cost-effectiveness, and improved accuracy, have made them the preferred choice for researchers and clinicians in various fields, including genomics, transcriptomics, and personalized medicine. However, the market faces challenges, primarily from the lack of clinical validation on direct-to-consumer genetic tests. As the use of NGS technology in consumer applications expands, ensuring the accuracy and reliability of results becomes crucial.
    The absence of standardized protocols and regulatory oversight in this area poses a significant challenge to market growth and trust. Companies seeking to capitalize on market opportunities must focus on addressing these challenges through collaborations, partnerships, and investments in research and development to ensure the clinical validity and reliability of their NGS-based RNA-Seq offerings.
    

    What will be the Size of the NGS-based RNA-Seq market during the forecast period?

    Explore in-depth regional segment analysis with market size data - historical 2018-2022 and forecasts 2024-2028 - in the full report.
    Request Free Sample

    The market continues to evolve, driven by advancements in NGS technology and its applications across various sectors. Spatial transcriptomics, a novel approach to studying gene expression in its spatial context, is gaining traction in disease research and precision medicine. Splice junction detection, a critical component of RNA-seq data analysis, enhances the accuracy of gene expression profiling and differential gene expression studies. Cloud computing plays a pivotal role in handling the massive amounts of data generated by NGS platforms, enabling real-time data analysis and storage. Enrichment analysis, gene ontology, and pathway analysis facilitate the interpretation of RNA-seq data, while data normalization and quality control ensure the reliability of results.

    Precision medicine and personalized therapy are key applications of RNA-seq, with single-cell RNA-seq offering unprecedented insights into the complexities of gene expression at the single-cell level. Read alignment and variant calling are essential steps in RNA-seq data analysis, while bioinformatics pipelines and RNA-seq software streamline the process. NGS technology is revolutionizing drug discovery by enabling the identification of biomarkers and gene fusion detection in various diseases, including cancer and neurological disorders. RNA-seq is also finding applications in infectious diseases, microbiome analysis, environmental monitoring, agricultural genomics, and forensic science. Sequencing costs are decreasing, making RNA-seq more accessible to researchers and clinicians.

    The ongoing development of sequencing platforms, library preparation, and sample preparation kits continues to drive innovation in the field. The dynamic nature of the market ensures that it remains a vibrant and evolving field, with ongoing research and development in areas such as data visualization, clinical trials, and sequencing depth.

    How is this NGS-based RNA-Seq industry segmented?

    The NGS-based RNA-seq industry research report provides comprehensive data (region-wise segment analysis), with forecasts and estimates in 'USD million' for the period 2024-2028, as well as historical data from 2018-2022 for the following segments.

    End-user
    
      Acamedic and research centers
      Clinical research
      Pharma companies
      Hospitals
    
    
    Technology
    
      Sequencing by synthesis
      Ion semiconductor sequencing
      Single-molecule real-time sequencing
      Others
    
    
    Geography
    
      North America
    
        US
    
    
      Europe
    
        Germany
        UK
    
    
      APAC
    
        China
        Singapore
    
    
      Rest of World (ROW)
    

    .

    By End-user Insights

    The acamedic and research centers segment is estimated to witness significant growth during the forecast period.

    The global next-generation sequencing (NGS) market for RNA sequencing (RNA-Seq) is primarily driven by academic and research institutions, including those from universities, research institutes, government entities, biotechnology organizations, and pharmaceutical companies. These institutions utilize NGS technology for various research applications, such as whole-genome sequencing, epigenetics, and emerging fields like agrigenomics and animal research, to enhance crop yield and nutritional composition. NGS-based RNA-Seq plays a pivotal role in translational research, with significant investments from both private and public organizations fueling its growth. The technology is instrumental in disease research, enabling the identification

  6. Z

    Results of "Curare and GenExVis: A versatile toolkit for analyzing and...

    • data.niaid.nih.gov
    • zenodo.org
    Updated Apr 12, 2024
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Diedrich, Sonja (2024). Results of "Curare and GenExVis: A versatile toolkit for analyzing and visualizing RNA-Seq data" [Dataset]. https://data.niaid.nih.gov/resources?id=zenodo_10362479
    Explore at:
    Dataset updated
    Apr 12, 2024
    Dataset provided by
    Pfister, Max
    Diedrich, Sonja
    Jaenicke, Sebastian
    Blumenkamp, Patrick
    Brinkrolf, Karina
    Goesmann, Alexander
    Description

    Even though high-throughput transcriptome sequencing is routinely performed in many laboratories, computational analysis of such data remains a cumbersome process often executed manually, hence error-prone and lacking reproducibility. For corresponding data processing, we introduce Curare, an easy-to-use yet versatile workflow builder for analyzing high-throughput RNA-Seq data focusing on differential gene expression experiments. Data analysis with Curare is customizable and subdivided into preprocessing, quality control, mapping, and downstream analysis stages, providing multiple options for each step while ensuring the reproducibility of the workflow. For a fast and straightforward exploration and visualization of differential gene expression results, we provide the gene expression visualizer software GenExVis. GenExVis can create various charts and tables from simple gene expression tables and DESeq2 results without the requirement to upload data or install software packages.

  7. Z

    Supporting data for "Software pipelines for RNA-Seq, ChIP-Seq and Germline...

    • data.niaid.nih.gov
    • zenodo.org
    Updated Sep 27, 2023
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Nikolaos Pechlivanis (2023). Supporting data for "Software pipelines for RNA-Seq, ChIP-Seq and Germline Variant calling analyses in Common Workflow Language (CWL)" [Dataset]. https://data.niaid.nih.gov/resources?id=zenodo_8116555
    Explore at:
    Dataset updated
    Sep 27, 2023
    Dataset provided by
    Nikolaos Pechlivanis
    Konstantinos Kyritsis
    Fotis Psomopoulos
    License

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

    Description

    Datasets produced during the validation of CWL-based pipelines, designed for the analysis of data from RNA-Seq, ChIP-Seq and germline variant calling experiments. Specifically, the workflows were tested using publicly available High-throughput (HTS) data from published studies on Chronic Lymphocytic Leukemia (CLL) (accession numbers: E-MTAB-6962, GSE115772) and Genome in a Bottle (GIAB) project samples (accession numbers: SRR6794144, SRR22476789, SRR22476790, SRR22476791).

    The supporting data include:

    Differential transcript and gene expression results produced during the analysis with the CWL-based RNA-Seq pipeline

    Bigwig and narrowPeak files, differential binding results, table of consensus peaks and read counts of EZH2 and H3K27me3, produced during the analysis with the CWL-based ChIP-Seq pipeline

    VCF files containing the detected and filtered variants, along with the respective hap.py () results regarding comparisons against the GIAB golden standard truth sets for both CWL-based germline variant calling pipelines

  8. o

    Transcription profiling by high throughput sequencing of two subspecies of...

    • omicsdi.org
    xml
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Miguel Ramos,Joao Coito,Margarida Rocheta,Helena Silva,Manuela Costa,Jorge Cunha, Transcription profiling by high throughput sequencing of two subspecies of grapevine at four flower developmental stages [Dataset]. https://www.omicsdi.org/dataset/arrayexpress-repository/E-GEOD-56844
    Explore at:
    xmlAvailable download formats
    Authors
    Miguel Ramos,Joao Coito,Margarida Rocheta,Helena Silva,Manuela Costa,Jorge Cunha
    Variables measured
    Transcriptomics,Multiomics
    Description

    Purpose: Next-generation sequencing (NGS) has revolutionized systems-based analysis of cellular pathways. The goals of this study are to compare NGS-derived flower development transcriptome profiling (RNA-seq) of two subspecies Methods: Flower mRNA profiles of wild-type (WT) four developmental stages and the same stages of Vitis vinifera subp vinifera were generated by deep sequencing using Illumina. Initial quality assessment was based on data passing the Illumina Chastity filtering. Subsequently, reads containing adapters and/or PhiX control signal were removed using an in-house filtering protocol. The second quality assessment was based on the remaining reads using the FASTQC quality control tool version 0.10.0. qRT–PCR validation was performed using EvaGreen assays. Results: Using an optimized data analysis workflow, we mapped about 13 to 19 million sequence reads per Vitis sample, 50 bp in length equivalent to 1.5 Gb of total sequence data by each sample. The exception was male stage G (M_G) were only 7 to 8 million sequence reads were obtained. Five genes (VvTFL1, VvLFY, VvAP1, Vv AP3, VvPI), related to flowering development, were used to validate RNA-Seq data and to test for data reproducibility through qRT–PCR. The coefficient of correlation (r) obtained between the log2 of RPKM (RNA-Seq) versus log2 of mRNA average number (RT-qPCR), varied from ≈ 0.97 (VvTLF) to ≈ 0.73 (VvPI) indicating a good correlation between both techniques and thus validating our RNA-Seq results. Conclusions: Our study represents the first detailed transcriptome analysis of four Vitis flower developmental stages, with the same individual, in three genders, generated by RNA-seq technology. The optimized data analysis workflows reported here should provide a framework for comparative investigations of expression profiles. Our results show that NGS offers a comprehensive and accurate quantitative and qualitative evaluation of mRNA contentper developmental stage. We conclude that RNA-seq based transcriptome characterization would expedite genetic network analyses and permit the dissection of complex biologic functions. Flowering mRNA profiles of four developmental stages of Vitis wild type (WT) and the domesticated Vitis were generated by deep sequencing using Illumina HiSeq 2500.

  9. Simulated RNA-seq data

    • zenodo.org
    • explore.openaire.eu
    application/gzip, csv
    Updated Jan 26, 2021
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Taavi Päll; Ülo Maiväli; Tanel Tenson; Hannes Luidalepp; Taavi Päll; Ülo Maiväli; Tanel Tenson; Hannes Luidalepp (2021). Simulated RNA-seq data [Dataset]. http://doi.org/10.5281/zenodo.4463804
    Explore at:
    application/gzip, csvAvailable download formats
    Dataset updated
    Jan 26, 2021
    Dataset provided by
    Zenodohttp://zenodo.org/
    Authors
    Taavi Päll; Ülo Maiväli; Tanel Tenson; Hannes Luidalepp; Taavi Päll; Ülo Maiväli; Tanel Tenson; Hannes Luidalepp
    License

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

    Description

    Simulated RNA-seq data shows that histograms from p value sets with around one hundred true effects out of 20,000 features can be classified as 'uniform'. RNA-seq data was simulated with polyester R package (Frazee, 2015) on 20,000 transcripts from human transcriptome using grid of 3, 6, and 10 replicates and 100, 200, 400, and 800 effects for two groups. Fold changes were set to 0.5 and 2. Differential expression was assessed using DESeq2 R package (Love, 2014) using default settings and group 1 versus group 2 contrast. Effects denotes in facet labels the number of true effects and N denotes number of replicates. Red line denotes QC threshold used for dividing p histograms into discrete classes. Workflow and code used to run this simulation is available on rstats-tartu/simulate-rnaseq.

    Files

    • de_simulation_results.csv -- merged and processed DE analysis results of simulated data.
    • simulate-reads-2021-01-25.tar.gz -- raw DE analysis results on 20,000 transcripts from human transcriptome using grid of 3, 6, and 10 replicates and 100, 200, 400, and 800 effects for two groups. Fold changes were set to 0.5, 1, and 2. Differential expression was assessed using DESeq2 with default settings.
    • simulate-rnaseq.tar.gz -- snakemake workflow and input fasta file to simulate RNA-seq data with polyester and analyse results with DESeq2. Adjust settings in config.yaml to customise simulation. Includes software to run workflow on Linux, given that Conda and snakemake are installed.

    The simulate-rnaseq.tar.gz archive can be re-executed on a vanilla machine that only has Conda and Snakemake installed via:

    tar -xf simulate-rnaseq.tar.gz
    snakemake --use-conda -n

  10. w

    Global Rna Sequencing Technologies Market Research Report: By Technology...

    • wiseguyreports.com
    Updated Sep 12, 2024
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    wWiseguy Research Consultants Pvt Ltd (2024). Global Rna Sequencing Technologies Market Research Report: By Technology (Next-Generation Sequencing (NGS), Third-Generation Sequencing (TGS), Single-Cell Sequencing, Long-Read Sequencing, Direct RNA Sequencing), By Application Type (Transcriptome profiling, Gene expression profiling, Genome-wide association studies, Cancer research, Infectious disease diagnosis, Drug discovery and development), By Sample Type (RNA from cells, RNA from tissues, RNA from body fluids, RNA from environmental samples), By Workflow (Sample preparation, Library preparation, Sequencing, Data analysis and interpretation), By End User (Academic and research institutions, Pharmaceutical and biotechnology companies, Diagnostic laboratories, Clinical research organizations, Government agencies and public health organizations) and By Regional (North America, Europe, South America, Asia Pacific, Middle East and Africa) - Forecast to 2032. [Dataset]. https://www.wiseguyreports.com/reports/rna-sequencing-technologies-market
    Explore at:
    Dataset updated
    Sep 12, 2024
    Dataset authored and provided by
    wWiseguy Research Consultants Pvt Ltd
    License

    https://www.wiseguyreports.com/pages/privacy-policyhttps://www.wiseguyreports.com/pages/privacy-policy

    Time period covered
    Jan 9, 2024
    Area covered
    Global
    Description
    BASE YEAR2024
    HISTORICAL DATA2019 - 2024
    REPORT COVERAGERevenue Forecast, Competitive Landscape, Growth Factors, and Trends
    MARKET SIZE 202324.62(USD Billion)
    MARKET SIZE 202432.42(USD Billion)
    MARKET SIZE 2032292.42(USD Billion)
    SEGMENTS COVEREDTechnology, Application Type, Sample Type, Workflow, End User, Regional
    COUNTRIES COVEREDNorth America, Europe, APAC, South America, MEA
    KEY MARKET DYNAMICSRising demand for RNA sequencing Advancements in technology Growing adoption of personalized medicine Increasing awareness of RNAs role in disease Government funding and initiatives
    MARKET FORECAST UNITSUSD Billion
    KEY COMPANIES PROFILEDThermo Fisher Scientific, Inc., BGI Group, Oxford Nanopore Technologies, Ltd., Genea Biomarkers, Roche Holding AG, Illumina, Inc., 10x Genomics, Inc., MGI Tech Co., Ltd., Novogene Corporation Limited, BioRad Laboratories, Inc., Agilent Technologies, Inc., PerkinElmer, Inc., QIAGEN, Pacific Biosciences of California, Inc., NanoString Technologies, Inc.
    MARKET FORECAST PERIOD2025 - 2032
    KEY MARKET OPPORTUNITIESNextgeneration sequencing Precision medicine Singlecell RNA sequencing Liquid biopsy Spatial transcriptomics
    COMPOUND ANNUAL GROWTH RATE (CAGR) 31.64% (2025 - 2032)
  11. E

    Data from: RaScALL: Rapid (Ra) screening (Sc) of RNA-seq data for...

    • ega-archive.org
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    RaScALL: Rapid (Ra) screening (Sc) of RNA-seq data for prognostically significant genomic alterations in acute lymphoblastic leukaemia (ALL) [Dataset]. https://ega-archive.org/datasets/EGAD00001009087
    Explore at:
    License

    https://ega-archive.org/dacs/EGAC00001002790https://ega-archive.org/dacs/EGAC00001002790

    Description

    RNA-sequencing (RNA-seq) efforts in acute lymphoblastic leukaemia (ALL) have identified numerous prognostically significant genomic alterations which can guide diagnostic risk stratification and treatment choices when detected early. However, a full RNA-seq Bioinformatics workflow is time-consuming and costly in a clinical setting where rapid detection and accurate reporting of clinically relevant alterations are essential. To accelerate the identification of ALL-specific alterations (including gene fusions, single nucleotide variants and focal gene deletions), we developed the rapid screening tool RaScALL, capable of identifying more than 100 prognostically significant lesions directly from raw sequencing reads. RaScALL uses the k-mer based targeted detection tool km and known ALL variant information to achieve a high degree of accuracy for reporting subtype defining genomic alterations compared to standard alignment-based pipelines. Gene fusions, including difficult to detect fusions involving EPOR and DUX4, were accurately identified in 98% (164 samples) of reported cases in a 180-patient Australian study cohort and 95% (n=63) of samples in a North American validation cohort. Pathogenic sequence variants were correctly identified in 75% of tested samples, including all cases involving subtype defining variants PAX5 p.P80R (n=12) and IKZF1 p.N159Y (n=4). Accurate detection of intragenic IKZF1 deletions resulting in aberrant transcript isoforms was also detectable with 98% accuracy. Importantly, the median analysis time for detection of all targeted alterations averaged 22 minutes per sample, significantly shorter than standard alignment-based approaches, ensuring accelerated risk-stratification and therapeutic triage.

  12. S

    RNA Sequencing (KYSE450SLC8A1KO vs KYSE450WT)

    • scidb.cn
    Updated Jun 6, 2025
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Li Xinxin; Liu Kuancan (2025). RNA Sequencing (KYSE450SLC8A1KO vs KYSE450WT) [Dataset]. http://doi.org/10.57760/sciencedb.26122
    Explore at:
    CroissantCroissant is a format for machine-learning datasets. Learn more about this at mlcommons.org/croissant.
    Dataset updated
    Jun 6, 2025
    Dataset provided by
    Science Data Bank
    Authors
    Li Xinxin; Liu Kuancan
    License

    CC0 1.0 Universal Public Domain Dedicationhttps://creativecommons.org/publicdomain/zero/1.0/
    License information was derived automatically

    Description

    The data uploaded herein represents the transcriptomic sequencing results from the human esophageal squamous cell carcinoma cell line KYSE450, specifically comparing SLC8A1-knockout cells (KYSE450SLC8A1KO) with wild-type controls (KYSE450WT). The data generation process began with the separate cultivation of KYSE450SLC8A1KO and KYSE450WT cells until they reached a specific growth state. Qualified RNA samples were used to construct sequencing libraries, involving steps such as RNA fragmentation, cDNA synthesis, end repair, addition of A-tails, and adapter ligation. Finally, high-throughput sequencing was performed using the Illumina platform, generating a large volume of short-read sequence data.The data processing and analysis workflow started with quality control of the raw sequencing data (in .fastq format). Tools such as Fastp were employed to remove low-quality bases, adapter sequences, and ambiguous reads, thereby obtaining high-quality clean reads. Subsequently, these clean reads were aligned to the human reference genome using alignment software. After alignment, tools like featureCounts or HTSeq-count were used to count the number of reads mapping to each gene or transcript, thereby quantifying gene expression levels and generating a gene expression matrix. This matrix records the read counts (Read Count) for each gene within each sample and was ultimately organized and saved in .xls format. The uploaded data files primarily consist of this gene expression matrix, encompassing expression data from two samples (KYSE450SLC8A1KO and KYSE450WT). The data covers changes in gene expression levels across the entire genome. Temporal and spatial resolution are not applicable, as this is an in vitro cell line study. This dataset provides a foundation for studying the regulatory role of the SLC8A1 gene in esophageal squamous cell carcinoma and facilitates subsequent analyses such as differential expression analysis and pathway enrichment.

  13. Comparison of Fixed Single Cell RNA-seq Methods to Enable Transcriptome...

    • zenodo.org
    application/gzip, bin +1
    Updated Dec 31, 2025
    + more versions
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Klas Hatje; Emma Bell; Klas Hatje; Emma Bell (2025). Comparison of Fixed Single Cell RNA-seq Methods to Enable Transcriptome Profiling of Neutrophils in Clinical Samples - Time course data [Dataset]. http://doi.org/10.5281/zenodo.13750776
    Explore at:
    html, bin, application/gzipAvailable download formats
    Dataset updated
    Dec 31, 2025
    Dataset provided by
    Zenodohttp://zenodo.org/
    Authors
    Klas Hatje; Emma Bell; Klas Hatje; Emma Bell
    License

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

    Description

    Monitoring neutrophil gene expression is a powerful tool for understanding disease mechanisms, developing new diagnostics, therapies and optimizing clinical trials. Neutrophils are sensitive to the processing, storage and transportation steps that are involved in clinical sample analysis. This study is the first to evaluate the capabilities of technologies from 10X Genomics, PARSE Biosciences, and HIVE (Honeycomb Biotechnologies) to generate high-quality RNA data from human blood-derived neutrophils. Our comparative analysis shows that all methods produced high quality data, importantly capturing the transcriptomes of neutrophils. 10X FLEX cell populations in particular showed a close concordance with the flow cytometry data. Here, we establish a reliable single-cell RNA sequencing workflow for neutrophils in clinical trials: we offer guidelines on sample collection to preserve RNA quality and demonstrate how each method performs in capturing sensitive cell populations in clinical practice.

    This dataset includes only the 10X Flex time course data and analysis.

  14. S

    RNA Sequencing (KYSE450SOX2KO vs KYSE450WT)

    • scidb.cn
    Updated Jun 6, 2025
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Li Xinxin; Liu Kuancan (2025). RNA Sequencing (KYSE450SOX2KO vs KYSE450WT) [Dataset]. http://doi.org/10.57760/sciencedb.26117
    Explore at:
    CroissantCroissant is a format for machine-learning datasets. Learn more about this at mlcommons.org/croissant.
    Dataset updated
    Jun 6, 2025
    Dataset provided by
    Science Data Bank
    Authors
    Li Xinxin; Liu Kuancan
    License

    CC0 1.0 Universal Public Domain Dedicationhttps://creativecommons.org/publicdomain/zero/1.0/
    License information was derived automatically

    Description

    The data uploaded herein represents the transcriptomic sequencing results from the human esophageal squamous cell carcinoma cell line KYSE450, specifically comparing SOX2-knockout cells (KYSE450SOX2KO) with wild-type controls (KYSE450WT). The data generation process began with the separate cultivation of KYSE450SOX2KO and KYSE450WT cells until they reached a specific growth state. Total RNA was then extracted using commercially available RNA extraction kits. Following extraction, the quality of the isolated total RNA was assessed to ensure it met the requirements for library construction. Qualified RNA samples were used to construct sequencing libraries, involving steps such as RNA fragmentation, cDNA synthesis, end repair, addition of A-tails, and adapter ligation. Finally, high-throughput sequencing was performed using the Illumina platform, generating a large volume of short-read sequence data.The data processing and analysis workflow started with quality control of the raw sequencing data (in .fastq format). Tools such as Fastp were employed to remove low-quality bases, adapter sequences, and ambiguous reads, thereby obtaining high-quality clean reads. Subsequently, these clean reads were aligned to the human reference genome using alignment software. After alignment, tools like featureCounts or HTSeq-count were used to count the number of reads mapping to each gene or transcript, thereby quantifying gene expression levels and generating a gene expression matrix. This matrix records the read counts (Read Count) for each gene within each sample and was ultimately organized and saved in .xls format. The uploaded data files primarily consist of this gene expression matrix, encompassing expression data from two samples (KYSE450SOX2KO and KYSE450WT). The data covers changes in gene expression levels across the entire genome. Temporal and spatial resolution are not applicable, as this is an in vitro cell line study. This dataset provides a foundation for studying the regulatory role of the SOX2 gene in esophageal squamous cell carcinoma and facilitates subsequent analyses such as differential expression analysis and pathway enrichment.

  15. o

    Gene-level read counts from bulk RNA-seq data for 38 follicular lymphoma...

    • explore.openaire.eu
    Updated Aug 21, 2022
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Andrew Weng; Christian Steidl; David Scott (2022). Gene-level read counts from bulk RNA-seq data for 38 follicular lymphoma diagnostic biopsies [Dataset]. http://doi.org/10.5281/zenodo.7013884
    Explore at:
    Dataset updated
    Aug 21, 2022
    Authors
    Andrew Weng; Christian Steidl; David Scott
    Description

    Conventional (bulk) RNA-sequencing was performed on unfractionated cell suspension or snap frozen whole tissue material. Total RNA was isolated with TRIzol reagent followed by purification over PureLink RNA Mini Kit columns (Invitrogen). RNA-seq was performed using a polyA-enriched strand-specific library construction protocol (doi: 10.1016/j.ccell.2016.02.009) and paired-end 75bp sequencing on an Illumina HiSeq 2500 instrument. Raw reads were aligned to the reference human genome assembly GRCh37 (hg19) using STAR (v2.5.2.a). To improve spliced alignment, STAR was provided with exon junction coordinates from the reference annotations (Gencode v19). We applied a modified version of a bioinformatics workflow for normalization of raw read counts and differential gene expression analysis (doi: 10.12688/f1000research.9005.3). Gene-level read counts were quantified using HTSEQ-count (v0.11.0; intersection-strict, reverse mode) (doi: 10.1093/bioinformatics/btu638). Genes showing low read counts (i.e., genes not showing counts per million (cpm) > 1.0 in at least 10% of samples) were removed from further analysis. Raw counts from expressed genes were then TMM-normalized and scaled to counts per million (CPM) using the edgeR (v3.22.2) package (doi: 10.1093/bioinformatics/btp616). Sample IDs correspond to those referenced in Wang X et al, Nature Communications (2022).

  16. N

    NGS-based RNA-seq Report

    • datainsightsmarket.com
    doc, pdf, ppt
    Updated May 10, 2025
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Data Insights Market (2025). NGS-based RNA-seq Report [Dataset]. https://www.datainsightsmarket.com/reports/ngs-based-rna-seq-1723482
    Explore at:
    pdf, ppt, docAvailable download formats
    Dataset updated
    May 10, 2025
    Dataset authored and provided by
    Data Insights Market
    License

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

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

    The Next-Generation Sequencing (NGS)-based RNA sequencing (RNA-seq) market is experiencing robust growth, driven by advancements in sequencing technologies, increasing research funding for genomic studies, and the expanding application of RNA-seq in various fields. The market's value in 2025 is estimated to be around $5 billion, with a Compound Annual Growth Rate (CAGR) projected at approximately 15% from 2025 to 2033. This substantial growth is fueled by the rising demand for personalized medicine, the increasing prevalence of chronic diseases necessitating improved diagnostics, and the growing adoption of RNA-seq in drug discovery and development. High-throughput sequencing currently dominates the market due to its cost-effectiveness and high throughput, but third-generation sequencing technologies are gaining traction due to their longer read lengths and potential for improved accuracy and reduced bias. Key market segments include hospitals and clinics, biopharmaceutical companies, and academic research organizations, with North America and Europe representing the largest regional markets. The market is highly competitive, with key players such as Illumina, Thermo Fisher Scientific, and Pacific Biosciences leading the innovation and market share. Despite the significant growth, the market faces certain restraints including the high cost of NGS platforms and data analysis, the complexity of RNA-seq workflows requiring specialized expertise, and ethical considerations related to data privacy and informed consent. However, these challenges are being actively addressed through technological advancements, the development of user-friendly software, and the establishment of clear ethical guidelines. Ongoing research into novel RNA biomarkers and their application in disease diagnosis and treatment is expected to further drive market growth. The diverse applications of NGS-based RNA-seq, ranging from cancer research and personalized oncology to infectious disease surveillance and agricultural genomics, ensures the market's long-term sustainability and future potential. The continued investment in research and development, coupled with a growing awareness of the clinical and research applications of this technology, will ensure sustained expansion in this rapidly evolving market segment.

  17. Enhanced Protein Isoform Characterization Through Long-Read Proteogenomics -...

    • zenodo.org
    • data.niaid.nih.gov
    application/gzip, bin +1
    Updated Jul 17, 2024
    + more versions
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Rachel Miller; Rachel Miller; Ben Jordan; Ben Jordan; Madison Mehlferber; Madison Mehlferber; Erin Jeffery; Erin Jeffery; Christina Chatzipantsiou; Christina Chatzipantsiou; Simran Kaur; Simran Kaur; Robert Millikin; Robert Millikin; Michael Shortreed; Michael Shortreed; Simone Tiberi; Simone Tiberi; Ana Conesa; Ana Conesa; Lloyd Smith; Lloyd Smith; Anne Deslattes Mays; Anne Deslattes Mays; Gloria Sheynkman; Gloria Sheynkman (2024). Enhanced Protein Isoform Characterization Through Long-Read Proteogenomics - Workflow Results [Dataset]. http://doi.org/10.5281/zenodo.5987905
    Explore at:
    application/gzip, bin, txtAvailable download formats
    Dataset updated
    Jul 17, 2024
    Dataset provided by
    Zenodohttp://zenodo.org/
    Authors
    Rachel Miller; Rachel Miller; Ben Jordan; Ben Jordan; Madison Mehlferber; Madison Mehlferber; Erin Jeffery; Erin Jeffery; Christina Chatzipantsiou; Christina Chatzipantsiou; Simran Kaur; Simran Kaur; Robert Millikin; Robert Millikin; Michael Shortreed; Michael Shortreed; Simone Tiberi; Simone Tiberi; Ana Conesa; Ana Conesa; Lloyd Smith; Lloyd Smith; Anne Deslattes Mays; Anne Deslattes Mays; Gloria Sheynkman; Gloria Sheynkman
    License

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

    Description
     

    The detection of physiologically relevant protein isoforms encoded by the human genome is critical to biomedicine. Mass spectrometry (MS)-based proteomics is the preeminent method for protein detection, but isoform-resolved proteomic analysis relies on accurate reference databases that match the sample; neither a subset nor a superset database is ideal. Long-read RNA sequencing (e.g. PacBio, Oxford Nanopore) provides full-length transcript sequencing, which can be used to predict full-length proteins. Here, we describe a long-read proteogenomics approach for integrating matched long-read RNA-seq and MS-based proteomics data to enhance isoform characterization. We introduce a classification scheme for protein isoforms, discover novel protein isoforms, and present the first protein inference algorithm for the direct incorporation of long-read transcriptome data in protein inference to enable detection of protein isoforms that are intractable to MS detection. We have released an open-source Nextflow pipeline that integrates long-read sequencing in a proteomic workflow for isoform-resolved analysis.

    Companion Repositories:

    1. Long-Read-Proteogenomics Workflow GitHub Repository Release
    2. Long-Read-Proteogenomics Analysis GitHub Repository Release

    Companion Datasets

    1. Long-Read-Proteogenomics Workflow Sample and Reference Data
    2. TEST Data for Long-Read-Proteogenomics Workflow GitHub Actions

    This Repository contains the complete output from the execution of the Long-Read-Proteogenomics Workflow, using the input from Jurkat Samples and Reference Data.

    The file jurkat.flnc.bam was 6.5 GB had to be split into 13 separate files and for use should be rejoined -- here are the steps that were used to split the file up.

    1. Convert jurkat.flnc.bam (binary format) to sam file (text format) without header: samtools view jurkat.flnc.bam > jurkat.flnc.sam

    2. Capture the header: samtools view -H jurkat.flnc.bam > jurkat.flnc.header.sam

    3. Split jurkat.flnc.sam into smaller files (aim to get final size under 2GB): split -l 400000 jurkat.flnc.sam jurkat.flnc.chunk.

    4. Convert each of these files back to bam for uploading: samtools view -b jurkat.flnc.chunk.a* -o jurkat.flnc.chunk.a*.bam (*=a,b,c,d,e,f,g,h,i,j,k,l,m)

    After downloading, reverse this process including using the header file which is found in the LRPG-Manuscript-Results-results-results-jurkat-isoseq3-companion-files.tar.gz file>

    1. Convert the bam files back to sam files: samtools view jurkat.flnc.chunk.a*.bam > jurkat.flnc.chunk.a*.sam (*=a,b,c,d,e,f,g,h,i,j,k,l,m)

    2. Combine the header together with the sam files: cat jurkat.flnc.chunk.a*sam > jurkcat.flnc.sam (verified the same number of lines of the sam files is identical to the number of lines of the original without header: 4,956,761. Header file is 13 lines.

    3. Convert to bam files if desired: samtools view -b jurkat.flnc.sam -o jurkat.flnc.bam

    4. Rehead with the header file: samtools reheader -P -i jurkat.flnc.header.sam jurkat.flnc.bam

  18. N

    Data from: RNA-Seq Reveals Age- and Species Differences of CAR-targeted...

    • data.niaid.nih.gov
    Updated Jul 25, 2021
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Cheng SL; Frazar C (2021). RNA-Seq Reveals Age- and Species Differences of CAR-targeted Drug-Processing Genes in Liver [Dataset]. https://data.niaid.nih.gov/resources?id=gse98666
    Explore at:
    Dataset updated
    Jul 25, 2021
    Dataset provided by
    universtiy of washington
    Authors
    Cheng SL; Frazar C
    Description

    Purpose: Next-generation sequencing (NGS) has been utilized for systems-based analysis of all liver samples. The goals of this study are to use NGS-derived mouse CAR and human CAR initiated transcriptome profiling (RNA-seq) and find out similarity and difference drug processing gene (DPG) pattern after CAR activation in different genotype include WT (C57BL/6 and human CAR transgenic mice with C57BL/6 background)Methods: Liver mRNA profiles of wild-type (WT) and human CAR knockin (hCAR-TG) mice at the age of day 5 and day 60 treated with mouse CAR activator (TCPOBOP) and human CAR activator (CITCO) respectively were generated by deep sequencing, in triplicate, using HiSeq 2000 sequencer. The sequence reads that passed quality filters were analyzed at the transcript level with followed method: HISAT followed by Cufflinks.Results: Using an optimized data analysis workflow,RNA-Seq generated approximately 47 to 68 million reads per sample, among which approximately 40 to 60 million reads were uniquely mapped to the mouse reference genome (NCBI GRCm/38/mm10). And we identified 393 drug processing genes in the livers of WT and hCAR-TG with with HISAT workflow. RNA-seq data confirmed that among all the 393 DPGs with known important functions in xenobiotic biotransformation, 90 DPGs were not expressed in livers of any groups (threshold: average FPKM < 1 in all treatment groups); whereas a total of 303 genes were expressed in livers of at least one groups, among which 258 DPGs were differentially regulated by mCAR or hCAR activation in either Day 5 or Day 60 (FDR-BH<0.05), and 45 genes were stably expressed among all treatment groups.Conclusions: Our study represents the first detailed analysis of drug processing genes, with 3 biologic replicates, generated by RNA-seq technology. The optimized data analysis reported here should provide a framework for comparative investigations of expression profiles by mouse CAR activation and human CAR activation. Our results show that NGS offers a comprehensive and accurate quantitative and qualitative evaluation of mRNA content within tissues. Liver mRNA profiles of wild type (WT) and human CAR knockin (hCAR-TG) mice at the age of day 5 and day 60 treated with TCPOBOP and CITCO respectively were generated by deep sequencing, in triplicate, using HiSeq 2000 sequencer.

  19. w

    Global Rna Seq Library Prep Kits Market Research Report: By Workflow Type...

    • wiseguyreports.com
    Updated Sep 12, 2024
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    wWiseguy Research Consultants Pvt Ltd (2024). Global Rna Seq Library Prep Kits Market Research Report: By Workflow Type (Sample Preparation, Library Construction, Sequencing, Data Analysis), By Sample Type (RNA, DNA, NGS), By Application (Genomics, Transcriptomics, Epigenomics, Metagenomics, CRISPR-Cas9 Editing), By Throughput (Low-Throughput, Medium-Throughput, High-Throughput), By Technology (PCR-Based, Non-PCR-Based, Isothermal Amplification, Next-Generation Sequencing (NGS)) and By Regional (North America, Europe, South America, Asia Pacific, Middle East and Africa) - Forecast to 2032. [Dataset]. https://www.wiseguyreports.com/fr/reports/rna-seq-library-prep-kits-market
    Explore at:
    Dataset updated
    Sep 12, 2024
    Dataset authored and provided by
    wWiseguy Research Consultants Pvt Ltd
    License

    https://www.wiseguyreports.com/pages/privacy-policyhttps://www.wiseguyreports.com/pages/privacy-policy

    Time period covered
    Jan 9, 2024
    Area covered
    Global
    Description
    BASE YEAR2024
    HISTORICAL DATA2019 - 2024
    REPORT COVERAGERevenue Forecast, Competitive Landscape, Growth Factors, and Trends
    MARKET SIZE 202311.57(USD Billion)
    MARKET SIZE 202413.49(USD Billion)
    MARKET SIZE 203245.96(USD Billion)
    SEGMENTS COVEREDWorkflow Type, Sample Type, Application, Throughput, Technology, Regional
    COUNTRIES COVEREDNorth America, Europe, APAC, South America, MEA
    KEY MARKET DYNAMICSTechnological advancements Rising demand for personalized medicine Increasing prevalence of chronic diseases Growing adoption of NGS Expansion into emerging markets
    MARKET FORECAST UNITSUSD Billion
    KEY COMPANIES PROFILEDIllumina, Agilent Technologies, BioRad Laboratories, Thermo Fisher Scientific, PerkinElmer, NEB, QIAGEN, Pacific Biosciences, Novogene, DaAn Gene, Oxford Nanopore Technologies, Takara Bio, Genapsys, Roche
    MARKET FORECAST PERIOD2025 - 2032
    KEY MARKET OPPORTUNITIESSinglecell RNA sequencing Spatial transcriptomics Longread RNA sequencing Cancer genomics Infectious disease research
    COMPOUND ANNUAL GROWTH RATE (CAGR) 16.56% (2025 - 2032)
  20. z

    Single-cell RNA-Seq and TCR-Seq analysis of PD-1+ CD8+ T-cells responding to...

    • zenodo.org
    bin, csv, zip
    Updated Oct 24, 2024
    Share
    FacebookFacebook
    TwitterTwitter
    Email
    Click to copy link
    Link copied
    Close
    Cite
    Bertram Bengsch; Bertram Bengsch; Sagar; Sagar; Zhen Zhang; Zhen Zhang (2024). Single-cell RNA-Seq and TCR-Seq analysis of PD-1+ CD8+ T-cells responding to anti-PD-1 and anti-PD-1/CTLA-4 immunotherapy in melanoma [Dataset]. http://doi.org/10.5281/zenodo.13971562
    Explore at:
    bin, csv, zipAvailable download formats
    Dataset updated
    Oct 24, 2024
    Dataset provided by
    Zenodo
    Authors
    Bertram Bengsch; Bertram Bengsch; Sagar; Sagar; Zhen Zhang; Zhen Zhang
    License

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

    Description

    This dataset details the scRNASeq and TCR-Seq analysis of sorted PD-1+ CD8+ T cells from patients with melanoma treated with checkpoint therapy (anti-PD-1 monotherapy and anti-PD-1 & anti-CTLA-4 combination therapy) at baseline and after the first cycle of therapy. A major publication using this dataset is accessible here: (reference)

    *experimental design

    Single-cell RNA sequencing was performed using 10x Genomics with feature barcoding technology to multiplex cell samples from different patients undergoing mono or dual therapy so that they can be loaded on one well to reduce costs and minimize technical variability. Hashtag oligomers (oligos) were obtained as purified and already oligo-conjugated in TotalSeq-C format from BioLegend. Cells were thawed, counted and 20 million cells per patient and time point were used for staining. Cells were stained with barcoded antibodies together with a staining solution containing antibodies against CD3, CD4, CD8, PD-1/IgG4 and fixable viability dye (eBioscience) prior to FACS sorting. Barcoded antibody concentrations used were 0.5 µg per million cells, as recommended by the manufacturer (BioLegend) for flow cytometry applications. After staining, cells were washed twice in PBS containing 2% BSA and 0.01% Tween 20, followed by centrifugation (300 xg 5 min at 4 °C) and supernatant exchange. After the final wash, cells were resuspended in PBS and filtered through 40 µm cell strainers and proceeded for sorting. Sorted cells were counted and approximately 75,000 cells were processed through 10x Genomics single-cell V(D)J workflow according to the manufacturer’s instructions. Gene expression, hashing and TCR libraries were pooled to desired quantities to obtain the sequencing depths of 15,000 reads per cell for gene expression libraries and 5,000 reads per cell for hashing and TCR libraries. Libraries were sequenced on a NovaSeq 6000 flow cell in a 2X100 paired-end format.

    *extract protocol

    PBMCs were thawed, counted and 20 million cells per patient and time point were used for staining. Cells were stained with barcoded antibodies together with a staining solution containing antibodies against CD3, CD4, CD8, PD-1/IgG4 and fixable viability dye (eBioscience) prior to FACS sorting. Barcoded antibody concentrations used were 0.5 µg per million cells, as recommended by the manufacturer (BioLegend) for flow cytometry applications. After staining, cells were washed twice in PBS containing 2% BSA and 0.01% Tween 20, followed by centrifugation (300 xg 5 min at 4 °C) and supernatant exchange. After the final wash, cells were resuspended in PBS and filtered through 40 µm cell strainers and proceeded for sorting. Sorted cells were counted and approximately 75,000 cells were processed through 10x Genomics single-cell V(D)J workflow according to the manufacturer’s instructions.

    *library construction protocol

    Sorted cells were counted and approximately 75,000 cells were processed through 10x Genomics single-cell V(D)J workflow according to the manufacturer’s instructions. Gene expression, hashing and TCR libraries were pooled to desired quantities to obtain the sequencing depths of 15,000 reads per cell for gene expression libraries and 5,000 reads per cell for hashing and TCR libraries. Libraries were sequenced on a NovaSeq 6000 flow cell in a 2X100 paired-end format.

    *library strategy

    scRNA-seq and scTCR-seq

    *data processing step

    Pre-processing of sequencing results to generate count matrices (gene expression and HTO barcode counts) was performed using the 10x genomics Cell Ranger pipeline.

    Further processing was done with Seurat (cell and gene filtering, hashtag identification, clustering, differential gene expression analysis based on gene expression).

    *genome build/assembly

    Alignment was performed using prebuilt Cell Ranger human reference GRCh38.

    *processed data files format and content

    RNA counts and HTO counts are in sparse matrix format and TCR clonotypes are in csv format.

    Datasets were merged and analyzed by Seurat and the analyzed objects are in rds format.

    file name

    file checksum

    PD1CD8_160421_filtered_feature_bc_matrix.zip

    da2e006d2b39485fd8cf8701742c6d77

    PD1CD8_190421_filtered_feature_bc_matrix.zip

    e125fc5031899bba71e1171888d78205

    PD1CD8_160421_filtered_contig_annotations.csv

    927241805d507204fbe9ef7045d0ccf4

    PD1CD8_190421_filtered_contig_annotations.csv

    8ca544d27f06e66592b567d3ab86551e

    *processed data file

    antibodies/tags

    PD1CD8_160421_filtered_feature_bc_matrix.zip

    none

    PD1CD8_160421_filtered_feature_bc_matrix.zip

    TotalSeq™-C0251 anti-human Hashtag 1 Antibody - (HASH_1) - M1_base_monotherapy
    TotalSeq™-C0252 anti-human Hashtag 2 Antibody - (HASH_2) - M1_post_monotherapy
    TotalSeq™-C0253 anti-human Hashtag 3 Antibody - (HASH_3) - C1_base_combined_therapy
    TotalSeq™-C0254 anti-human Hashtag 4 Antibody - (HASH_4) - C1_post_combined_therapy
    TotalSeq™-C0255 anti-human Hashtag 5 Antibody - (HASH_5) - C2_base_combined_therapy
    TotalSeq™-C0256 anti-human Hashtag 6 Antibody - (HASH_6) - C2_post_combined_therapy

    PD1CD8_160421_filtered_contig_annotations.csv

    none

    PD1CD8_190421_filtered_feature_bc_matrix.zip

    none

    PD1CD8_190421_filtered_feature_bc_matrix.zip

    TotalSeq™-C0251 anti-human Hashtag 1 Antibody - (HASH_1) - M2_base_monotherapy
    TotalSeq™-C0252 anti-human Hashtag 2 Antibody - (HASH_2) - M2_post_monotherapy
    TotalSeq™-C0253 anti-human Hashtag 3 Antibody - (HASH_3) - M3_base_monotherapy
    TotalSeq™-C0254 anti-human Hashtag 4 Antibody - (HASH_4) - M3_post_monotherapy
    TotalSeq™-C0255 anti-human Hashtag 5 Antibody - (HASH_5) - C3_base_combined_therapy
    TotalSeq™-C0256 anti-human Hashtag 6 Antibody - (HASH_6) - C3_post_combined_therapy

    PD1CD8_190421_filtered_contig_annotations.csv

    none

Share
FacebookFacebook
TwitterTwitter
Email
Click to copy link
Link copied
Close
Cite
Sateesh Peri; Sarah Roberts; Isabella R. Kreko; Lauren B. McHan; Alexandra Naron; Archana Ram; Rebecca L. Murphy; Eric Lyons; Brian D. Gregory; Upendra K. Devisetty; Andrew D. L. Nelson (2023). Table_1_Read Mapping and Transcript Assembly: A Scalable and High-Throughput Workflow for the Processing and Analysis of Ribonucleic Acid Sequencing Data.xlsx [Dataset]. http://doi.org/10.3389/fgene.2019.01361.s002

Table_1_Read Mapping and Transcript Assembly: A Scalable and High-Throughput Workflow for the Processing and Analysis of Ribonucleic Acid Sequencing Data.xlsx

Related Article
Explore at:
xlsxAvailable download formats
Dataset updated
Jun 1, 2023
Dataset provided by
Frontiers
Authors
Sateesh Peri; Sarah Roberts; Isabella R. Kreko; Lauren B. McHan; Alexandra Naron; Archana Ram; Rebecca L. Murphy; Eric Lyons; Brian D. Gregory; Upendra K. Devisetty; Andrew D. L. Nelson
License

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

Description

Next-generation RNA-sequencing is an incredibly powerful means of generating a snapshot of the transcriptomic state within a cell, tissue, or whole organism. As the questions addressed by RNA-sequencing (RNA-seq) become both more complex and greater in number, there is a need to simplify RNA-seq processing workflows, make them more efficient and interoperable, and capable of handling both large and small datasets. This is especially important for researchers who need to process hundreds to tens of thousands of RNA-seq datasets. To address these needs, we have developed a scalable, user-friendly, and easily deployable analysis suite called RMTA (Read Mapping, Transcript Assembly). RMTA can easily process thousands of RNA-seq datasets with features that include automated read quality analysis, filters for lowly expressed transcripts, and read counting for differential expression analysis. RMTA is containerized using Docker for easy deployment within any compute environment [cloud, local, or high-performance computing (HPC)] and is available as two apps in CyVerse's Discovery Environment, one for normal use and one specifically designed for introducing undergraduates and high school to RNA-seq analysis. For extremely large datasets (tens of thousands of FASTq files) we developed a high-throughput, scalable, and parallelized version of RMTA optimized for launching on the Open Science Grid (OSG) from within the Discovery Environment. OSG-RMTA allows users to utilize the Discovery Environment for data management, parallelization, and submitting jobs to OSG, and finally, employ the OSG for distributed, high throughput computing. Alternatively, OSG-RMTA can be run directly on the OSG through the command line. RMTA is designed to be useful for data scientists, of any skill level, interested in rapidly and reproducibly analyzing their large RNA-seq data sets.

Search
Clear search
Close search
Google apps
Main menu