Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
2.
Nucleic Acids Res ; 50(6): 3475-3489, 2022 04 08.
Article in English | MEDLINE | ID: mdl-35244721

ABSTRACT

The SARS-CoV-2 virus has a complex transcriptome characterised by multiple, nested subgenomic RNAsused to express structural and accessory proteins. Long-read sequencing technologies such as nanopore direct RNA sequencing can recover full-length transcripts, greatly simplifying the assembly of structurally complex RNAs. However, these techniques do not detect the 5' cap, thus preventing reliable identification and quantification of full-length, coding transcript models. Here we used Nanopore ReCappable Sequencing (NRCeq), a new technique that can identify capped full-length RNAs, to assemble a complete annotation of SARS-CoV-2 sgRNAs and annotate the location of capping sites across the viral genome. We obtained robust estimates of sgRNA expression across cell lines and viral isolates and identified novel canonical and non-canonical sgRNAs, including one that uses a previously un-annotated leader-to-body junction site. The data generated in this work constitute a useful resource for the scientific community and provide important insights into the mechanisms that regulate the transcription of SARS-CoV-2 sgRNAs.


Subject(s)
COVID-19 , Nanopores , RNA, Guide, Kinetoplastida/chemistry , COVID-19/genetics , Genome, Viral/genetics , Humans , RNA Caps , RNA, Viral/genetics , RNA, Viral/metabolism , SARS-CoV-2/genetics
3.
RNA ; 28(2): 162-176, 2022 02.
Article in English | MEDLINE | ID: mdl-34728536

ABSTRACT

Nanopore sequencing devices read individual RNA strands directly. This facilitates identification of exon linkages and nucleotide modifications; however, using conventional direct RNA nanopore sequencing, the 5' and 3' ends of poly(A) RNA cannot be identified unambiguously. This is due in part to RNA degradation in vivo and in vitro that can obscure transcription start and end sites. In this study, we aimed to identify individual full-length human RNA isoforms among ∼4 million nanopore poly(A)-selected RNA reads. First, to identify RNA strands bearing 5' m7G caps, we exchanged the biological cap for a modified cap attached to a 45-nt oligomer. This oligomer adaptation method improved 5' end sequencing and ensured correct identification of the 5' m7G capped ends. Second, among these 5'-capped nanopore reads, we screened for features consistent with a 3' polyadenylation site. Combining these two steps, we identified 294,107 individual high-confidence full-length RNA scaffolds from human GM12878 cells, most of which (257,721) aligned to protein-coding genes. Of these, 4876 scaffolds indicated unannotated isoforms that were often internal to longer, previously identified RNA isoforms. Orthogonal data for m7G caps and open chromatin, such as CAGE and DNase-HS seq, confirmed the validity of these high-confidence RNA scaffolds.


Subject(s)
RNA Isoforms/chemistry , RNA, Messenger/chemistry , Cell Line, Tumor , Humans , Nanopore Sequencing/methods , RNA 3' Polyadenylation Signals , RNA Isoforms/genetics , RNA, Messenger/genetics , Transcriptome
4.
ACS Nano ; 15(10): 16642-16653, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34618430

ABSTRACT

We describe a method for direct tRNA sequencing using the Oxford Nanopore MinION. The principal technical advance is custom adapters that facilitate end-to-end sequencing of individual transfer RNA (tRNA) molecules at subnanometer precision. A second advance is a nanopore sequencing pipeline optimized for tRNA. We tested this method using purified E. coli tRNAfMet, tRNALys, and tRNAPhe samples. 76-92% of individual aligned tRNA sequence reads were full length. As a proof of concept, we showed that nanopore sequencing detected all 43 expected isoacceptors in total E. coli MRE600 tRNA as well as isodecoders that further define that tRNA population. Alignment-based comparisons between the three purified tRNAs and their synthetic controls revealed systematic nucleotide miscalls that were diagnostic of known modifications. Systematic miscalls were also observed proximal to known modifications in total E. coli tRNA alignments, including a highly conserved pseudouridine in the T loop. This work highlights the potential of nanopore direct tRNA sequencing as well as improvements needed to implement tRNA sequencing for human healthcare applications.


Subject(s)
Nanopore Sequencing , Nanopores , Escherichia coli/genetics , High-Throughput Nucleotide Sequencing , Humans , Nucleotides
5.
RNA ; 27(12): 1497-1511, 2021 12.
Article in English | MEDLINE | ID: mdl-34446532

ABSTRACT

Understanding transcriptomes requires documenting the structures, modifications, and abundances of RNAs as well as their proximity to other molecules. The methods that make this possible depend critically on enzymes (including mutant derivatives) that act on nucleic acids for capturing and sequencing RNA. We tested two 3' nucleotidyl transferases, Saccharomyces cerevisiae poly(A) polymerase and Schizosaccharomyces pombe Cid1, for the ability to add base and sugar modified rNTPs to free RNA 3' ends, eventually focusing on Cid1. Although unable to polymerize ΨTP or 1meΨTP, Cid1 can use 5meUTP and 4thioUTP. Surprisingly, Cid1 can use inosine triphosphate to add poly(I) to the 3' ends of a wide variety of RNA molecules. Most poly(A) mRNAs efficiently acquire a uniform tract of about 50 inosine residues from Cid1, whereas non-poly(A) RNAs acquire longer, more heterogeneous tails. Here we test these activities for use in direct RNA sequencing on nanopores, and find that Cid1-mediated poly(I)-tailing permits detection and quantification of both mRNAs and non-poly(A) RNAs simultaneously, as well as enabling the analysis of nascent RNAs associated with RNA polymerase II. Poly(I) produces a different current trace than poly(A), enabling recognition of native RNA 3' end sequence lost by in vitro poly(A) addition. Addition of poly(I) by Cid1 offers a broadly useful alternative to poly(A) capture for direct RNA sequencing on nanopores.


Subject(s)
Nanopores , Nucleotides/chemistry , Nucleotidyltransferases/metabolism , Polymers/chemistry , Polynucleotide Adenylyltransferase/metabolism , Saccharomyces cerevisiae/enzymology , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/enzymology , Sequence Analysis, RNA/methods , Nucleotidyltransferases/genetics , Polynucleotide Adenylyltransferase/genetics , Schizosaccharomyces pombe Proteins/genetics
6.
Methods Mol Biol ; 2298: 53-74, 2021.
Article in English | MEDLINE | ID: mdl-34085238

ABSTRACT

Historically, RNA has been sequenced as cDNA copies derived from reverse transcription of cellular RNA followed by PCR amplification. Recently, RNA sequencing using nanopores has emerged as an alternative. Using this technology, individual cellular RNA strands are read directly as they are driven through nanoscale pores by an applied voltage. The speed of translocation is regulated by a helicase that is loaded onto each RNA strand by an adapter that also facilitates capture by the nanopore electric field. Here we describe a technique for adapting human ribosomal RNA subunits for nanopore sequencing. Using this strategy, a single Oxford Nanopore MinION run delivered 470,907 sequence reads of which 396,048 aligned to ribosomal RNA, with 28S, 18S, 5.8S, and 5S coverage of 6053, 369,472, 16,058, and 4465 reads, respectively. Example alignments that reveal putative nucleotide modifications are provided.


Subject(s)
Nanopore Sequencing/methods , Nucleotides/genetics , RNA, Ribosomal/genetics , Sequence Analysis, RNA/methods , Humans , Nanopores , Sequence Analysis, DNA/methods
7.
Cell Syst ; 12(8): 827-838.e5, 2021 08 18.
Article in English | MEDLINE | ID: mdl-34146471

ABSTRACT

The accurate identification and quantitation of RNA isoforms present in the cancer transcriptome is key for analyses ranging from the inference of the impacts of somatic variants to pathway analysis to biomarker development and subtype discovery. The ICGC-TCGA DREAM Somatic Mutation Calling in RNA (SMC-RNA) challenge was a crowd-sourced effort to benchmark methods for RNA isoform quantification and fusion detection from bulk cancer RNA sequencing (RNA-seq) data. It concluded in 2018 with a comparison of 77 fusion detection entries and 65 isoform quantification entries on 51 synthetic tumors and 32 cell lines with spiked-in fusion constructs. We report the entries used to build this benchmark, the leaderboard results, and the experimental features associated with the accurate prediction of RNA species. This challenge required submissions to be in the form of containerized workflows, meaning each of the entries described is easily reusable through CWL and Docker containers at https://github.com/SMC-RNA-challenge. A record of this paper's transparent peer review process is included in the supplemental information.


Subject(s)
Neoplasms , Humans , Neoplasms/genetics , Protein Isoforms/genetics , RNA/genetics , RNA-Seq , Sequence Analysis, RNA
8.
Elife ; 102021 05 28.
Article in English | MEDLINE | ID: mdl-34047695

ABSTRACT

Determining the layers of gene regulation within the innate immune response is critical to our understanding of the cellular responses to infection and dysregulation in disease. We identified a conserved mechanism of gene regulation in human and mouse via changes in alternative first exon (AFE) usage following inflammation, resulting in changes to the isoforms produced. Of these AFE events, we identified 95 unannotated transcription start sites in mice using a de novo transcriptome generated by long-read native RNA-sequencing, one of which is in the cytosolic receptor for dsDNA and known inflammatory inducible gene, Aim2. We show that this unannotated AFE isoform of Aim2 is the predominant isoform expressed during inflammation and contains an iron-responsive element in its 5'UTR enabling mRNA translation to be regulated by iron levels. This work highlights the importance of examining alternative isoform changes and translational regulation in the innate immune response and uncovers novel regulatory mechanisms of Aim2.


Subject(s)
Alternative Splicing , DNA-Binding Proteins/genetics , Exons , Immunity, Innate/genetics , Inflammation/genetics , Macrophages/metabolism , 5' Untranslated Regions , Animals , Cells, Cultured , DNA-Binding Proteins/metabolism , Gene Expression Profiling , Humans , Inflammation/immunology , Inflammation/metabolism , Macrophages/immunology , Mice , Promoter Regions, Genetic , Transcriptome
9.
bioRxiv ; 2021 Apr 06.
Article in English | MEDLINE | ID: mdl-33851162

ABSTRACT

We report a SARS-CoV-2 lineage that shares N501Y, P681H, and other mutations with known variants of concern, such as B.1.1.7. This lineage, which we refer to as B.1.x (COG-UK sometimes references similar samples as B.1.324.1), is present in at least 20 states across the USA and in at least six countries. However, a large deletion causes the sequence to be automatically rejected from repositories, suggesting that the frequency of this new lineage is underestimated using public data. Recent dynamics based on 339 samples obtained in Santa Cruz County, CA, USA suggest that B.1.x may be increasing in frequency at a rate similar to that of B.1.1.7 in Southern California. At present the functional differences between this variant B.1.x and other circulating SARS-CoV-2 variants are unknown, and further studies on secondary attack rates, viral loads, immune evasion and/or disease severity are needed to determine if it poses a public health concern. Nonetheless, given what is known from well-studied circulating variants of concern, it seems unlikely that the lineage could pose larger concerns for human health than many already globally distributed lineages. Our work highlights a need for rapid turnaround time from sequence generation to submission and improved sequence quality control that removes submission bias. We identify promising paths toward this goal.

10.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Article in English | MEDLINE | ID: mdl-33758101

ABSTRACT

Among the large, diverse set of mammalian long noncoding RNAs (lncRNAs), long noncoding primary microRNAs (lnc-pri-miRNAs) are those that host miRNAs. Whether lnc-pri-miRNA loci have important biological function independent of their cognate miRNAs is poorly understood. From a genome-scale lncRNA screen, lnc-pri-miRNA loci were enriched for function in cell proliferation, and in glioblastoma (i.e., GBM) cells with DGCR8 or DROSHA knockdown, lnc-pri-miRNA screen hits still regulated cell growth. To molecularly dissect the function of a lnc-pri-miRNA locus, we studied LOC646329 (also known as MIR29HG), which hosts the miR-29a/b1 cluster. In GBM cells, LOC646329 knockdown reduced miR-29a/b1 levels, and these cells exhibited decreased growth. However, genetic deletion of the miR-29a/b1 cluster (LOC646329-miR29Δ) did not decrease cell growth, while knockdown of LOC646329-miR29Δ transcripts reduced cell proliferation. The miR-29a/b1-independent activity of LOC646329 corresponded to enhancer-like activation of a neighboring oncogene (MKLN1), regulating cell propagation. The LOC646329 locus interacts with the MKLN1 promoter, and antisense oligonucleotide knockdown of the lncRNA disrupts these interactions and reduces the enhancer-like activity. More broadly, analysis of genome-wide data from multiple human cell types showed that lnc-pri-miRNA loci are significantly enriched for DNA looping interactions with gene promoters as well as genomic and epigenetic characteristics of transcriptional enhancers. Functional studies of additional lnc-pri-miRNA loci demonstrated cognate miRNA-independent enhancer-like activity. Together, these data demonstrate that lnc-pri-miRNA loci can regulate cell biology via both miRNA-dependent and miRNA-independent mechanisms.


Subject(s)
Cell Proliferation/genetics , Genetic Loci , RNA, Long Noncoding/metabolism , Apoptosis/genetics , Gene Knockdown Techniques , HEK293 Cells , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , RNA-Seq
11.
Genes (Basel) ; 12(1)2021 01 16.
Article in English | MEDLINE | ID: mdl-33467183

ABSTRACT

For the past two decades, microbial monitoring of the International Space Station (ISS) has relied on culture-dependent methods that require return to Earth for analysis. This has a number of limitations, with the most significant being bias towards the detection of culturable organisms and the inherent delay between sample collection and ground-based analysis. In recent years, portable and easy-to-use molecular-based tools, such as Oxford Nanopore Technologies' MinION™ sequencer and miniPCR bio's miniPCR™ thermal cycler, have been validated onboard the ISS. Here, we report on the development, validation, and implementation of a swab-to-sequencer method that provides a culture-independent solution to real-time microbial profiling onboard the ISS. Method development focused on analysis of swabs collected in a low-biomass environment with limited facility resources and stringent controls on allowed processes and reagents. ISS-optimized procedures included enzymatic DNA extraction from a swab tip, bead-based purifications, altered buffers, and the use of miniPCR and the MinION. Validation was conducted through extensive ground-based assessments comparing current standard culture-dependent and newly developed culture-independent methods. Similar microbial distributions were observed between the two methods; however, as expected, the culture-independent data revealed microbial profiles with greater diversity. Protocol optimization and verification was established during NASA Extreme Environment Mission Operations (NEEMO) analog missions 21 and 22, respectively. Unique microbial profiles obtained from analog testing validated the swab-to-sequencer method in an extreme environment. Finally, four independent swab-to-sequencer experiments were conducted onboard the ISS by two crewmembers. Microorganisms identified from ISS swabs were consistent with historical culture-based data, and primarily consisted of commonly observed human-associated microbes. This simplified method has been streamlined for high ease-of-use for a non-trained crew to complete in an extreme environment, thereby enabling environmental and human health diagnostics in real-time as future missions take us beyond low-Earth orbit.


Subject(s)
Bacteria/genetics , DNA, Bacterial/genetics , Nanopore Sequencing , Sequence Analysis, DNA , Spacecraft , Specimen Handling , Humans
12.
Methods Mol Biol ; 2186: 145-155, 2021.
Article in English | MEDLINE | ID: mdl-32918735

ABSTRACT

Proteins present a significant challenge for nanopore-based sequence analysis. This is partly due to their stable tertiary structures that must be unfolded for linear translocation, and the absence of regular charge density. To address these challenges, here we describe how ClpXP, an ATP-dependent protein unfoldase, can be harnessed to unfold and processively translocate multi-domain protein substrates through an alpha-hemolysin nanopore sensor. This process results in ionic current patterns that are diagnostic of protein sequence and structure at the single-molecule level.


Subject(s)
Endopeptidase Clp/metabolism , Hemolysin Proteins/chemistry , Hemolysin Proteins/metabolism , Lipid Bilayers/metabolism , Nanopores , Protein Unfolding , Protein Transport
13.
Nat Biotechnol ; 38(9): 1044-1053, 2020 09.
Article in English | MEDLINE | ID: mdl-32686750

ABSTRACT

De novo assembly of a human genome using nanopore long-read sequences has been reported, but it used more than 150,000 CPU hours and weeks of wall-clock time. To enable rapid human genome assembly, we present Shasta, a de novo long-read assembler, and polishing algorithms named MarginPolish and HELEN. Using a single PromethION nanopore sequencer and our toolkit, we assembled 11 highly contiguous human genomes de novo in 9 d. We achieved roughly 63× coverage, 42-kb read N50 values and 6.5× coverage in reads >100 kb using three flow cells per sample. Shasta produced a complete haploid human genome assembly in under 6 h on a single commercial compute node. MarginPolish and HELEN polished haploid assemblies to more than 99.9% identity (Phred quality score QV = 30) with nanopore reads alone. Addition of proximity-ligation sequencing enabled near chromosome-level scaffolds for all 11 genomes. We compare our assembly performance to existing methods for diploid, haploid and trio-binned human samples and report superior accuracy and speed.


Subject(s)
Genome, Human/genetics , High-Throughput Nucleotide Sequencing/methods , Nanopore Sequencing , Sequence Analysis, DNA/methods , Algorithms , Benchmarking , Chromosomes, Human/genetics , Deep Learning , Genomics , HLA Antigens/genetics , Haploidy , High-Throughput Nucleotide Sequencing/standards , Humans , Sequence Analysis, DNA/standards
14.
Genome Biol ; 21(1): 83, 2020 03 31.
Article in English | MEDLINE | ID: mdl-32234056

ABSTRACT

BACKGROUND: Long non-coding RNAs (lncRNAs) exhibit highly cell type-specific expression and function, making this class of transcript attractive for targeted cancer therapy. However, the vast majority of lncRNAs have not been tested as potential therapeutic targets, particularly in the context of currently used cancer treatments. Malignant glioma is rapidly fatal, and ionizing radiation is part of the current standard-of-care used to slow tumor growth in both adult and pediatric patients. RESULTS: We use CRISPR interference (CRISPRi) to screen 5689 lncRNA loci in human glioblastoma (GBM) cells, identifying 467 hits that modify cell growth in the presence of clinically relevant doses of fractionated radiation. Thirty-three of these lncRNA hits sensitize cells to radiation, and based on their expression in adult and pediatric gliomas, nine of these hits are prioritized as lncRNA Glioma Radiation Sensitizers (lncGRS). Knockdown of lncGRS-1, a primate-conserved, nuclear-enriched lncRNA, inhibits the growth and proliferation of primary adult and pediatric glioma cells, but not the viability of normal brain cells. Using human brain organoids comprised of mature neural cell types as a three-dimensional tissue substrate to model the invasive growth of glioma, we find that antisense oligonucleotides targeting lncGRS-1 selectively decrease tumor growth and sensitize glioma cells to radiation therapy. CONCLUSIONS: These studies identify lncGRS-1 as a glioma-specific therapeutic target and establish a generalizable approach to rapidly identify novel therapeutic targets in the vast non-coding genome to enhance radiation therapy.


Subject(s)
Brain Neoplasms/therapy , CRISPR-Cas Systems , Glioblastoma/therapy , RNA, Long Noncoding/antagonists & inhibitors , Adult , Astrocytes , Brain , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/radiotherapy , Cell Line, Tumor , Combined Modality Therapy , Glioblastoma/genetics , Glioblastoma/pathology , Glioblastoma/radiotherapy , Humans , Oligonucleotides, Antisense , Organoids , Radiation Tolerance
15.
Genes (Basel) ; 11(1)2020 01 09.
Article in English | MEDLINE | ID: mdl-31936690

ABSTRACT

The MinION sequencer has made in situ sequencing feasible in remote locations. Following our initial demonstration of its high performance off planet with Earth-prepared samples, we developed and tested an end-to-end, sample-to-sequencer process that could be conducted entirely aboard the International Space Station (ISS). Initial experiments demonstrated the process with a microbial mock community standard. The DNA was successfully amplified, primers were degraded, and libraries prepared and sequenced. The median percent identities for both datasets were 84%, as assessed from alignment of the mock community. The ability to correctly identify the organisms in the mock community standard was comparable for the sequencing data obtained in flight and on the ground. To validate the process on microbes collected from and cultured aboard the ISS, bacterial cells were selected from a NASA Environmental Health Systems Surface Sample Kit contact slide. The locations of bacterial colonies chosen for identification were labeled, and a small number of cells were directly added as input into the sequencing workflow. Prepared DNA was sequenced, and the data were downlinked to Earth. Return of the contact slide to the ground allowed for standard laboratory processing for bacterial identification. The identifications obtained aboard the ISS, Staphylococcus hominis and Staphylococcus capitis, matched those determined on the ground down to the species level. This marks the first ever identification of microbes entirely off Earth, and this validated process could be used for in-flight microbial identification, diagnosis of infectious disease in a crewmember, and as a research platform for investigators around the world.


Subject(s)
Nanopore Sequencing/methods , RNA, Ribosomal, 16S/genetics , Specimen Handling/methods , Bacteria/genetics , DNA, Bacterial/genetics , DNA, Ribosomal/genetics , Exobiology/methods , Extraterrestrial Environment , Genome, Bacterial/genetics , Microbiota/genetics , Nanopores , Sequence Analysis, DNA/methods , Spacecraft/instrumentation
17.
Nat Methods ; 16(12): 1297-1305, 2019 12.
Article in English | MEDLINE | ID: mdl-31740818

ABSTRACT

High-throughput complementary DNA sequencing technologies have advanced our understanding of transcriptome complexity and regulation. However, these methods lose information contained in biological RNA because the copied reads are often short and modifications are not retained. We address these limitations using a native poly(A) RNA sequencing strategy developed by Oxford Nanopore Technologies. Our study generated 9.9 million aligned sequence reads for the human cell line GM12878, using thirty MinION flow cells at six institutions. These native RNA reads had a median length of 771 bases, and a maximum aligned length of over 21,000 bases. Mitochondrial poly(A) reads provided an internal measure of read-length quality. We combined these long nanopore reads with higher accuracy short-reads and annotated GM12878 promoter regions to identify 33,984 plausible RNA isoforms. We describe strategies for assessing 3' poly(A) tail length, base modifications and transcript haplotypes.


Subject(s)
Nanopore Sequencing/methods , Poly A/genetics , Sequence Analysis, RNA/methods , Transcriptome , Cells, Cultured , Humans
18.
PLoS One ; 14(5): e0216709, 2019.
Article in English | MEDLINE | ID: mdl-31095620

ABSTRACT

The ribosome small subunit is expressed in all living cells. It performs numerous essential functions during translation, including formation of the initiation complex and proofreading of base-pairs between mRNA codons and tRNA anticodons. The core constituent of the small ribosomal subunit is a ~1.5 kb RNA strand in prokaryotes (16S rRNA) and a homologous ~1.8 kb RNA strand in eukaryotes (18S rRNA). Traditional sequencing-by-synthesis (SBS) of rRNA genes or rRNA cDNA copies has achieved wide use as a 'molecular chronometer' for phylogenetic studies, and as a tool for identifying infectious organisms in the clinic. However, epigenetic modifications on rRNA are erased by SBS methods. Here we describe direct MinION nanopore sequencing of individual, full-length 16S rRNA absent reverse transcription or amplification. As little as 5 picograms (~10 attomole) of purified E. coli 16S rRNA was detected in 4.5 micrograms of total human RNA. Nanopore ionic current traces that deviated from canonical patterns revealed conserved E. coli 16S rRNA 7-methylguanosine and pseudouridine modifications, and a 7-methylguanosine modification that confers aminoglycoside resistance to some pathological E. coli strains.


Subject(s)
Nanopores , RNA, Ribosomal, 16S/genetics , Sequence Analysis, RNA/methods , Escherichia coli/genetics , RNA, Bacterial/genetics
19.
Nat Biotechnol ; 36(4): 321-323, 2018 04.
Article in English | MEDLINE | ID: mdl-29553574

ABSTRACT

The human genome reference sequence remains incomplete owing to the challenge of assembling long tracts of near-identical tandem repeats in centromeres. We implemented a nanopore sequencing strategy to generate high-quality reads that span hundreds of kilobases of highly repetitive DNA in a human Y chromosome centromere. Combining these data with short-read variant validation, we assembled and characterized the centromeric region of a human Y chromosome.


Subject(s)
Centromere/genetics , Chromosomes, Human, Y/genetics , High-Throughput Nucleotide Sequencing , Tandem Repeat Sequences/genetics , Genome, Human/genetics , Humans , Nanopores , Repetitive Sequences, Nucleic Acid/genetics
20.
Nat Commun ; 8: 16027, 2017 07 19.
Article in English | MEDLINE | ID: mdl-28722025

ABSTRACT

Understanding gene regulation and function requires a genome-wide method capable of capturing both gene expression levels and isoform diversity at the single-cell level. Short-read RNAseq is limited in its ability to resolve complex isoforms because it fails to sequence full-length cDNA copies of RNA molecules. Here, we investigate whether RNAseq using the long-read single-molecule Oxford Nanopore MinION sequencer is able to identify and quantify complex isoforms without sacrificing accurate gene expression quantification. After benchmarking our approach, we analyse individual murine B1a cells using a custom multiplexing strategy. We identify thousands of unannotated transcription start and end sites, as well as hundreds of alternative splicing events in these B1a cells. We also identify hundreds of genes expressed across B1a cells that display multiple complex isoforms, including several B cell-specific surface receptors. Our results show that we can identify and quantify complex isoforms at the single cell level.


Subject(s)
B-Lymphocytes/metabolism , Gene Expression Profiling/methods , Receptors, Cell Surface/metabolism , Single-Cell Analysis/methods , Animals , Benchmarking , Mice, Inbred C57BL , Protein Isoforms/metabolism , Sequence Analysis, DNA , Sequence Analysis, RNA , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL
...