Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
STAR Protoc ; 5(2): 102966, 2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38512867

ABSTRACT

Studying RNA splicing factor mutations is challenging due to difficulties in distinguishing wild-type and mutant cells within complex human tissues and inaccuracies associated with reconstructing splicing signals from short-read sequencing data. Here, we present Genotyping of Transcriptomes (GoT)-Splice, a protocol that overcomes these limitations by combining GoT with enhanced long-read single-cell transcriptome and cell-surface proteomics profiling. We describe steps for long-read library preparation and analysis, followed by cDNA re-amplification, enrichment of mutation of interest, sample indexing, and GoT library preparation. For complete details on the use and execution of this protocol, please refer to Cortés-López et al.1.

2.
Cell Stem Cell ; 30(12): 1658-1673.e10, 2023 12 07.
Article in English | MEDLINE | ID: mdl-38065069

ABSTRACT

Stem cells regulate their self-renewal and differentiation fate outcomes through both symmetric and asymmetric divisions. m6A RNA methylation controls symmetric commitment and inflammation of hematopoietic stem cells (HSCs) through unknown mechanisms. Here, we demonstrate that the nuclear speckle protein SON is an essential m6A target required for murine HSC self-renewal, symmetric commitment, and inflammation control. Global profiling of m6A identified that m6A mRNA methylation of Son increases during HSC commitment. Upon m6A depletion, Son mRNA increases, but its protein is depleted. Reintroduction of SON rescues defects in HSC symmetric commitment divisions and engraftment. Conversely, Son deletion results in a loss of HSC fitness, while overexpression of SON improves mouse and human HSC engraftment potential by increasing quiescence. Mechanistically, we found that SON rescues MYC and suppresses the METTL3-HSC inflammatory gene expression program, including CCL5, through transcriptional regulation. Thus, our findings define a m6A-SON-CCL5 axis that controls inflammation and HSC fate.


Subject(s)
DNA-Binding Proteins , Hematopoietic Stem Cells , Inflammation , RNA Methylation , Animals , Humans , Mice , Cell Differentiation/genetics , Hematopoietic Stem Cells/metabolism , Methylation , Methyltransferases/genetics , Methyltransferases/metabolism , RNA, Messenger/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , RNA Methylation/genetics
3.
Cell Stem Cell ; 30(9): 1262-1281.e8, 2023 09 07.
Article in English | MEDLINE | ID: mdl-37582363

ABSTRACT

RNA splicing factors are recurrently mutated in clonal blood disorders, but the impact of dysregulated splicing in hematopoiesis remains unclear. To overcome technical limitations, we integrated genotyping of transcriptomes (GoT) with long-read single-cell transcriptomics and proteogenomics for single-cell profiling of transcriptomes, surface proteins, somatic mutations, and RNA splicing (GoT-Splice). We applied GoT-Splice to hematopoietic progenitors from myelodysplastic syndrome (MDS) patients with mutations in the core splicing factor SF3B1. SF3B1mut cells were enriched in the megakaryocytic-erythroid lineage, with expansion of SF3B1mut erythroid progenitor cells. We uncovered distinct cryptic 3' splice site usage in different progenitor populations and stage-specific aberrant splicing during erythroid differentiation. Profiling SF3B1-mutated clonal hematopoiesis samples revealed that erythroid bias and cell-type-specific cryptic 3' splice site usage in SF3B1mut cells precede overt MDS. Collectively, GoT-Splice defines the cell-type-specific impact of somatic mutations on RNA splicing, from early clonal outgrowths to overt neoplasia, directly in human samples.


Subject(s)
Myelodysplastic Syndromes , RNA Splice Sites , Humans , Multiomics , RNA Splicing/genetics , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , Mutation/genetics , Phosphoproteins/genetics , Phosphoproteins/metabolism
4.
Oncoimmunology ; 12(1): 2184143, 2023.
Article in English | MEDLINE | ID: mdl-36875548

ABSTRACT

Despite massive improvements in the treatment of B-ALL through CART-19 immunotherapy, a large number of patients suffer a relapse due to loss of the targeted epitope. Mutations in the CD19 locus and aberrant splicing events are known to account for the absence of surface antigen. However, early molecular determinants suggesting therapy resistance as well as the time point when first signs of epitope loss appear to be detectable are not enlightened so far. By deep sequencing of the CD19 locus, we identified a blast-specific 2-nucleotide deletion in intron 2 that exists in 35% of B-ALL samples at initial diagnosis. This deletion overlaps with the binding site of RNA binding proteins (RBPs) including PTBP1 and might thereby affect CD19 splicing. Moreover, we could identify a number of other RBPs that are predicted to bind to the CD19 locus being deregulated in leukemic blasts, including NONO. Their expression is highly heterogeneous across B-ALL molecular subtypes as shown by analyzing 706 B-ALL samples accessed via the St. Jude Cloud. Mechanistically, we show that downregulation of PTBP1, but not of NONO, in 697 cells reduces CD19 total protein by increasing intron 2 retention. Isoform analysis in patient samples revealed that blasts, at diagnosis, express increased amounts of CD19 intron 2 retention compared to normal B cells. Our data suggest that loss of RBP functionality by mutations altering their binding motifs or by deregulated expression might harbor the potential for the disease-associated accumulation of therapy-resistant CD19 isoforms.


Subject(s)
Antigens, CD19 , Heterogeneous-Nuclear Ribonucleoproteins , Leukemia, B-Cell , Polypyrimidine Tract-Binding Protein , RNA-Binding Proteins , Humans , Binding Sites , Epitopes , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Mutation , Polypyrimidine Tract-Binding Protein/genetics , RNA-Binding Proteins/genetics , Leukemia, B-Cell/genetics
5.
Nat Commun ; 13(1): 5570, 2022 09 22.
Article in English | MEDLINE | ID: mdl-36138008

ABSTRACT

Following CART-19 immunotherapy for B-cell acute lymphoblastic leukaemia (B-ALL), many patients relapse due to loss of the cognate CD19 epitope. Since epitope loss can be caused by aberrant CD19 exon 2 processing, we herein investigate the regulatory code that controls CD19 splicing. We combine high-throughput mutagenesis with mathematical modelling to quantitatively disentangle the effects of all mutations in the region comprising CD19 exons 1-3. Thereupon, we identify ~200 single point mutations that alter CD19 splicing and thus could predispose B-ALL patients to developing CART-19 resistance. Furthermore, we report almost 100 previously unknown splice isoforms that emerge from cryptic splice sites and likely encode non-functional CD19 proteins. We further identify cis-regulatory elements and trans-acting RNA-binding proteins that control CD19 splicing (e.g., PTBP1 and SF3B4) and validate that loss of these factors leads to pervasive CD19 mis-splicing. Our dataset represents a comprehensive resource for identifying predictive biomarkers for CART-19 therapy.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma , RNA Splice Sites , Alternative Splicing/genetics , Antigens, CD19/genetics , Antigens, CD19/metabolism , Epitopes/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , Mutagenesis/genetics , Mutation , Neoplasm Recurrence, Local/genetics , Polypyrimidine Tract-Binding Protein/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Protein Isoforms/genetics , RNA Splicing , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
6.
Genome Biol ; 22(1): 190, 2021 06 28.
Article in English | MEDLINE | ID: mdl-34183059

ABSTRACT

Resistance to CD19-directed immunotherapies in lymphoblastic leukemia has been attributed, among other factors, to several aberrant CD19 pre-mRNA splicing events, including recently reported excision of a cryptic intron embedded within CD19 exon 2. While "exitrons" are known to exist in hundreds of human transcripts, we discovered, using reporter assays and direct long-read RNA sequencing (dRNA-seq), that the CD19 exitron is an artifact of reverse transcription. Extending our analysis to publicly available datasets, we identified dozens of questionable exitrons, dubbed "falsitrons," that appear only in cDNA-seq, but never in dRNA-seq. Our results highlight the importance of dRNA-seq for transcript isoform validation.


Subject(s)
Alternative Splicing , Artifacts , RNA, Messenger/genetics , Receptors, Antigen, T-Cell/genetics , Reverse Transcription , Antibodies, Bispecific/pharmacology , Antineoplastic Agents, Immunological/pharmacology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Base Pairing , Base Sequence , Cell Line, Tumor , Datasets as Topic , Exons , High-Throughput Nucleotide Sequencing , Humans , Immunotherapy/methods , Introns , Models, Biological , Nucleic Acid Conformation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/immunology , RNA, Messenger/chemistry , RNA, Messenger/immunology , Receptors, Antigen, T-Cell/immunology
7.
Nat Commun ; 9(1): 3315, 2018 08 17.
Article in English | MEDLINE | ID: mdl-30120239

ABSTRACT

Mutations causing aberrant splicing are frequently implicated in human diseases including cancer. Here, we establish a high-throughput screen of randomly mutated minigenes to decode the cis-regulatory landscape that determines alternative splicing of exon 11 in the proto-oncogene MST1R (RON). Mathematical modelling of splicing kinetics enables us to identify more than 1000 mutations affecting RON exon 11 skipping, which corresponds to the pathological isoform RON∆165. Importantly, the effects correlate with RON alternative splicing in cancer patients bearing the same mutations. Moreover, we highlight heterogeneous nuclear ribonucleoprotein H (HNRNPH) as a key regulator of RON splicing in healthy tissues and cancer. Using iCLIP and synergy analysis, we pinpoint the functionally most relevant HNRNPH binding sites and demonstrate how cooperative HNRNPH binding facilitates a splicing switch of RON exon 11. Our results thereby offer insights into splicing regulation and the impact of mutations on alternative splicing in cancer.


Subject(s)
Alternative Splicing/genetics , Mutagenesis/genetics , Neoplasms/genetics , Receptor Protein-Tyrosine Kinases/genetics , Base Sequence , Binding Sites , Exons/genetics , HEK293 Cells , Heterogeneous-Nuclear Ribonucleoprotein Group F-H/metabolism , Humans , Introns/genetics , Linear Models , MCF-7 Cells , Mutation/genetics , Proto-Oncogene Mas , RNA-Binding Proteins/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Sequence Analysis, RNA
8.
Methods Mol Biol ; 1724: 27-41, 2018.
Article in English | MEDLINE | ID: mdl-29322438

ABSTRACT

The genome-wide expression patterns of circular RNAs (circRNAs) are of increasing interest for their potential roles in normal cellular homeostasis, development, and disease. Thousands of circRNAs have been annotated from various species in recent years. Analysis of publically available or user-generated rRNA-depleted total RNA-seq data can be performed to uncover new circRNA expression trends. Here we provide a primer for profiling circRNAs from RNA-seq datasets. The description is tailored for the wet lab scientist with limited or no experience in analyzing RNA-seq data. We begin by describing how to access and interpret circRNA annotations. Next, we cover converting circRNA annotations into junction sequences that are used as scaffolds to align RNA-seq reads. Lastly, we visit quantifying circRNA expression trends from the alignment data.


Subject(s)
Computational Biology/methods , Gene Expression Regulation , Genome, Human , High-Throughput Nucleotide Sequencing/methods , RNA/genetics , Sequence Analysis, RNA/methods , Humans , RNA, Circular
9.
BMC Genomics ; 19(1): 8, 2018 01 03.
Article in English | MEDLINE | ID: mdl-29298683

ABSTRACT

BACKGROUND: Circular RNAs (CircRNAs) are a newly appreciated class of RNAs that lack free 5' and 3' ends, are expressed by the thousands in diverse forms of life, and are mostly of enigmatic function. Ostensibly due to their resistance to exonucleases, circRNAs are known to be exceptionally stable. Previous work in Drosophila and mice have shown that circRNAs increase during aging in neural tissues. RESULTS: Here, we examined the global profile of circRNAs in C. elegans during aging by performing ribo-depleted total RNA-seq from the fourth larval stage (L4) through 10-day old adults. Using stringent bioinformatic criteria and experimental validation, we annotated a high-confidence set of 1166 circRNAs, including 575 newly discovered circRNAs. These circRNAs were derived from 797 genes with diverse functions, including genes involved in the determination of lifespan. A massive accumulation of circRNAs during aging was uncovered. Many hundreds of circRNAs were significantly increased among the aging time-points and increases of select circRNAs by over 40-fold during aging were quantified by RT-qPCR. The expression of 459 circRNAs was determined to be distinct from the expression of linear RNAs from the same host genes, demonstrating host gene independence of circRNA age-accumulation. CONCLUSIONS: We attribute the global scale of circRNA age-accumulation to the high composition of post-mitotic cells in adult C. elegans, coupled with the high resistance of circRNAs to decay. These findings suggest that the exceptional stability of circRNAs might explain age-accumulation trends observed from neural tissues of other organisms, which also have a high composition of post-mitotic cells. Given the suitability of C. elegans for aging research, it is now poised as an excellent model system to determine whether there are functional consequences of circRNA accumulation during aging.


Subject(s)
Aging/genetics , Caenorhabditis elegans/genetics , RNA/metabolism , Animals , Caenorhabditis elegans/metabolism , Gene Expression Profiling , Genomics , High-Throughput Nucleotide Sequencing , RNA, Circular , Sequence Analysis, RNA
10.
Elife ; 62017 06 06.
Article in English | MEDLINE | ID: mdl-28583253

ABSTRACT

Neuromuscular junction degeneration is a prominent aspect of sarcopenia, the age-associated loss of skeletal muscle integrity. Previously, we showed that muscle stem cells activate and contribute to mouse neuromuscular junction regeneration in response to denervation (Liu et al., 2015). Here, we examined gene expression profiles and neuromuscular junction integrity in aged mouse muscles, and unexpectedly found limited denervation despite a high level of degenerated neuromuscular junctions. Instead, degenerated neuromuscular junctions were associated with reduced contribution from muscle stem cells. Indeed, muscle stem cell depletion was sufficient to induce neuromuscular junction degeneration at a younger age. Conversely, prevention of muscle stem cell and derived myonuclei loss was associated with attenuation of age-related neuromuscular junction degeneration, muscle atrophy, and the promotion of aged muscle force generation. Our observations demonstrate that deficiencies in muscle stem cell fate and post-synaptic myogenesis provide a cellular basis for age-related neuromuscular junction degeneration and associated skeletal muscle decline.


Subject(s)
Aging/pathology , Muscle, Skeletal/pathology , Neuromuscular Junction/pathology , Sarcopenia/pathology , Stem Cells/physiology , Animals , Mice
11.
Yale J Biol Med ; 89(4): 527-537, 2016 12.
Article in English | MEDLINE | ID: mdl-28018143

ABSTRACT

Many thousands of Circular RNAs (circRNAs) have recently been identified in metazoan genomes by transcriptome-wide sequencing. Most circRNAs are generated by back-splicing events from exons of protein-coding genes. A great deal of progress has recently been made in understanding the genome-wide expression patterns, biogenesis, and regulation of circRNAs. To date, however, few functions of circRNAs have been identified. CircRNAs are preferentially expressed in neural tissues and some are found at synapses, suggesting possible functions in the nervous system. Several circRNAs have been shown to function as microRNA "sponges" to counteract microRNA mediated repression of mRNA. New functions for circRNAs are arising, including protein sequestration, transcriptional regulation, and potential functions in cancer. Here, we highlight the recent progress made in understanding the biogenesis and regulation of circRNAs, discuss newly uncovered circRNA functions, and explain the methodological approaches that could reveal more exciting and unexpected roles for these RNAs.


Subject(s)
RNA/genetics , Alternative Splicing/genetics , Animals , Exons/genetics , Gene Expression Regulation/genetics , Humans , MicroRNAs/genetics , RNA, Circular , RNA, Untranslated/genetics
12.
Sci Rep ; 6: 38907, 2016 12 13.
Article in English | MEDLINE | ID: mdl-27958329

ABSTRACT

Circular RNAs (circRNAs) are a newly appreciated class of RNAs expressed across diverse phyla. These enigmatic transcripts are most commonly generated by back-splicing events from exons of protein-coding genes. This results in highly stable RNAs due to the lack of free 5' and 3' ends. CircRNAs are enriched in neural tissues, suggesting that they might have neural functions. Here, we sought to determine whether circRNA accumulation occurs during aging in mice. Total RNA-seq profiling of young (1 month old) and aged (22 month old) cortex, hippocampus and heart samples was performed. This led to the confident detection of 6,791 distinct circRNAs across these samples, including 675 novel circRNAs. Analysis uncovered a strong bias for circRNA upregulation during aging in neural tissues. These age-accumulation trends were verified for individual circRNAs by RT-qPCR and Northern analysis. In contrast, comparison of aged versus young hearts failed to reveal a global trend for circRNA upregulation. Age-accumulation of circRNAs in brain tissues was found to be largely independent from linear RNA expression of host genes. These findings suggest that circRNAs might play biological roles relevant to the aging nervous system.


Subject(s)
Aging , Cerebral Cortex/metabolism , Hippocampus/metabolism , Myocardium/metabolism , RNA/metabolism , Animals , Exons , Gene Expression , Mice, Inbred C57BL , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...