Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters










Publication year range
1.
RNA Biol ; 19(1): 829-840, 2022 01.
Article in English | MEDLINE | ID: mdl-35723015

ABSTRACT

Alternative splicing enables higher eukaryotes to expand mRNA diversity from a finite number of genes through highly combinatorial splice site selection mechanisms that are influenced by the sequence of competing splice sites, cis-regulatory elements binding trans-acting factors, the length of exons and introns harbouring alternative splice sites and RNA secondary structures at putative splice junctions. To test the hypothesis that the intron definition or exon definition modes of splice site recognition direct the selection of alternative splice patterns, we created a database of alternative splice site usage (ALTssDB). When alternative splice sites are embedded within short introns (intron definition), the 5' and 3' splice sites closest to each other across the intron preferentially pair, consistent with previous observations. However, when alternative splice sites are embedded within large flanking introns (exon definition), the 5' and 3' splice sites closest to each other across the exon are preferentially selected. Thus, alternative splicing decisions are influenced by the intron and exon definition modes of splice site recognition. The results demonstrate that the spliceosome pairs splice sites that are closest in proximity within the unit of initial splice site selection.


Subject(s)
RNA Splice Sites , RNA Splicing , Alternative Splicing , Exons , Introns
2.
PLoS One ; 16(12): e0257503, 2021.
Article in English | MEDLINE | ID: mdl-34898625

ABSTRACT

Metabolic labeling is a widely used tool to investigate different aspects of pre-mRNA splicing and RNA turnover. The labeling technology takes advantage of native cellular machineries where a nucleotide analog is readily taken up and incorporated into nascent RNA. One such analog is 4-thiouridine (4sU). Previous studies demonstrated that the uptake of 4sU at elevated concentrations (>50µM) and extended exposure led to inhibition of rRNA synthesis and processing, presumably induced by changes in RNA secondary structure. Thus, it is possible that 4sU incorporation may also interfere with splicing efficiency. To test this hypothesis, we carried out splicing analyses of pre-mRNA substrates with varying levels of 4sU incorporation (0-100%). We demonstrate that increased incorporation of 4sU into pre-mRNAs decreased splicing efficiency. The overall impact of 4sU labeling on pre-mRNA splicing efficiency negatively correlates with the strength of splice site signals such as the 3' and the 5' splice sites. Introns with weaker splice sites are more affected by the presence of 4sU. We also show that transcription by T7 polymerase and pre-mRNA degradation kinetics were impacted at the highest levels of 4sU incorporation. Increased incorporation of 4sU caused elevated levels of abortive transcripts, and fully labeled pre-mRNA is more stable than its uridine-only counterpart. Cell culture experiments show that a small number of alternative splicing events were modestly, but statistically significantly influenced by metabolic labeling with 4sU at concentrations considered to be tolerable (40 µM). We conclude that at high 4sU incorporation rates small, but noticeable changes in pre-mRNA splicing can be detected when splice sites deviate from consensus. Given these potential 4sU artifacts, we suggest that appropriate controls for metabolic labeling experiments need to be included in future labeling experiments.


Subject(s)
Alternative Splicing/drug effects , RNA Precursors/metabolism , RNA Splice Sites , Thiouridine/pharmacology , HEK293 Cells , Humans , Nucleic Acid Conformation , RNA Precursors/genetics , RNA Stability/drug effects , Staining and Labeling
4.
Commun Biol ; 3(1): 453, 2020 08 19.
Article in English | MEDLINE | ID: mdl-32814826

ABSTRACT

Intestinal stem cells are non-quiescent, dividing epithelial cells that rapidly differentiate into progenitor cells of the absorptive and secretory cell lineages. The kinetics of this process is rapid such that the epithelium is replaced weekly. To determine how the transcriptome and proteome keep pace with rapid differentiation, we developed a new cell sorting method to purify mouse colon epithelial cells. Here we show that alternative mRNA splicing and polyadenylation dominate changes in the transcriptome as stem cells differentiate into progenitors. In contrast, as progenitors differentiate into mature cell types, changes in mRNA levels dominate the transcriptome. RNA processing targets regulators of cell cycle, RNA, cell adhesion, SUMOylation, and Wnt and Notch signaling. Additionally, global proteome profiling detected >2,800 proteins and revealed RNA:protein patterns of abundance and correlation. Paired together, these data highlight new potentials for autocrine and feedback regulation and provide new insights into cell state transitions in the crypt.


Subject(s)
Cell Differentiation , Cell Self Renewal , Colon , Enterocytes/metabolism , Proteome , Stem Cells/metabolism , Transcriptome , Animals , Biomarkers , Cell Self Renewal/genetics , Computational Biology/methods , Enterocytes/cytology , Gene Expression Profiling , Gene Expression Regulation, Developmental , Immunophenotyping , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Mice , Proteomics , RNA Processing, Post-Transcriptional , Stem Cells/cytology
5.
RNA ; 26(10): 1389-1399, 2020 10.
Article in English | MEDLINE | ID: mdl-32522889

ABSTRACT

Alternative splicing is responsible for much of the transcriptomic and proteomic diversity observed in eukaryotes and involves combinatorial regulation by many cis-acting elements and trans-acting factors. SR and hnRNP splicing regulatory proteins often have opposing effects on splicing efficiency depending on where they bind the pre-mRNA relative to the splice site. Position-dependent splicing repression occurs at spliceosomal E-complex, suggesting that U1 snRNP binds but cannot facilitate higher order spliceosomal assembly. To test the hypothesis that the structure of U1 snRNA changes during activation or repression, we developed a method to structure-probe native U1 snRNP in enriched conformations that mimic activated or repressed spliceosomal E-complexes. While the core of U1 snRNA is highly structured, the 5' end of U1 snRNA shows different SHAPE reactivities and psoralen crosslinking efficiencies depending on where splicing regulatory elements are located relative to the 5' splice site. A motif within the 5' splice site binding region of U1 snRNA is more reactive toward SHAPE electrophiles when repressors are bound, suggesting U1 snRNA is bound, but less base-paired. These observations demonstrate that splicing regulators modulate splice site selection allosterically.


Subject(s)
Allosteric Regulation/genetics , Alternative Splicing/genetics , RNA, Small Nuclear/genetics , Ribonucleoprotein, U1 Small Nuclear/genetics , Spliceosomes/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Proteomics/methods , RNA Precursors/genetics , RNA Splice Sites/genetics , RNA, Messenger/genetics
6.
PLoS One ; 14(10): e0223132, 2019.
Article in English | MEDLINE | ID: mdl-31581208

ABSTRACT

Alternative splicing diversifies mRNA transcripts in human cells. While the spliceosome pairs exons with a high degree of accuracy, the rates of rare aberrant and non-canonical pre-mRNA splicing have not been evaluated at the nucleotide level to determine the quantity and identity of these events across splice junctions. Using ultra-deep sequencing the frequency of aberrant and non-canonical splicing events for three splice junctions flanking exon 7 of SMN1 were determined at single nucleotide resolution. After correction for background noise introduced by PCR amplification and sequencing steps, pre-mRNA splicing was shown to maintain a low overall rate of aberrant and non-canonically spliced events. Several previously unannotated splicing events across 3 exon|intron junctions in SMN1 were identified. Mutations within SMN exon 7 were shown to affect splicing fidelity by modulating RNA secondary structures, by altering the binding site of regulatory proteins and by changing the 5' splice site strength. Mutations also create a truncated SMN1 exon 7 through the introduction of a de novo non-canonical 5' splice site. The results from the ultra-deep sequencing approach highlight the impressive fidelity of pre-mRNA splicing and demonstrate that the immediate sequence context around splice sites is the main driving force behind non-canonical splice site pairing.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , RNA Precursors/genetics , RNA Splicing/genetics , Base Sequence , Binding Sites , DNA/genetics , Exons/genetics , Humans , Introns/genetics , Mutation/genetics , Mutation Rate , RNA Precursors/metabolism , RNA Splice Sites/genetics , RNA, Messenger/genetics , Ribonucleoprotein, U1 Small Nuclear/metabolism
7.
RNA ; 25(12): 1793-1805, 2019 12.
Article in English | MEDLINE | ID: mdl-31554659

ABSTRACT

Pre-mRNA splicing is regulated through multiple trans-acting splicing factors. These regulators interact with the pre-mRNA at intronic and exonic positions. Given that most exons are protein coding, the evolution of exons must be modulated by a combination of selective coding and splicing pressures. It has previously been demonstrated that selective splicing pressures are more easily deconvoluted when phylogenetic comparisons are made for exons of identical size, suggesting that exon size-filtered sequence alignments may improve identification of nucleotides evolved to mediate efficient exon ligation. To test this hypothesis, an exon size database was created, filtering 76 vertebrate sequence alignments based on exon size conservation. In addition to other genomic parameters, such as splice-site strength, gene position, or flanking intron length, this database permits the identification of exons that are size- and/or sequence-conserved. Highly size-conserved exons are always sequence-conserved. However, sequence conservation does not necessitate exon size conservation. Our analysis identified evolutionarily young exons and demonstrated that length conservation is a strong predictor of alternative splicing. A published data set of approximately 5000 exonic SNPs associated with disease was analyzed to test the hypothesis that exon size-filtered sequence comparisons increase detection of splice-altering nucleotides. Improved splice predictions could be achieved when mutations occur at the third codon position, especially when a mutation decreases exon inclusion efficiency. The results demonstrate that coding pressures dominate nucleotide composition at invariable codon positions and that exon size-filtered sequence alignments permit identification of splice-altering nucleotides at wobble positions.


Subject(s)
Alternative Splicing , Base Sequence , Conserved Sequence , Exons , Humans , Nucleotides , Phylogeny , Polymorphism, Single Nucleotide , RNA Precursors/genetics
8.
Biochim Biophys Acta Gene Regul Mech ; 1862(11-12): 194392, 2019.
Article in English | MEDLINE | ID: mdl-31276857

ABSTRACT

The generation of protein coding mRNAs from pre-mRNA is a fundamental biological process that is required for gene expression. Alternative pre-mRNA splicing is responsible for much of the transcriptomic and proteomic diversity observed in higher order eukaryotes. Aberrations that disrupt regular alternative splicing patterns are known to cause human diseases, including various cancers. Alternative splicing is a combinatorial process, meaning many factors affect which two splice sites are ligated together. The features that dictate exon inclusion are comprised of splice site strength, intron-exon architecture, RNA secondary structure, splicing regulatory elements, promoter use and transcription speed by RNA polymerase and the presence of post-transcriptional nucleotide modifications. A comprehensive view of all of the factors that influence alternative splicing decisions is necessary to predict splicing outcomes and to understand the molecular basis of disease. This article is part of a Special Issue entitled: RNA structure and splicing regulation edited by Francisco Baralle, Ravindra Singh and Stefan Stamm.


Subject(s)
Alternative Splicing , RNA, Messenger/metabolism , DNA-Directed RNA Polymerases/metabolism , Gene Expression Regulation , Humans , Nucleic Acid Conformation , RNA Splice Sites , RNA, Messenger/chemistry , Regulatory Elements, Transcriptional
9.
Dev Biol ; 449(1): 14-20, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30771302

ABSTRACT

The embryonic origin of pericytes is heterogeneous, both between and within organs. While pericytes of coelomic organs were proposed to differentiate from the mesothelium, a single-layer squamous epithelium, the embryonic origin of pancreatic pericytes has yet to be reported. Here, we show that adult pancreatic pericytes originate from the embryonic pancreatic mesenchyme. Our analysis indicates that pericytes of the adult mouse pancreas originate from cells expressing the transcription factor Nkx3.2. In the embryonic pancreas, Nkx3.2-expressing cells constitute the multilayered mesenchyme, which surrounds the pancreatic epithelium and supports multiple events in its development. Thus, we traced the fate of the pancreatic mesenchyme. Our analysis reveals that pancreatic mesenchymal cells acquire various pericyte characteristics, including gene expression, typical morphology, and periendothelial location, during embryogenesis. Importantly, we show that the vast majority of pancreatic mesenchymal cells differentiate into pericytes already at embryonic day 13.5 and progressively acquires a more mature pericyte phenotype during later stages of pancreas organogenesis. Thus, our study indicates the embryonic pancreatic mesenchyme as the primary origin to adult pancreatic pericytes. As pericytes of other coelomic organs were suggested to differentiate from the mesothelium, our findings point to a distinct origin of these cells in the pancreas. Thus, our study proposes a complex ontogeny of pericytes of coelomic organs.


Subject(s)
Mesoderm/cytology , Mesoderm/embryology , Pancreas/cytology , Pancreas/embryology , Pericytes/cytology , Animals , Biomarkers/metabolism , Embryonic Development/genetics , Endothelial Cells/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Mice , Receptor, Platelet-Derived Growth Factor beta/metabolism , Transcription Factors/metabolism
10.
Mol Cell ; 69(1): 62-74.e4, 2018 01 04.
Article in English | MEDLINE | ID: mdl-29276085

ABSTRACT

Alternative mRNA processing is a critical mechanism for proteome expansion and gene regulation in higher eukaryotes. The SR family proteins play important roles in splicing regulation. Intriguingly, mammalian genomes encode many poorly characterized SR-like proteins, including subunits of the mRNA 3'-processing factor CFIm, CFIm68 and CFIm59. Here we demonstrate that CFIm functions as an enhancer-dependent activator of mRNA 3' processing. CFIm regulates global alternative polyadenylation (APA) by specifically binding and activating enhancer-containing poly(A) sites (PASs). Importantly, the CFIm activator functions are mediated by the arginine-serine repeat (RS) domains of CFIm68/59, which bind specifically to an RS-like region in the CPSF subunit Fip1, and this interaction is inhibited by CFIm68/59 hyper-phosphorylation. The remarkable functional similarities between CFIm and SR proteins suggest that interactions between RS-like domains in regulatory and core factors may provide a common activation mechanism for mRNA 3' processing, splicing, and potentially other steps in RNA metabolism.


Subject(s)
Alternative Splicing/genetics , Gene Expression Regulation/genetics , Polyadenylation , RNA, Messenger/metabolism , mRNA Cleavage and Polyadenylation Factors/metabolism , Animals , Cell Line , Enhancer Elements, Genetic/genetics , Gene Knockout Techniques , HEK293 Cells , Humans , Phosphorylation , Poly A/metabolism , Protein Domains/genetics , RNA-Binding Proteins/metabolism , Sf9 Cells , Spodoptera
11.
Methods Mol Biol ; 1648: 11-26, 2017.
Article in English | MEDLINE | ID: mdl-28766286

ABSTRACT

The ability to perform in vitro splicing assays has paved the way for in-depth studies of the mechanisms and machinery involved in the process of splicing. The in vitro splicing assay is a valuable experimental approach that combines the complexity of the spliceosome and regulatory systems with the flexibility of performing endless splicing and alternative splicing reactions. Through the use of crude nuclear extract and radiolabeled pre-mRNA, spliced mRNAs can be visualized using autoradiography for downstream analysis. This chapter describes the necessary steps to perform an in vitro splicing reaction, including the generation of the key components necessary for the splicing reaction; nuclear extract.


Subject(s)
Cell Nucleus/chemistry , RNA Precursors/chemistry , RNA Splicing , Spliceosomes/chemistry , Animals , Cell Nucleus/metabolism , Cell-Free System/chemistry , Cell-Free System/metabolism , Complex Mixtures/chemistry , HeLa Cells , Humans , RNA Precursors/metabolism , Spliceosomes/metabolism
12.
Methods Mol Biol ; 1648: 169-176, 2017.
Article in English | MEDLINE | ID: mdl-28766297

ABSTRACT

Isolation of newly transcribed RNA is an invaluable approach that can be used to study the dynamic life of RNA in cellulo. Traditional methods of whole-cell RNA extraction limit subsequent gene expression analyses to the steady-state levels of RNA abundance, which often masks changes in RNA synthesis and processing. This chapter describes a methodology with low cytotoxicity that permits the labeling and isolation of nascent pre-mRNA in cell culture. The resulting isolate is suitable for use in a series of downstream applications aimed at studying changes in RNA synthesis, processing, or stability.


Subject(s)
RNA Precursors , Staining and Labeling/methods , Thiouridine/chemistry , Transcription, Genetic , Animals , Cell Line , Humans , RNA Precursors/biosynthesis , RNA Precursors/chemistry , RNA Precursors/isolation & purification
13.
Mol Cell ; 63(2): 218-228, 2016 07 21.
Article in English | MEDLINE | ID: mdl-27397683

ABSTRACT

Phosphorylation has been generally thought to activate the SR family of splicing factors for efficient splice-site recognition, but this idea is incompatible with an early observation that overexpression of an SR protein kinase, such as the CDC2-like kinase 1 (CLK1), weakens splice-site selection. Here, we report that CLK1 binds SR proteins but lacks the mechanism to release phosphorylated SR proteins, thus functionally inactivating the splicing factors. Interestingly, CLK1 overcomes this dilemma through a symbiotic relationship with the serine-arginine protein kinase 1 (SRPK1). We show that SRPK1 interacts with an RS-like domain in the N terminus of CLK1 to facilitate the release of phosphorylated SR proteins, which then promotes efficient splice-site recognition and subsequent spliceosome assembly. These findings reveal an unprecedented signaling mechanism by which two protein kinases fulfill separate catalytic features that are normally encoded in single kinases to institute phosphorylation control of pre-mRNA splicing in the nucleus.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , RNA Precursors/metabolism , RNA Splicing , RNA, Messenger/metabolism , Spliceosomes/enzymology , Catalysis , HeLa Cells , Humans , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , RNA Interference , RNA Precursors/genetics , RNA, Messenger/genetics , Ribonucleoprotein, U1 Small Nuclear/metabolism , Spliceosomes/genetics , Time Factors , Transfection , beta-Globins/genetics , beta-Globins/metabolism
14.
RNA Biol ; 13(7): 646-55, 2016 07 02.
Article in English | MEDLINE | ID: mdl-27245359

ABSTRACT

Alternative polyadenylation has been implicated as an important regulator of gene expression. In some cases, alternative polyadenylation is known to couple with alternative splicing to influence last intron removal. However, it is unknown whether alternative polyadenylation events influence alternative splicing decisions at upstream exons. Knockdown of the polyadenylation factors CFIm25 or CstF64 in HeLa cells was used as an approach in identifying alternative polyadenylation and alternative splicing events on a genome-wide scale. Although hundreds of alternative splicing events were found to be differentially spliced in the knockdown of CstF64, genes associated with alternative polyadenylation did not exhibit an increased incidence of alternative splicing. These results demonstrate that the coupling between alternative polyadenylation and alternative splicing is usually limited to defining the last exon. The striking influence of CstF64 knockdown on alternative splicing can be explained through its effects on UTR selection of known splicing regulators such as hnRNP A2/B1, thereby indirectly influencing splice site selection. We conclude that changes in the expression of the polyadenylation factor CstF64 influences alternative splicing through indirect effects.


Subject(s)
Alternative Splicing/physiology , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Introns/physiology , Polyadenylation/physiology , mRNA Cleavage and Polyadenylation Factors/metabolism , HeLa Cells , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Humans , mRNA Cleavage and Polyadenylation Factors/genetics
15.
J Biol Chem ; 290(46): 27700-11, 2015 Nov 13.
Article in English | MEDLINE | ID: mdl-26424794

ABSTRACT

Alternative splicing diversifies mRNA transcripts in human cells. This sequence-driven process can be influenced greatly by mutations, even those that do not change the protein coding potential of the transcript. Synonymous mutations have been shown to alter gene expression through modulation of splicing, mRNA stability, and translation. Using a synonymous position mutation library in SMN1 exon 7, we show that 23% of synonymous mutations across the exon decrease exon inclusion, suggesting that nucleotide identity across the entire exon has been evolutionarily optimized to support a particular exon inclusion level. Although phylogenetic conservation scores are insufficient to identify synonymous positions important for exon inclusion, an alignment of organisms filtered based on similar exon/intron architecture is highly successful. Although many of the splicing neutral mutations are observed to occur, none of the exon inclusion reducing mutants was found in the filtered alignment. Using the modified phylogenetic comparison as an approach to evaluate the impact on pre-mRNA splicing suggests that up to 45% of synonymous SNPs are likely to alter pre-mRNA splicing. These results demonstrate that coding and pre-mRNA splicing pressures co-evolve and that a modified phylogenetic comparison based on the exon/intron architecture is a useful tool in identifying splice altering SNPs.


Subject(s)
Alternative Splicing/genetics , Mutation , RNA, Messenger/genetics , Animals , Base Sequence , DNA Mutational Analysis , Exons , Gene Library , HeLa Cells , Humans , Introns , Molecular Sequence Data , Phylogeny , RNA Precursors/genetics , Survival of Motor Neuron 1 Protein/genetics
16.
RNA ; 21(5): 813-23, 2015 May.
Article in English | MEDLINE | ID: mdl-25805853

ABSTRACT

Alternative splicing is a key player in the creation of complex mammalian transcriptomes and its misregulation is associated with many human diseases. Multiple mRNA isoforms are generated from most human genes, a process mediated by the interplay of various RNA signature elements and trans-acting factors that guide spliceosomal assembly and intron removal. Here, we introduce a splicing predictor that evaluates hundreds of RNA features simultaneously to successfully differentiate between exons that are constitutively spliced, exons that undergo alternative 5' or 3' splice-site selection, and alternative cassette-type exons. Surprisingly, the splicing predictor did not feature strong discriminatory contributions from binding sites for known splicing regulators. Rather, the ability of an exon to be involved in one or multiple types of alternative splicing is dictated by its immediate sequence context, mainly driven by the identity of the exon's splice sites, the conservation around them, and its exon/intron architecture. Thus, the splicing behavior of human exons can be reliably predicted based on basic RNA sequence elements.


Subject(s)
Alternative Splicing , Computational Biology/methods , RNA Splice Sites/genetics , Sequence Analysis, RNA , Animals , Exons , Genetic Code , Humans , Mammals/genetics , Reproducibility of Results
17.
Nucleic Acids Res ; 42(22): 13615-32, 2014 Dec 16.
Article in English | MEDLINE | ID: mdl-25414359

ABSTRACT

LEF/TCFs direct the final step in Wnt/ß-catenin signalling by recruiting ß-catenin to genes for activation of transcription. Ancient, non-vertebrate TCFs contain two DNA binding domains, a High Mobility Group box for recognition of the Wnt Response Element (WRE; 5'-CTTTGWWS-3') and the C-clamp domain for recognition of the GC-rich Helper motif (5'-RCCGCC-3'). Two vertebrate TCFs (TCF-1/TCF7 and TCF-4/TCF7L2) use the C-clamp as an alternatively spliced domain to regulate cell-cycle progression, but how the C-clamp influences TCF binding and activity genome-wide is not known. Here, we used a doxycycline inducible system with ChIP-seq to assess how the C-clamp influences human TCF1 binding genome-wide. Metabolic pulse-labeling of nascent RNA with 4'Thiouridine was used with RNA-seq to connect binding to the Wnt transcriptome. We find that the C-clamp enables targeting to a greater number of gene loci for stronger occupancy and transcription regulation. The C-clamp uses Helper sites concurrently with WREs for gene targeting, but it also targets TCF1 to sites that do not have readily identifiable canonical WREs. The coupled ChIP-seq/4'Thiouridine-seq analysis identified new Wnt target genes, including additional regulators of cell proliferation. Thus, C-clamp containing isoforms of TCFs are potent transcriptional regulators with an expanded transcriptome directed by C-clamp-Helper site interactions.


Subject(s)
Hepatocyte Nuclear Factor 1-alpha/metabolism , Transcriptome , Wnt Signaling Pathway , Animals , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Chromatin Immunoprecipitation , DNA/chemistry , DNA/metabolism , Gene Expression Regulation , Genetic Loci , Hepatocyte Nuclear Factor 1-alpha/chemistry , Hepatocyte Nuclear Factor 1-alpha/genetics , Humans , Mutation , Nucleotide Motifs , Protein Binding , Protein Structure, Tertiary , Response Elements , Sequence Analysis, DNA , Thiouridine
18.
PLoS One ; 9(5): e95486, 2014.
Article in English | MEDLINE | ID: mdl-24788257

ABSTRACT

miRNA levels are altered in pancreatic ductal adenocarcinoma (PDA), the most common and lethal pancreatic malignancy, and intact miRNA processing is essential for lineage specification during pancreatic development. However, the role of miRNA processing in PDA has not been explored. Here we study the role of miRNA biogenesis in PDA development by deleting the miRNA processing enzyme Dicer in a PDA mouse model driven by oncogenic Kras. We find that loss of Dicer accelerates Kras driven acinar dedifferentiation and acinar to ductal metaplasia (ADM), a process that has been shown to precede and promote the specification of PDA precursors. However, unconstrained ADM also displays high levels of apoptosis. Dicer loss does not accelerate development of Kras driven PDA precursors or PDA, but surprisingly, we observe that mouse PDA can develop without Dicer, although at the expense of proliferative capacity. Our data suggest that intact miRNA processing is involved in both constraining pro-tumorigenic changes in pancreatic differentiation as well as maintaining viability during PDA initiation.


Subject(s)
Cell Transformation, Neoplastic/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Ribonuclease III/genetics , Acinar Cells/metabolism , Acinar Cells/pathology , Animals , Carcinoma in Situ/genetics , Carcinoma in Situ/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Differentiation/genetics , Cell Survival/genetics , Disease Models, Animal , Gene Deletion , Metaplasia , Mice , Pancreatic Ducts/metabolism , Pancreatic Ducts/pathology , Proto-Oncogene Proteins p21(ras)/genetics
19.
Methods Mol Biol ; 1126: 117-21, 2014.
Article in English | MEDLINE | ID: mdl-24549659

ABSTRACT

Splicing components play an essential role in mediating accurate and efficient splicing. The complexity of the spliceosome and its regulatory networks increase the difficulty of studying the splicing reaction in detail. Nuclear extracts derived from HeLa cells provide all of the obligatory components to carry out intron removal in vitro. This chapter describes the large-scale preparation of nuclear extract from HeLa cells.


Subject(s)
Molecular Biology/methods , RNA Splicing , Spliceosomes , Cell Extracts/genetics , Cell Nucleus/genetics , HeLa Cells , Humans
20.
Methods Mol Biol ; 1126: 151-60, 2014.
Article in English | MEDLINE | ID: mdl-24549662

ABSTRACT

The in vitro splicing assay is a valuable technique that can be used to study the mechanism and machinery involved in the splicing process. The ability to investigate various aspects of splicing and alternative splicing appears to be endless due to the flexibility of this assay. Here, we describe the tools and techniques necessary to carry out an in vitro splicing assay. Through the use of radiolabeled pre-mRNA and crude nuclear extract, spliced mRNAs can be purified and visualized by autoradiography for downstream analysis.


Subject(s)
Alternative Splicing/genetics , Molecular Biology/methods , RNA Precursors/genetics , Animals , Cell Extracts/genetics , Cell Extracts/isolation & purification , Cell Nucleus/genetics , HeLa Cells , Humans , Mammals
SELECTION OF CITATIONS
SEARCH DETAIL
...