101 results on '"Hertel KJ"'
Search Results
2. Kinetic analysis of in vitro Pre-mRNA splicing in HeLa nuclear extract
- Author
-
Mueller, WF and Hertel, KJ
- Abstract
Kinetic analysis of in vitro splicing is a valuable technique for understanding splicing regulation. It allows the determination of specific contributions from functional elements for the efficient removal of introns. This chapter will describe the rationale and approach employed to use kinetic analysis to evaluate an in vitro splicing reaction using radiolabeled pre-mRNA incubated in splicing-competent HeLa nuclear extract (NE). © 2014 Springer Science+Business Media, LLC.
- Published
- 2014
3. In vitro assay of pre-mRNA splicing in mammalian nuclear extract
- Author
-
Movassat, M, Mueller, WF, and Hertel, KJ
- 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. © 2014 Springer Science+Business Media, LLC.
- Published
- 2014
4. Linking splicing to Pol II transcription stabilizes pre-mRNAs and influences splicing patterns.
- Author
-
Hicks, MJ, Yang, CR, Kotlajich, MV, and Hertel, KJ
- Subjects
Biological Sciences ,Agricultural and Veterinary Sciences ,Medical and Health Sciences ,Developmental Biology - Abstract
RNA processing is carried out in close proximity to the site of transcription, suggesting a regulatory link between transcription and pre-mRNA splicing. Using an in vitro transcription/splicing assay, we demonstrate that an association of RNA polymerase II (Pol II) transcription and pre-mRNA splicing is required for efficient gene expression. Pol II-synthesized RNAs containing functional splice sites are protected from nuclear degradation, presumably because the local concentration of the splicing machinery is sufficiently high to ensure its association over interactions with nucleases. Furthermore, the process of transcription influences alternative splicing of newly synthesized pre-mRNAs. Because other RNA polymerases do not provide similar protection from nucleases, and their RNA products display altered splicing patterns, the link between transcription and RNA processing is RNA Pol II-specific. We propose that the connection between transcription by Pol II and pre-mRNA splicing guarantees an extended half-life and proper processing of nascent pre-mRNAs.
- Published
- 2006
5. The architecture of pre-mRNAs affects mechanisms of splice-site pairing
- Author
-
Fox-Walsh, KL, Dou, YM, Lam, BJ, Hung, SP, Baldi, PF, and Hertel, KJ
- Subjects
alternative splicing ,bioinformatics ,EST database ,intron length - Published
- 2005
6. Modulation of survival motor neuron pre-mRNA splicing by inhibition of alternative 3' splice site pairing.
- Author
-
Lim, SR and Hertel, KJ
- Subjects
Cell Line ,Tumor Cells ,Cultured ,Humans ,RNA-Binding Proteins ,Nerve Tissue Proteins ,Oligonucleotides ,Antisense ,RNA ,Messenger ,Protein Biosynthesis ,RNA Splicing ,Base Sequence ,Dose-Response Relationship ,Drug ,Alleles ,Introns ,Exons ,Models ,Genetic ,Time Factors ,Molecular Sequence Data ,Cyclic AMP Response Element-Binding Protein ,SMN Complex Proteins ,Survival of Motor Neuron 1 Protein ,Survival of Motor Neuron 2 Protein ,Biochemistry & Molecular Biology ,Biological Sciences ,Medical and Health Sciences ,Chemical Sciences - Abstract
Spinal muscular atrophy is caused by the loss of functional survival motor neuron (SMN1) alleles. A translationally silent nucleotide transition in the duplicated copy of the gene (SMN2) leads to exon 7 skipping and expression of a nonfunctional gene product. It has been suggested that differential SMN2 splicing is caused by the disruption of an exonic splicing enhancer. Here we show that the single nucleotide difference reduces the intrinsic strength of the 3' splice site of exon 7 2-fold, whereas the strength of the 5' splice site of the exon 7 is not affected. Thus, a decrease in splice site strength is magnified in the context of competing exons. These data suggest that lower levels of exon 7 definition not only reduce intron 6 removal but, more importantly, increase the efficiency of the competing exon 7 skipping pathway. Antisense oligonucleotides were tested to modulate exon 7 inclusion, which contains the authentic translation stop codon. Oligonucleotides directed toward the 3' splice site of exon 8 were shown to alter SMN2 splicing in favor of exon 7 inclusion. These results suggest that antisense oligonucleotides could be used as a therapeutic strategy to counteract the progression of SMA.
- Published
- 2001
7. The role of U2AF35 and U2AF65 in enhancer-dependent splicing.
- Author
-
Graveley, BR, Hertel, KJ, and Maniatis, T
- Subjects
Genetics ,Generic health relevance ,Base Sequence ,Enhancer Elements ,Genetic ,Immunoglobulin M ,Mutation ,Nuclear Proteins ,Pyrimidines ,RNA Precursors ,RNA Splicing ,RNA ,Messenger ,Ribonucleoproteins ,Splicing Factor U2AF ,recruitment ,splicing enhancer ,SR protein ,U2AF ,Biochemistry and Cell Biology ,Developmental Biology - Abstract
Splicing enhancers are RNA sequence elements that promote the splicing of nearby introns. The mechanism by which these elements act is still unclear. Some experiments support a model in which serine-arginine (SR)-rich proteins function as splicing activators by binding to enhancers and recruiting the splicing factor U2AF to an adjacent weak 3' splice site. In this model, recruitment requires interactions between the SR proteins and the 35-kDa subunit of U2AF (U2AF35). However, more recent experiments have not supported the U2AF recruitment model. Here we provide additional evidence for the recruitment model. First, we confirm that base substitutions that convert weak 3' splice sites to a consensus sequence, and therefore increase U2AF binding, relieve the requirement for a splicing activator. Second, we confirm that splicing activators are required for the formation of early spliceosomal complexes on substrates containing weak 3' splice sites. Most importantly, we find that splicing activators promote the binding of both U2AF65 and U2AF35 to weak 3' splice sites under splicing conditions. Finally, we show that U2AF35 is required for maximum levels of activator-dependent splicing. We conclude that a critical function of splicing activators is to recruit U2AF to the weak 3' splice sites of enhancer-dependent introns, and that efficient enhancer-dependent splicing requires U2AF35.
- Published
- 2001
8. SR proteins induce alternative exon skipping through their activities on the flanking constitutive exons
- Author
-
Han, J, Ding, JH, Byeon, CW, Kim, JH, Hertel, KJ, Jeong, S, and Fu, XD
- Abstract
SR proteins are well known to promote exon inclusion in regulated splicing through exonic splicing enhancers. SR proteins have also been reported to cause exon skipping, but little is known about the mechanism. We previously characterized SRSF1 (SF2/ASF)-dependent exon skipping of the CaMKIIδ gene during heart remodeling. By using mouse embryo fibroblasts derived from conditional SR protein knockout mice, we now show that SR protein-induced exon skipping depends on their prevalent actions on a flanking constitutive exon and requires collaboration of more than one SR protein. These findings, coupled with other established rules for SR proteins, provide a theoretical framework to understand the complex effect of SR protein-regulated splicing in mammalian cells. We further demonstrate that heart-specific CaMKIIδ splicing can be reconstituted in fibroblasts by downregulating SR proteins and upregulating a RBFOX protein and that SR protein overexpression impairs regulated CaMKIIδ splicing and neuronal differentiation in P19 cells, illustrating that SR protein-dependent exon skipping may constitute a key strategy for synergism with other splicing regulators in establishing tissue-specific alternative splicing critical for cell differentiation programs. Copyright © 2011, American Society for Microbiology. All Rights Reserved.
- Published
- 2011
9. Spliceosomes walk the line: splicing errors and their impact on cellular function
- Author
-
Hsu, SN and Hertel, KJ
- Abstract
The splicing of nuclear pre-mRNAs is a fundamental process required for the expression of most metazoan genes. The majority of the approximately 25,000 genes encoded by the human genome has been shown to produce more than one kind of transcripts through alternative splicing. Alternative splicing of pre-mRNAs can lead to the production of multiple protein isoforms from a single gene, significantly enriching the proteomic diversity of higher eukaryotic organisms. Because regulation of this process determines the timing and location that a particular protein isoform is produced, changes of alternative splicing patterns have the potential to modulate many cellular activities. Consequently, pre-mRNA splicing must occur with a high degree of specificity and fidelity to ensure the appropriate expression of functional mRNAs. Here we review recent progress made in understanding the extent of alternative splicing within the human genome with particular emphasis on splicing fidelity.
- Published
- 2009
10. The role of U2AF35and U2AF65in enhancer-dependent splicing
- Author
-
Graveley, BR, Hertel, KJ, and Maniatis, TOM
- Abstract
Splicing enhancers are RNA sequence elements that promote the splicing of nearby introns. The mechanism by which these elements act is still unclear. Some experiments support a model in which serine-arginine (SR)-rich proteins function as splicing activators by binding to enhancers and recruiting the splicing factor U2AF to an adjacent weak 3′ splice site. In this model, recruitment requires interactions between the SR proteins and the 35-kDa subunit of U2AF (U2AF35). However, more recent experiments have not supported the U2AF recruitment model. Here we provide additional evidence for the recruitment model. First, we confirm that base substitutions that convert weak 3′ splice sites to a consensus sequence, and therefore increase U2AF binding, relieve the requirement for a splicing activator. Second, we confirm that splicing activators are required for the formation of early spliceosomal complexes on substrates containing weak 3′ splice sites. Most importantly, we find that splicing activators promote the binding of both U2AF65and U2AF35to weak 3′ splice sites under splicing conditions. Finally, we show that U2AF35is required for maximum levels of activator-dependent splicing. We conclude that a critical function of splicing activators is to recruit U2AF to the weak 3′ splice sites of enhancer-dependent introns, and that efficient enhancer-dependent splicing requires U2AF35.
- Published
- 2001
11. Interdependent regulation of alternative splicing by SR and hnRNP proteins.
- Author
-
Holmes ME and Hertel KJ
- Abstract
Alternative pre-mRNA splicing is a combinatorial process involving SR and hnRNP splicing factors. These proteins can silence or enhance splicing based on their expression levels and binding positions. To better understand their combinatorial and interdependent regulation, computational analyses were performed using HepG2 and K562 cell knockdown and binding datasets from the ENCODE Project. Analyses of diMerential splicing for 6 SR proteins and 13 hnRNP knockdowns revealed statistically significant exon overlap among most RBP combinations, albeit at diMerent levels. Neither SR proteins nor hnRNPs showed strong preferences for collaborating with specific RBP classes in mediating exon inclusion. While SRSF1, hnRNPK, and hnRNPC stand out as major influencers of alternative splicing, they do so predominantly independent of other RBPs. Meanwhile, minor influencers of alternative splicing such as hnRNPAB and hnRNPA0 predominantly regulate exon inclusion in concert with other RBPs, indicating that inclusion can be mediated by both single and multiple RBPs. Interestingly, the higher the number of RBPs that regulate the inclusion of an exon, the more variable exon inclusion preferences become. Interdependently regulated exons are more modular and have diMerent physical characteristics such as reduced exon length compared to their independent counterparts. A comparison of RBP interdependence between HepG2 and K562 cells provides the framework that explains cell-type-specific alternative splicing. Our study highlights the importance of the interdependent regulation of alternative exons and identifies characteristics of interdependently regulated exons that diMer from independently regulated exons.
- Published
- 2024
- Full Text
- View/download PDF
12. Splice site proximity influences alternative exon definition.
- Author
-
Carranza F, Shenasa H, and Hertel KJ
- Subjects
- Alternative Splicing, Exons, Introns, RNA Splice Sites, RNA Splicing
- 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.
- Published
- 2022
- Full Text
- View/download PDF
13. The influence of 4-thiouridine labeling on pre-mRNA splicing outcomes.
- Author
-
Altieri JAC and Hertel KJ
- Subjects
- HEK293 Cells, Humans, Nucleic Acid Conformation, RNA Precursors genetics, RNA Stability drug effects, Staining and Labeling, Alternative Splicing drug effects, RNA Precursors metabolism, RNA Splice Sites, Thiouridine pharmacology
- 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., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2021
- Full Text
- View/download PDF
14. Allosteric regulation of U1 snRNP by splicing regulatory proteins controls spliceosomal assembly.
- Author
-
Shenasa H, Movassat M, Forouzmand E, and Hertel KJ
- Subjects
- Heterogeneous-Nuclear Ribonucleoproteins genetics, Proteomics methods, RNA Precursors genetics, RNA Splice Sites genetics, RNA, Messenger genetics, Allosteric Regulation genetics, Alternative Splicing genetics, RNA, Small Nuclear genetics, Ribonucleoprotein, U1 Small Nuclear genetics, Spliceosomes metabolism
- 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., (© 2020 Shenasa et al.; Published by Cold Spring Harbor Laboratory Press for the RNA Society.)
- Published
- 2020
- Full Text
- View/download PDF
15. Publisher Correction: Transcriptomic and proteomic signatures of stemness and differentiation in the colon crypt.
- Author
-
Habowski AN, Flesher JL, Bates JM, Tsai CF, Martin K, Zhao R, Ganesan AK, Edwards RA, Shi T, Wiley HS, Shi Y, Hertel KJ, and Waterman ML
- Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2020
- Full Text
- View/download PDF
16. Transcriptomic and proteomic signatures of stemness and differentiation in the colon crypt.
- Author
-
Habowski AN, Flesher JL, Bates JM, Tsai CF, Martin K, Zhao R, Ganesan AK, Edwards RA, Shi T, Wiley HS, Shi Y, Hertel KJ, and Waterman ML
- Subjects
- Animals, Biomarkers, 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, Cell Differentiation, Cell Self Renewal genetics, Colon, Enterocytes metabolism, Proteome, Stem Cells metabolism, Transcriptome
- 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.
- Published
- 2020
- Full Text
- View/download PDF
17. Exon size and sequence conservation improves identification of splice-altering nucleotides.
- Author
-
Movassat M, Forouzmand E, Reese F, and Hertel KJ
- Subjects
- Humans, Nucleotides, Phylogeny, Polymorphism, Single Nucleotide, RNA Precursors genetics, Alternative Splicing, Base Sequence, Conserved Sequence, Exons
- 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., (© 2019 Movassat et al.; Published by Cold Spring Harbor Laboratory Press for the RNA Society.)
- Published
- 2019
- Full Text
- View/download PDF
18. Combinatorial regulation of alternative splicing.
- Author
-
Shenasa H and Hertel KJ
- Subjects
- DNA-Directed RNA Polymerases metabolism, Gene Expression Regulation, Humans, Nucleic Acid Conformation, RNA Splice Sites, RNA, Messenger chemistry, Regulatory Elements, Transcriptional, Alternative Splicing, RNA, Messenger metabolism
- 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., (Copyright © 2019 Elsevier B.V. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
19. Ultra-deep sequencing reveals pre-mRNA splicing as a sequence driven high-fidelity process.
- Author
-
Reynolds DJ and Hertel KJ
- Subjects
- 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, High-Throughput Nucleotide Sequencing methods, RNA Precursors genetics, RNA Splicing genetics
- 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., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2019
- Full Text
- View/download PDF
20. Pancreatic pericytes originate from the embryonic pancreatic mesenchyme.
- Author
-
Harari N, Sakhneny L, Khalifa-Malka L, Busch A, Hertel KJ, Hebrok M, and Landsman L
- Subjects
- 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, Mesoderm cytology, Mesoderm embryology, Pancreas cytology, Pancreas embryology, Pericytes cytology
- 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., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
21. Molecular Mechanisms for CFIm-Mediated Regulation of mRNA Alternative Polyadenylation.
- Author
-
Zhu Y, Wang X, Forouzmand E, Jeong J, Qiao F, Sowd GA, Engelman AN, Xie X, Hertel KJ, and Shi Y
- Subjects
- 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, Alternative Splicing genetics, Gene Expression Regulation genetics, Polyadenylation, RNA, Messenger metabolism, mRNA Cleavage and Polyadenylation Factors metabolism
- 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., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
22. Isolation of Newly Transcribed RNA Using the Metabolic Label 4-Thiouridine.
- Author
-
Garibaldi A, Carranza F, and Hertel KJ
- Subjects
- Animals, Cell Line, Humans, RNA Precursors biosynthesis, RNA Precursors chemistry, RNA Precursors isolation & purification, Staining and Labeling methods, Thiouridine chemistry, Transcription, Genetic
- 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.
- Published
- 2017
- Full Text
- View/download PDF
23. Preparation of Splicing Competent Nuclear Extract from Mammalian Cells and In Vitro Pre-mRNA Splicing Assay.
- Author
-
Movassat M, Shenasa H, and Hertel KJ
- Subjects
- Animals, Cell Nucleus metabolism, Cell-Free System chemistry, Cell-Free System metabolism, Complex Mixtures chemistry, HeLa Cells, Humans, RNA Precursors metabolism, Spliceosomes metabolism, Cell Nucleus chemistry, RNA Precursors chemistry, RNA Splicing, Spliceosomes chemistry
- 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.
- Published
- 2017
- Full Text
- View/download PDF
24. Release of SR Proteins from CLK1 by SRPK1: A Symbiotic Kinase System for Phosphorylation Control of Pre-mRNA Splicing.
- Author
-
Aubol BE, Wu G, Keshwani MM, Movassat M, Fattet L, Hertel KJ, Fu XD, and Adams JA
- Subjects
- 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, Protein Serine-Threonine Kinases metabolism, Protein-Tyrosine Kinases metabolism, RNA Precursors metabolism, RNA Splicing, RNA, Messenger metabolism, Spliceosomes enzymology
- 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., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
25. Coupling between alternative polyadenylation and alternative splicing is limited to terminal introns.
- Author
-
Movassat M, Crabb TL, Busch A, Yao C, Reynolds DJ, Shi Y, and Hertel KJ
- Subjects
- HeLa Cells, Heterogeneous-Nuclear Ribonucleoprotein Group A-B genetics, Humans, mRNA Cleavage and Polyadenylation Factors genetics, Alternative Splicing physiology, Heterogeneous-Nuclear Ribonucleoprotein Group A-B metabolism, Introns physiology, Polyadenylation physiology, mRNA Cleavage and Polyadenylation Factors metabolism
- 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.
- Published
- 2016
- Full Text
- View/download PDF
26. The Silent Sway of Splicing by Synonymous Substitutions.
- Author
-
Mueller WF, Larsen LS, Garibaldi A, Hatfield GW, and Hertel KJ
- Subjects
- 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, Alternative Splicing genetics, Mutation, RNA, Messenger genetics
- 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., (© 2015 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
27. Splicing predictions reliably classify different types of alternative splicing.
- Author
-
Busch A and Hertel KJ
- Subjects
- Animals, Exons, Genetic Code, Humans, Mammals genetics, Reproducibility of Results, Alternative Splicing, Computational Biology methods, RNA Splice Sites genetics, Sequence Analysis, RNA
- 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., (© 2015 Busch and Hertel; Published by Cold Spring Harbor Laboratory Press for the RNA Society.)
- Published
- 2015
- Full Text
- View/download PDF
28. The TCF C-clamp DNA binding domain expands the Wnt transcriptome via alternative target recognition.
- Author
-
Hoverter NP, Zeller MD, McQuade MM, Garibaldi A, Busch A, Selwan EM, Hertel KJ, Baldi P, and Waterman ML
- Subjects
- 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, Hepatocyte Nuclear Factor 1-alpha metabolism, Transcriptome, Wnt Signaling Pathway
- Abstract
LEF/TCFs direct the final step in Wnt/β-catenin signalling by recruiting β-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., (© The Author(s) 2014. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2014
- Full Text
- View/download PDF
29. Dicer regulates differentiation and viability during mouse pancreatic cancer initiation.
- Author
-
Morris JP 4th, Greer R, Russ HA, von Figura G, Kim GE, Busch A, Lee J, Hertel KJ, Kim S, McManus M, and Hebrok M
- Subjects
- 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, Cell Transformation, Neoplastic genetics, Pancreatic Neoplasms genetics, Pancreatic Neoplasms pathology, Ribonuclease III genetics
- 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.
- Published
- 2014
- Full Text
- View/download PDF
30. The chromatin regulator Brg1 suppresses formation of intraductal papillary mucinous neoplasm and pancreatic ductal adenocarcinoma.
- Author
-
von Figura G, Fukuda A, Roy N, Liku ME, Morris Iv JP, Kim GE, Russ HA, Firpo MA, Mulvihill SJ, Dawson DW, Ferrer J, Mueller WF, Busch A, Hertel KJ, and Hebrok M
- Subjects
- Adenocarcinoma, Mucinous pathology, Animals, Carcinogenesis metabolism, Carcinogenesis pathology, Carcinoma, Pancreatic Ductal pathology, Cell Line, Tumor, Cell Proliferation, Chromatin Assembly and Disassembly, Female, Humans, Male, Mice, Mice, Inbred NOD, Mice, SCID, Mice, Transgenic, Pancreas pathology, Pancreatic Neoplasms pathology, Proto-Oncogene Proteins p21(ras) genetics, Adenocarcinoma, Mucinous metabolism, Carcinoma, Pancreatic Ductal metabolism, DNA Helicases physiology, Nuclear Proteins physiology, Pancreatic Neoplasms metabolism, Transcription Factors physiology
- Abstract
Pancreatic ductal adenocarcinoma (PDA) develops through distinct precursor lesions, including pancreatic intraepithelial neoplasia (PanIN) and intraductal papillary mucinous neoplasia (IPMN). However, genetic features resulting in IPMN-associated PDA (IPMN-PDA) versus PanIN-associated PDA (PanIN-PDA) are largely unknown. Here we find that loss of Brg1, a core subunit of SWI/SNF chromatin remodelling complexes, cooperates with oncogenic Kras to form cystic neoplastic lesions that resemble human IPMN and progress to PDA. Although Brg1-null IPMN-PDA develops rapidly, it possesses a distinct transcriptional profile compared with PanIN-PDA driven by mutant Kras and hemizygous p53 deletion. IPMN-PDA also is less lethal, mirroring prognostic trends in PDA patients. In addition, Brg1 deletion inhibits Kras-dependent PanIN development from adult acinar cells, but promotes Kras-driven preneoplastic transformation in adult duct cells. Therefore, this study implicates Brg1 as a determinant of context-dependent Kras-driven pancreatic tumorigenesis and suggests that chromatin remodelling may underlie the development of distinct PDA subsets.
- Published
- 2014
- Full Text
- View/download PDF
31. HEXEvent: a database of Human EXon splicing Events.
- Author
-
Busch A and Hertel KJ
- Subjects
- Humans, Internet, User-Computer Interface, Alternative Splicing, Databases, Nucleic Acid, Exons
- Abstract
HEXEvent (http://hexevent.mmg.uci.edu) is a new database that permits the user to compile genome-wide exon data sets of human internal exons showing selected splicing events. User queries can be customized based on the type and the frequency of alternative splicing events. For each splicing version of an exon, an ESTs count is given, specifying the frequency of the event. A user-specific definition of constitutive exons can be entered to designate an exon exclusion level still acceptable for an exon to be considered as constitutive. Similarly, the user has the option to define a maximum inclusion level for an exon to be called an alternatively spliced exon. Unlike other existing splicing databases, HEXEvent permits the user to easily extract alternative splicing information for individual, multiple or genome-wide human internal exons. Importantly, the generated data sets are downloadable for further analysis.
- Published
- 2013
- Full Text
- View/download PDF
32. Position-dependent splicing activation and repression by SR and hnRNP proteins rely on common mechanisms.
- Author
-
Erkelenz S, Mueller WF, Evans MS, Busch A, Schöneweis K, Hertel KJ, and Schaal H
- Subjects
- Exons, HeLa Cells, Heterogeneous-Nuclear Ribonucleoproteins genetics, Humans, Introns, RNA Splice Sites genetics, RNA Splice Sites physiology, RNA Splicing genetics, Heterogeneous-Nuclear Ribonucleoproteins metabolism, RNA Splicing physiology
- Abstract
Alternative splicing is regulated by splicing factors that modulate splice site selection. In some cases, however, splicing factors show antagonistic activities by either activating or repressing splicing. Here, we show that these opposing outcomes are based on their binding location relative to regulated 5' splice sites. SR proteins enhance splicing only when they are recruited to the exon. However, they interfere with splicing by simply relocating them to the opposite intronic side of the splice site. hnRNP splicing factors display analogous opposing activities, but in a reversed position dependence. Activation by SR or hnRNP proteins increases splice site recognition at the earliest steps of exon definition, whereas splicing repression promotes the assembly of nonproductive complexes that arrest spliceosome assembly prior to splice site pairing. Thus, SR and hnRNP splicing factors exploit similar mechanisms to positively or negatively influence splice site selection.
- Published
- 2013
- Full Text
- View/download PDF
33. Extensive regulation of NAGNAG alternative splicing: new tricks for the spliceosome?
- Author
-
Busch A and Hertel KJ
- Subjects
- Exons, High-Throughput Nucleotide Sequencing, Humans, Introns, Organ Specificity genetics, Protein Isoforms genetics, Alternative Splicing genetics, RNA Splice Sites genetics, Spliceosomes genetics
- Abstract
A recent study using massive parallel sequencing demonstrates unequivocally that alternative tandem acceptor splicing is tissue-specifically regulated.
- Published
- 2012
- Full Text
- View/download PDF
34. Evolution of SR protein and hnRNP splicing regulatory factors.
- Author
-
Busch A and Hertel KJ
- Subjects
- Animals, Humans, Phylogeny, Nuclear Proteins genetics, RNA Precursors genetics, RNA Splicing, RNA-Binding Proteins genetics
- Abstract
The splicing of pre-mRNAs is an essential step of gene expression in eukaryotes. Introns are removed from split genes through the activities of the spliceosome, a large ribonuclear machine that is conserved throughout the eukaryotic lineage. While unicellular eukaryotes are characterized by less complex splicing, pre-mRNA splicing of multicellular organisms is often associated with extensive alternative splicing that significantly enriches their proteome. The alternative selection of splice sites and exons permits multicellular organisms to modulate gene expression patterns in a cell type-specific fashion, thus contributing to their functional diversification. Alternative splicing is a regulated process that is mainly influenced by the activities of splicing regulators, such as SR proteins or hnRNPs. These modular factors have evolved from a common ancestor through gene duplication events to a diverse group of splicing regulators that mediate exon recognition through their sequence-specific binding to pre-mRNAs. Given the strong correlations between intron expansion, the complexity of pre-mRNA splicing, and the emergence of splicing regulators, it is argued that the increased presence of SR and hnRNP proteins promoted the evolution of alternative splicing through relaxation of the sequence requirements of splice junctions., (Copyright © 2011 John Wiley & Sons, Ltd.)
- Published
- 2012
- Full Text
- View/download PDF
35. Efficient internal exon recognition depends on near equal contributions from the 3' and 5' splice sites.
- Author
-
Shepard PJ, Choi EA, Busch A, and Hertel KJ
- Subjects
- HeLa Cells, Humans, Alternative Splicing, Exons, RNA Splice Sites
- Abstract
Pre-mRNA splicing is carried out by the spliceosome, which identifies exons and removes intervening introns. In vertebrates, most splice sites are initially recognized by the spliceosome across the exon, because most exons are small and surrounded by large introns. This gene architecture predicts that efficient exon recognition depends largely on the strength of the flanking 3' and 5' splice sites. However, it is unknown if the 3' or the 5' splice site dominates the exon recognition process. Here, we test the 3' and 5' splice site contributions towards efficient exon recognition by systematically replacing the splice sites of an internal exon with sequences of different splice site strengths. We show that the presence of an optimal splice site does not guarantee exon inclusion and that the best predictor for exon recognition is the sum of both splice site scores. Using a genome-wide approach, we demonstrate that the combined 3' and 5' splice site strengths of internal exons provide a much more significant separator between constitutive and alternative exons than either the 3' or the 5' splice site strength alone.
- Published
- 2011
- Full Text
- View/download PDF
36. Complex and dynamic landscape of RNA polyadenylation revealed by PAS-Seq.
- Author
-
Shepard PJ, Choi EA, Lu J, Flanagan LA, Hertel KJ, and Shi Y
- Subjects
- Animals, Embryonic Stem Cells metabolism, Gene Expression Profiling, HeLa Cells, Histones chemistry, Humans, Mice, Neural Stem Cells metabolism, Neurons metabolism, RNA, Messenger genetics, High-Throughput Nucleotide Sequencing methods, Polyadenylation, RNA, Messenger chemistry, Sequence Analysis, RNA methods
- Abstract
Alternative polyadenylation (APA) of mRNAs has emerged as an important mechanism for post-transcriptional gene regulation in higher eukaryotes. Although microarrays have recently been used to characterize APA globally, they have a number of serious limitations that prevents comprehensive and highly quantitative analysis. To better characterize APA and its regulation, we have developed a deep sequencing-based method called Poly(A) Site Sequencing (PAS-Seq) for quantitatively profiling RNA polyadenylation at the transcriptome level. PAS-Seq not only accurately and comprehensively identifies poly(A) junctions in mRNAs and noncoding RNAs, but also provides quantitative information on the relative abundance of polyadenylated RNAs. PAS-Seq analyses of human and mouse transcriptomes showed that 40%-50% of all expressed genes produce alternatively polyadenylated mRNAs. Furthermore, our study detected evolutionarily conserved polyadenylation of histone mRNAs and revealed novel features of mitochondrial RNA polyadenylation. Finally, PAS-Seq analyses of mouse embryonic stem (ES) cells, neural stem/progenitor (NSP) cells, and neurons not only identified more poly(A) sites than what was found in the entire mouse EST database, but also detected significant changes in the global APA profile that lead to lengthening of 3' untranslated regions (UTR) in many mRNAs during stem cell differentiation. Together, our PAS-Seq analyses revealed a complex landscape of RNA polyadenylation in mammalian cells and the dynamic regulation of APA during stem cell differentiation.
- Published
- 2011
- Full Text
- View/download PDF
37. G Run-mediated recognition of proteolipid protein and DM20 5' splice sites by U1 small nuclear RNA is regulated by context and proximity to the splice site.
- Author
-
Wang E, Mueller WF, Hertel KJ, and Cambi F
- Subjects
- HeLa Cells, Heterogeneous-Nuclear Ribonucleoprotein Group F-H genetics, Humans, Myelin Proteolipid Protein genetics, RNA, Small Nuclear genetics, Heterogeneous-Nuclear Ribonucleoprotein Group F-H metabolism, Myelin Proteolipid Protein metabolism, RNA Splice Sites physiology, RNA Splicing physiology, RNA, Small Nuclear metabolism
- Abstract
Highly conserved G runs, G1M2 and ISE, regulate the proteolipid protein (PLP)/DM20 ratio. We have investigated recruitment of U1 small nuclear ribonuclear protein (snRNP) by G1M2 and ISE and examined the effect of splice site strength, distance, and context on G run function. G1M2 is necessary for initial recruitment of U1snRNP to the DM20 5' splice site independent of the strength of the splice site. G1M2 regulates E complex formation and supports DM20 splicing when functional U1snRNP is reduced. By contrast, the ISE is not required for the initial recruitment of U1snRNP to the PLP 5' splice site. However, in close proximity to either the DM20 or the PLP 5' splice site, the ISE recruits U1snRNP to both splice sites. The ISE enhances DM20 splicing, whereas close to the PLP 5' splice site, it inhibits PLP splicing. Splicing enhancement and inhibition are mediated by heterogeneous nuclear ribonuclear protein (hnRNP)H/F. The data show that recognition of the DM20 5' splice site depends on G run-mediated recruitment of U1snRNA, whereas a complex interaction between the ISE G runs, context and position determines the functional outcome on splicing. The data suggest that different mechanisms underlie G run-mediated recognition of 5' splice sites and that context and position play a critical role.
- Published
- 2011
- Full Text
- View/download PDF
38. SR proteins induce alternative exon skipping through their activities on the flanking constitutive exons.
- Author
-
Han J, Ding JH, Byeon CW, Kim JH, Hertel KJ, Jeong S, and Fu XD
- Subjects
- Animals, Base Sequence, Calcium-Calmodulin-Dependent Protein Kinase Type 2 genetics, Calcium-Calmodulin-Dependent Protein Kinase Type 2 metabolism, Cell Differentiation, Cells, Cultured, Mice, Mice, Knockout, Models, Biological, Mutagenesis, Neurons cytology, Neurons metabolism, Nuclear Proteins deficiency, Nuclear Proteins genetics, RNA-Binding Proteins genetics, Recombinant Proteins genetics, Recombinant Proteins metabolism, Serine-Arginine Splicing Factors, Alternative Splicing, Exons, Nuclear Proteins metabolism, RNA-Binding Proteins metabolism
- Abstract
SR proteins are well known to promote exon inclusion in regulated splicing through exonic splicing enhancers. SR proteins have also been reported to cause exon skipping, but little is known about the mechanism. We previously characterized SRSF1 (SF2/ASF)-dependent exon skipping of the CaMKIIδ gene during heart remodeling. By using mouse embryo fibroblasts derived from conditional SR protein knockout mice, we now show that SR protein-induced exon skipping depends on their prevalent actions on a flanking constitutive exon and requires collaboration of more than one SR protein. These findings, coupled with other established rules for SR proteins, provide a theoretical framework to understand the complex effect of SR protein-regulated splicing in mammalian cells. We further demonstrate that heart-specific CaMKIIδ splicing can be reconstituted in fibroblasts by downregulating SR proteins and upregulating a RBFOX protein and that SR protein overexpression impairs regulated CaMKIIδ splicing and neuronal differentiation in P19 cells, illustrating that SR protein-dependent exon skipping may constitute a key strategy for synergism with other splicing regulators in establishing tissue-specific alternative splicing critical for cell differentiation programs.
- Published
- 2011
- Full Text
- View/download PDF
39. Retention of spliceosomal components along ligated exons ensures efficient removal of multiple introns.
- Author
-
Crabb TL, Lam BJ, and Hertel KJ
- Subjects
- Animals, Drosophila genetics, Drosophila Proteins genetics, Gene Knockdown Techniques, HeLa Cells, Humans, Nerve Tissue Proteins genetics, Transcription Factors genetics, beta-Globins genetics, Exons, Introns, RNA Splicing, Spliceosomes metabolism
- Abstract
The majority of mammalian pre-mRNAs contains multiple introns that are excised prior to export and translation. After intron excision, ligated exon intermediates participate in subsequent intron excisions. However, exon ligation generates an exon of increased size, a feature of pre-mRNA splicing that can interfere with downstream splicing events. These considerations raise the question of whether unique mechanisms exist that permit efficient removal of introns neighboring ligated exons. Kinetic analyses of multiple intron-containing pre-mRNAs revealed that splicing is more efficient following an initial intron removal event, suggesting that either the recruitment of the exon junction complex (EJC) to ligated exons increases the efficiency of multiple intron excisions or that the initial definition of splice sites is sufficient to permit efficient splicing of introns neighboring ligated exons. Knockdown experiments show that the deposition of the EJC does not affect subsequent splicing kinetics. Instead, spliceosomal components that are not involved in the initial splicing event remain associated with the pre-mRNA to ensure efficient removal of neighboring introns. Thus, ligated exons do not require redefinition, providing an additional kinetic advantage for exon defined splice sites.
- Published
- 2010
- Full Text
- View/download PDF
40. Embracing the complexity of pre-mRNA splicing.
- Author
-
Shepard PJ and Hertel KJ
- Subjects
- RNA Precursors chemistry, RNA, Messenger chemistry, Alternative Splicing, RNA Precursors metabolism, RNA, Messenger metabolism, Regulatory Sequences, Ribonucleic Acid
- Published
- 2010
- Full Text
- View/download PDF
41. Competing upstream 5' splice sites enhance the rate of proximal splicing.
- Author
-
Hicks MJ, Mueller WF, Shepard PJ, and Hertel KJ
- Subjects
- Base Sequence, Enhancer Elements, Genetic, Exons, Humans, Molecular Sequence Data, RNA, Messenger genetics, RNA, Messenger metabolism, RNA, Small Nuclear genetics, beta-Globins genetics, 5' Untranslated Regions genetics, Alternative Splicing, RNA Splice Sites, RNA, Small Nuclear metabolism
- Abstract
Alternative 5' splice site selection is one of the major pathways resulting in mRNA diversification. Regulation of this type of alternative splicing depends on the presence of regulatory elements that activate or repress the use of competing splice sites, usually leading to the preferential use of the proximal splice site. However, the mechanisms involved in proximal splice site selection and the thermodynamic advantage realized by proximal splice sites are not well understood. Here, we have carried out a systematic analysis of alternative 5' splice site usage using in vitro splicing assays. We show that observed rates of splicing correlate well with their U1 snRNA base pairing potential. Weak U1 snRNA interactions with the 5' splice site were significantly rescued by the proximity of the downstream exon, demonstrating that the intron definition mode of splice site recognition is highly efficient. In the context of competing splice sites, the proximity to the downstream 3' splice site was more influential in dictating splice site selection than the actual 5' splice site/U1 snRNA base pairing potential. Surprisingly, the kinetic analysis also demonstrated that an upstream competing 5' splice site enhances the rate of proximal splicing. These results reveal the discovery of a new splicing regulatory element, an upstream 5' splice site functioning as a splicing enhancer.
- Published
- 2010
- Full Text
- View/download PDF
42. Identification of a novel nuclear localization signal and speckle-targeting sequence of tuftelin-interacting protein 11, a splicing factor involved in spliceosome disassembly.
- Author
-
Tannukit S, Crabb TL, Hertel KJ, Wen X, Jans DA, and Paine ML
- Subjects
- Amino Acid Sequence, Animals, Cell Line, Cell Nucleus metabolism, Humans, Mice, Molecular Sequence Data, Nuclear Localization Signals genetics, Nuclear Proteins genetics, Protein Structure, Tertiary, RNA Helicases genetics, RNA Splicing, RNA Splicing Factors, RNA-Binding Proteins, Vesicular Transport Proteins genetics, Nuclear Localization Signals metabolism, Nuclear Proteins metabolism, RNA Helicases metabolism, Spliceosomes metabolism, Vesicular Transport Proteins metabolism
- Abstract
Tuftelin-interacting protein 11 (TFIP11) is a protein component of the spliceosome complex that promotes the release of the lariat-intron during late-stage splicing through a direct recruitment and interaction with DHX15/PRP43. Expression of TFIP11 is essential for cell and organismal survival. TFIP11 contains a G-patch domain, a signature motif of RNA-processing proteins that is responsible for TFIP11-DHX15 interactions. No other functional domains within TFIP11 have been described. TFIP11 is localized to distinct speckled regions within the cell nucleus, although excluded from the nucleolus. In this study sequential C-terminal deletions and mutational analyses have identified two novel protein elements in mouse TFIP11. The first domain covers amino acids 701-706 (VKDKFN) and is an atypical nuclear localization signal (NLS). The second domain is contained within amino acids 711-735 and defines TFIP11's distinct speckled nuclear localization. The identification of a novel TFIP11 nuclear speckle-targeting sequence (TFIP11-STS) suggests that this domain directly interacts with additional spliceosomal components. These data help define the mechanism of nuclear/nuclear speckle localization of the splicing factor TFIP11, with implications for it's function.
- Published
- 2009
- Full Text
- View/download PDF
43. Restoration of full-length SMN promoted by adenoviral vectors expressing RNA antisense oligonucleotides embedded in U7 snRNAs.
- Author
-
Geib T and Hertel KJ
- Subjects
- Alternative Splicing genetics, Base Sequence, Fibroblasts metabolism, Fibroblasts pathology, HeLa Cells, Humans, Molecular Sequence Data, Adenoviridae genetics, Genetic Therapy, Genetic Vectors genetics, Oligonucleotides, Antisense genetics, RNA, Small Nuclear genetics, Survival of Motor Neuron 1 Protein genetics
- Abstract
Background: Spinal Muscular Atrophy (SMA) is an autosomal recessive disease that leads to specific loss of motor neurons. It is caused by deletions or mutations of the survival of motor neuron 1 gene (SMN1). The remaining copy of the gene, SMN2, generates only low levels of the SMN protein due to a mutation in SMN2 exon 7 that leads to exon skipping., Methodology/principal Findings: To correct SMN2 splicing, we use Adenovirus type 5-derived vectors to express SMN2-antisense U7 snRNA oligonucleotides targeting the SMN intron 7/exon 8 junction. Infection of SMA type I-derived patient fibroblasts with these vectors resulted in increased levels of exon 7 inclusion, upregulating the expression of SMN to similar levels as in non-SMA control cells., Conclusions/significance: These results show that Adenovirus type 5-derived vectors delivering U7 antisense oligonucleotides can efficiently restore full-length SMN protein and suggest that the viral vector-mediated oligonucleotide application may be a suitable therapeutic approach to counteract SMA.
- Published
- 2009
- Full Text
- View/download PDF
44. Spliceosomes walk the line: splicing errors and their impact on cellular function.
- Author
-
Hsu SN and Hertel KJ
- Subjects
- Alternative Splicing, Animals, Genome, Human, Humans, RNA Splicing, Spliceosomes physiology
- Abstract
The splicing of nuclear pre-mRNAs is a fundamental process required for the expression of most metazoan genes. The majority of the approximately 25,000 genes encoded by the human genome has been shown to produce more than one kind of transcripts through alternative splicing. Alternative splicing of pre-mRNAs can lead to the production of multiple protein isoforms from a single gene, significantly enriching the proteomic diversity of higher eukaryotic organisms. Because regulation of this process determines the timing and location that a particular protein isoform is produced, changes of alternative splicing patterns have the potential to modulate many cellular activities. Consequently, pre-mRNA splicing must occur with a high degree of specificity and fidelity to ensure the appropriate expression of functional mRNAs. Here we review recent progress made in understanding the extent of alternative splicing within the human genome with particular emphasis on splicing fidelity.
- Published
- 2009
- Full Text
- View/download PDF
45. Dangerous play--splitting the message may leave you empty handed.
- Author
-
Klaue Y and Hertel KJ
- Subjects
- Animals, Exons, Introns, RNA Polymerase II metabolism, RNA, Catalytic metabolism, Gene Expression Regulation, RNA Precursors genetics, RNA Precursors metabolism, Transcription, Genetic
- Published
- 2009
- Full Text
- View/download PDF
46. Splice-site pairing is an intrinsically high fidelity process.
- Author
-
Fox-Walsh KL and Hertel KJ
- Subjects
- Cell Line, DNA Polymerase II, Exons, Female, Humans, Male, Muscular Atrophy, Spinal pathology, RNA Precursors, Sequence Deletion, RNA Splice Sites, RNA Splicing, Survival of Motor Neuron 1 Protein physiology
- Abstract
The extensive alternative splicing in higher eukaryotes has initiated a debate whether alternative mRNA isoforms are generated by an inaccurate spliceosome or are the consequence of highly degenerate splice sites within the human genome. Here, we established a quantitative assay to evaluate the accuracy of splice-site pairing by determining the number of incorrect exon-skipping events made from constitutively spliced pre-mRNA transcripts. We demonstrate that the spliceosome pairs exons with an astonishingly high degree of accuracy that may be limited by the quality of pre-mRNAs generated by RNA pol II. The error rate of exon pairing is increased by the effects of the neurodegenerative disorder spinal muscular atrophy because of reduced levels of Survival of Motor Neuron, a master assembler of spliceosomal components. We conclude that all multi-intron-containing genes are alternatively spliced and that the reduction of SMN results in a general splicing defect that is mediated through alterations in the fidelity of splice-site pairing.
- Published
- 2009
- Full Text
- View/download PDF
47. Spliceosome assembly pathways for different types of alternative splicing converge during commitment to splice site pairing in the A complex.
- Author
-
Kotlajich MV, Crabb TL, and Hertel KJ
- Subjects
- Adenosine Triphosphate, Exons genetics, HeLa Cells, Humans, Hydrolysis, Introns genetics, Kinetics, Models, Genetic, Time Factors, Alternative Splicing genetics, Multiprotein Complexes metabolism, RNA Splice Sites genetics, Spliceosomes metabolism
- Abstract
Differential splice site pairing establishes alternative splicing patterns resulting in the generation of multiple mRNA isoforms. This process is carried out by the spliceosome, which is activated by a series of sequential structural rearrangements of its five core snRNPs. To determine when splice sites become functionally paired, we carried out a series of kinetic trap experiments using pre-mRNAs that undergo alternative 5' splice site selection or alternative exon inclusion. We show that commitment to splice site pairing in both cases occurs in the A complex, which is characterized by the ATP-dependent association of the U2 snRNP with the branch point. Interestingly, the timing of splice site pairing is independent of the intron or exon definition modes of splice site recognition. Using the ATP analog ATPgammaS, we showed that ATP hydrolysis is required for splice site pairing independent from U2 snRNP binding to the pre-mRNA. These results identify the A complex as the spliceosomal assembly step dedicated to splice site pairing and suggest that ATP hydrolysis locks splice sites into a splicing pattern after stable U2 snRNP association to the branch point.
- Published
- 2009
- Full Text
- View/download PDF
48. The SR protein family.
- Author
-
Shepard PJ and Hertel KJ
- Subjects
- Animals, Evolution, Molecular, Humans, Protein Biosynthesis, Protein Transport, Serine-Arginine Splicing Factors, Multigene Family, Nuclear Proteins metabolism, RNA-Binding Proteins metabolism
- Abstract
Summary: The processing of pre-mRNAs is a fundamental step required for the expression of most metazoan genes. Members of the family of serine/arginine (SR)-rich proteins are critical components of the machineries carrying out these essential processing events, highlighting their importance in maintaining efficient gene expression. SR proteins are characterized by their ability to interact simultaneously with RNA and other protein components via an RNA recognition motif (RRM) and through a domain rich in arginine and serine residues, the RS domain. Their functional roles in gene expression are surprisingly diverse, ranging from their classical involvement in constitutive and alternative pre-mRNA splicing to various post-splicing activities, including mRNA nuclear export, nonsense-mediated decay, and mRNA translation. These activities point up the importance of SR proteins during the regulation of mRNA metabolism.
- Published
- 2009
- Full Text
- View/download PDF
49. Death by splicing: tumor suppressor RBM5 freezes splice-site pairing.
- Author
-
Kotlajich MV and Hertel KJ
- Subjects
- Exons, Humans, Introns, Models, Genetic, Protein Isoforms metabolism, RNA Precursors metabolism, RNA, Messenger metabolism, fas Receptor genetics, Alternative Splicing, Apoptosis genetics, Cell Cycle Proteins physiology, DNA-Binding Proteins physiology, RNA Splice Sites, RNA-Binding Proteins physiology, Tumor Suppressor Proteins physiology, fas Receptor metabolism
- Abstract
In a recent issue of Molecular Cell, Bonnal et al. (2008) demonstrate that the tumor suppressor gene RBM5 regulates alternative splicing of Fas pre-mRNA by interfering with splice-site pairing.
- Published
- 2008
- Full Text
- View/download PDF
50. Conserved RNA secondary structures promote alternative splicing.
- Author
-
Shepard PJ and Hertel KJ
- Subjects
- Algorithms, Animals, Base Sequence, Humans, RNA chemistry, RNA Splice Sites, Sequence Alignment, Alternative Splicing, Genome, Human, Nucleic Acid Conformation, RNA metabolism
- Abstract
Pre-mRNA splicing is carried out by the spliceosome, which identifies exons and removes intervening introns. Alternative splicing in higher eukaryotes results in the generation of multiple protein isoforms from gene transcripts. The extensive alternative splicing observed implies a flexibility of the spliceosome to identify exons within a given pre-mRNA. To reach this flexibility, splice-site selection in higher eukaryotes has evolved to depend on multiple parameters such as splice-site strength, splicing regulators, the exon/intron architecture, and the process of pre-mRNA synthesis itself. RNA secondary structures have also been proposed to influence alternative splicing as stable RNA secondary structures that mask splice sites are expected to interfere with splice-site recognition. Using structural and functional conservation, we identified RNA structure elements within the human genome that associate with alternative splice-site selection. Their frequent involvement with alternative splicing demonstrates that RNA structure formation is an important mechanism regulating gene expression and disease.
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.