54 results on '"Samuel I. Gunderson"'
Search Results
2. Supplemental Figures and Table from U1 Adaptors Suppress the KRAS-MYC Oncogenic Axis in Human Pancreatic Cancer Xenografts
- Author
-
Darren R. Carpizo, Samuel I. Gunderson, Eric S. Ho, Mark A. Brenneman, Samuel Kogan, Kristen Donohue, Rafal Goraczniak, Xin Yu, Lan Yi, Crissy Dudgeon, and Ashley T. Tsang
- Abstract
Supplemental Figures 1-4 with figure legends and Supplemental Table
- Published
- 2023
- Full Text
- View/download PDF
3. Systematic profiling of poly(A)+ transcripts modulated by core 3' end processing and splicing factors reveals regulatory rules of alternative cleavage and polyadenylation.
- Author
-
Wencheng Li, Bei You, Mainul Hoque, Dinghai Zheng, Wenting Luo, Zhe Ji, Ji Yeon Park, Samuel I Gunderson, Auinash Kalsotra, James L Manley, and Bin Tian
- Subjects
Genetics ,QH426-470 - Abstract
Alternative cleavage and polyadenylation (APA) results in mRNA isoforms containing different 3' untranslated regions (3'UTRs) and/or coding sequences. How core cleavage/polyadenylation (C/P) factors regulate APA is not well understood. Using siRNA knockdown coupled with deep sequencing, we found that several C/P factors can play significant roles in 3'UTR-APA. Whereas Pcf11 and Fip1 enhance usage of proximal poly(A) sites (pAs), CFI-25/68, PABPN1 and PABPC1 promote usage of distal pAs. Strong cis element biases were found for pAs regulated by CFI-25/68 or Fip1, and the distance between pAs plays an important role in APA regulation. In addition, intronic pAs are substantially regulated by splicing factors, with U1 mostly inhibiting C/P events in introns near the 5' end of gene and U2 suppressing those in introns with features for efficient splicing. Furthermore, PABPN1 inhibits expression of transcripts with pAs near the transcription start site (TSS), a property possibly related to its role in RNA degradation. Finally, we found that groups of APA events regulated by C/P factors are also modulated in cell differentiation and development with distinct trends. Together, our results support an APA code where an APA event in a given cellular context is regulated by a number of parameters, including relative location to the TSS, splicing context, distance between competing pAs, surrounding cis elements and concentrations of core C/P factors.
- Published
- 2015
- Full Text
- View/download PDF
4. U1 Adaptor Oligonucleotides Targeting BCL2 and GRM1 Suppress Growth of Human Melanoma Xenografts In Vivo
- Author
-
Rafal Goraczniak, Brian A Wall, Mark A Behlke, Kim A Lennox, Eric S Ho, Nikolas H Zaphiros, Christopher Jakubowski, Neil R Patel, Steven Zhao, Carlo Magaway, Stacey A Subbie, Lumeng Jenny Yu, Stephanie LaCava, Kenneth R Reuhl, Suzie Chen, and Samuel I Gunderson
- Subjects
cancer therapy ,dendrimer ,gene silencing ,oligonucleotide therapeutic ,tumor targeting ,Therapeutics. Pharmacology ,RM1-950 - Abstract
U1 Adaptor is a recently discovered oligonucleotide-based gene-silencing technology with a unique mechanism of action that targets nuclear pre-mRNA processing. U1 Adaptors have two distinct functional domains, both of which must be present on the same oligonucleotide to exert their gene-silencing function. Here, we present the first in vivo use of U1 Adaptors by targeting two different human genes implicated in melanomagenesis, B-cell lymphoma 2 (BCL2) and metabotropic glutamate receptor 1 (GRM1), in a human melanoma cell xenograft mouse model system. Using a newly developed dendrimer delivery system, anti-BCL2 U1 Adaptors were very potent and suppressed tumor growth at doses as low as 34 µg/kg with twice weekly intravenous (iv) administration. Anti-GRM1 U1 Adaptors suppressed tumor xenograft growth with similar potency. Mechanism of action was demonstrated by showing target gene suppression in tumors and by observing that negative control U1 Adaptors with just one functional domain show no tumor suppression activity. The anti-BCL2 and anti-GRM1 treatments were equally effective against cell lines harboring either wild-type or a mutant V600E B-RAF allele, the most common mutation in melanoma. Treatment of normal immune-competent mice (C57BL6) indicated no organ toxicity or immune stimulation. These proof-of-concept studies represent an in-depth (over 800 mice in ~108 treatment groups) validation that U1 Adaptors are a highly potent gene-silencing therapeutic and open the way for their further development to treat other human diseases.
- Published
- 2013
- Full Text
- View/download PDF
5. U1 Adaptors Suppress the KRAS-MYC Oncogenic Axis in Human Pancreatic Cancer Xenografts
- Author
-
Eric S. Ho, Mark Brenneman, Crissy Dudgeon, Rafal Goraczniak, Lan Yi, Xin Yu, Samuel I. Gunderson, Ashley T. Tsang, Kristen Donohue, Darren R. Carpizo, and Samuel Kogan
- Subjects
0301 basic medicine ,Cancer Research ,Cancer ,Biology ,medicine.disease ,medicine.disease_cause ,Molecular biology ,digestive system diseases ,In vitro ,03 medical and health sciences ,030104 developmental biology ,Oncology ,Cell culture ,In vivo ,Apoptosis ,Pancreatic cancer ,medicine ,Cancer research ,Gene silencing ,KRAS - Abstract
Targeting KRAS and MYC has been a tremendous challenge in cancer drug development. Genetic studies in mouse models have validated the efficacy of silencing expression of both KRAS and MYC in mutant KRAS-driven tumors. We investigated the therapeutic potential of a new oligonucleotide-mediated gene silencing technology (U1 Adaptor) targeting KRAS and MYC in pancreatic cancer. Nanoparticles in complex with anti-KRAS U1 Adaptors (U1-KRAS) showed remarkable inhibition of KRAS in different human pancreatic cancer cell lines in vitro and in vivo. As a nanoparticle-free approach is far easier to develop into a drug, we refined the formulation of U1 Adaptors by conjugating them to tumor-targeting peptides (iRGD and cRGD). Peptides coupled to fluorescently tagged U1 Adaptors showed selective tumor localization in vivo. Efficacy experiments in pancreatic cancer xenograft models showed highly potent (>90%) antitumor activity of both iRGD and (cRGD)2-KRAS Adaptors. U1 Adaptors targeting MYC inhibited pancreatic cancer cell proliferation caused by apoptosis in vitro (40%–70%) and tumor regressions in vivo. Comparison of iRGD-conjugated U1 KRAS and U1 MYC Adaptors in vivo revealed a significantly greater degree of cleaved caspase-3 staining and decreased Ki67 staining as compared with controls. There was no significant difference in efficacy between the U1 KRAS and U1 MYC Adaptor groups. Our results validate the value in targeting both KRAS and MYC in pancreatic cancer therapeutics and provide evidence that the U1 Adaptor technology can be successfully translated using a nanoparticle-free delivery system to target two undruggable genes in cancer. Mol Cancer Ther; 16(8); 1445–55. ©2017 AACR.
- Published
- 2017
- Full Text
- View/download PDF
6. A multispecies polyadenylation site model.
- Author
-
Eric S. Ho, Samuel I. Gunderson, and Siobain Duffy
- Published
- 2013
- Full Text
- View/download PDF
7. iTriplet, a rule-based nucleic acid sequence motif finder.
- Author
-
Eric S. Ho, Christopher D. Jakubowski, and Samuel I. Gunderson
- Published
- 2009
- Full Text
- View/download PDF
8. U1 Adaptors Suppress the
- Author
-
Ashley T, Tsang, Crissy, Dudgeon, Lan, Yi, Xin, Yu, Rafal, Goraczniak, Kristen, Donohue, Samuel, Kogan, Mark A, Brenneman, Eric S, Ho, Samuel I, Gunderson, and Darren R, Carpizo
- Subjects
Oligonucleotides ,Mice, Nude ,Reproducibility of Results ,Oncogenes ,Xenograft Model Antitumor Assays ,digestive system diseases ,Article ,Pancreatic Neoplasms ,Proto-Oncogene Proteins c-myc ,Proto-Oncogene Proteins p21(ras) ,Cell Line, Tumor ,Mutation ,Animals ,Humans ,Female ,Peptides ,neoplasms ,Cell Proliferation - Abstract
Targeting KRAS and MYC has been a tremendous challenge in cancer drug development. Genetic studies in mouse models have validated the efficacy of silencing expression of both KRAS and MYC in mutant KRAS driven tumors. We investigated the therapeutic potential of a new oligonucleotide-mediated gene silencing technology (U1 Adaptor) targeting KRAS and MYC in pancreatic cancer. Nanoparticles in complex with anti-KRAS U1 Adaptors (U1-KRAS) showed remarkable inhibition of KRAS in different human pancreatic cancer cell lines in vitro and in vivo. As a nanoparticle-free approach is far easier to develop into a drug, we refined the formulation of U1 Adaptors by conjugating them to tumor targeting peptides (iRGD and cRGD). Peptides coupled to fluorescently tagged U1 Adaptors showed selective tumor localization in vivo. Efficacy experiments in pancreatic cancer xenograft models showed highly potent (>90%) anti-tumor activity of both iRGD and (cRGD)2-KRAS Adaptors. U1 Adaptors targeting MYC inhibited pancreatic cancer cell proliferation caused apoptosis in vitro (40–70%) and tumor regressions in vivo. Comparison of iRGD conjugated U1 KRAS and U1 MYC Adaptors in vivo revealed a significantly greater degree of cleaved caspase-3 staining and decreased Ki67 staining as compared with controls. There was no significant difference in efficacy between the U1 KRAS and U1 MYC Adaptor groups. Our results validate the value in targeting both KRAS and MYC in pancreatic cancer therapeutics and provide evidence that the U1 Adaptor technology can be successfully translated using a nanoparticle free delivery system to target two undruggable genes in cancer.
- Published
- 2016
9. Long Conserved Fragments Upstream of Mammalian Polyadenylation Sites
- Author
-
Samuel I. Gunderson and Eric S. Ho
- Subjects
RNA, Untranslated ,Polyadenylation ,conserved fragments ,Biology ,Conserved sequence ,Evolution, Molecular ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Transcription (biology) ,RNA polymerase ,Databases, Genetic ,RNA Precursors ,Genetics ,Animals ,Deoxyribonuclease I ,Humans ,Selection, Genetic ,Platypus ,Conserved Sequence ,Research Articles ,Ecology, Evolution, Behavior and Systematics ,030304 developmental biology ,Expressed Sequence Tags ,Regulation of gene expression ,0303 health sciences ,Computational Biology ,RNA ,bioinformatics ,Chromatin ,Rats ,Gene Expression Regulation ,chemistry ,cis-regulatory elements ,RNA splicing ,Cattle ,030217 neurology & neurosurgery - Abstract
Polyadenylation is a cotranscriptional nuclear RNA processing event involving endonucleolytic cleavage of the nascent, emerging pre-messenger RNA (pre-mRNA) from the RNA polymerase, immediately followed by the polymerization of adenine ribonucleotides, called the poly(A) tail, to the cleaved 3′ end of the polyadenylation site (PAS). This apparently simple molecular processing step has been discovered to be connected to transcription and splicing therefore increasing its potential for regulation of gene expression. Here, through a bioinformatic analysis of cis-PAS–regulatory elements in mammals that includes taking advantage of multiple evolutionary time scales, we find unexpected selection pressure much further upstream, up to 200 nt, from the PAS than previously thought. Strikingly, close to 3,000 long (30–500 nt) noncoding conserved fragments (CFs) were discovered in the PAS flanking region of three remotely related mammalian species, human, mouse, and cow. When an even more remote transitional mammal, platypus, was included, still over a thousand CFs were found in the proximity of the PAS. Even though the biological function of these CFs remains unknown, their considerable sizes makes them unlikely to serve as protein recognition sites, which are typically ≤15 nt. By harnessing genome wide DNaseI hypersensitivity data, we have discovered that the presence of CFs correlates with chromatin accessibility. Our study is important in highlighting novel experimental targets, which may provide new understanding about the regulatory aspects of polyadenylation.
- Published
- 2011
- Full Text
- View/download PDF
10. Next-generation SELEX identifies sequence and structural determinants of splicing factor binding in human pre-mRNA sequence
- Author
-
Lauren Alpert, William G. Fairbrother, Brian L. Chang, William Thompson, Samuel I. Gunderson, and Daniel C. Reid
- Subjects
Base Sequence ,RNA Splicing ,Method ,Sequence Analysis, DNA ,Computational biology ,Biology ,Ligands ,Ribonucleoproteins, Small Nuclear ,Molecular biology ,Exon ,Polypyrimidine tract ,RNA splicing ,RNA Precursors ,Splicing factor binding ,biology.protein ,Humans ,Nucleic Acid Conformation ,Polypyrimidine tract-binding protein ,Precursor mRNA ,Molecular Biology ,Systematic evolution of ligands by exponential enrichment ,Polypyrimidine Tract-Binding Protein ,Protein Binding ,Ribonucleoprotein - Abstract
Many splicing factors interact with both mRNA and pre-mRNA. The identification of these interactions has been greatly improved by the development of in vivo cross-linking immunoprecipitation. However, the output carries a strong sampling bias in favor of RNPs that form on more abundant RNA species like mRNA. We have developed a novel in vitro approach for surveying binding on pre-mRNA, without cross-linking or sampling bias. Briefly, this approach entails specifically designed oligonucleotide pools that tile through a pre-mRNA sequence. The pool is then partitioned into bound and unbound fractions, which are quantified by a two-color microarray. We applied this approach to locating splicing factor binding sites in and around ∼4000 exons. We also quantified the effect of secondary structure on binding. The method is validated by the finding that U1snRNP binds at the 5′ splice site (5′ss) with a specificity that is nearly identical to the splice donor motif. In agreement with prior reports, we also show that U1snRNP appears to have some affinity for intronic G triplets that are proximal to the 5′ss. Both U1snRNP and the polypyrimidine tract binding protein (PTB) avoid exonic binding, and the PTB binding map shows increased enrichment at the polypyrimidine tract. For PTB, we confirm polypyrimidine specificity and are also able to identify structural determinants of PTB binding. We detect multiple binding motifs enriched in the PTB bound fraction of oligonucleotides. These motif combinations augment binding in vitro and are also enriched in the vicinity of exons that have been determined to be in vivo targets of PTB.
- Published
- 2009
- Full Text
- View/download PDF
11. Gene silencing by synthetic U1 Adaptors
- Author
-
Samuel I. Gunderson, Mark A. Behlke, and Rafal Goraczniak
- Subjects
Small interfering RNA ,Molecular Sequence Data ,Trans-acting siRNA ,Biomedical Engineering ,Bioengineering ,Biology ,Transfection ,Applied Microbiology and Biotechnology ,Cell Line ,Ribonucleoprotein, U1 Small Nuclear ,Exon ,Genes, Reporter ,RNA interference ,RNA, Small Nuclear ,Animals ,Humans ,Gene Silencing ,RNA, Small Interfering ,Luciferases ,Base Sequence ,RNA ,Sequence Analysis, DNA ,Molecular biology ,Proto-Oncogene Proteins c-raf ,RNA silencing ,Gene Expression Regulation ,Molecular Medicine ,Small nuclear ribonucleoprotein ,Small nuclear RNA ,HeLa Cells ,Biotechnology - Abstract
We describe a gene silencing method that employs a mechanism of action distinct from those of antisense and RNA interference. U1 Adaptors are bifunctional oligonucleotides with a 'target domain' complementary to a site in the target gene's terminal exon and a 'U1 domain' that binds to the U1 small nuclear RNA component of the U1 small nuclear ribonucleoprotein (U1 snRNP) splicing factor. Tethering of U1 snRNP to the target pre-mRNA inhibits poly(A)-tail addition, causing degradation of that RNA species in the nucleus. U1 Adaptors can inhibit both endogenous and reporter genes in a sequence-specific manner. Comparison of U1 Adaptors with small interfering RNA (siRNA) using a genome-wide microarray analysis indicates that U1 Adaptors have limited off-target effects and no detectable adverse effects on splicing. Further, targeting the same gene either with multiple U1 Adaptors or with a U1 Adaptor and siRNA strongly enhances gene silencing.
- Published
- 2009
- Full Text
- View/download PDF
12. The Regulatory Element in the 3′-Untranslated Region of Human Papillomavirus 16 Inhibits Expression by Binding CUG-binding Protein 1
- Author
-
Rafal Goraczniak and Samuel I. Gunderson
- Subjects
Gene Expression Regulation, Viral ,Small interfering RNA ,Ultraviolet Rays ,Negative regulatory element ,RNA-binding protein ,Biology ,Biochemistry ,SnRNP binding ,RNA, Small Nuclear ,polycyclic compounds ,Humans ,Point Mutation ,RNA, Small Interfering ,Binding site ,3' Untranslated Regions ,Molecular Biology ,CELF1 Protein ,Bovine papillomavirus 1 ,Human papillomavirus 16 ,Binding Sites ,Three prime untranslated region ,RNA-Binding Proteins ,RNA ,RNA 3' Polyadenylation Signals ,Cell Biology ,Molecular biology ,RNA, Viral ,Small nuclear ribonucleoprotein ,HeLa Cells - Abstract
The 3'-untranslated regions (UTRs) of human papillomavirus 16 (HPV16) and bovine papillomavirus 1 (BPV1) contain a negative regulatory element (NRE) that inhibits viral late gene expression. The BPV1 NRE consists of a single 9-nucleotide (nt) U1 small nuclear ribonucleoprotein (snRNP) base pairing site (herein called a U1 binding site) that via U1 snRNP binding leads to inhibition of the late poly(A) site. The 79-nt HPV16 NRE is far more complicated, consisting of 4 overlapping very weak U1 binding sites followed by a poorly understood GU-rich element (GRE). We undertook a molecular dissection of the HPV16 GRE and identify via UV cross-linking, RNA affinity chromatography, and mass spectrometry that is bound by the CUG-binding protein 1 (CUGBP1). Reporter assays coupled with knocking down CUGBP1 levels by small interfering RNA and Dox-regulated shRNA, demonstrate CUGBP1 is inhibitory in vivo. CUGBP1 is the first GRE-binding protein to have RNA interfering knockdown evidence in support of its role in vivo. Several fine-scale GRE mutations that inactivate GRE activity in vivo and GRE binding to CUGBP1 in vitro are identified. The CUGBP1.GRE complex has no activity on its own but specifically synergizes with weak U1 binding sites to inhibit expression in vivo. No synergy is seen if the U1 binding sites are made weaker by a 1-nt down-mutation or made stronger by a 1-nt up-mutation, underscoring that the GRE operates only on weak sites. Interestingly, inhibition occurs at multiple levels, in particular at the level of poly(A) site activity, nuclear-cytoplasmic export, and translation of the mRNA. Implications for understanding the HPV16 life cycle are discussed.
- Published
- 2008
- Full Text
- View/download PDF
13. The transcription factor RUNX2 regulates receptor tyrosine kinase expression in melanoma
- Author
-
Anna Rabkin, Marina Chekmareva, Samuel I. Gunderson, Suzie Chen, David J. Foran, Wenjin Chen, Rajeev K. Boregowda, Michael A. Bryan, Ahmed Lasfar, Daniel J. Medina, James S. Goydos, Karine A. Cohen-Solal, Michael J. Vido, and Elke Markert
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,RUNX2 ,Core Binding Factor Alpha 1 Subunit ,Receptor tyrosine kinase ,resistance to targeted therapy ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,medicine ,melanoma ,Humans ,Autocrine signalling ,Protein kinase B ,neoplasms ,PI3K/AKT/mTOR pathway ,transcription factor ,biology ,AXL receptor tyrosine kinase ,Melanoma ,fungi ,Receptor Protein-Tyrosine Kinases ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Autocrine Communication ,030104 developmental biology ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,ROR1 ,biology.protein ,Cancer research ,receptor tyrosine kinase ,Research Paper - Abstract
Receptor tyrosine kinases-based autocrine loops largely contribute to activate the MAPK and PI3K/AKT pathways in melanoma. However, the molecular mechanisms involved in generating these autocrine loops are still largely unknown. In the present study, we examine the role of the transcription factor RUNX2 in the regulation of receptor tyrosine kinase (RTK) expression in melanoma. We have demonstrated that RUNX2-deficient melanoma cells display a significant decrease in three receptor tyrosine kinases, EGFR, IGF-1R and PDGFRβ. In addition, we found co-expression of RUNX2 and another RTK, AXL, in both melanoma cells and melanoma patient samples. We observed a decrease in phosphoAKT2 (S474) and phosphoAKT (T308) levels when RUNX2 knock down resulted in significant RTK down regulation. Finally, we showed a dramatic up regulation of RUNX2 expression with concomitant up-regulation of EGFR, IGF-1R and AXL in melanoma cells resistant to the BRAF V600E inhibitor PLX4720. Taken together, our results strongly suggest that RUNX2 might be a key player in RTK-based autocrine loops and a mediator of resistance to BRAF V600E inhibitors involving RTK up regulation in melanoma.
- Published
- 2015
14. Systematic profiling of poly(A)+ transcripts modulated by core 3' end processing and splicing factors reveals regulatory rules of alternative cleavage and polyadenylation
- Author
-
Wenting Luo, Dinghai Zheng, Bei You, Mainul Hoque, Samuel I. Gunderson, Bin Tian, Zhe Ji, James L. Manley, Wencheng Li, Ji Yeon Park, and Auinash Kalsotra
- Subjects
Untranslated region ,Cancer Research ,Polyadenylation ,lcsh:QH426-470 ,RNA Splicing ,RNA Stability ,education ,Biology ,Poly(A)-Binding Protein I ,03 medical and health sciences ,Exon ,0302 clinical medicine ,Gene expression ,Genetics ,Humans ,RNA, Messenger ,Molecular Biology ,3' Untranslated Regions ,Genetics (clinical) ,Ecology, Evolution, Behavior and Systematics ,030304 developmental biology ,Regulation of gene expression ,0303 health sciences ,Three prime untranslated region ,Intron ,High-Throughput Nucleotide Sequencing ,Cell Differentiation ,Exons ,Introns ,lcsh:Genetics ,Gene Expression Regulation ,RNA splicing ,030217 neurology & neurosurgery ,Research Article - Abstract
Alternative cleavage and polyadenylation (APA) results in mRNA isoforms containing different 3’ untranslated regions (3’UTRs) and/or coding sequences. How core cleavage/polyadenylation (C/P) factors regulate APA is not well understood. Using siRNA knockdown coupled with deep sequencing, we found that several C/P factors can play significant roles in 3’UTR-APA. Whereas Pcf11 and Fip1 enhance usage of proximal poly(A) sites (pAs), CFI-25/68, PABPN1 and PABPC1 promote usage of distal pAs. Strong cis element biases were found for pAs regulated by CFI-25/68 or Fip1, and the distance between pAs plays an important role in APA regulation. In addition, intronic pAs are substantially regulated by splicing factors, with U1 mostly inhibiting C/P events in introns near the 5’ end of gene and U2 suppressing those in introns with features for efficient splicing. Furthermore, PABPN1 inhibits expression of transcripts with pAs near the transcription start site (TSS), a property possibly related to its role in RNA degradation. Finally, we found that groups of APA events regulated by C/P factors are also modulated in cell differentiation and development with distinct trends. Together, our results support an APA code where an APA event in a given cellular context is regulated by a number of parameters, including relative location to the TSS, splicing context, distance between competing pAs, surrounding cis elements and concentrations of core C/P factors., Author Summary A gene can express multiple isoforms varying in the 3’ end, a phenomenon called alternative cleavage and polyadenylation, or APA. Previous studies have indicated that most eukaryotic genes display APA and the APA profile changes under different physiological and pathological conditions. However, how APA is regulated in the cell is unclear. Here using gene knockdown and high throughput sequencing we examine how APA is regulated by factors in the machinery responsible for cleavage and polyadenylation as well as factors that play essential roles in splicing. We identify several factors that play significant roles in APA in the last exon, including CFI-25/68, PABPN1, PABPC1, Fip1 and Pcf11. We also elucidate how cleavage and polyadenylation events are regulated in introns and near the transcription start site. We uncover a group of APA events that are highly regulated by core factors as well as in cell differentiation and development. We present an APA code where an APA event in a given cellular context is regulated by a number of parameters, including relative location to the transcription start site, splicing context, distance between competing pAs, surrounding cis elements and concentrations of core cleavage and polyadenylation factors.
- Published
- 2015
15. Non-snRNP U1A levels decrease during mammalian B-cell differentiation and release the IgM secretory poly(A) site from repression
- Author
-
Catherine Phillips, Jianglin Ma, and Samuel I. Gunderson
- Subjects
Polyadenylation ,Cellular differentiation ,Cell ,RNA-binding protein ,Models, Biological ,Article ,Cell Line ,Ribonucleoprotein, U1 Small Nuclear ,Mice ,RNA, Small Nuclear ,RNA Precursors ,medicine ,Animals ,Humans ,RNA, Messenger ,Molecular Biology ,B cell ,B-Lymphocytes ,Messenger RNA ,biology ,RNA-Binding Proteins ,Cell Differentiation ,Ribonucleoproteins, Small Nuclear ,Molecular biology ,medicine.anatomical_structure ,Immunoglobulin M ,Cell culture ,biology.protein ,HeLa Cells - Abstract
A regulated shift from the production of membrane to secretory forms of Immunoglobulin M (IgM) mRNA occurs during B cell differentiation due to the activation of an upstream secretory poly(A) site. U1A plays a key role in inhibiting the expression of the secretory poly(A) site by inhibiting both cleavage at the poly(A) site and subsequent poly(A) tail addition. However, how the inhibitory effect of U1A is alleviated in differentiated cells, which express the secretory poly(A) site, is not known. Using B cell lines representing different stages of B cell differentiation, we show that the amount of U1A available to inhibit the secretory poly(A) site is reduced in differentiated cells. Undifferentiated B cells have more total U1A than differentiated cells and a greater proportion of this is not associated with the U1snRNP. We show that this is available to inhibit poly(A) addition at the secretory poly(A) site using cold competitor RNA oligos to de-repress poly(A) addition in nuclear extracts from the respective cell lines. In addition, endogenous non-snRNP associated U1A—immunopurified from the different cell lines—inhibits poly(A) polymerase activity proportional to U1A recovered, suggesting that available U1A level alone is responsible for changes in its inhibitory effect at the secretory IgM poly (A) site.
- Published
- 2005
- Full Text
- View/download PDF
16. PRMT7, a New Protein Arginine Methyltransferase That Synthesizes Symmetric Dimethylarginine
- Author
-
Jin-Hyung Lee, Arthur M. Felix, Olga Mirochnitchenko, Ralf Hoffmann, Zhi-Hong Yang, Samuel I. Gunderson, Sidney Pestka, Nicole Herth, and Jeffry R. Cook
- Subjects
Fibrillarin ,Protein-Arginine N-Methyltransferases ,Methyltransferase ,Protein arginine methyltransferase 5 ,Molecular Sequence Data ,Methyltransferases ,Cell Biology ,Biology ,Arginine ,Biochemistry ,Myelin basic protein ,Histone H4 ,chemistry.chemical_compound ,chemistry ,Complementary DNA ,Escherichia coli ,biology.protein ,Humans ,Amino Acid Sequence ,Asymmetric dimethylarginine ,Molecular Biology ,HeLa Cells - Abstract
The cDNA for PRMT7, a recently discovered human protein-arginine methyltransferase (PRMT), was cloned and expressed in Escherichia coli and mammalian cells. Immunopurified PRMT7 actively methylated histones, myelin basic protein, a fragment of human fibrillarin (GAR) and spliceosomal protein SmB. Amino acid analysis showed that the modifications produced were predominantly monomethylarginine and symmetric dimethylarginine (SDMA). Examination of PRMT7 expressed in E. coli demonstrated that peptides corresponding to sequences contained in histone H4, myelin basic protein, and SmD3 were methylated. Furthermore, analysis of the methylated proteins showed that symmetric dimethylarginine and relatively small amounts of monomethylarginine and asymmetric dimethylarginine were produced. SDMA was also formed when a GRG tripeptide was methylated by PRMT7, indicating that a GRG motif is by itself sufficient for symmetric dimethylation to occur. Symmetric dimethylation is reduced dramatically compared with monomethylation as the concentration of the substrate is increased. The data demonstrate that PRMT7 (like PRMT5) is a Type II methyltransferase capable of producing SDMA modifications in proteins.
- Published
- 2005
- Full Text
- View/download PDF
17. U1A Inhibits Cleavage at the Immunoglobulin M Heavy-Chain Secretory Poly(A) Site by Binding between the Two Downstream GU-Rich Regions
- Author
-
Catherine Phillips, Samuel I. Gunderson, and Niseema Pachikara
- Subjects
Polyadenylation ,Gene Expression ,RNA-binding protein ,Plasma protein binding ,Biology ,Cleavage (embryo) ,Ribonucleoprotein, U1 Small Nuclear ,Conserved sequence ,RNA Precursors ,Animals ,Humans ,RNA Processing, Post-Transcriptional ,Molecular Biology ,Conserved Sequence ,Base Composition ,Cleavage stimulation factor ,RNA-Binding Proteins ,Cell Biology ,Molecular biology ,Protein Structure, Tertiary ,Protein Subunits ,A-site ,Cleavage Stimulation Factor ,Immunoglobulin M ,Mutation ,Nucleic Acid Conformation ,Immunoglobulin heavy chain ,Immunoglobulin Heavy Chains ,HeLa Cells ,Protein Binding - Abstract
The immunoglobulin M heavy-chain locus contains two poly(A) sites which are alternatively expressed during B-cell differentiation. Despite its promoter proximal location, the secretory poly(A) site is not expressed in undifferentiated cells. Crucial to the activation of the secretory poly(A) site during B-cell differentiation are changes in the binding of cleavage stimulatory factor 64K to GU-rich elements downstream of the poly(A) site. What regulates this change is not understood. The secretory poly(A) site contains two downstream GU-rich regions separated by a 29-nucleotide sequence. Both GU-rich regions are necessary for binding of the specific cleavage-polyadenylation complex. We demonstrate here that U1A binds two (AUGCN(1-3)C) motifs within the 29-nucleotide sequence and inhibits the binding of cleavage stimulatory factor 64K and cleavage at the secretory poly(A) site.
- Published
- 2004
- Full Text
- View/download PDF
18. Sequences Adjacent to the 5′ Splice Site Control U1A Binding Upstream of the IgM Heavy Chain Secretory Poly(A) Site
- Author
-
Samuel I. Gunderson and Catherine Phillips
- Subjects
RNase P ,Amino Acid Motifs ,Biology ,Biochemistry ,Ribonucleoprotein, U1 Small Nuclear ,Ribonucleases ,Escherichia coli ,RNA, Messenger ,Binding site ,Molecular Biology ,Polymerase ,Messenger RNA ,Binding Sites ,Models, Genetic ,RNA-Binding Proteins ,Cell Biology ,Molecular biology ,Recombinant Proteins ,Protein Structure, Tertiary ,Cell biology ,DNA binding site ,Alternative Splicing ,A-site ,Immunoglobulin M ,Lead ,Mutation ,RNA splicing ,biology.protein ,Nucleic Acid Conformation ,RNA ,Small nuclear RNA ,Plasmids ,Protein Binding - Abstract
We have recently shown that the stability of the alternatively expressed immunoglobulin M heavy chain secretory mRNA is developmentally regulated by U1A. U1A binds novel non-consensus sites upstream of the secretory poly(A) site and inhibits poly(A) tail addition in undifferentiated cells. U1A's dependence for binding and function upon a stem-loop structure has been extensively characterized for the consensus sites. We therefore probed the structure surrounding the novel U1A binding sites. We show that two of the three novel binding sites represent the major single-stranded regions upstream of the secretory poly(A) site, consistent with a major role at this site. The strength of binding and ability of U1A to inhibit poly(A) polymerase correlate with the accessibility of the novel sites. However, long range interactions are responsible for maintaining them in an open configuration. Mutation of an RNase V1-sensitive site 102 nucleotides upstream, directly adjacent to the competing 5' splice site, changes the structure of one the U1A binding sites and thus abolishes the binding of the second U1A molecule and the ability of U1A ability to inhibit poly(A) polymerase activity at this site. These sites bind U1A via its N-terminal domain but with a 10-fold lower affinity than U1 small nuclear RNA. This lower binding affinity is more conducive to U1A's regulation of poly(A) tail addition to heterologous mRNA.
- Published
- 2003
- Full Text
- View/download PDF
19. Determinants within an 18-Amino-Acid U1A Autoregulatory Domain That Uncouple Cooperative RNA Binding, Inhibition of Polyadenylation, and Homodimerization
- Author
-
Reem I. Hussein, Samuel I. Gunderson, Fei Guan, and Daphne Palacios
- Subjects
Models, Molecular ,Polyadenylation ,DNA Mutational Analysis ,Molecular Sequence Data ,Mutant ,Gene Expression ,Ribonucleoprotein, U1 Small Nuclear ,Structure-Activity Relationship ,Protein structure ,Homeostasis ,Humans ,Amino Acid Sequence ,Amino Acids ,Binding site ,Molecular Biology ,Peptide sequence ,Polymerase ,Binding Sites ,biology ,Polynucleotide Adenylyltransferase ,RNA-Binding Proteins ,RNA ,Cooperative binding ,Cell Biology ,Recombinant Proteins ,Protein Structure, Tertiary ,Biochemistry ,Mutation ,biology.protein ,Dimerization ,HeLa Cells ,Protein Binding - Abstract
The human U1 snRNP-specific U1A protein autoregulates its own production by binding to and inhibiting the polyadenylation of its own pre-mRNA. Previous work demonstrated that a short sequence of U1A protein is essential for autoregulation and contains three distinct activities, which are (i) cooperative binding of two U1A proteins to a 50-nucleotide region of U1A pre-mRNA called polyadenylation-inhibitory element RNA, (ii) formation of a novel homodimerization surface, and (iii) inhibition of polyadenylation by inhibition of poly(A) polymerase (PAP). In this study, we purified and analyzed 11 substitution mutant proteins, each having one or two residues in this region mutated. In 5 of the 11 mutant proteins, we found that particular amino acids associate with one activity but not another, indicating that they can be uncoupled. Surprisingly, in three mutant proteins, these activities were improved upon, suggesting that U1A autoregulation is selected for suboptimal inhibitory efficiency. The effects of these mutations on autoregulatory activity in vivo were also determined. Only U1A and U170K are known to regulate nuclear polyadenylation by PAP inhibition; thus, these results will aid in determining how widespread this type of regulation is. Our molecular dissection of the consequences of conformational changes within an RNP complex presents a powerful example to those studying more complicated pre-mRNA-regulatory systems.
- Published
- 2003
- Full Text
- View/download PDF
20. Identification of New Poly(A) Polymerase-inhibitory Proteins Capable of Regulating Pre-mRNA Polyadenylation
- Author
-
Samuel I. Gunderson and Bom Ko
- Subjects
Polyadenylation ,RNA-binding protein ,Transfection ,SR protein ,Structural Biology ,Gene expression ,Escherichia coli ,RNA Precursors ,Humans ,RNA Processing, Post-Transcriptional ,Molecular Biology ,Polymerase ,Serine-Arginine Splicing Factors ,biology ,Nuclear Proteins ,Polynucleotide Adenylyltransferase ,RNA-Binding Proteins ,RNA ,Splicing Factor U2AF ,Molecular biology ,Cell biology ,Ribonucleoproteins ,RNA splicing ,biology.protein ,Precursor mRNA ,HeLa Cells - Abstract
The 3' ends of nearly all eukaryotic pre-mRNAs undergo cleavage and polyadenylation, thereby acquiring a poly(A) tail added by the enzyme poly(A) polymerase (PAP). Two well-characterized examples of regulated poly(A) tail addition in the nucleus consist of spliceosomal proteins, either the U1A or U170K proteins, binding to the pre-mRNA and inhibiting PAP via their PAP regulatory domains (PRDs). These two proteins are the only known examples of this type of gene regulation. On the basis of sequence comparisons, it was predicted that many other proteins, including some members of the SR family of splicing proteins, contain functional PRDs. Here we demonstrate that the putative PRDs found in the SR domains of the SR proteins SRP75 and U2AF65, via fusion to a heterologous MS2 RNA binding protein, specifically and efficiently inhibit PAP in vitro and pre-mRNA polyadenylation in vitro and in vivo. A similar region from the SR domain of SRP40 does not exhibit these activities, indicating that this is not a general property of SR domains. We find that the polyadenylation- and PAP-inhibitory activity of a given polypeptide can be accurately predicted based on sequence similarity to known PRDs and can be measured even if the polypeptides' RNA target is unknown. Our results also indicate that PRDs function as part of a network of interactions within the pre-mRNA processing complex and suggest that this type of regulation will be more widespread than previously thought.
- Published
- 2002
- Full Text
- View/download PDF
21. Regulation of nuclear poly(A) addition controls the expression of immunoglobulin M secretory mRNA
- Author
-
Catherine Phillips, Stephen P. Jung, and Samuel I. Gunderson
- Subjects
Polyadenylation ,Cellular differentiation ,Amino Acid Motifs ,DNA Mutational Analysis ,Molecular Sequence Data ,Biology ,Transfection ,Article ,General Biochemistry, Genetics and Molecular Biology ,Ribonucleases ,Escherichia coli ,medicine ,Animals ,Humans ,RNA, Messenger ,Binding site ,Molecular Biology ,Cell Nucleus ,Regulation of gene expression ,B-Lymphocytes ,Messenger RNA ,Binding Sites ,Base Sequence ,Dose-Response Relationship, Drug ,Models, Genetic ,General Immunology and Microbiology ,General Neuroscience ,Cell Differentiation ,Molecular biology ,Recombinant Proteins ,Cell nucleus ,Secretory protein ,medicine.anatomical_structure ,Gene Expression Regulation ,Immunoglobulin M ,Mutation ,Cattle ,Poly A ,HeLa Cells ,Plasmids ,Protein Binding - Abstract
B-cell differentiation is accompanied by a dramatic increase in cytoplasmic accumulation and stability of the IgM heavy chain (mu) secretory mRNA. Despite considerable effort, the mechanism is unknown. We have identified three short motifs upstream of the secretory poly(A) site, which, when mutated in the mu heavy chain gene, significantly increase the accumulation of the secretory form of poly(A)(+) mRNA relative to the membrane form and regulate the expression of the secretory poly(A) site in a developmental manner. We show that these motifs bind U1A and inhibit polyadenylation in vitro and in vivo. Overexpression of U1A in vivo results in the selective inhibition of the secretory form. Thus, this novel mechanism selectively controls post-cleavage expression of the mu secretory mRNA. We present evidence that this mechanism is used to regulate alternative expression of other genes.
- Published
- 2001
- Full Text
- View/download PDF
22. Reduction of Target Gene Expression by a Modified U1 snRNA
- Author
-
Alexander C. Lichtler, S. A. Beckley, Peng Liu, David W. Rowe, Samuel I. Gunderson, and Mary Louise Stover
- Subjects
Chloramphenicol O-Acetyltransferase ,Green Fluorescent Proteins ,Gene Expression ,Biology ,Transfection ,Green fluorescent protein ,Mice ,Genes, Reporter ,RNA, Small Nuclear ,Gene expression ,Animals ,Molecular Biology ,Reporter gene ,Base Sequence ,Intron ,Gene targeting ,RNA ,3T3 Cells ,DNA ,Cell Biology ,beta-Galactosidase ,Molecular biology ,Luminescent Proteins ,Antitarget ,Gene Targeting ,Small nuclear RNA - Abstract
Although the primary function of U1 snRNA is to define the 5' donor site of an intron, it can also block the accumulation of a specific RNA transcript when it binds to a donor sequence within its terminal exon. This work was initiated to investigate if this property of U1 snRNA could be exploited as an effective method for inactivating any target gene. The initial 10-bp segment of U1 snRNA, which is complementary to the 5' donor sequence, was modified to recognize various target mRNAs (chloramphenicol acetyltransferase [CAT], beta-galactosidase, or green fluorescent protein [GFP]). Transient cotransfection of reporter genes and appropriate U1 antitarget vectors resulted in >90% reduction of transgene expression. Numerous sites within the CAT transcript were suitable for targeting. The inhibitory effect of the U1 antitarget vector is directly related to the hybrid formed between the U1 vector and target transcripts and is dependent on an intact 70,000-molecular-weight binding domain within the U1 gene. The effect is long lasting when the target (CAT or GFP) and U1 antitarget construct are inserted into fibroblasts by stable transfection. Clonal cell lines derived from stable transfection with a pOB4GFP target construct and subsequently stably transfected with the U1 anti-GFP construct were selected. The degree to which GFP fluorescence was inhibited by U1 anti-GFP in the various clonal cell lines was assessed by fluorescence-activated cell sorter analysis. RNA analysis demonstrated reduction of the GFP mRNA in the nuclear and cytoplasmic compartment and proper 3' cleavage of the GFP residual transcript. An RNase protection strategy demonstrated that the transfected U1 antitarget RNA level varied between 1 to 8% of the endogenous U1 snRNA level. U1 antitarget vectors were demonstrated to have potential as effective inhibitors of gene expression in intact cells.
- Published
- 2001
- Full Text
- View/download PDF
23. [Untitled]
- Author
-
Luca Varani, Lewis E. Kay, Gabriele Varani, Iain W. Mattaj, David Neuhaus, and Samuel I. Gunderson
- Subjects
Messenger RNA ,Polyadenylation ,Biochemistry ,Structural Biology ,Genetics ,RNA ,Cooperativity ,RNA-binding protein ,Biology ,Non-coding RNA ,Small nuclear RNA ,Post-transcriptional modification - Abstract
The status of the poly(A) tail at the 3'-end of mRNAs controls the expression of numerous genes in response to developmental and extracellular signals. Poly(A) tail regulation requires cooperative binding of two human U1A proteins to an RNA regulatory region called the polyadenylation inhibition element (PIE). When bound to PIE RNA, U1A proteins also bind to the enzyme responsible for formation of the mature 3'-end of most eukaryotic mRNAs, poly(A) polymerase (PAP). The NMR structure of the 38 kDa complex formed between two U1A molecules and PIE RNA shows that binding cooperativity depends on helix C located at the end of the RNA-binding domain and just adjacent to the PAP-interacting domain of U1A. Since helix C undergoes a conformational change upon RNA binding, the structure shows that binding cooperativity and interactions with PAP occur only when U1A is bound to its cognate RNA. This mechanism ensures that the activity of PAP enzyme, which is essential to the cell, is only down regulated when U1A is bound to the U1A mRNA.
- Published
- 2000
- Full Text
- View/download PDF
24. Fourteen Residues of the U1 snRNP-Specific U1A Protein Are Required for Homodimerization, Cooperative RNA Binding, and Inhibition of Polyadenylation
- Author
-
Samuel I. Gunderson, Reem I. Hussein, Walther J. van Venrooij, Yvonne van Aarssen, Jacqueline M. T. Klein Gunnewiek, Rob N. de Jong, and Daphne Palacios
- Subjects
Polyadenylation ,Molecular Sequence Data ,Gene Expression ,RNA-binding protein ,Saccharomyces cerevisiae ,Biology ,Ribonucleoprotein, U1 Small Nuclear ,Humans ,snRNP ,Amino Acid Sequence ,Molecular Biology ,Ribonucleoprotein ,Binding Sites ,RNA-Binding Proteins ,RNA ,Cell Biology ,Molecular biology ,Post-transcriptional modification ,Biochemistry ,Mutation ,RNA splicing ,Dimerization ,Sequence Alignment ,Small nuclear RNA ,Protein Binding - Abstract
The U1 small nuclear ribonucleoprotein (snRNP) particle is the most abundant member of the spliceosomal snRNPs. Human U1 snRNP is comprised of 10 proteins and the 164-nucleotide U1 small nuclear RNA (U1RNA) and is required for splicing of pre-mRNA (38). One of the U1 snRNP-specific proteins, the U1A protein, contains two evolutionarily conserved RNA recognition motifs (RRMs) characteristic of a large family of proteins involved in the biosynthesis of cellular RNA (reviewed in reference 37). The signature motifs for the RRM family consist of two ribonucleoprotein (RNP) sequences, RNP1 and RNP2, which are the most conserved features of this family. The N-terminal RRM of U1A is, together with some flanking amino acids, necessary and sufficient for binding to the loop part of stem-loop 2 (SL2) sequence AUUGCAC of U1RNA (22, 27, 28). The structure of the N-terminal RRM of the U1A protein (amino acids [aa] 2 to 95) has been solved both by X-ray crystallography and by nuclear magnetic resonance (NMR) and consists of a β1α1β2β3α2β4 structure in which the β strands form a sheet with the highly conserved RNP1 and RNP2 motifs located in the two central β strands, β3 and β1, respectively (14, 23). An additional α helix (helix 3; hereafter referred to as helix C) is present when a longer fragment of the U1A protein is analyzed (aa 2 to 102; reference 15). Using both NMR and X-ray crystallography, the structure of U1A aa 2 to 98 in complex with SL2 of U1RNA has also been solved (1, 2, 15, 24). In this structure, the RNA loop lies across the β sheet, fitting into a groove formed between loop 3 (connecting β2 and β3) and the C-terminal portion of the RRM domain. In spite of intensive investigation, the C-terminal RRM (aa 202 to 283) of U1A does not seem to have any affinity for RNA (21). The U1 snRNP particle is involved in the first step of spliceosome formation, in which it binds to the 5′ splice site of the pre-mRNA (reviewed in reference 18). It is possible that U1A is not essential for the splicing reaction, since in vitro splicing can still proceed in the absence of U1A. It has been suggested, however, that the U1A protein might play an important role in 5′ and 3′ splice site communication (33). In vertebrates, the U1A protein is able to regulate the polyadenylation of U1A pre-mRNA, thereby regulating its own expression level (4). The 3′ untranslated region (UTR) of the human U1A pre-mRNA contains a 50-nucleotide region, designated the polyadenylation-inhibitory element (PIE) RNA, whose sequence and structure are conserved in vertebrates. Located within the PIE RNA are two stretches of seven unpaired nucleotides designated loops 1 and 2, each being able to bind one molecule of U1A protein. Although one of the loops, when studied in isolation, has 27-fold lower affinity for U1A than the other, it was demonstrated that two molecules of U1A bind with high affinity (Kd, ∼0.1 nM) to PIE RNA, indicative of cooperative RNA binding (4, 35). The resulting (U1A)2-PIE RNA complex inhibits addition of the poly(A) tail to the U1A pre-mRNA by specifically inhibiting the enzyme poly(A) polymerase (PAP) (10). Inhibition of polyadenylation requires both the C-terminal 20 residues of PAP and aa 103 to 115 of U1A. A model has been proposed in which the U1A autoregulatory complex inhibits PAP by bringing two copies of U1A aa 103 to 115 into close proximity (11, 34). In support of this model, it was found that two molecules, but not one molecule, of U1A bound to PIE RNA inhibit PAP. Likewise, a monomeric peptide consisting of U1A aa 103 to 115 is unable to inhibit PAP; however, upon increasing its local concentration by conjugation to bovine serum albumin (BSA), this same peptide becomes a potent inhibitor of PAP (11). PAP inhibition by the BSA-peptide conjugate does not require PIE RNA, suggesting that the main role of PIE RNA in PAP inhibition is to bring the two U1A proteins into close proximity. Indeed, the unusual secondary structure of PIE RNA is not essential for inhibition (11, 34). Independent of the biochemical analysis, the determination of the structure by NMR of one molecule of U1A (aa 2 to 98) bound to PIE RNA (1) has also led to the proposal (based on modeling) that the two PIE-RNA-bound U1A proteins make extensive protein-protein interactions throughout the N-terminal 100 residues (16). Here we show, by using both the yeast two-hybrid system and in vitro assays, that U1A is able to homodimerize in the absence of RNA sequences that specifically bind U1A (i.e., SL2 of U1RNA and PIE RNA). Dimerization requires two regions, both located in the N-terminal 115 residues. Mutations of the second region (aa 103 to 115) which abolish dimerization also result in either reduction or complete loss of cooperative binding to PIE RNA but with no effect on U1A binding as a monomer to PIE RNA. These same mutations also result in loss of U1A's ability to inhibit polyadenylation. A dimeric form of a peptide containing these residues also inhibits polyadenylation. A model integrating these results will be presented explaining how the U1A autoregulatory complex functions.
- Published
- 2000
- Full Text
- View/download PDF
25. Position-dependent inhibition of the cleavage step of pre-mRNA 3′-end processing by U1 snRNP
- Author
-
Samuel I. Gunderson, Ursula Rüegsegger, Stéphan Vagner, Walter Keller, and Iain W. Mattaj
- Subjects
Cleavage factor ,Polyadenylation ,RNA Splicing ,Cleavage and polyadenylation specificity factor ,Biology ,Cleavage (embryo) ,Ribonucleoprotein, U1 Small Nuclear ,SnRNP binding ,RNA Precursors ,Animals ,Humans ,snRNP ,RNA Processing, Post-Transcriptional ,Molecular Biology ,Bovine papillomavirus 1 ,HIV Long Terminal Repeat ,Cleavage stimulation factor ,Binding Sites ,Base Sequence ,Polynucleotide Adenylyltransferase ,Molecular biology ,Cell biology ,Polynucleotide adenylyltransferase ,Mutation ,HIV-1 ,Cattle ,HeLa Cells ,Research Article - Abstract
The 3' ends of most eukaryotic pre-mRNAs are generated by 3' endonucleolytic cleavage and subsequent polyadenylation. 3'-end formation can be influenced positively or negatively by various factors. In particular, U1 snRNP acts as an inhibitor when bound to a 5' splice site located either upstream of the 3'-end formation signals of bovine papilloma virus (BPV) late transcripts or downstream of the 3'-end processing signals in the 5' LTR of the HIV-1 provirus. Previous work showed that in BPV it is not the first step, 3' cleavage, that is affected by U1 snRNP, but rather the second step, polyadenylation, that is inhibited. Since in HIV-1 the biological requirement is to produce transcripts that read through the 5' LTR cleavage site rather than being cleaved there, this mechanism seemed unlikely to apply. The obvious difference between the two examples was the relative orientation of the 3'-end formation signals and the U1 snRNP-binding site. In vitro assays were therefore used to assess the effect of U1 snRNP bound at various locations relative to a cleavage/polyadenylation site on the 3' cleavage reaction. U1 snRNP was found to inhibit cleavage when bound to a 5' splice site downstream of the cleavage/polyadenylation site, as in the HIV-1 LTR. U1 snRNP binding at this location was shown not to affect the recruitment of multiple cleavage/polyadenylation factors to the cleavage substrate, indicating that inhibition is unlikely to be due to steric hindrance. Interactions between U1A, U1 70K, and poly(A) polymerase, which mediate the effect of U1 snRNP on polyadenylation of other pre-mRNAs, were shown not to be required for cleavage inhibition. Therefore, U1 snRNP bound to a 5' splice site can inhibit cleavage and polyadenylation in two mechanistically different ways depending on whether the 5' splice site is located upstream or downstream of the cleavage site.
- Published
- 2000
- Full Text
- View/download PDF
26. 694. Targeting KRAS in Pancreatic Cancer by Gene Silencing with U1 Adaptors
- Author
-
Samuel I. Gunderson, Rafal Goraczniak, Ashley T. Tsang, Mark Brenneman, Xin Yu, and Darren R. Carpizo
- Subjects
Small nuclear ribonucleoprotein complex ,Pharmacology ,Gene knockdown ,endocrine system diseases ,Cell growth ,Biology ,medicine.disease ,medicine.disease_cause ,Molecular biology ,digestive system diseases ,In vivo ,Pancreatic cancer ,Drug Discovery ,Neuropilin ,medicine ,Cancer research ,Genetics ,Gene silencing ,Molecular Medicine ,KRAS ,Molecular Biology - Abstract
Activating mutations of the KRAS gene are key drivers of pancreatic cancer, but the KRAS protein has been refractory to small-molecule drugging. U1 Adaptors are oligonucleotides that enable the U1 small nuclear ribonucleoprotein complex to stably bind within the terminal exon of a specific pre-mRNA. This interferes with the obligatory polyadenylation step in mRNA maturation, causing selective destruction of the targeted mRNA in the nucleus. Unlike siRNA or antisense oligos, U1 Adaptors can be extensively modified for nuclease resistance or targeted delivery without loss of silencing activity, offering important advantages as therapeutic agents.We sought to translate U1 Adaptor technology to suppress KRAS in pancreatic cancer. A panel of U1 Adaptors targeting human KRAS (KRAS Adaptors) was screened in vitro using the human pancreatic cancer cell line MiaPaca-2 (KRAS G12D mutant). Candidate KRAS Adaptors reduced KRAS mRNA expression by up to 76%, as effectively as an siRNA control. Knockdown of KRAS protein expression was confirmed by western blot. Inhibition of cell growth in vitro was demonstrated for MiaPaca-2 and two additional pancreatic cancer cell lines, Panc1 (KRAS G12D mutant) and BXPC3 (KRAS wildtype).We evaluated KRAS Adaptors in mice bearing subcutaneous MiaPaca-2 xenograft tumors. For in-vivo delivery, the Adaptors were initially complexed with PAMAM dendrimers linked to a tumor-targeting, cyclic RGD peptide (cRGD), and administered by tail vein injection twice weekly for three weeks. Tumor growth was inhibited by as much as 68% compared to vehicle controls (p=0.0002). Excised tumors were analyzed by qPCR and IHC, which confirmed reductions of KRAS mRNA and protein.Although U1 Adaptors complexed with cRGD-dendrimers have been efficacious in this study and others, dendrimers have technical drawbacks and reported toxicities, and dendrimer-free formulations are preferable. As an alternative, we conjugated KRAS Adaptorsdirectly to cRGD peptide or to internalizing RGD (iRGD), which is a variant RGD peptide that triggers permeabilization of tumor endothelium and internalization by cells through secondary binding to neuropilin. The cRGD- and iRGD-conjugated KRAS Adaptors were tested for efficacy against subcutaneous MiaPaca-2 xenografts, and tumor growth was inhibited to equal or greater extent as with the original cRGD-dendrimer system. iRGD may be of particular benefit for pancreatic adenocarcinomas, which have a densely fibrotic stroma that impedes drug delivery. Enhanced delivery of small- and large-molecule therapeutics into primary pancreatic adenocarcinomas in KPC mice has been achieved previously by conjugating or coinjecting iRGD peptide.We have shown that U1 Adaptors can successfully target human KRAS both in vitro and in vivo. These results support the continued development of U1 Adaptor technology as a strategy for therapeutic suppression of KRAS in pancreatic cancer.
- Published
- 2015
- Full Text
- View/download PDF
27. U1 snRNP Inhibits Pre-mRNA Polyadenylation through a Direct Interaction between U1 70K and Poly(A) Polymerase
- Author
-
Maria Polycarpou-Schwarz, Samuel I. Gunderson, and Iain W. Mattaj
- Subjects
Saccharomyces cerevisiae Proteins ,Polyadenylation ,RNA Splicing ,Molecular Sequence Data ,Ribonuclease H ,Saccharomyces cerevisiae ,Gene Expression Regulation, Enzymologic ,Ribonucleoprotein, U1 Small Nuclear ,Bacterial Proteins ,SnRNP binding ,RNA Precursors ,Animals ,Humans ,snRNP ,Amino Acid Sequence ,Molecular Biology ,Polymerase ,Ribonucleoprotein ,Cell Nucleus ,biology ,Adenine ,Escherichia coli Proteins ,Polynucleotide Adenylyltransferase ,Cell Biology ,Molecular biology ,Enzyme Activation ,Polynucleotide adenylyltransferase ,biology.protein ,Cattle ,Precursor mRNA ,Small nuclear RNA ,HeLa Cells - Abstract
It has previously been shown in vivo that bovine papillomavirus represses its late gene expression via a 5' splice site sequence located upstream of the late polyadenylation signal. Here, the mechanism of repression is determined by in vitro analysis. U1 snRNP binding to the 5' splice site results in inhibition of polyadenylation via a direct interaction with poly(A) polymerase (PAP). Although the inhibitory mechanism is similar to that used in U1A autoregulation, U1A within the U1 snRNP does not contribute to PAP inhibition. Instead the U1 70K protein, when bound to U1 snRNA, both interacts with and inhibits PAP. Conservation of the U1 70K inhibitory domains suggests that polyadenylation regulation via PAP inhibition may be more widespread than previously thought.
- Published
- 1998
- Full Text
- View/download PDF
28. Involvement of the carboxyl terminus of vertebrate poly(A) polymerase in U1A autoregulation and in the coupling of splicing and polyadenylation
- Author
-
Maria Polycarpou-Schwarz, Iain W. Mattaj, Samuel I. Gunderson, and Stéphan Vagner
- Subjects
Polyadenylation ,RNA Splicing ,Recombinant Fusion Proteins ,Molecular Sequence Data ,Peptide ,Ribonucleoprotein, U1 Small Nuclear ,Fungal Proteins ,Species Specificity ,Genetics ,Protein biosynthesis ,Animals ,Homeostasis ,Humans ,Amino Acid Sequence ,Conserved Sequence ,Polymerase ,chemistry.chemical_classification ,Binding Sites ,Base Sequence ,biology ,Polynucleotide Adenylyltransferase ,RNA-Binding Proteins ,RNA ,Fusion protein ,Amino acid ,chemistry ,Biochemistry ,RNA splicing ,biology.protein ,Nucleic Acid Conformation ,Cattle ,Developmental Biology - Abstract
Interactions required for inhibition of poly(A) polymerase (PAP) by the U1 snRNP-specific U1A protein, a reaction whose function is to autoregulate U1A protein production, are examined. PAP inhibition requires a substrate RNA to which at least two molecules of U1A protein can bind tightly, but we demonstrate that the secondary structure of the RNA is not highly constrained. A mutational analysis reveals that the carboxy-terminal 20 amino acids of PAP are essential for its inhibition by the U1A-RNA complex. Remarkably, transfer of these amino acids to yeast PAP, which is otherwise not affected by U1A protein, is sufficient to confer U1A-mediated inhibition onto the yeast enzyme. A glutathione S-transferase fusion protein containing only these 20 PAP residues can interact in vitro with an RNA-U1A protein complex containing two U1A molecules, but not with one containing a single U1A protein, explaining the requirement for two U1A-binding sites on the autoregulatory RNA element. A mutational analysis of the U1A protein demonstrates that amino acids 103-119 are required for PAP inhibition. A monomeric synthetic peptide consisting of the conserved U1A amino acids from this region has no detectable effect on PAP activity. However, the same U1A peptide, when conjugated to BSA, inhibits vertebrate PAP. In addition to this activity, the U1A peptide-BSA conjugate specifically uncouples splicing and 3'-end formation in vitro without affecting uncoupled splicing or 3'-end cleavage efficiencies. This suggests that the carboxy-terminal region of PAP with which it interacts is involved not only in U1A autoregulation but also in the coupling of splicing and 3'-end formation.
- Published
- 1997
- Full Text
- View/download PDF
29. The Conserved Intronic Cleavage and Polyadenylation Site of CstF-77 Gene Imparts Control of 3′ End Processing Activity through Feedback Autoregulation and by U1 snRNP
- Author
-
Samuel I. Gunderson, Wencheng Li, Wenting Luo, Zhenhua Pan, Bei You, Mainul Hoque, Bin Tian, and Zhe Ji
- Subjects
Cancer Research ,lcsh:QH426-470 ,Polyadenylation ,Biology ,Ribonucleoprotein, U1 Small Nuclear ,03 medical and health sciences ,Exon ,0302 clinical medicine ,Molecular cell biology ,Genetics ,Humans ,snRNP ,RNA, Messenger ,Molecular Biology ,3' Untranslated Regions ,Genetics (clinical) ,Ecology, Evolution, Behavior and Systematics ,030304 developmental biology ,Cell Proliferation ,Regulation of gene expression ,0303 health sciences ,Cleavage stimulation factor ,Three prime untranslated region ,Genome, Human ,Systems Biology ,Intron ,Cell Differentiation ,Exons ,Molecular biology ,Introns ,Nucleic acids ,lcsh:Genetics ,RNA processing ,Cleavage Stimulation Factor ,RNA splicing ,RNA ,Gene expression ,RNA Splice Sites ,030217 neurology & neurosurgery ,Research Article ,HeLa Cells - Abstract
The human gene encoding the cleavage/polyadenylation (C/P) factor CstF-77 contains 21 exons. However, intron 3 (In3) accounts for nearly half of the gene region, and contains a C/P site (pA) with medium strength, leading to short mRNA isoforms with no apparent protein products. This intron contains a weak 5′ splice site (5′SS), opposite to the general trend for large introns in the human genome. Importantly, the intron size and strengths of 5′SS and pA are all highly conserved across vertebrates, and perturbation of these parameters drastically alters intronic C/P. We found that the usage of In3 pA is responsive to the expression level of CstF-77 as well as several other C/P factors, indicating it attenuates the expression of CstF-77 via a negative feedback mechanism. Significantly, intronic C/P of CstF-77 pre-mRNA correlates with global 3′UTR length across cells and tissues. In addition, inhibition of U1 snRNP also leads to regulation of the usage of In3 pA, suggesting that the C/P activity in the cell can be cross-regulated by splicing, leading to coordination between these two processes. Importantly, perturbation of CstF-77 expression leads to widespread alternative cleavage and polyadenylation (APA) and disturbance of cell proliferation and differentiation. Thus, the conserved intronic pA of the CstF-77 gene may function as a sensor for cellular C/P and splicing activities, controlling the homeostasis of CstF-77 and C/P activity and impacting cell proliferation and differentiation., Author Summary Autoregulation is commonly used in biological systems to control the homeostasis of certain activity, and cross-regulation coordinates multiple processes. We show that vertebrate genes encoding the cleavage/polyadenylation (C/P) factor CstF-77 contain a conserved intronic C/P site (pA) which regulates CstF-77 expression through a negative feedback loop. Since the usage of this intronic pA is also responsive to the expression of other C/P factors, the pA can function as a sensor for the cellular C/P activity. Because the CstF-77 level is important for the usage of a large number of pAs in the genome and is particularly critical for expression of genes involved in cell cycle, this autoregulatory mechanism has far-reaching implications for cell proliferation and differentiation. The human intron harboring the pA is large and has a weak 5′ splice site, both of which are also highly conserved in other vertebrates. Inhibition of U1 snRNP, which recognizes the 5′ splice site of intron, leads to upregulation of the intronic pA isoform of CstF-77 gene, suggesting that the C/P activity in the cell can be cross-regulated by splicing, leading to coordination between these two processes.
- Published
- 2013
30. A multispecies polyadenylation site model
- Author
-
Siobain Duffy, Eric S. Ho, and Samuel I. Gunderson
- Subjects
Polyadenylation ,Computational biology ,Biology ,lcsh:Computer applications to medicine. Medical informatics ,Sensitivity and Specificity ,Biochemistry ,Conserved sequence ,03 medical and health sciences ,Artificial Intelligence ,Structural Biology ,Phylogenetics ,Animals ,RNA, Messenger ,lcsh:QH301-705.5 ,Molecular Biology ,Conserved Sequence ,Phylogeny ,030304 developmental biology ,Genetics ,Principal Component Analysis ,0303 health sciences ,Phylogenetic tree ,Applied Mathematics ,030302 biochemistry & molecular biology ,Discriminant Analysis ,Models, Theoretical ,Plants ,Matthews correlation coefficient ,Linear discriminant analysis ,Nucleosomes ,Computer Science Applications ,Logistic Models ,Proceedings ,lcsh:Biology (General) ,RNA splicing ,Principal component analysis ,lcsh:R858-859.7 ,Poly A - Abstract
Background Polyadenylation is present in all three domains of life, making it the most conserved post-transcriptional process compared with splicing and 5'-capping. Even though most mammalian poly(A) sites contain a highly conserved hexanucleotide in the upstream region and a far less conserved U/GU-rich sequence in the downstream region, there are many exceptions. Furthermore, poly(A) sites in other species, such as plants and invertebrates, exhibit high deviation from this genomic structure, making the construction of a general poly(A) site recognition model challenging. We surveyed nine poly(A) site prediction methods published between 1999 and 2011. All methods exploit the skewed nucleotide profile across the poly(A) sites, and the highly conserved poly(A) signal as the primary features for recognition. These methods typically use a large number of features, which increases the dimensionality of the models to crippling degrees, and typically are not validated against many kinds of genomes. Results We propose a poly(A) site model that employs minimal features to capture the essence of poly(A) sites, and yet, produces better prediction accuracy across diverse species. Our model consists of three dior-trinucleotide profiles identified through principle component analysis, and the predicted nucleosome occupancy flanking the poly(A) sites. We validated our model using two machine learning methods: logistic regression and linear discriminant analysis. Results show that models achieve 85-92% sensitivity and 85-96% specificity in seven animals and plants. When we applied one model from one species to predict poly(A) sites from other species, the sensitivity scores correlate with phylogenetic distances. Conclusions A four-feature model geared towards small motifs was sufficient to accurately learn and predict poly(A) sites across eukaryotes.
- Published
- 2013
- Full Text
- View/download PDF
31. 259. Targeted Gene Silencing by U1 Adaptor Oligonucleotides in Preclinical Models of Parkinson's Disease and Huntington's Disease
- Author
-
Samuel I. Gunderson, Kavita Prasad, Eric K. Richfield, Mark Brenneman, and Rafal Goraczniak
- Subjects
Pharmacology ,Gene isoform ,Messenger RNA ,Small RNA ,Huntingtin ,Oligonucleotide ,Biology ,SCNA ,Molecular biology ,Drug Discovery ,Genetics ,Molecular Medicine ,Gene silencing ,Northern blot ,Molecular Biology - Abstract
Many candidate genes are implicated in neurodegenerative disease, but to study potential therapeutic effects of modifying their expression in the central nervous system of animal models has been difficult, often requiring slow, expensive transgenic methods. Transient gene silencing with synthetic oligonucleotides can be a fast, inexpensive alternative to making new transgenic animal models, and a complimentary technique to extend the utility of existing ones. For genes with products that have been validated as therapeutic targets, but are not amenable to small molecule drugs, gene silencing may also be the therapeutic modality of choice.U1 Adaptors are a third generation of oligonucleotide-mediated gene silencing technology, mechanistically distinct from antisense or siRNA. U1 Adaptors act by selectively interfering with a key step in mRNA maturation: the addition of a 3’ polyadenosine (polyA) tail. Nearly all protein-coding mRNAs require a polyA tail, and failure to add one results in rapid degradation of the nascent mRNA inside the nucleus, preventing expression of a protein product. U1 Adaptor oligonucleotides are well suited to in vivo applications because they can accept extensive chemical modifications to improve nuclease resistance and the attachment of bulky groups, such as tags for imaging or ligands for receptor-mediated uptake by target cells, without loss of silencing activity.To explore the feasibility of U1 Adaptor technology for CNS targets, we designed panels of candidate U1 Adaptor oligos for mouse Scna (alpha-synuclein) and human HTT (Huntingtin), and screened them in cell culture. We identified U1 Adaptors that robustly suppress mouse Scna mRNA and reduce alpha-synuclein protein levels in mouse cells. Similarly, we identified U1 Adaptors that suppress the predominant, full length human HTT mRNA and reduce HTT protein levels in human cells. We also identified U1 Adaptors that suppress the HTT exon-1 truncation isoform recently implicated in HD pathogenesis. For in-vivo PK/PD studies, U1 Adaptors were delivered into the CNS of mice, by intracerebroventricular (ICV) injection or by direct stereotaxic injections into the striatum. To examine distribution, cellular uptake and persistence over time, fluorescently tagged U1 Adaptors were administered, then visualized by confocal microscopy in brain sections. ICV injection achieved broad distribution of fluorescent U1 Adaptors throughout the brain, with uptake visible in most cells. Subcellular distribution 24 hours after injection was diffuse in both cytoplasmic and nuclear compartments, but became more punctate and perinuclear by 48 hours. U1 Adaptor oligonucleotides were detected on northern blots of small RNA recovered from brain tissue specimens. Their levels in tissue were estimated by comparison to a standard loading curve, and correlated well with ICV-injected dose. After direct stereotaxic injection to the striatum, U1 Adaptors diffused rapidly and widely, and were taken up by all striatial cells, though preferentially by neurons. Adaptors persisted in tissue for at least five days (the last time point assayed) and reached the nuclei of striatial cells. We then did a series of studies with U1 Adaptors specific for mouse Scna mRNA. RNAScope was used to visualize relative levels of mRNA in situ. Injection of U1 Adaptors directly into the striatum resulted in clearly reduced expression of Scna mRNA and also of mRNA for synaptophysin, known to be down-regulated when α-synuclein expression is reduced.
- Published
- 2016
- Full Text
- View/download PDF
32. 262. Therapeutic Suppression of the KRAS-MYC Oncogenic Axis in Human Pancreatic Cancer Xenografts with U1 Adaptor Oligonucleotide / RGD Peptide Conjugates
- Author
-
Kristen Donohue, Rafal Goraczniak, Mark Brenneman, Samuel I. Gunderson, Crissy Dudgeon, Lan Yi, Darren R. Carpizo, Ashley T. Tsang, and Xin Yu
- Subjects
Pharmacology ,Oncogene ,Cell growth ,Biology ,medicine.disease ,medicine.disease_cause ,Molecular biology ,Cell culture ,Apoptosis ,Pancreatic cancer ,Drug Discovery ,Genetics ,medicine ,Cancer research ,Molecular Medicine ,Gene silencing ,KRAS ,Molecular Biology ,Small nuclear ribonucleoprotein - Abstract
U1 Adaptors are synthetic oligonucleotides that enable the U1 small nuclear ribonucleoprotein (U1 snRNP) complex to stably bind within the terminal exon of a specific pre-mRNA. This interferes with the obligatory polyadenylation step in mRNA maturation, causing selective destruction of the targeted mRNA inside the nucleus. In contrast to siRNA or antisense oligos, U1 Adaptors can accept extensive covalent modifications for nuclease resistance, targeted delivery or in-vivo imaging without loss of silencing activity, offering important advantages as therapeutic agents. A panel of candidate U1 Adaptors targeting human KRAS (KRAS Adaptors) was screened in vitro using the human pancreatic cancer cell line MIA-PaCa2. The best candidates reduced KRAS mRNA expression by up to 76% - as effectively as siRNA controls. Reduced KRAS protein expression was confirmed by western blot. Inhibition of cell growth in vitro and increased apoptosis were seen for both the MIA-PaCa2 (KRASG12C) and Panc1(KRASG12D) cell lines, but not in BxPC3, a KRASwildtype pancreatic cancer cell line. In a parallel project, U1 Adaptors targeting human MYC mRNA were designed and screened in B-cell lymphoma lines, where the best candidates reduced MYC mRNA levels by over 95%. Because of the observed relationship between activating KRAS mutation and MYC overexpression in pancreatic cancers, MYC Adaptors were tested in MIA-PaCa2 cells. MYC Adaptors also inhibited cell growth and increased apoptosis in vitro. U1 Adaptors were tested for efficacy in mice bearing subcutaneous MIA-PaCa2 xenograft tumors. For in-vivo delivery, Adaptor oligos were directly conjugated to a cyclic RGD-motif peptide (cRGD), which is a targeting ligand for specific integrin-family receptors overexpressed on parenchyma and endothelial cells of many solid tumors. Alternately, U1 Adaptor oligos were linked to “internalizing” RGD (iRGD), a variant RGD peptide that also triggers permeabilization of tumor endothelium and internalization by cells through secondary binding to neuropilin-1. KRAS Adaptors linked to cRGD or iRGD were administered by tail vein injections twice weekly for three to four weeks. Over a series of experiments, tumor growth was inhibited by averages of 68% to 93%. Tumor stasis or regression occurred in some treated mice. In a pilot study, MYC Adaptors conjugated to iRGD peptide were also highly effective in suppressing tumor growth and inducing tumor regression. Excised tumors were analyzed by qPCR and western blot, which confirmed reductions of the targeted mRNAs and proteins. We have shown that U1 Adaptors conjugated to tumor-targeting / tumor-penetrating peptides can effectively target human KRAS and MYC oncogenes in vivo. These results support the continued development of U1 Adaptor technology as a strategy for therapeutic suppression of KRAS, MYC and possibly other oncogene targets in pancreatic cancer.
- Published
- 2016
- Full Text
- View/download PDF
33. The human U1A snRNP protein regulates polyadenylation via a direct interaction with poly(A) polymerase
- Author
-
Georges Martin, Katrin Beyer, Water Keller, Iain W. Mattaj, Samuel I. Gunderson, and Wilbert C. Boelens
- Subjects
Cleavage factor ,Polyadenylation ,Saccharomyces cerevisiae ,Cleavage and polyadenylation specificity factor ,General Biochemistry, Genetics and Molecular Biology ,Ribonucleoprotein, U1 Small Nuclear ,Substrate Specificity ,RNA Precursors ,Animals ,Homeostasis ,Humans ,snRNP ,Conserved Sequence ,Polymerase ,Ribonucleoprotein ,Mammals ,Cleavage stimulation factor ,Binding Sites ,Base Sequence ,biology ,Polynucleotide Adenylyltransferase ,Biochemistry ,Polynucleotide adenylyltransferase ,biology.protein ,Cattle ,Poly A ,Protein Binding - Abstract
The human U1 snRNP-specific U1A protein autoregulates its production by binding its own pre-mRNA and inhibiting polyadenylation. The mechanism of this regulation has been elucidated by in vitro studies. U1A protein is shown not to prevent either binding of cleavage and polyadenylation specificity factor (CPSF) to its recognition sequence (AUUAAA) or to prevent cleavage of U1A pre-mRNA. Instead, U1A protein bound to U1A pre-mRNA inhibits both specific and nonspecific polyadenylation by mammalian, but not by yeast, poly(A) polymerase (PAP). Domains are identified in both proteins whose removal uncouples the polyadenylation activity of mammalian PAP from its inhibition via RNA-bound U1A protein. Finally, U1A protein is shown to specifically interact with mammalian PAP in vitro. The possibility that this interaction may reflect a broader role of the U1A protein in polyadenylation is discussed.
- Published
- 1994
- Full Text
- View/download PDF
34. Ectopic 5' splice sites inhibit gene expression by engaging RNA surveillance and silencing pathways in plants
- Author
-
Samuel I. Gunderson, Rafal Goraczniak, Christophe Lacomme, Jennifer Stephens, Krzysztof Wypijewski, Jane A. Shaw, and Csaba Hornyik
- Subjects
Small interfering RNA ,RNA-induced transcriptional silencing ,Physiology ,RNA-induced silencing complex ,Trans-acting siRNA ,Green Fluorescent Proteins ,Plant Science ,Biology ,Genes, Plant ,Polyadenylation ,Tombusvirus ,Suppression, Genetic ,RNA interference ,Gene Expression Regulation, Plant ,Tobacco ,Genetics ,Gene silencing ,RNA, Messenger ,RNA, Small Interfering ,3' Untranslated Regions ,Argonaute ,RNA silencing ,Mutagenesis, Insertional ,RNA, Plant ,Gene Knockdown Techniques ,Nucleic Acid Conformation ,RNA Interference ,RNA Splice Sites ,Research Article - Abstract
The quality control of mRNA maturation is a highly regulated process that surveys pre-mRNA integrity and eliminates improperly matured pre-mRNAs. In nature, certain viruses regulate the expression of their genes by hijacking the endogenous RNA quality control machinery. We demonstrate that the inclusion of 5′ splice sites within the 3′-untranslated region of a reporter gene in plants alters the pre-mRNA cleavage and polyadenylation process, resulting in pre-mRNA degradation, exemplifying a regulatory mechanism conserved between kingdoms. Altered pre-mRNA processing was associated with an inhibition of homologous gene expression in trans and the preferential accumulation of 24-nucleotide (nt) short-interfering RNAs (siRNAs) as opposed to 21-nt siRNA subspecies, suggesting that degradation of the aberrant pre-mRNA involves the silencing machinery. However, gene expression was not restored by coexpression of a silencing suppressor or in an RNA-dependent RNA polymerase (RDR6)-deficient background despite reduced 24-nt siRNA accumulation. Our data highlight a complex cross talk between the quality control RNA machinery, 3′-end pre-mRNA maturation, and RNA-silencing pathways capable of discriminating among different types of aberrant RNAs.
- Published
- 2009
35. Mutational analysis of a Dcp2-binding element reveals general enhancement of decapping by 5'-end stem-loop structures
- Author
-
You Li, Samuel I. Gunderson, Megerditch Kiledjian, and Eric S. Ho
- Subjects
RNA Caps ,DNA Mutational Analysis ,Molecular Sequence Data ,RNA-binding protein ,Plasma protein binding ,Biology ,03 medical and health sciences ,P-bodies ,Gene expression ,Endoribonucleases ,Genetics ,Humans ,030304 developmental biology ,0303 health sciences ,Messenger RNA ,Base Sequence ,030302 biochemistry & molecular biology ,RNA ,RNA-Binding Proteins ,Stem-loop ,Molecular biology ,Cell biology ,Mutation testing ,Nucleic Acid Conformation ,5' Untranslated Regions ,Protein Binding - Abstract
mRNA decapping is a critical step in the control of mRNA stability and gene expression and is carried out by the Dcp2 protein. Dcp2 is an RNA-binding protein that must bind the RNA in order to recognize the cap for hydrolysis. We previously demonstrated that a 60 nucleotide (nt) element at the 5' end of the mRNA encoding Rrp41 is preferentially bound and decapped by Dcp2. Here, we demonstrate that enhanced decapping of this element is dependent on the structural integrity of its first 33 nt and not its primary sequence. The structure consists of a stem-loop positioned
- Published
- 2009
36. Reduction of human chorionic gonadotropin beta subunit expression by modified U1 snRNA caused apoptosis in cervical cancer cells
- Author
-
Anna Szczerba, Samuel I. Gunderson, Jerzy B. Warchol, Mirosław Andrusiewicz, Artur Jarmolowski, Beata Burczynska, Anna Jankowska, and Ewa Nowak-Markwitz
- Subjects
Cancer Research ,Uterine Cervical Neoplasms ,Apoptosis ,Transfection ,lcsh:RC254-282 ,Human chorionic gonadotropin ,HeLa ,03 medical and health sciences ,Exon ,0302 clinical medicine ,RNA, Small Nuclear ,Humans ,Gene silencing ,Chorionic Gonadotropin, beta Subunit, Human ,Gene Silencing ,RNA, Messenger ,030304 developmental biology ,Regulation of gene expression ,0303 health sciences ,Messenger RNA ,biology ,Research ,Cell Cycle ,biology.organism_classification ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immunohistochemistry ,Molecular biology ,3. Good health ,Gene Expression Regulation, Neoplastic ,Oncology ,Cell culture ,030220 oncology & carcinogenesis ,Molecular Medicine ,Female ,HeLa Cells - Abstract
Background Secretion of human chorionic gonadotropin, especially its beta subunit by malignant trophoblastic tumors and varieties of tumors of different origin is now well documented; however the role of hCG in tumorogenesis is still unknown. Results This study documents the molecular presence of human chorionic gonadotropin beta subunit in uterine cervix cancer tissues and investigates a novel technique to reduce hCGβ levels based on expression of a modified U1 snRNA as a method to study the hormone's role in biology of human cervical cancer cells cultured in vitro. The property of U1 snRNA to block the accumulation of specific RNA transcript when it binds to its donor sequence within the 3' terminal exon was used. The first 10 nucleotides of the human U1 snRNA gene, which normally binds to the 5'ss in pre-mRNA were replaced by a sequence complementary to a 10-nt segment in the terminal exon of the hCGβ mRNA. Three different 5' end-mutated U1 snRNA expression plasmids were tested, each targeting a different sequence in the hCGβ mRNA, and we found each one blocked the expression of hCGβ in HeLa cells, a cervix carcinoma cell line, as shown by immunohistochemistry and qRT-PCR. Reduction of hCGβ levels resulted in a significantly increased apoptosis rate with almost 90% of cells transfected with modified anti-hCGβ U1 snRNAs showing morphological changes characteristic of the apoptotic process. Conclusion These data suggest that human chorionic gonadotropin beta subunit may act as a tumor growth-stimulating factor.
- Published
- 2008
37. Requirements for gene silencing mediated by U1 snRNA binding to a target sequence
- Author
-
Xabi Abad, Stephen P. Jung, Maria Vera, Vaibhav Amin, Inés Romero, Evelyn Oswald, Puri Fortes, and Samuel I. Gunderson
- Subjects
Polyadenylation ,RNA, Small Nuclear/chemistry ,Base Pair Mismatch ,RNA Splicing ,Biology ,Regulatory Sequences, Ribonucleic Acid ,U1 snRNA binding ,RNA interference ,SnRNP binding ,RNA, Small Nuclear ,Genetics ,RNA Precursors ,Gene silencing ,Humans ,Point Mutation ,snRNP ,RNA, Messenger ,RNA Precursors/chemistry ,Binding Sites ,Serine-Arginine Splicing Factors ,Nuclear Proteins ,RNA-Binding Proteins ,Hydrogen Bonding ,Cell biology ,Protein Structure, Tertiary ,RNA splicing ,Nucleic Acid Conformation ,RNA ,RNA, Messenger/chemistry ,RNA Interference ,Small nuclear RNA ,HeLa Cells - Abstract
U1 interference (U1i) is a novel method to block gene expression. U1i requires expression of a 5'-end-mutated U1 snRNA designed to base pair to the 3'-terminal exon of the target gene's pre-mRNA that leads to inhibition of polyadenylation. Here, we show U1i is robust (> or =95%) and a 10-nt target length is sufficient for good silencing. Surprisingly, longer U1 snRNAs, which could increase annealing to the target, fail to improve silencing. Extensive mutagenesis of the 10-bp U1 snRNA:target duplex shows that any single mismatch different from GU at positions 3-8, destroys silencing. However, mismatches within the other positions give partial silencing, suggesting that off-target inhibition could occur. The specificity of U1i may be enhanced, however, by the fact that silencing is impaired by RNA secondary structure or by splicing factors binding nearby, the latter mediated by Arginine-Serine (RS) domains. U1i inhibition can be reconstituted in vivo by tethering of RS domains of U1-70K and U2AF65. These results help to: (i) define good target sites for U1i; (ii) identify and understand natural cellular examples of U1i; (iii) clarify the contribution of hydrogen bonding to U1i and to U1 snRNP binding to 5' splice sites and (iv) understand the mechanism of U1i.
- Published
- 2008
38. The human U1 snRNA promoter correctly initiates transcription in vitro and is activated by PSE1
- Author
-
Samuel I. Gunderson, Mark W. Knuth, and Richard R. Burgess
- Subjects
Transcription, Genetic ,Molecular Sequence Data ,Response element ,RNA polymerase II ,Prp24 ,chemistry.chemical_compound ,Transcription (biology) ,RNA, Small Nuclear ,RNA polymerase ,Genetics ,Humans ,snRNP ,Ribonuclease T1 ,Cloning, Molecular ,Promoter Regions, Genetic ,Base Sequence ,biology ,Proteins ,Promoter ,Templates, Genetic ,Molecular biology ,DNA-Binding Proteins ,chemistry ,Mutation ,SnRNA transcription ,biology.protein ,Guanosine Triphosphate ,RNA Polymerase II ,HeLa Cells ,Developmental Biology - Abstract
A DNA-dependent in vitro transcription system for the human U1 small nuclear RNA (snRNA) promoter has been developed. This in vitro transcription system uses extracts of tissue culture cells to drive transcription of an RNA polymerase II-transcribed snRNA gene. A U1 promoter (-393 to +192) template was constructed in which the sequences from +10 to +171 were replaced with a 179-bp sequence from a G-less cassette. This DNA template thus retained all of the known U1 promoter elements, including the U1 3'-end box (positions +175 to +191), which is responsible for snRNA 3'-end formation. HeLa cell nuclear extracts were shown to drive specific transcription of this promoter by RNA polymerase II. This transcription system has many of the properties observed for wild-type snRNA promoters in vivo. Transcription was shown to initiate at +1 (and -2) relative to the U1 promoter and to efficiently (greater than 90%) form a 3' end corresponding to the 3' end found in the primary transcript of U1 in vivo. The transcription signal is responsive to either deletion or replacement of the U1 distal sequence (enhancer-like) and proximal sequence (TATA-like) elements, as well as the 3'-end box. Additionally, the signal was shown by depletion/repletion experiments to be responsive to a protein called PSE1 (related to Ku), which has recently been shown to specifically bind sequences in the U1 promoter. This in vitro snRNA transcription system should facilitate the biochemical analysis of the human U1 snRNA promoter and lead to a better understanding of the differences between snRNA and mRNA promoters.
- Published
- 1990
- Full Text
- View/download PDF
39. A bipartite U1 site represses U1A expression by synergizing with PIE to inhibit nuclear polyadenylation
- Author
-
Rafal Goraczniak, Rose M. Caratozzolo, Samuel I. Gunderson, Fei Guan, and Eric S. Ho
- Subjects
DNA, Complementary ,Polyadenylation ,Molecular Sequence Data ,RNA-binding protein ,Biology ,Article ,Conserved sequence ,Ribonucleoprotein, U1 Small Nuclear ,Sequence Homology, Nucleic Acid ,Animals ,Homeostasis ,Humans ,snRNP ,RNA, Messenger ,Binding site ,Cloning, Molecular ,Molecular Biology ,Gene ,3' Untranslated Regions ,Conserved Sequence ,Regulation of gene expression ,Cell Nucleus ,Mammals ,Binding Sites ,Base Sequence ,Three prime untranslated region ,RNA-Binding Proteins ,Molecular biology ,Cell biology ,Gene Expression Regulation ,Poly A ,Sequence Alignment ,Plasmids - Abstract
U1A protein negatively autoregulates itself by polyadenylation inhibition of its own pre-mRNA by binding as two molecules to a 3′UTR-located Polyadenylation Inhibitory Element (PIE). The (U1A)2-PIE complex specifically blocks U1A mRNA biosynthesis by inhibiting polyA tail addition, leading to lower mRNA levels. U1 snRNP bound to a 5′ss-like sequence, which we call a U1 site, in the 3′UTRs of certain papillomaviruses leads to inhibition of viral late gene expression via a similar mechanism. Although such U1 sites can also be artificially used to potently silence reporter and endogenous genes, no naturally occurring U1 sites have been found in eukaryotic genes. Here we identify a conserved U1 site in the human U1A gene that is, unexpectedly, within a bipartite element where the other part represses the U1 site via a base-pairing mechanism. The bipartite element inhibits U1A expression via a synergistic action with the nearby PIE. Unexpectedly, synergy is not based on stabilizing binding of the inhibitory factors to the 3′UTR, but rather is a property of the larger ternary complex. Inhibition targets the biosynthetic step of polyA tail addition rather than altering mRNA stability. This is the first example of a functional U1 site in a cellular gene and of a single gene containing two dissimilar elements that inhibit nuclear polyadenylation. Parallels with other examples where U1 snRNP inhibits expression are discussed. We expect that other cellular genes will harbor functional U1 sites.
- Published
- 2007
40. Spliceosome Sm proteins D1, D3, and B/B' are asymmetrically dimethylated at arginine residues in the nucleus
- Author
-
Samuel I. Gunderson, Jin Hyung Lee, Permanan R. Khusial, Sidney Pestka, Steven Clarke, Gary W. Zieve, Tina B. Miranda, and Jeffry R. Cook
- Subjects
Tris ,Cell Extracts ,Arginine ,Stereochemistry ,Ion chromatography ,Molecular Sequence Data ,Biophysics ,Biochemistry ,Methylation ,Cell Line ,chemistry.chemical_compound ,Mice ,Structure-Activity Relationship ,Animals ,Humans ,Amino Acid Sequence ,Molecular Biology ,Polyacrylamide gel electrophoresis ,Peptide sequence ,chemistry.chemical_classification ,Cell Nucleus ,Methionine ,Binding Sites ,Cell Biology ,Fibroblasts ,Ribonucleoproteins, Small Nuclear ,Amino acid ,Cytosol ,chemistry ,Spliceosomes ,HeLa Cells ,Protein Binding - Abstract
We report a novel modification of spliceosome proteins Sm D1, Sm D3, and Sm B/B'. L292 mouse fibroblasts were labeled in vivo with [3H]methionine. Sm D1, Sm D3, and Sm B/B' were purified from either nuclear extracts, cytosolic extracts or a cytosolic 6S complex by immunoprecipitation of the Sm protein-containing complexes and then separation by electrophoresis on a polyacrylamide gel containing urea. The isolated Sm D1, Sm D3 or Sm B/B' proteins were hydrolyzed to amino acids and the products were analyzed by high-resolution cation exchange chromatography. Sm D1, Sm D3, and Sm B/B' isolated from nuclear fractions were all found to contain omega-NG-monomethylarginine and symmetric omega-NG,NG'-dimethylarginine, modifications that have been previously described. In addition, Sm D1, Sm D3, and Sm B/B' were also found to contain asymmetric omega-NG,NG-dimethylarginine in these nuclear fractions. Analysis of Sm B/B' from cytosolic fractions and Sm B/B' and Sm D1 from cytosolic 6S complexes showed only the presence of omega-NG-monomethylarginine and symmetric omega-NG,NG'-dimethylarginine. These results indicate that Sm D1, Sm D3, and Sm B/B' are asymmetrically dimethylated and that these modified proteins are located in the nucleus. In reactions in which Sm D1 or Sm D3 was methylated in vitro with a hemagglutinin-tagged PRMT5 purified from HeLa cells, we detected both symmetric omega-NG,NG'-dimethylarginine and asymmetric omega-NG,NG-dimethylarginine when reactions were done in a Tris/HCl buffer, but only detected symmetric omega-NG,NG'-dimethylarginine when a sodium phosphate buffer was used. These results suggest that the activity responsible for the formation of asymmetric dimethylated arginine residues in Sm proteins is either PRMT5 or a protein associated with it in the immunoprecipitated complex.
- Published
- 2004
41. Cypin regulates dendrite patterning in hippocampal neurons by promoting microtubule assembly
- Author
-
Monica M Scerri-Hansen, Barbara F. Akum, Bonnie L. Firestein, Maxine Chen, Samuel I. Gunderson, and Gary M. Riefler
- Subjects
Mutant ,Dendrite ,Biology ,Transfection ,Hippocampus ,Microtubules ,Microtubule polymerization ,RNA, Small Nuclear ,medicine ,Animals ,Cells, Cultured ,Body Patterning ,Guanine Deaminase ,Messenger RNA ,General Neuroscience ,Neurodegeneration ,Dendrites ,medicine.disease ,Embryo, Mammalian ,Immunohistochemistry ,Rats ,medicine.anatomical_structure ,Tubulin ,Mutation ,biology.protein ,Guanine deaminase activity ,Neuron ,Carrier Proteins ,Neuroscience - Abstract
Dendrite branching has an important role in normal brain function. Here we report that overexpression of cypin, a protein that has guanine deaminase activity and is expressed in developing processes in rat hippocampal neurons, results in increased dendrite branching in primary culture. Mutant cypin proteins that lack guanine deaminase activity act in a dominant-negative manner when expressed in primary neurons. Furthermore, we knocked down cypin protein levels using a new strategy: expressing a 5' end-mutated U1 small nuclear RNA (snRNA) to inhibit maturation of cypin mRNA. Neurons that express this mutant snRNA show little or no detectable cypin protein and fewer dendrites than normal. In addition, we found that cypin binds directly to tubulin heterodimers and promotes microtubule polymerization. Thus, our results demonstrate a new pathway by which dendrite patterning is regulated, and we also introduce a new method for decreasing endogenous protein expression in neurons.
- Published
- 2003
42. Inhibiting expression of specific genes in mammalian cells with 5' end-mutated U1 small nuclear RNAs targeted to terminal exons of pre-mRNA
- Author
-
Stephen P. Jung, Fei Guan, Jesús Prieto, Peng Liu, Puri Fortes, Samuel I. Gunderson, María L. Martínez-Chantar, Sara Pentlicky, Yolanda Cuevas, and David W. Rowe
- Subjects
Polyadenylation ,RNA Splicing ,Prp24 ,Simian virus 40 ,Biology ,U1 snRNA binding ,Exon ,Genes, Reporter ,RNA, Small Nuclear ,RNA Precursors ,Humans ,snRNP ,RNA Processing, Post-Transcriptional ,Luciferases ,Promoter Regions, Genetic ,3' Untranslated Regions ,Base Pairing ,Gene expression inhibition ,Reporter gene ,Multidisciplinary ,Binding Sites ,Mutant U1 snRNAs ,Three prime untranslated region ,Nucleic Acid Hybridization ,Drug Synergism ,Biological Sciences ,Fibroblasts ,Molecular biology ,Introns ,Enhancer Elements, Genetic ,Gene Expression Regulation ,Nucleic Acid Conformation ,Precursor mRNA ,5' Untranslated Regions ,Poly A ,Polyadenylation inhibition ,HeLa Cells - Abstract
Reducing or eliminating expression of a given gene is likely to require multiple methods to ensure coverage of all of the genes in a given mammalian cell. We and others [Furth, P. A., Choe, W. T., Rex, J. H., Byrne, J. C., and Baker, C. C. (1994) Mol. Cell. Biol. 14, 5278–5289] have previously shown that U1 small nuclear (sn) RNA, both natural or with 5′ end mutations, can specifically inhibit reporter gene expression in mammalian cells. This inhibition occurs when the U1 snRNA 5′ end base pairs near the polyadenylation signal of the reporter gene's pre-mRNA. This base pairing inhibits poly(A) tail addition, a key, nearly universal step in mRNA biosynthesis, resulting in degradation of the mRNA. Here we demonstrate that expression of endogenous mammalian genes can be efficiently inhibited by transiently or stably expressed 5′ end-mutated U1 snRNA. Also, we determine the inhibitory mechanism and establish a set of rules to use this technique and to improve the efficiency of inhibition. Two U1 snRNAs base paired to a single pre-mRNA act synergistically, resulting in up to 700-fold inhibition of the expression of specific reporter genes and 25-fold inhibition of endogenous genes. Surprisingly, distance from the U1 snRNA binding site to the poly(A) signal is not critical for inhibition, instead the U1 snRNA must be targeted to the terminal exon of the pre-mRNA. This could reflect a disruption by the 5′ end-mutated U1 snRNA of the definition of the terminal exon as described by the exon definition model.
- Published
- 2003
43. Abstract B52: Therapeutic targeting of human KRAS in pancreatic cancer using a novel method of gene-silencing: U1 adaptors
- Author
-
Xin Yu, Samuel I. Gunderson, Darren R. Carpizo, Rafal Goraczniak, Ashley T. Tsang, and Mark Brenneman
- Subjects
Small nuclear ribonucleoprotein complex ,Cancer Research ,Messenger RNA ,Gene knockdown ,Polyadenylation ,Biology ,medicine.disease_cause ,Exon ,Oncology ,RNA interference ,Cancer research ,medicine ,Gene silencing ,KRAS ,Molecular Biology - Abstract
Background: While genetic knockdown of RAS in mouse tumor models has substantiated it as a therapeutic target, there is no effective means of targeting RAS currently available in the clinic today. Numerous RNA interference-based studies targeting RAS have demonstrated therapeutic effects, however, effective delivery has been a major obstacle that has impeded this approach. U1 Adaptors are a novel technology for oligonucleotide-mediated gene silencing that act by selectively interfering with polyadenylation of messenger RNA (mRNA) inside the cell nucleus. Polyadenosine (PolyA) tail addition is an obligatory step in mRNA maturation and function, and its failure results in rapid degradation of the nascent message by endogenous nucleases. The eukaryotic U1 small nuclear ribonucleoprotein complex (U1 snRNP) is best known for its role as a pre-mRNA splicing factor, but also acts naturally to silence some genes by suppressing polyadenylation. U1 Adaptors are synthetic oligonucleotides that enable the U1 snRNP complex to stably bind to the terminal exon of any chosen pre-mRNA target, thereby interfering with polyA tail addition and causing it to be selectively degraded in the nucleus. The silencing mechanism of U1 Adaptors is distinct from those of siRNA or antisense oligonucleotides and this distinction confers an important advantage for their use as therapeutic agents. We have validated this technology in vivo demonstrating an 85% tumor growth inhibition by targeting BCL-2 and GRM-1 in human melanoma xenografts. Our in vivo proof-of-concept study relied on delivery of the U1 Adaptors non-covalently complexed with a nanoparticle comprised of a positively charged dendrimer covalently linked to a cyclic penta-peptide containing Arginine-Glycine-Aspartate referred to as the cRGD peptide, a widely-used tumor-targeting moiety. Methods/Results: We sought to translate the U1 Adaptor technology to target human KRAS. We first designed a set of U1 Adaptors for screening purposes targeting human KRAS at eight different positions along the human KRAS pre-mRNA located at the junction of the terminal exon (position 632) and untranslated region (UTR). We have evaluated these adaptors in vitro using the human pancreatic cancer cell line MIA Paca-2 (KRASG12D). Screening of these eight U1 Adaptors reveals a range of KRAS gene silencing as measured by quantitative PCR. Notably, Adaptors 2 and 3 silenced KRAS down to 27 and 24% respectively, as effective as the siRNA control. We then evaluated Adaptors 2 and 3 in human MIA Paca-2 xenografts. These adaptors were coupled to the cRGD-nanoparticle complex and administered by tail vein injection twice weekly. We observed significant tumor growth inhibition (37.3% by KRAS Adaptor 2, p=0.025, and 68.3% by KRAS Adaptor 3, p=0.0002, as compared to the vehicle control by Day 34. We also observed significant tumor growth inhibition with a U1 Adaptor targeting BCL-2 albeit to a lesser extent than KRAS Adaptor 3. Conclusion: We have demonstrated that the U1 Adaptor method of gene silencing can be successfully applied to target human KRAS both in vitro and in vivo. These results support the continued investigation of U1 Adaptor technology as a strategy for therapeutic targeting of RAS oncogenes. Citation Format: Ashley T. Tsang, Xin Yu, Rafal Goraczniak, Mark Brenneman, Samuel Gunderson, Darren R. Carpizo. Therapeutic targeting of human KRAS in pancreatic cancer using a novel method of gene-silencing: U1 adaptors. [abstract]. In: Proceedings of the AACR Special Conference on RAS Oncogenes: From Biology to Therapy; Feb 24-27, 2014; Lake Buena Vista, FL. Philadelphia (PA): AACR; Mol Cancer Res 2014;12(12 Suppl):Abstract nr B52. doi: 10.1158/1557-3125.RASONC14-B52
- Published
- 2014
- Full Text
- View/download PDF
44. The influence of 5' and 3' end structures on pre-mRNA metabolism
- Author
-
Joe Lewis, Iain W. Mattaj, and Samuel I. Gunderson
- Subjects
Genetics ,RNA Caps ,biology ,Polyadenylation ,Mechanism (biology) ,RNA Splicing ,RNA-Binding Proteins ,RNA polymerase II ,Cell Biology ,Metabolism ,Cell biology ,Ribonucleoprotein, U1 Small Nuclear ,Gene Expression Regulation ,Gene expression ,RNA splicing ,biology.protein ,RNA Precursors ,Directionality ,Animals ,Humans ,RNA Processing, Post-Transcriptional ,Precursor mRNA ,Poly A - Abstract
SUMMARY The 5′ cap structure of RNA polymerase II transcripts and the poly(A) tail found at the 3′ end of most mRNAs have been demonstrated to play multiple roles in gene expression and its regulation. In the first part of this review we will concentrate on the role played by the cap in pre-mRNA splicing and how it may contribute to efficient and specific substrate recognition. In the second half, we will discuss the roles that polyadenylation has been demonstated to play in RNA metabolism and will concentrate in particular on an elegant mechanism where regulation of polyadenylation is used to control gene expression.
- Published
- 1995
45. A complex secondary structure in U1A pre-mRNA that binds two molecules of U1A protein is required for regulation of polyadenylation
- Author
-
Iain W. Mattaj, M. Polycarpou-Schwarz, Wilbert C. Boelens, Eric Jansen, W. J. Van Venrooij, Samuel I. Gunderson, and C. W.G. Van Gelder
- Subjects
Polyadenylation ,Molecular Sequence Data ,RNA-binding protein ,Cleavage and polyadenylation specificity factor ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Ribonucleoprotein, U1 Small Nuclear ,RNA, Small Nuclear ,Sequence Homology, Nucleic Acid ,Humans ,snRNP ,RNA, Messenger ,Binding site ,RNA Processing, Post-Transcriptional ,Molecular Biology ,Protein secondary structure ,Binding Sites ,General Immunology and Microbiology ,Base Sequence ,Three prime untranslated region ,General Neuroscience ,Binding protein ,Nucleic Acid Precursors ,RNA-Binding Proteins ,Molecular biology ,Cell biology ,Nucleic Acid Conformation ,Poly A ,Sequence Alignment ,Research Article - Abstract
The human U1A protein-U1A pre-mRNA complex and the relationship between its structure and function in inhibition of polyadenylation in vitro were investigated. Two molecules of U1A protein were shown to bind to a conserved region in the 3' untranslated region of U1A pre-mRNA. The secondary structure of this region was determined by a combination of theoretical prediction, phylogenetic sequence alignment, enzymatic structure probing and molecular genetics. The U1A binding sites form (part of) a complex secondary structure which is significantly different from the binding site of U1A protein on U1 snRNA. Studies with mutant pre-mRNAs showed that the integrity of much of this structure is required for both high affinity binding to U1A protein and specific inhibition of polyadenylation in vitro. In particular, binding of a single molecule of U1A protein to U1A pre-mRNA is not sufficient to produce efficient inhibition of polyadenylation.
- Published
- 1993
46. The human U1 snRNP-specific U1A protein inhibits polyadenylation of its own pre-mRNA
- Author
-
Wilbert C. Boelens, Renata Stripecke, Samuel I. Gunderson, Walther J. van Venrooij, Iain W. Mattaj, and Eric Jansen
- Subjects
Untranslated region ,Polyadenylation ,Molecular Sequence Data ,Xenopus ,In Vitro Techniques ,General Biochemistry, Genetics and Molecular Biology ,Ribonucleoprotein, U1 Small Nuclear ,Mice ,Animals ,Humans ,snRNP ,RNA, Messenger ,RNA Processing, Post-Transcriptional ,Ribonucleoprotein ,Messenger RNA ,biology ,Base Sequence ,RNA-Binding Proteins ,Hydrogen Bonding ,biology.organism_classification ,Molecular biology ,Gene Expression Regulation ,Oligodeoxyribonucleotides ,Protein Biosynthesis ,Nucleic Acid Conformation ,Precursor mRNA ,Poly A ,Small nuclear RNA ,Protein Binding - Abstract
Human, mouse, and Xenopus mRNAs encoding the U1 snRNP-specific U1 A protein contain a conserved 47 nt region in their 3′ untranslated regions (UTRs). In vitro studies show that human U1A protein binds to two sites within the conserved region that resemble, in part, the previously characterized UlA-binding site on U1 snRNA. Overexpression of human U1A protein in mouse cells results in down-regulation of endogenous mouse UlA mRNA accumulation. In vitro and in vivo experiments demonstrate that excess UIA protein specifically inhibits polyadenylation of pre-mRNAs that contain the conserved 3′ UTR from human UlA mRNA. Thus, U1 A protein regulates the production of its own mRNA via a mechanism that involves pre-mRNA binding and inhibition of polyadenylation.
- Published
- 1993
47. Polymerase Selectivity and the Promoters of U snRNA Genes
- Author
-
Jordi Bernués, Samuel I. Gunderson, Iain W. Mattaj, and Kenneth A. Simmen
- Subjects
Genetics ,Messenger RNA ,Nucleoplasm ,urogenital system ,Transcription (biology) ,Nucleolus ,Intron ,Ribosomal RNA ,Biology ,Gene ,humanities ,Small nuclear RNA - Abstract
Capítulo en: Fritz Eckstein; David M. J. Lilley (eds.). Nucleic Acids and Molecular Biology. Berlin: Springer, 1992, p.174-186. (Nucleic Acids and Molecular Biology ; 6), The U small nuclear RNAs (snRNAs) form a functionally conserved family of RNAs found in eukaryotic cells. A subset of these RNAs, the spliceosomal snRNAs, function in the removal of introns from messenger RNA precursors. In most eukaryotes this group of snRNAs are U1, U2, U4, U5 and U6 but in trypanosomes, which produce mature mRNAs by transrather than cis-splicing, no homologues of U1 or U5 have been found (see Guthrie and Patterson 1988; Lamond et al. 1990 for reviews). Another ubiquitous member of the U snRNA family which will be discussed here is U3. U3 is found in the nucleolus rather than the nucleoplasm and is involved in the processing of ribosomal RNA precursors (Kass et al. 1990). The topic of this review will not, however, be the function of these RNAs, what will interest us is their transcription
- Published
- 1992
- Full Text
- View/download PDF
48. iTriplet, a rule-based nucleic acid sequence motif finder
- Author
-
Samuel I. Gunderson, Christopher D Jakubowski, and Eric S. Ho
- Subjects
0303 health sciences ,lcsh:QH426-470 ,Computer science ,Applied Mathematics ,Research ,Sequencing data ,Nucleic acid sequence ,Rule-based system ,Computational biology ,DNA sequencing ,03 medical and health sciences ,lcsh:Genetics ,0302 clinical medicine ,lcsh:Biology (General) ,Computational Theory and Mathematics ,Structural Biology ,030220 oncology & carcinogenesis ,Enumeration ,Leverage (statistics) ,Motif (music) ,Algorithm ,lcsh:QH301-705.5 ,Molecular Biology ,Combinatorial explosion ,030304 developmental biology - Abstract
Background With the advent of high throughput sequencing techniques, large amounts of sequencing data are readily available for analysis. Natural biological signals are intrinsically highly variable making their complete identification a computationally challenging problem. Many attempts in using statistical or combinatorial approaches have been made with great success in the past. However, identifying highly degenerate and long (>20 nucleotides) motifs still remains an unmet challenge as high degeneracy will diminish statistical significance of biological signals and increasing motif size will cause combinatorial explosion. In this report, we present a novel rule-based method that is focused on finding degenerate and long motifs. Our proposed method, named iTriplet, avoids costly enumeration present in existing combinatorial methods and is amenable to parallel processing. Results We have conducted a comprehensive assessment on the performance and sensitivity-specificity of iTriplet in analyzing artificial and real biological sequences in various genomic regions. The results show that iTriplet is able to solve challenging cases. Furthermore we have confirmed the utility of iTriplet by showing it accurately predicts polyA-site-related motifs using a dual Luciferase reporter assay. Conclusion iTriplet is a novel rule-based combinatorial or enumerative motif finding method that is able to process highly degenerate and long motifs that have resisted analysis by other methods. In addition, iTriplet is distinguished from other methods of the same family by its parallelizability, which allows it to leverage the power of today's readily available high-performance computing systems.
- Published
- 2009
49. Bacteriophage T7 late promoters with point mutations: quantitative footprinting andin vivoexpression
- Author
-
Samuel I. Gunderson, Michael Anello, Kenneth A. Chapman, Robert D. Wells, and Richard R. Burgess
- Subjects
Genetics ,Base Sequence ,Genes, Viral ,Transcription, Genetic ,biology ,Point mutation ,Molecular Sequence Data ,DNA footprinting ,Promoter ,DNA-Directed DNA Polymerase ,Templates, Genetic ,biology.organism_classification ,Molecular biology ,Footprinting ,Bacteriophage ,In vivo ,Transcription (biology) ,Mutation ,Gene expression ,Escherichia coli ,T-Phages ,Promoter Regions, Genetic - Published
- 1988
- Full Text
- View/download PDF
50. Binding of transcription factors to the promoter of the human U1 RNA gene studied by footprinting
- Author
-
J T Murphy, J H Dahlberg, Mark W. Knuth, Richard R. Burgess, T H Steinberg, and Samuel I. Gunderson
- Subjects
General transcription factor ,biology ,Response element ,Promoter ,RNA polymerase II ,Cell Biology ,Biochemistry ,Trans-regulatory element ,Molecular biology ,Transcription (biology) ,biology.protein ,Enhancer ,Molecular Biology ,Transcription factor - Abstract
The promoter structure of the known small nuclear RNA (snRNA) genes contains two major effectors of transcriptional activity: a proximal sequence element and a distal sequence element. In addition to these two functional elements (called elements B and D), the human U1 snRNA gene contains at least three minor elements (elements A, C, and E) that contribute to overall transcriptional efficiency (Murphy, J.T., Skuzeski, J.M., Lund, E., Steinberg, T.H., Burgess, R.R., and Dahlberg, J.E. (1987) J. Biol. Chem. 262, 1795-1803). To elucidate further the function of these transcription elements, we carried out a computer search to look for sequences in the U1 gene homologous to known transcription factor consensus sequences. Where such homology was found, DNase I and MPE-Fe(II) (methidiumpropyl-EDTA-Fe(II] footprinting was employed to study the interactions of these promoter regions with proteins partially purified from extracts of HeLa cells or human placenta. Footprints were observed over element D (the distal element) corresponding to sequences homologous to the octanucleotide binding protein (OCTA) and activator protein 1 (AP1). Protection was also observed over element B (the proximal element) corresponding to possible sites for stimulatory protein 1 (Sp1), enhancer core, major late transcription factor (MLTF), and a U1-specific transcription factor. Prior to this study, no specific transcription factor footprints had been observed over proximal elements of any snRNA gene. Footprints were also found over elements A and E. The results of the computer search and the footprinting are discussed in light of what is known about snRNA promoter activity.
- Published
- 1988
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.