Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Oncogene ; 41(20): 2811-2823, 2022 05.
Article in English | MEDLINE | ID: mdl-35418690

ABSTRACT

The cytoplasmic phosphatase DUSP6 and its nuclear counterpart DUSP5 are negative regulators of RAS/ERK signalling. Here we use deletion of either Dusp5 or Dusp6 to explore the roles of these phosphatases in a murine model of KRASG12D-driven pancreatic cancer. By 56-days, loss of either DUSP5 or DUSP6 causes a significant increase in KRASG12D-driven pancreatic hyperplasia. This is accompanied by increased pancreatic acinar to ductal metaplasia (ADM) and the development of pre-neoplastic pancreatic intraepithelial neoplasia (PanINs). In contrast, by 100-days, pancreatic hyperplasia is reversed with significant atrophy of pancreatic tissue and weight loss observed in animals lacking either DUSP5 or DUSP6. On further ageing, Dusp6-/- mice display accelerated development of metastatic pancreatic ductal adenocarcinoma (PDAC), while in Dusp5-/- animals, although PDAC development is increased this process is attenuated by atrophy of pancreatic acinar tissue and severe weight loss in some animals before cancer could progress. Our data suggest that despite a common target in the ERK MAP kinase, DUSP5 and DUSP6 play partially non-redundant roles in suppressing oncogenic KRASG12D signalling, thus retarding both tumour initiation and progression. Our data suggest that loss of either DUSP5 or DUSP6, as observed in certain human tumours, including the pancreas, could promote carcinogenesis.


Subject(s)
Carcinoma, Pancreatic Ductal , Dual Specificity Phosphatase 6 , Dual-Specificity Phosphatases , Pancreatic Neoplasms , Animals , Atrophy/pathology , Carcinogenesis/genetics , Carcinogenesis/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Dual Specificity Phosphatase 6/genetics , Dual-Specificity Phosphatases/genetics , Hyperplasia , Mice , Pancreas/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Weight Loss , Pancreatic Neoplasms
2.
PLoS One ; 11(4): e0154759, 2016.
Article in English | MEDLINE | ID: mdl-27128805

ABSTRACT

The snoMEN (snoRNA Modulator of gene ExpressioN) vector technology was developed from a human box C/D snoRNA, HBII-180C, which contains an internal sequence that can be manipulated to make it complementary to RNA targets, allowing knock-down of targeted genes. Here we have screened additional human nucleolar snoRNAs and assessed their application for gene specific knock-downs to improve the efficiency of snoMEN vectors. We identify and characterise a new snoMEN vector, termed 47snoMEN, that is derived from box C/D snoRNA U47, demonstrating its use for knock-down of both endogenous cellular proteins and G/YFP-fusion proteins. Using multiplex 47snoMEM vectors that co-express multiple 47snoMEN in a single transcript, each of which can target different sites in the same mRNA, we document >3-fold increase in knock-down efficiency when compared with the original HBII-180C based snoMEN. The multiplex 47snoMEM vector allowed the construction of human protein replacement cell lines with improved efficiency, including the establishment of novel GFP-HIF-1α replacement cells. Quantitative mass spectrometry analysis confirmed the enhanced efficiency and specificity of protein replacement using the 47snoMEN-PR vectors. The 47snoMEN vectors expand the potential applications for snoMEN technology in gene expression studies, target validation and gene therapy.


Subject(s)
Genetic Vectors , Green Fluorescent Proteins/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , RNA, Small Nucleolar/genetics , Base Sequence , Cell Line , Gene Expression , Gene Knockdown Techniques/methods , HeLa Cells , Humans , Nucleic Acid Conformation , Protein Binding , RNA, Small Nucleolar/chemistry , RNA, Small Nucleolar/metabolism , Recombinant Fusion Proteins/genetics , Ribonucleoproteins, Small Nucleolar/metabolism
3.
PLoS One ; 10(9): e0138668, 2015.
Article in English | MEDLINE | ID: mdl-26405811

ABSTRACT

We have previously reported an antisense technology, 'snoMEN vectors', for targeted knock-down of protein coding mRNAs using human snoRNAs manipulated to contain short regions of sequence complementarity with the mRNA target. Here we characterise the use of snoMEN vectors to target the knock-down of micro RNA primary transcripts. We document the specific knock-down of miR21 in HeLa cells using plasmid vectors expressing miR21-targeted snoMEN RNAs and show this induces apoptosis. Knock-down is dependent on the presence of complementary sequences in the snoMEN vector and the induction of apoptosis can be suppressed by over-expression of miR21. Furthermore, we have also developed lentiviral vectors for delivery of snoMEN RNAs and show this increases the efficiency of vector transduction in many human cell lines that are difficult to transfect with plasmid vectors. Transduction of lentiviral vectors expressing snoMEN targeted to pri-miR21 induces apoptosis in human lung adenocarcinoma cells, which express high levels of miR21, but not in human primary cells. We show that snoMEN-mediated suppression of miRNA expression is prevented by siRNA knock-down of Ago2, but not by knock-down of Ago1 or Upf1. snoMEN RNAs colocalise with Ago2 in cell nuclei and nucleoli and can be co-immunoprecipitated from nuclear extracts by antibodies specific for Ago2.


Subject(s)
Adenocarcinoma/genetics , Gene Knockdown Techniques/methods , Lung Neoplasms/genetics , MicroRNAs/genetics , RNA Precursors/genetics , RNA, Antisense/genetics , Adenocarcinoma of Lung , Apoptosis , Argonaute Proteins/genetics , Eukaryotic Initiation Factors/genetics , Genetic Vectors/pharmacology , HEK293 Cells , HeLa Cells , Humans , Lentivirus/genetics , Plasmids/genetics , RNA Helicases , Trans-Activators/genetics
4.
Cell Cycle ; 12(15): 2348-54, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23839046

ABSTRACT

We have identified the human FMN2 gene as a novel target regulated by induction of p14ARF and by multiple other stress responses, including DNA damage and hypoxia, which have in common activation of cell cycle arrest. We showed that increased expression of the FMN2 gene following p14ARF induction is caused, at the transcriptional level, by relief of repression by RelA and E2F1, which, under non-induced conditions, bind the FMN2 promoter. Increased FMN2 protein levels promote cell cycle arrest by inhibiting the degradation of p21, and our data show that control of p21 stability is a key part of the mechanism that regulates p21 induction. Consistent with this model, we have shown that transient expression of exogenous FMN2 protein alone is sufficient to increase p21 protein levels in cells, without altering p21 mRNA levels. Here, we provide additional evidence for the role of the N terminus of FMN2 as being the important domain required for p21 stability. In addition, we also investigate the role of RelA's threonine 505 residue in the control of FMN2. Our results identify FMN2 as a crucial protein involved in the control of p21.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Microfilament Proteins/physiology , Nuclear Proteins/physiology , Animals , Cell Hypoxia , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA Damage , Formins , Gene Expression Regulation , Humans , Phosphorylation , Protein Processing, Post-Translational , Proteolysis , Signal Transduction , Transcription Factor RelA/metabolism
5.
PLoS One ; 8(4): e62305, 2013.
Article in English | MEDLINE | ID: mdl-23638031

ABSTRACT

The study of the function of many human proteins is often hampered by technical limitations, such as cytotoxicity and phenotypes that result from overexpression of the protein of interest together with the endogenous version. Here we present the snoMEN (snoRNA Modulator of gene ExpressioN) vector technology for generating stable cell lines where expression of the endogenous protein can be reduced and replaced by an exogenous protein, such as a fluorescent protein (FP)-tagged version. SnoMEN are snoRNAs engineered to contain complementary sequences that can promote knock-down of targeted RNAs. We have established and characterised two such partial protein replacement human cell lines (snoMEN-PR). Quantitative mass spectrometry was used to analyse the specificity of knock-down and replacement at the protein level and also showed an increased pull-down efficiency of protein complexes containing exogenous, tagged proteins in the protein replacement cell lines, as compared with conventional co-expression strategies. The snoMEN approach facilitates the study of mammalian proteins, particularly those that have so far been difficult to investigate by exogenous expression and has wide applications in basic and applied gene-expression research.


Subject(s)
Genetic Engineering/methods , Genetic Vectors/genetics , Proteins/metabolism , RNA, Small Nucleolar/genetics , Cell Line , Gene Expression , Humans , Proteins/genetics , RNA Interference , RNA, Small Interfering/genetics
6.
Mol Cell ; 49(5): 922-33, 2013 Mar 07.
Article in English | MEDLINE | ID: mdl-23375502

ABSTRACT

The ARF tumor suppressor is a central component of the cellular defense against oncogene activation in mammals. p14ARF activates p53 by binding and inhibiting HDM2, resulting, inter alia, in increased transcription and expression of the cyclin-dependent kinase inhibitor p21 and consequent cell-cycle arrest. We analyzed the effect of p14ARF induction on nucleolar protein dynamics using SILAC mass spectrometry and have identified the human Formin-2 (FMN2) protein as a component of the p14ARF tumor suppressor pathway. We show that FMN2 is increased upon p14ARF induction at both the mRNA and the protein level via a NF-κB-dependent mechanism that is independent of p53. FMN2 enhances expression of the cell-cycle inhibitor p21 by preventing its degradation. FMN2 is also induced by activation of other oncogenes, hypoxia, and DNA damage. These results identify FMN2 as a crucial component in the regulation of p21 and consequent oncogene/stress-induced cell-cycle arrest in human cells.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/genetics , Nerve Tissue Proteins/genetics , Cell Cycle Checkpoints , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Nucleolus , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Humans , Mass Spectrometry , NF-kappa B/genetics , Nerve Tissue Proteins/metabolism , Proteomics , RNA, Messenger/metabolism , Tumor Suppressor Protein p14ARF/genetics , Tumor Suppressor Protein p14ARF/metabolism
7.
Nucleic Acids Res ; 40(8): 3676-88, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22199253

ABSTRACT

Small nucleolar RNAs (snoRNAs) function mainly as guides for the post-transcriptional modification of ribosomal RNAs (rRNAs). In recent years, several studies have identified a wealth of small fragments (<35 nt) derived from snoRNAs (termed sdRNAs) that stably accumulate in the cell, some of which may regulate splicing or translation. A comparison of human small RNA deep sequencing data sets reveals that box C/D sdRNA accumulation patterns are conserved across multiple cell types although the ratio of the abundance of different sdRNAs from a given snoRNA varies. sdRNA profiles of many snoRNAs are specific and resemble the cleavage profiles of miRNAs. Many do not show characteristics of general RNA degradation, as seen for the accumulation of small fragments derived from snRNA or rRNA. While 53% of the sdRNAs contain an snoRNA box C motif and boxes D and D' are also common in sdRNAs (54%), relatively few (12%) contain a full snoRNA guide region. One box C/D snoRNA, HBII-180C, was analysed in greater detail, revealing the presence of C' box-containing sdRNAs complementary to several pre-messenger RNAs (pre-mRNAs) including FGFR3. Functional analyses demonstrated that this region of HBII-180C can influence the alternative splicing of FGFR3 pre-mRNA, supporting a role for some snoRNAs in the regulation of splicing.


Subject(s)
RNA Processing, Post-Transcriptional , RNA, Small Nucleolar/metabolism , Alternative Splicing , Base Sequence , Cell Line , Conserved Sequence , Humans , Molecular Sequence Data , RNA, Small Nucleolar/chemistry , Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptor, Fibroblast Growth Factor, Type 3/metabolism
8.
Biochimie ; 93(11): 1987-92, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21664409

ABSTRACT

Small nucleolar RNAs (snoRNAs) are an ancient class of small non-coding RNAs present in all eukaryotes and a subset of archaea that carry out a fundamental role in the modification and processing of ribosomal RNA. In recent years, however, a large proportion of snoRNAs have been found to be further processed into smaller molecules, some of which display different functionality. In parallel, several studies have uncovered extensive similarities between snoRNAs and other types of small non-coding RNAs, and in particular microRNAs. Here, we explore the extent of the relationship between these types of non-coding RNA and the possible underlying evolutionary forces that shaped this subset of the current non-coding RNA landscape.


Subject(s)
Archaea/genetics , MicroRNAs/genetics , RNA, Small Nucleolar/genetics , Animals , Evolution, Molecular , Gene Expression Regulation , Genomics , Humans , MicroRNAs/metabolism , Protein Biosynthesis , RNA Isoforms/genetics , RNA, Ribosomal/genetics , RNA, Small Nucleolar/metabolism
9.
Nucleic Acids Res ; 39(9): 3879-91, 2011 May.
Article in English | MEDLINE | ID: mdl-21247878

ABSTRACT

There are two main classes of small nucleolar RNAs (snoRNAs): the box C/D snoRNAs and the box H/ACA snoRNAs that function as guide RNAs to direct sequence-specific modification of rRNA precursors and other nucleolar RNA targets. A previous computational and biochemical analysis revealed a possible evolutionary relationship between miRNA precursors and some box H/ACA snoRNAs. Here, we investigate a similar evolutionary relationship between a subset of miRNA precursors and box C/D snoRNAs. Computational analyses identified 84 intronic miRNAs that are encoded within either box C/D snoRNAs, or in precursors showing similarity to box C/D snoRNAs. Predictions of the folded structures of these box C/D snoRNA-like miRNA precursors resemble the structures of known box C/D snoRNAs, with the boxes C and D often in close proximity in the folded molecule. All five box C/D snoRNA-like miRNA precursors tested (miR-27b, miR-16-1, mir-28, miR-31 and let-7g) bind to fibrillarin, a specific protein component of functional box C/D snoRNP complexes. The data suggest that a subset of small regulatory RNAs may have evolved from box C/D snoRNAs.


Subject(s)
MicroRNAs/chemistry , RNA Precursors/chemistry , RNA, Small Nucleolar/chemistry , Base Sequence , Cell Nucleolus/genetics , Chromosomal Proteins, Non-Histone/metabolism , Humans , MicroRNAs/analysis , MicroRNAs/metabolism , Molecular Sequence Data , RNA Precursors/analysis , RNA Precursors/metabolism , RNA, Small Nucleolar/metabolism
10.
Mol Biol Cell ; 21(9): 1569-84, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20219969

ABSTRACT

Human small nucleolar RNAs (snoRNAs) that copurify with nucleoli isolated from HeLa cells have been characterized. Novel fibrillarin-associated snoRNAs were detected that allowed the creation of a new vector system for the targeted knockdown of one or more genes in mammalian cells. The snoMEN (snoRNA modulator of gene expressioN) vector technology is based on snoRNA HBII-180C, which contains an internal sequence that can be manipulated to make it complementary to RNA targets. Gene-specific knockdowns are demonstrated for endogenous cellular proteins and for G/YFP-fusion proteins. Multiplex snoMEN vectors coexpress multiple snoRNAs in one transcript, targeted either to different genes or to different sites in the same gene. Protein replacement snoMEN vectors can express a single transcript combining cDNA for a tagged protein with introns containing cognate snoRNAs targeted to knockdown the endogenous cellular protein. We foresee applications for snoMEN vectors in basic gene expression research, target validation, and gene therapy.


Subject(s)
Gene Expression , Gene Knockdown Techniques/methods , Genetic Vectors/genetics , RNA, Small Nucleolar/genetics , Cell Nucleolus/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Cloning, Molecular , Gene Library , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , In Situ Hybridization, Fluorescence , Microscopy, Fluorescence , Mutation , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 1 Protein/metabolism , Transfection
11.
PLoS Comput Biol ; 5(9): e1000507, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19763159

ABSTRACT

MicroRNAs (miRNAs) and small nucleolar RNAs (snoRNAs) are two classes of small non-coding regulatory RNAs, which have been much investigated in recent years. While their respective functions in the cell are distinct, they share interesting genomic similarities, and recent sequencing projects have identified processed forms of snoRNAs that resemble miRNAs. Here, we investigate a possible evolutionary relationship between miRNAs and box H/ACA snoRNAs. A comparison of the genomic locations of reported miRNAs and snoRNAs reveals an overlap of specific members of these classes. To test the hypothesis that some miRNAs might have evolved from snoRNA encoding genomic regions, reported miRNA-encoding regions were scanned for the presence of box H/ACA snoRNA features. Twenty miRNA precursors show significant similarity to H/ACA snoRNAs as predicted by snoGPS. These include molecules predicted to target known ribosomal RNA pseudouridylation sites in vivo for which no guide snoRNA has yet been reported. The predicted folded structures of these twenty H/ACA snoRNA-like miRNA precursors reveal molecules which resemble the structures of known box H/ACA snoRNAs. The genomic regions surrounding these predicted snoRNA-like miRNAs are often similar to regions around snoRNA retroposons, including the presence of transposable elements, target site duplications and poly (A) tails. We further show that the precursors of five H/ACA snoRNA-like miRNAs (miR-151, miR-605, mir-664, miR-215 and miR-140) bind to dyskerin, a specific protein component of functional box H/ACA small nucleolar ribonucleoprotein complexes suggesting that these molecules have retained some H/ACA snoRNA functionality. The detection of small RNA molecules that share features of miRNAs and snoRNAs suggest that these classes of RNA may have an evolutionary relationship.


Subject(s)
Evolution, Molecular , MicroRNAs/chemistry , RNA Precursors/chemistry , RNA, Small Nucleolar/chemistry , Base Sequence , Cell Cycle Proteins/metabolism , Conserved Sequence/genetics , HeLa Cells , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Molecular Sequence Data , Nuclear Proteins/metabolism , Nucleic Acid Conformation , RNA Precursors/genetics , RNA Precursors/metabolism , RNA, Small Nucleolar/genetics , RNA, Small Nucleolar/metabolism , Repetitive Sequences, Nucleic Acid , Subcellular Fractions/metabolism
12.
Zoolog Sci ; 21(4): 393-6, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15118226

ABSTRACT

The hibernation-specific HP-27 gene is expressed specifically in the liver of the chipmunk, a hibernating species of the squirrel family, and exists as a pseudogene in the tree squirrel, a nonhibernating species. In the promoter region, the chipmunk gene has a potential HNF-1 binding site, and the tree squirrel gene has two base substitutions in the corresponding sequence. In this paper, we investigated the role of HNF-1 in the HP-27 gene promoter activity. Gel retardation assays with in vitro-translated HNF-1 and super-shift assays using HepG2 nuclear extracts and an anti-HNF-1 antibody revealed that HNF-1 bound to the chipmunk gene sequence. HNF-1 also bound to the tree squirrel sequence, but with much lower affinity. In HepG2 cells, HNF-1 activated transcription from the chipmunk HP-27 gene, but not from the tree squirrel gene. In addition, the tree squirrel-type base substitutions in the HNF-1 binding site greatly reduced the promoter activity of the chipmunk HP-27 gene. These results indicate that HNF-1 is required for the promoter activity of the chipmunk HP-27 gene, and that the base substitutions in the HNF-1 binding site are involved in the lack of HP-27 gene expression in the tree squirrel.


Subject(s)
Blood Proteins/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Nuclear Proteins/metabolism , Promoter Regions, Genetic/genetics , Sciuridae/genetics , Sciuridae/metabolism , Transcription Factors/metabolism , Animals , Autoradiography , Base Sequence , Binding Sites , Cells, Cultured , DNA Primers , Electrophoretic Mobility Shift Assay , Hepatocyte Nuclear Factor 1 , Liver/metabolism , Luciferases/metabolism , Plasmids/genetics , Sequence Alignment , Transfection
13.
Gene ; 302(1-2): 193-9, 2003 Jan 02.
Article in English | MEDLINE | ID: mdl-12527211

ABSTRACT

The chipmunk hibernation-specific protein HP-27 is a component of the 140-kDa complex that decreases in the blood during hibernation. Although the HP-27 gene is detected in both the chipmunk, a hibernating species of the squirrel family, and the tree squirrel, a nonhibernating species, it is expressed only in the chipmunk, in a liver-specific manner. To understand the difference in HP-27 gene expression between the chipmunk and tree squirrel, we isolated chipmunk and tree squirrel HP-27 genomic clones, and compared their promoter activities. Transient transfection studies in HepG2 cells revealed that the 170 bp 5'-flanking sequence of the chipmunk HP-27 gene was sufficient for liver-specific promoter activity and that deletion of the sequence from -170 to -140 reduced the promoter activity by 90%. Although the corresponding 170 bp 5'-flanking sequence of the tree squirrel HP-27 gene had 89% nucleotide sequence homology to that of the chipmunk, it showed almost no promoter activity in HepG2 cells. In a gel retardation assay using HepG2 or chipmunk liver nuclear extracts, the 5'-flanking sequence of the chipmunk HP-27 gene from -170 to -140 showed a shifted band, but the corresponding tree squirrel sequence did not. Taken together, these data indicate that a transcription factor that binds to this 5'-flanking sequence of the chipmunk HP-27 gene plays an important role in HP-27 gene expression, and the failure of this factor to bind in the case of the tree squirrel HP-27 gene could be responsible for this animal's lack of HP-27 gene expression.


Subject(s)
Blood Proteins/genetics , Promoter Regions, Genetic/genetics , Sciuridae/genetics , 5' Flanking Region/genetics , Amino Acid Sequence , Animals , Base Sequence , DNA/chemistry , DNA/genetics , DNA/metabolism , DNA, Complementary/chemistry , DNA, Complementary/genetics , DNA-Binding Proteins/metabolism , Exons , Genes/genetics , Humans , Introns , Luciferases/genetics , Luciferases/metabolism , Molecular Sequence Data , Protein Binding , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...