Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters










Publication year range
1.
Mech Ageing Dev ; 199: 111552, 2021 10.
Article in English | MEDLINE | ID: mdl-34363832

ABSTRACT

Exercise training seems to promote healthy biological ageing partly by inducing telomere maintenance, yet the molecular mechanisms are not fully understood. Recent studies have emphasised the importance of microRNAs (miRNAs) in ageing and their ability to mirror pathophysiological alterations associated with age-related diseases. We examined the association between aerobic fitness and leukocyte telomere length before determining the influence of vigorous exercise training on the regulation of leukocyte miRNA networks. Telomere length was positively correlated to aerobic fitness (r = 0.32, p = 0.02). 104 miRNAs were differentially expressed after six weeks of thrice-weekly sprint interval training (SIT) in healthy men (q < 0.05). Gene co-expression analysis (WGCNA) detected biologically meaningful miRNA networks, five of which were significantly correlated with pre-SIT and post-SIT expression profiles (p < 0.001) and telomere length. Enrichment analysis revealed that the immune response, T cell differentiation and lipid metabolism associated miRNAs clusters were significantly down-regulated after SIT. Using data acquired from the Gene Expression Omnibus (GEO), we also identified two co-expressed miRNAs families that were modulated by exercise training in previous investigations. Collectively, our findings highlight the miRNA networks implicated in exercise adaptations and telomere regulation, and suggest that SIT may attenuate biological ageing through the control of the let-7 and miR-320 miRNA families.


Subject(s)
Aging/physiology , Exercise/genetics , High-Intensity Interval Training/methods , Leukocytes/physiology , MicroRNAs/physiology , Physical Fitness/physiology , Telomere Homeostasis/physiology , Adult , Female , Gene Expression Profiling/methods , Healthy Aging/genetics , Humans , Male , MicroRNAs/genetics , Regulatory Sequences, Ribonucleic Acid/physiology , Signal Transduction
2.
PLoS Biol ; 19(1): e3001053, 2021 01.
Article in English | MEDLINE | ID: mdl-33439856

ABSTRACT

Myelin is a specialized membrane produced by oligodendrocytes that insulates and supports axons. Oligodendrocytes extend numerous cellular processes, as projections of the plasma membrane, and simultaneously wrap multiple layers of myelin membrane around target axons. Notably, myelin sheaths originating from the same oligodendrocyte are variable in size, suggesting local mechanisms regulate myelin sheath growth. Purified myelin contains ribosomes and hundreds of mRNAs, supporting a model that mRNA localization and local protein synthesis regulate sheath growth and maturation. However, the mechanisms by which mRNAs are selectively enriched in myelin sheaths are unclear. To investigate how mRNAs are targeted to myelin sheaths, we tested the hypothesis that transcripts are selected for myelin enrichment through consensus sequences in the 3' untranslated region (3' UTR). Using methods to visualize mRNA in living zebrafish larvae, we identified candidate 3' UTRs that were sufficient to localize mRNA to sheaths and enriched near growth zones of nascent membrane. We bioinformatically identified motifs common in 3' UTRs from 3 myelin-enriched transcripts and determined that these motifs are required and sufficient in a context-dependent manner for mRNA transport to myelin sheaths. Finally, we show that 1 motif is highly enriched in the myelin transcriptome, suggesting that this sequence is a global regulator of mRNA localization during developmental myelination.


Subject(s)
3' Untranslated Regions/physiology , Myelin Sheath/metabolism , RNA Transport/genetics , RNA, Messenger/metabolism , Animals , Animals, Genetically Modified , Embryo, Nonmammalian , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/metabolism , Molecular Imaging/methods , Myelin Sheath/genetics , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Regulatory Sequences, Ribonucleic Acid/physiology , Sequence Analysis, RNA , Tissue Distribution , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
3.
PLoS Pathog ; 16(5): e1008488, 2020 05.
Article in English | MEDLINE | ID: mdl-32433693

ABSTRACT

There are approximately 20 million events of hepatitis E virus (HEV) infection worldwide annually. The genome of HEV is a single-strand, positive-sense RNA containing 5' and 3' untranslated regions and three open reading frames (ORF). HEV genome has 5' cap and 3' poly(A) tail to mimic host mRNA to escape the host innate immune surveillance and utilize host translational machineries for viral protein translation. The replication mechanism of HEV is poorly understood, especially how the viral polymerase distinguishes viral RNA from host mRNA to synthesize new viral genomes. We hypothesize that the HEV genome contains cis-acting elements that can be recognized by the virally encoded polymerase as "self" for replication. To identify functional cis-acting elements systematically across the HEV genome, we utilized an ORF1 transcomplementation system. Ultimately, we found two highly conserved cis-acting RNA elements within the ORF1 and ORF2 coding regions that are required for viral genome replication in a diverse panel of HEV genotypes. Synonymous mutations in the cis-acting RNA elements, not altering the ORF1 and ORF2 protein sequences, significantly impaired production of infectious viral particles. Mechanistic studies revealed that the cis-acting elements form secondary structures needed to interact with the HEV ORF1 protein to promote HEV replication. Thus, these cis-acting elements function as a scaffold, providing a specific "signal" that recruits viral and host factors to assemble the viral replication complex. Altogether, this work not only facilitates our understanding of the HEV life cycle and provides novel, RNA-directed targets for potential HEV treatments, but also sheds light on the development of HEV as a therapeutic delivery vector.


Subject(s)
Genome, Viral , Hepatitis E virus/physiology , RNA, Viral , Regulatory Sequences, Ribonucleic Acid/physiology , Virus Replication/physiology , Animals , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Mutation , Open Reading Frames/physiology , RNA, Viral/biosynthesis , RNA, Viral/genetics
4.
BMC Cell Biol ; 18(1): 23, 2017 05 25.
Article in English | MEDLINE | ID: mdl-28545392

ABSTRACT

BACKGROUND: In Dictyostelium discoideum, vesicular transport of the adenylyl cyclase A (ACA) to the posterior of polarized cells is essential to relay exogenous 3',5'-cyclic adenosine monophosphate (cAMP) signals during chemotaxis and for the collective migration of cells in head-to-tail arrangements called streams. RESULTS: Using fluorescence in situ hybridization (FISH), we discovered that the ACA mRNA is asymmetrically distributed at the posterior of polarized cells. Using both standard estimators and Monte Carlo simulation methods, we found that the ACA mRNA enrichment depends on the position of the cell within a stream, with the posterior localization of ACA mRNA being strongest for cells at the end of a stream. By monitoring the recovery of ACA-YFP after cycloheximide (CHX) treatment, we observed that ACA mRNA and newly synthesized ACA-YFP first emerge as fluorescent punctae that later accumulate to the posterior of cells. We also found that the ACA mRNA localization requires 3' ACA cis-acting elements. CONCLUSIONS: Together, our findings suggest that the asymmetric distribution of ACA mRNA allows the local translation and accumulation of ACA protein at the posterior of cells. These data represent a novel functional role for localized translation in the relay of chemotactic signal during chemotaxis.


Subject(s)
Adenylyl Cyclases , Chemotaxis/genetics , Dictyostelium/enzymology , Protozoan Proteins , RNA, Messenger/genetics , RNA, Messenger/metabolism , Adenylyl Cyclases/genetics , Adenylyl Cyclases/metabolism , Animals , Cell Polarity/drug effects , Cell Polarity/genetics , Cells, Cultured , Chemotaxis/drug effects , Cycloheximide/pharmacology , Cytoplasm/enzymology , Cytoplasmic Streaming/drug effects , Cytoplasmic Streaming/physiology , Dictyostelium/metabolism , In Situ Hybridization, Fluorescence , Protein Biosynthesis/drug effects , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , RNA Transport/physiology , RNA, Messenger/analysis , RNA, Protozoan/analysis , RNA, Protozoan/genetics , RNA, Protozoan/metabolism , Regulatory Sequences, Ribonucleic Acid/physiology , Signal Transduction
6.
Physiol Res ; 66(Suppl 1): S21-S38, 2017 04 05.
Article in English | MEDLINE | ID: mdl-28379027

ABSTRACT

MicroRNAs (miRNAs) are a class of short non-coding regulatory RNA molecules which play an important role in intracellular communication and cell signaling and which influence cellular processes such as proliferation, differentiation, and cellular death. Over the past two decades, the crucial role of microRNAs in controlling tissue homeostasis and disease in cardiovascular systems has become widely recognized. By controlling the expression levels of their targets, several miRNAs have been shown to modulate the function of endothelial cells (miR-221/222 and -126), vascular smooth muscle cells (miR-143/145) and macrophages (miR-33, -758, and -26), thereby regulating the development and progression of atherosclerosis. The stability of miRNAs within the blood suggests that circulating miRNAs may function as important biomarkers of disease development and progression. Numerous circulating miRNAs have been found to be dysregulated in a wide variety of different disease states, including diabetes, cancer, and cardiovascular disease.


Subject(s)
Cardiovascular Diseases/blood , Cardiovascular Diseases/genetics , MicroRNAs/blood , MicroRNAs/genetics , Regulatory Sequences, Ribonucleic Acid/physiology , Animals , Biomarkers/blood , Cardiovascular Diseases/diagnosis , Cholesterol/genetics , Cholesterol/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Humans
7.
Mol Biol Cell ; 28(3): 476-487, 2017 Feb 01.
Article in English | MEDLINE | ID: mdl-27903772

ABSTRACT

Cis-acting RNA structural elements govern crucial aspects of viral gene expression. How these structures and other posttranscriptional signals affect RNA trafficking and translation in the context of single cells is poorly understood. Herein we describe a multicolor, long-term (>24 h) imaging strategy for measuring integrated aspects of viral RNA regulatory control in individual cells. We apply this strategy to demonstrate differential mRNA trafficking behaviors governed by RNA elements derived from three retroviruses (HIV-1, murine leukemia virus, and Mason-Pfizer monkey virus), two hepadnaviruses (hepatitis B virus and woodchuck hepatitis virus), and an intron-retaining transcript encoded by the cellular NXF1 gene. Striking behaviors include "burst" RNA nuclear export dynamics regulated by HIV-1's Rev response element and the viral Rev protein; transient aggregations of RNAs into discrete foci at or near the nuclear membrane triggered by multiple elements; and a novel, pulsiform RNA export activity regulated by the hepadnaviral posttranscriptional regulatory element. We incorporate single-cell tracking and a data-mining algorithm into our approach to obtain RNA element-specific, high-resolution gene expression signatures. Together these imaging assays constitute a tractable, systems-based platform for studying otherwise difficult to access spatiotemporal features of viral and cellular gene regulation.


Subject(s)
Molecular Imaging/methods , Single-Cell Analysis/methods , Active Transport, Cell Nucleus/physiology , Cell Nucleus/metabolism , Gene Expression Regulation, Viral , Gene Products, rev/metabolism , Genes, env/physiology , HIV-1 , Mason-Pfizer monkey virus , RNA Processing, Post-Transcriptional/physiology , RNA, Messenger/metabolism , RNA, Viral , Regulatory Sequences, Nucleic Acid/genetics , Regulatory Sequences, Nucleic Acid/physiology , Regulatory Sequences, Ribonucleic Acid/genetics , Regulatory Sequences, Ribonucleic Acid/physiology
8.
Proc Natl Acad Sci U S A ; 112(45): E6243-52, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26494284

ABSTRACT

Glutamine synthetase (GS), a key enzyme in biological nitrogen assimilation, is regulated in multiple ways in response to varying nitrogen sources and levels. Here we show a small regulatory RNA, NsiR4 (nitrogen stress-induced RNA 4), which plays an important role in the regulation of GS in cyanobacteria. NsiR4 expression in the unicellular Synechocystis sp. PCC 6803 and in the filamentous, nitrogen-fixing Anabaena sp. PCC 7120 is stimulated through nitrogen limitation via NtcA, the global transcriptional regulator of genes involved in nitrogen metabolism. NsiR4 is widely conserved throughout the cyanobacterial phylum, suggesting a conserved function. In silico target prediction, transcriptome profiling on pulse overexpression, and site-directed mutagenesis experiments using a heterologous reporter system showed that NsiR4 interacts with the 5'UTR of gifA mRNA, which encodes glutamine synthetase inactivating factor (IF)7. In Synechocystis, we observed an inverse relationship between the levels of NsiR4 and the accumulation of IF7 in vivo. This NsiR4-dependent modulation of gifA (IF7) mRNA accumulation influenced the glutamine pool and thus [Formula: see text] assimilation via GS. As a second target, we identified ssr1528, a hitherto uncharacterized nitrogen-regulated gene. Competition experiments between WT and an ΔnsiR4 KO mutant showed that the lack of NsiR4 led to decreased acclimation capabilities of Synechocystis toward oscillating nitrogen levels. These results suggest a role for NsiR4 in the regulation of nitrogen metabolism in cyanobacteria, especially for the adaptation to rapid changes in available nitrogen sources and concentrations. NsiR4 is, to our knowledge, the first identified bacterial sRNA regulating the primary assimilation of a macronutrient.


Subject(s)
Bacterial Proteins/metabolism , DNA-Binding Proteins/metabolism , Glutamate-Ammonia Ligase/antagonists & inhibitors , Nitrogen Fixation/physiology , Regulatory Sequences, Ribonucleic Acid/physiology , Synechocystis/physiology , Transcription Factors/metabolism , Blotting, Northern , Gene Expression Profiling , Gene Knockout Techniques , Immunoblotting , Microarray Analysis , Mutagenesis, Site-Directed , Regulatory Sequences, Ribonucleic Acid/genetics
9.
J Biol Chem ; 289(30): 20979-90, 2014 Jul 25.
Article in English | MEDLINE | ID: mdl-24872408

ABSTRACT

Connexin43 (Cx43) is the most abundant gap junction protein in higher vertebrate organisms and has been shown to be involved in junctional and non-junctional functions. In addition to the expression of full-length Cx43, endogenously produced carboxyl-terminal segments of Cx43 have been described and have been suggested to be involved in manifold biological functions, such as hypoxic preconditioning and neuronal migration. Molecular aspects, however, behind the separate generation of carboxyl-terminal segments of Cx43 have remained elusive. Here we report on a mechanism that may play a key role in the separate production of these domains. First, stringent evidence derived from siRNA treatment and specific knockouts revealed significant loss of the low molecular weight fragments of Cx43. By applying a dicistronic vector strategy on transfected cell lines, we were able to identify putative IRES activity (nucleotides 442­637) in the coding region of Cx43, which resides upstream from the nucleotide sequence encoding the carboxyl terminus (nucleotides 637­1149). Functional responsiveness of the endogenous expression of Cx43 fragments to hypoxic/ischemic treatment was evaluated in in vitro and in vivo models, which led to a significant increase of the fastest migrating form (20 kDa) under conditions of metabolic deprivation. By nano-MS spectrometry, we achieved stringent evidence of the identity of the 20-kDa segment as part of the carboxyl-terminal domain of full-length Cx43. Our data prove the existence of endogenously expressed carboxyl-terminal domains, which may serve as valuable tools for further translational application in ischemic disorders.


Subject(s)
Connexin 43/biosynthesis , Models, Biological , Protein Biosynthesis/physiology , Regulatory Sequences, Ribonucleic Acid/physiology , Animals , Cell Hypoxia/physiology , Connexin 43/genetics , Mice , NIH 3T3 Cells , Protein Structure, Tertiary , Rats
10.
RNA Biol ; 10(12): 1778-97, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24356572

ABSTRACT

RNAs have many important functional properties, including that they are independently controllable and highly tunable. As a result of these advantageous properties, their use in a myriad of sophisticated devices has been widely explored. Yet, the exploitation of RNAs for synthetic applications is highly dependent on the ability to characterize the many new molecules that continue to be discovered by large-scale sequencing and high-throughput screening techniques. In this review, we present an exhaustive survey of the most recent synthetic bacterial riboswitches and small RNAs while emphasizing their virtues in gene expression management. We also explore the use of these RNA components as building blocks in the RNA synthetic biology toolbox and discuss examples of synthetic RNA components used to rewire bacterial regulatory circuitry. We anticipate that this field will expand its catalog of smart devices by mimicking and manipulating natural RNA mechanisms and functions.


Subject(s)
Aptamers, Nucleotide/chemical synthesis , RNA, Small Untranslated/metabolism , Regulatory Sequences, Ribonucleic Acid/physiology , Riboswitch , Gene Expression Regulation, Bacterial , RNA, Bacterial , Regulatory Sequences, Ribonucleic Acid/genetics , Synthetic Biology
11.
Cold Spring Harb Perspect Med ; 3(9): a010298, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-24003243

ABSTRACT

Pathogenic bacteria possess intricate regulatory networks that temporally control the production of virulence factors, and enable the bacteria to survive and proliferate after host infection. Regulatory RNAs are now recognized as important components of these networks, and their study may not only identify new approaches to combat infectious diseases but also reveal new general control mechanisms involved in bacterial gene expression. In this review, we illustrate the diversity of regulatory RNAs in bacterial pathogens, their mechanism of action, and how they can be integrated into the regulatory circuits that govern virulence-factor production.


Subject(s)
Bacteria/pathogenicity , RNA, Bacterial/physiology , RNA, Small Untranslated/physiology , Virulence Factors/biosynthesis , Bacterial Proteins/physiology , Genes, Bacterial/genetics , Quorum Sensing/physiology , RNA, Antisense/physiology , RNA, Small Interfering/metabolism , Regulatory Sequences, Ribonucleic Acid/physiology , Signal Transduction , Virulence Factors/genetics
13.
Trends Cogn Sci ; 16(10): 497-503, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22940578

ABSTRACT

The evolution of the human brain has resulted in the emergence of higher-order cognitive abilities, such as reasoning, planning and social awareness. Although there has been a concomitant increase in brain size and complexity, and component diversification, we argue that RNA regulation of epigenetic processes, RNA editing, and the controlled mobilization of transposable elements have provided the major substrates for cognitive advance. We also suggest that these expanded capacities and flexibilities have led to the collateral emergence of psychiatric fragilities and conditions.


Subject(s)
Cognition/physiology , DNA Transposable Elements/physiology , Epigenesis, Genetic/physiology , Evolution, Molecular , Regulatory Sequences, Ribonucleic Acid/physiology , Adaptation, Physiological , Animals , Epigenesis, Genetic/genetics , Humans
14.
RNA ; 18(10): 1910-20, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22915601

ABSTRACT

Adjusting translation is crucial for cells to rapidly adapt to changing conditions. While pro-proliferative signaling via the PI3K-mTOR-pathway is known to induce cap-dependent translation, stress conditions, such as nutrient deprivation or hypoxia often activate alternative modes of translation, e.g., via internal ribosome entry sites (IRESs). As the effects of inflammatory conditions on translation are only poorly characterized, we aimed at identifying translationally deregulated targets in inflammatory settings. For this purpose, we cocultured breast tumor cells with conditioned medium of activated monocyte-derived macrophages (CM). Polysome profiling and microarray analysis identified early growth response-2 (egr2) to be regulated at the level of translation. Using bicistronic reporter assays, we found that egr2 contains an IRES within its 5' UTR, which facilitated enhanced translation upon CM treatment. We further provide evidence that the activity of egr2-IRES was induced by IL-1ß and p38-MAPK signaling. In addition, we identified several potential IRES trans-acting factors (ITAFs) such as polypyrimidine tract binding protein (PTB) and hnRNP-A1 that directly bind to the egr2-5'UTR. In summary, our data provide evidence that egr2 expression is translationally regulated via an IRES element, which is responsive to an inflammatory environment.


Subject(s)
Early Growth Response Protein 2/metabolism , Inflammation/metabolism , Protein Biosynthesis , Regulatory Sequences, Ribonucleic Acid/physiology , Ribosomes/metabolism , Base Sequence , Binding Sites/drug effects , Binding Sites/genetics , Cells, Cultured , Early Growth Response Protein 2/genetics , Humans , Inflammation/genetics , Interleukin-1beta/pharmacology , Models, Biological , Molecular Sequence Data , Nucleic Acid Conformation , Protein Biosynthesis/drug effects , Protein Biosynthesis/physiology , U937 Cells , Up-Regulation/drug effects
15.
Cell Rep ; 2(1): 111-23, 2012 Jul 26.
Article in English | MEDLINE | ID: mdl-22840402

ABSTRACT

Genome-wide studies have identified thousands of long noncoding RNAs (lncRNAs) lacking protein-coding capacity. However, most lncRNAs are expressed at a very low level, and in most cases there is no genetic evidence to support their in vivo function. Malat1 (metastasis associated lung adenocarcinoma transcript 1) is among the most abundant and highly conserved lncRNAs, and it exhibits an uncommon 3'-end processing mechanism. In addition, its specific nuclear localization, developmental regulation, and dysregulation in cancer are suggestive of it having a critical biological function. We have characterized a Malat1 loss-of-function genetic model that indicates that Malat1 is not essential for mouse pre- and postnatal development. Furthermore, depletion of Malat1 does not affect global gene expression, splicing factor level and phosphorylation status, or alternative pre-mRNA splicing. However, among a small number of genes that were dysregulated in adult Malat1 knockout mice, many were Malat1 neighboring genes, thus indicating a potential cis-regulatory role of Malat1 gene transcription.


Subject(s)
Growth and Development/genetics , RNA, Long Noncoding/physiology , Regulatory Sequences, Ribonucleic Acid/physiology , Transcription, Genetic , Age Factors , Aging/genetics , Aging/metabolism , Animals , Female , Gene Expression Regulation, Developmental , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Models, Biological , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Regulatory Sequences, Ribonucleic Acid/genetics , Transcription, Genetic/genetics
16.
J Biol Chem ; 287(24): 20154-63, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22493486

ABSTRACT

GPR41 is a G protein-coupled receptor activated by short chain fatty acids. The gene encoding GPR41 is located immediately downstream of a related gene encoding GPR40, a receptor for long chain fatty acids. Expression of GPR41 has been reported in a small number of cell types, including gut enteroendocrine cells and sympathetic ganglia, where it may play a role in the maintenance of metabolic homeostasis. We now demonstrate that GPR41, like GPR40, is expressed in pancreatic beta cells. Surprisingly, we found no evidence for transcriptional control elements or transcriptional initiation in the intergenic GPR40-GPR41 region. Rather, using 5'-rapid amplification of cDNA ends analysis, we demonstrated that GPR41 is transcribed from the promoter of the GPR40 gene. We confirmed this finding by generating bicistronic luciferase reporter plasmids, and we were able to map a potential internal ribosome entry site-containing region to a 2474-nucleotide region of the intergenic sequence. Consistent with this, we observed m(7)G cap-independent reporter gene expression upon transfection of RNA containing this region. Thus, GPR41 expression is mediated via an internal ribosome entry site located in the intergenic region of a bicistronic mRNA. This novel sequence organization may be utilized to permit coordinated regulation of the fatty acid receptors GPR40 and GPR41.


Subject(s)
Gene Expression Regulation/physiology , Protein Biosynthesis/physiology , Receptors, G-Protein-Coupled/biosynthesis , Regulatory Sequences, Ribonucleic Acid/physiology , Animals , Cricetinae , DNA, Complementary/genetics , HeLa Cells , Humans , Mice , NIH 3T3 Cells , Pancreas/cytology , Pancreas/metabolism , RNA Caps/genetics , RNA Caps/metabolism , Receptors, G-Protein-Coupled/genetics
17.
Cell Mol Life Sci ; 69(1): 103-13, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21598019

ABSTRACT

Hepatitis C virus (HCV) translation is mediated by an internal ribosome entry site (IRES) located at the 5' end of the genomic RNA. The 3' untranslatable region (3'UTR) stimulates translation by the recruitment of protein factors that simultaneously bind to the 5' end of the viral genome. This leads to the formation of a macromolecular complex with a closed loop conformation, similar to that described for the cap-translated mRNAs. We previously demonstrated the existence of a long-range RNA-RNA interaction involving subdomain IIId of the IRES region and the stem-loop 5BSL3.2 of the CRE element at the 3' end of the viral genome. The present study provides evidence that the enhancement of HCV IRES-dependent translation mediated by the 3'UTR is negatively controlled by the CRE region in the human hepatoma cell lines Huh-7 and Hep-G2 in a time-dependent manner. Domain 5BSL3.2 is the major partner in this process. Mutations in this motif lead to an increase in IRES activity by up to eightfold. These data support the existence of a functional high order structure in the HCV genome that involves two evolutionarily conserved RNA elements, domain IIId in the IRES and stem-loop 5BSL3.2 in the CRE region. This interaction could have a role in the circularisation of the viral genome.


Subject(s)
3' Untranslated Regions/physiology , 5' Untranslated Regions/physiology , Genome, Viral/physiology , Hepacivirus , RNA, Viral/genetics , Base Sequence , Binding Sites , Cell Line, Tumor , Hepacivirus/genetics , Hepacivirus/metabolism , Humans , Nucleic Acid Conformation , Protein Biosynthesis/genetics , RNA, Viral/metabolism , Regulatory Sequences, Ribonucleic Acid/physiology , Ribosomes/genetics , Ribosomes/metabolism , Ribosomes/virology
18.
Mol Cancer Res ; 10(1): 167-80, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22049153

ABSTRACT

Commonly observed in colorectal cancer is the elevated expression of the prostaglandin (PG) synthase COX-2. In normal intestinal epithelium, the COX-2 mRNA is targeted for rapid decay through the 3'-untranslated region (3'-UTR) adenylate- and uridylate (AU)-rich element (ARE), whereas in tumors ARE-mediated decay is compromised. Here we show that the COX-2 ARE can mediate degradation through microRNA (miRNA)-mediated regulation. We identified miR-16 to bind the COX-2 3'-UTR and inhibit COX-2 expression by promoting rapid mRNA decay. In colorectal cancer cells and tumors, miR-16 levels were decreased approximately twofold and miR-16 expression in cancer cells attenuated COX-2 expression and PG synthesis. The COX-2 ARE is also bound by the RNA-binding protein HuR. In colorectal cancer tumors, HuR is overexpressed and localized within the cytoplasm, where it promotes ARE-mRNA stabilization. Under conditions of HuR overexpression, miR-16 was unable to promote rapid mRNA decay through the COX-2 ARE. Ribonucleoprotein immunoprecipitation of HuR showed direct association with miR-16 that was reversed when cytoplasmic trafficking of HuR was inhibited. Furthermore, this interaction between HuR and miR-16 promoted the downregulation of miR-16. These new results identify miR-16 as a central posttranscriptional regulator of COX-2 and show the ability of elevated levels of HuR to antagonize miR-16 function. Along with insight into altered ARE-mediated mRNA decay observed in colorectal cancer, these findings provide a new explanation for tumor-derived loss of miR-16.


Subject(s)
Cyclooxygenase 2/genetics , ELAV Proteins/physiology , MicroRNAs/antagonists & inhibitors , Animals , Caco-2 Cells , Carcinoma/genetics , Carcinoma/metabolism , Carcinoma/pathology , Cells, Cultured , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Cyclooxygenase 2/metabolism , Down-Regulation/genetics , ELAV Proteins/genetics , ELAV Proteins/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , HT29 Cells , HeLa Cells , Humans , Mice , Mice, Transgenic , MicroRNAs/genetics , MicroRNAs/metabolism , Protein Binding , Protein Transport/genetics , RNA Stability/genetics , Regulatory Sequences, Ribonucleic Acid/genetics , Regulatory Sequences, Ribonucleic Acid/physiology , Tumor Cells, Cultured
19.
PLoS Pathog ; 7(9): e1002248, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21960769

ABSTRACT

Replication of non-segmented negative-strand RNA viruses requires the continuous supply of the nucleoprotein (N) in the form of a complex with the phosphoprotein (P). Here, we present the structural characterization of a soluble, heterodimeric complex between a variant of vesicular stomatitis virus N lacking its 21 N-terminal residues (N(Δ21)) and a peptide of 60 amino acids (P(60)) encompassing the molecular recognition element (MoRE) of P that binds RNA-free N (N(0)). The complex crystallized in a decameric circular form, which was solved at 3.0 Å resolution, reveals how the MoRE folds upon binding to N and competes with RNA binding and N polymerization. Small-angle X-ray scattering experiment and NMR spectroscopy on the soluble complex confirms the binding of the MoRE and indicates that its flanking regions remain flexible in the complex. The structure of this complex also suggests a mechanism for the initiation of viral RNA synthesis.


Subject(s)
Multiprotein Complexes/chemistry , Nucleocapsid Proteins/chemistry , Phosphoproteins/chemistry , Vesiculovirus/chemistry , Viral Structural Proteins/chemistry , Crystallography, X-Ray , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Nuclear Magnetic Resonance, Biomolecular , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Binding , Protein Structure, Quaternary , RNA, Viral/biosynthesis , RNA, Viral/chemistry , RNA, Viral/genetics , Regulatory Sequences, Ribonucleic Acid/physiology , Vesiculovirus/genetics , Vesiculovirus/metabolism , Viral Structural Proteins/genetics , Viral Structural Proteins/metabolism
20.
Wiley Interdiscip Rev RNA ; 2(1): 92-105, 2011.
Article in English | MEDLINE | ID: mdl-21956971

ABSTRACT

The immunoglobulin heavy chain (IgH) genes, which encode one of the two chains of antibody molecules, were the first cellular genes shown to undergo developmentally regulated alternative RNA processing. These genes produce two different mRNAs from a single primary transcript. One mRNA is cleaved and polyadenylated at an upstream poly(A) signal while the other mRNA removes this poly(A) signal by RNA splicing and is cleaved and polyadenylated at a downstream poly(A) site. A broad range of studies have been performed to understand the mechanism of IgH RNA processing regulation during B lymphocyte development. The model that has emerged is much more complex than envisioned by the earliest view of regulation through poly(A) signal choice. Regulation requires that the IgH gene contain competing splice and cleavage-polyadenylation reactions with balanced efficiencies. Because non-IgH genes with these structural features also can be regulated, IgH gene-specific sequence elements are not required for regulation. Changes in cleavage-polyadenylation and RNA splicing, as well as pol II elongation, all contribute to IgH developmental RNA processing regulation. Multiple factors are likely involved in the regulation during B lymphocyte maturation. Additional biologically relevant factors that contribute to IgH regulation remain to be identified and incorporated into a mechanistic model for regulation. Much of the work to date confirms the complex nature of IgH mRNA regulation and suggests that a thorough understanding of this control will remain a challenge. However, it is also likely that such understanding will help elucidate novel mechanisms of RNA processing regulation.


Subject(s)
Alternative Splicing/physiology , Gene Expression Regulation , Genes, Immunoglobulin Heavy Chain/genetics , Polyadenylation/physiology , Alternative Splicing/genetics , Animals , Binding, Competitive/physiology , Gene Expression Regulation/physiology , Humans , Models, Biological , Polyadenylation/genetics , RNA Processing, Post-Transcriptional/genetics , RNA Processing, Post-Transcriptional/physiology , RNA-Binding Proteins/metabolism , Regulatory Sequences, Ribonucleic Acid/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...