Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Haematologica ; 109(6): 1893-1908, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38124661

ABSTRACT

REIIBP is a lysine methyltransferase aberrantly expressed through alternative promoter usage of NSD2 locus in t(4;14)-translocated multiple myeloma (MM). Clinically, t(4;14) translocation is an adverse prognostic factor found in approximately 15% of MM patients. The contribution of REIIBP relative to other NSD2 isoforms as a dependency gene in t(4;14)-translocated MM remains to be evaluated. Here, we demonstrated that despite homology with NSD2, REIIBP displayed distinct substrate specificity by preferentially catalyzing H3K4me3 and H3K27me3, with little activity on H3K36me2. Furthermore, REIIBP was regulated through microRNA by EZH2 in a Dicer-dependent manner, exemplifying a role of REIIBP in SET-mediated H3K27me3. Chromatin immunoprecipitation sequencing revealed chromatin remodeling characterized by changes in genome-wide and loci-specific occupancy of these opposing histone marks, allowing a bidirectional regulation of its target genes. Transcriptomics indicated that REIIBP induced a pro-inflammatory gene signature through upregulation of TLR7, which in turn led to B-cell receptor-independent activation of BTK and driving NFkB-mediated production of cytokines such as IL-6. Activation of this pathway is targetable using Ibrutinib and partially mitigated bortezomib resistance in a REIIBP xenograft model. Mechanistically, REIIBP upregulated TLR7 through eIF3E, and this relied on eIF3E RNA-binding function instead of its canonical protein synthesis activity, as demonstrated by direct binding to the 3'UTR of TLR7 mRNA. Altogether, we provided a rationale that co-existence of different NSD2 isoforms induced diversified oncogenic programs that should be considered in the strategies for t(4;14)-targeted therapy.


Subject(s)
Chromosomes, Human, Pair 14 , Epigenesis, Genetic , Histone-Lysine N-Methyltransferase , Multiple Myeloma , Translocation, Genetic , Humans , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Multiple Myeloma/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Animals , Mice , Chromosomes, Human, Pair 14/genetics , Chromosomes, Human, Pair 4/genetics , Gene Expression Regulation, Neoplastic , Cell Line, Tumor , Phenotype , Inflammation/genetics , Inflammation/metabolism , Histones/metabolism , Repressor Proteins
2.
Elife ; 112022 07 14.
Article in English | MEDLINE | ID: mdl-35834310

ABSTRACT

The hallmark event of the canonical transforming growth factor ß (TGFß) family signaling is the assembly of the Smad-complex, consisting of the common Smad, Smad4, and phosphorylated receptor-regulated Smads. How the Smad-complex is assembled and regulated is still unclear. Here, we report that active Arl15, an Arf-like small G protein, specifically binds to the MH2 domain of Smad4 and colocalizes with Smad4 at the endolysosome. The binding relieves the autoinhibition of Smad4, which is imposed by the intramolecular interaction between its MH1 and MH2 domains. Activated Smad4 subsequently interacts with phosphorylated receptor-regulated Smads, forming the Smad-complex. Our observations suggest that Smad4 functions as an effector and a GTPase activating protein (GAP) of Arl15. Assembly of the Smad-complex enhances the GAP activity of Smad4 toward Arl15, therefore dissociating Arl15 before the nuclear translocation of the Smad-complex. Our data further demonstrate that Arl15 positively regulates the TGFß family signaling.


Subject(s)
Trans-Activators , Transforming Growth Factor beta , Signal Transduction , Smad3 Protein/metabolism , Smad4 Protein/metabolism , Trans-Activators/metabolism , Transforming Growth Factor beta/metabolism
3.
Nat Cell Biol ; 24(6): 928-939, 2022 06.
Article in English | MEDLINE | ID: mdl-35618746

ABSTRACT

Most mammalian genes generate messenger RNAs with variable untranslated regions (UTRs) that are important post-transcriptional regulators. In cancer, shortening at 3' UTR ends via alternative polyadenylation can activate oncogenes. However, internal 3' UTR splicing remains poorly understood as splicing studies have traditionally focused on protein-coding alterations. Here we systematically map the pan-cancer landscape of 3' UTR splicing and present this in SpUR ( http://www.cbrc.kaust.edu.sa/spur/home/ ). 3' UTR splicing is widespread, upregulated in cancers, correlated with poor prognosis and more prevalent in oncogenes. We show that antisense oligonucleotide-mediated inhibition of 3' UTR splicing efficiently reduces oncogene expression and impedes tumour progression. Notably, CTNNB1 3' UTR splicing is the most consistently dysregulated event across cancers. We validate its upregulation in hepatocellular carcinoma and colon adenocarcinoma, and show that the spliced 3' UTR variant is the predominant contributor to its oncogenic functions. Overall, our study highlights the importance of 3' UTR splicing in cancer and may launch new avenues for RNA-based anti-cancer therapeutics.


Subject(s)
Adenocarcinoma , Colonic Neoplasms , 3' Untranslated Regions/genetics , Adenocarcinoma/genetics , Alternative Splicing/genetics , Animals , Carcinogenesis/genetics , Colonic Neoplasms/genetics , Mammals , Up-Regulation
4.
J Invest Dermatol ; 142(4): 1206-1216.e8, 2022 04.
Article in English | MEDLINE | ID: mdl-34710388

ABSTRACT

Nonhealing wounds are a major area of unmet clinical need remaining problematic to treat. Improved understanding of prohealing mechanisms is invaluable. The enzyme arginase1 (ARG1) is involved in prohealing responses, with its role in macrophages best characterized. ARG1 is also expressed by keratinocytes; however, ARG1 function in these critical wound repair cells is not understood. We characterized ARG1 expression in keratinocytes during normal cutaneous repair and reveal de novo temporal and spatial expression at the epidermal wound edge. Interestingly, epidermal ARG1 expression was decreased in both human and murine delayed healing wounds. We therefore generated a keratinocyte-specific ARG1-null mouse model (K14-cre;Arg1fl/fl) to explore arginase function. Wound repair, linked to changes in keratinocyte proliferation, migration, and differentiation, was significantly delayed in K14-cre;Arg1fl/fl mice. Similarly, using the arginase inhibitor N(omega)-hydroxy-nor-L-arginine, human in vitro and ex vivo models further confirmed this finding, revealing the importance of the downstream polyamine pathway in repair. Indeed, restoring the balance in ARG1 activity through the addition of putrescine proved beneficial in wound closure. In summary, we show that epidermal ARG1 plays, to our knowledge, a previously unreported intrinsic role in cutaneous healing, highlighting epidermal ARG1 and the downstream mediators as potential targets for the therapeutic modulation of wound repair.


Subject(s)
Arginase , Skin Abnormalities , Animals , Arginase/genetics , Arginase/metabolism , Epidermis/metabolism , Keratinocytes/metabolism , Macrophages/metabolism , Mice , Mice, Knockout , Skin/metabolism , Skin Abnormalities/metabolism
5.
Front Mol Biosci ; 8: 773866, 2021.
Article in English | MEDLINE | ID: mdl-34778380

ABSTRACT

Arginase (ARG) represents an important evolutionarily conserved enzyme that is expressed by multiple cell types in the skin. Arg acts as the mediator of the last step of the urea cycle, thus providing protection against excessive ammonia under homeostatic conditions through the production of L-ornithine and urea. L-ornithine represents the intersection point between the ARG-dependent pathways and the urea cycle, therefore contributing to cell detoxification, proliferation and collagen production. The ARG pathways help balance pro- and anti-inflammatory responses in the context of wound healing. However, local and systemic dysfunctionalities of the ARG pathways have been shown to contribute to the hindrance of the healing process and the occurrence of chronic wounds. This review discusses the functions of ARG in macrophages and fibroblasts while detailing the deleterious implications of a malfunctioning ARG enzyme in chronic skin conditions such as leg ulcers. The review also highlights how ARG links with the microbiota and how this impacts on infected chronic wounds. Lastly, the review depicts chronic wound treatments targeting the ARG pathway, alongside future diagnosis and treatment perspectives.

6.
J Invest Dermatol ; 141(9): 2178-2188.e6, 2021 09.
Article in English | MEDLINE | ID: mdl-33984347

ABSTRACT

Maintaining tissue homeostasis depends on a balance between cell proliferation, differentiation, and apoptosis. Within the epidermis, the levels of the polyamines putrescine, spermidine, and spermine are altered in many different skin conditions, yet their role in epidermal tissue homeostasis is poorly understood. We identify the polyamine regulator, Adenosylmethionine decarboxylase 1 (AMD1), as a crucial regulator of keratinocyte (KC) differentiation. AMD1 protein is upregulated on differentiation and is highly expressed in the suprabasal layers of the human epidermis. During KC differentiation, elevated AMD1 promotes decreased putrescine and increased spermine levels. Knockdown or inhibition of AMD1 results in reduced spermine levels and inhibition of KC differentiation. Supplementing AMD1-knockdown KCs with exogenous spermidine or spermine rescued aberrant differentiation. We show that the polyamine shift is critical for the regulation of key transcription factors and signaling proteins that drive KC differentiation, including KLF4 and ZNF750. These findings show that human KCs use controlled changes in polyamine levels to modulate gene expression to drive cellular behavior changes. Modulation of polyamine levels during epidermal differentiation could impact skin barrier formation or can be used in the treatment of hyperproliferative skin disorders.


Subject(s)
Adenosylmethionine Decarboxylase/metabolism , Epidermal Cells/metabolism , Spermine/metabolism , Adenosylmethionine Decarboxylase/genetics , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Epidermal Cells/pathology , Gene Knockdown Techniques , Humans , Kruppel-Like Factor 4/metabolism , Mice , Polyamines/metabolism , Signal Transduction , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Up-Regulation
9.
Nat Biotechnol ; 39(3): 336-346, 2021 03.
Article in English | MEDLINE | ID: mdl-33106685

ABSTRACT

Current methods for determining RNA structure with short-read sequencing cannot capture most differences between distinct transcript isoforms. Here we present RNA structure analysis using nanopore sequencing (PORE-cupine), which combines structure probing using chemical modifications with direct long-read RNA sequencing and machine learning to detect secondary structures in cellular RNAs. PORE-cupine also captures global structural features, such as RNA-binding-protein binding sites and reactivity differences at single-nucleotide variants. We show that shared sequences in different transcript isoforms of the same gene can fold into different structures, highlighting the importance of long-read sequencing for obtaining phase information. We also demonstrate that structural differences between transcript isoforms of the same gene lead to differences in translation efficiency. By revealing isoform-specific RNA structure, PORE-cupine will deepen understanding of the role of structures in controlling gene regulation.


Subject(s)
Nanopore Sequencing/methods , Nucleic Acid Conformation , RNA/chemistry , Sequence Analysis, RNA/methods , Human Embryonic Stem Cells/metabolism , Humans , Isomerism , RNA/genetics , Tetrahymena/genetics , Transcriptome
10.
J Invest Dermatol ; 140(10): 2032-2040.e1, 2020 10.
Article in English | MEDLINE | ID: mdl-32119868

ABSTRACT

Hyperpigmentary conditions can arise when melanogenesis in the epidermis is misregulated. Understanding the pathways underlying melanogenesis is essential for the development of effective treatments. Here, we report that a group of metabolites called polyamines are important in the control of melanogenesis in human skin. Polyamines are cationic molecules present in all cells and are essential for cellular function. We report that polyamine regulator ODC1 is upregulated in melanocytes from melasma lesional skin. We report that the polyamine putrescine can promote pigmentation in human skin explants and primary normal human epidermal melanocytes through induction of tyrosinase which is rate-limiting for the synthesis of melanin. Putrescine supplementation on normal human epidermal melanocytes results in the activation of polyamine catabolism, which results in increased intracellular H2O2. Polyamine catabolism is also increased in human skin explants that have been treated with putrescine. We further report that inhibition of polyamine catabolism prevents putrescine-induced promotion of tyrosinase levels and pigmentation in normal human epidermal melanocytes, showing that polyamine catabolism is responsible for the putrescine induction of melanogenesis. Our data showing that putrescine promotes pigmentation has important consequences for hyperpigmented and hypopigmented conditions. Further understanding of how polyamines control epidermal pigmentation could open the door for the development of new therapeutics.


Subject(s)
Epidermis/drug effects , Melanins/biosynthesis , Putrescine/pharmacology , Biogenic Polyamines/metabolism , Cells, Cultured , Dicarboxylic Acid Transporters/physiology , Epidermis/metabolism , Humans , Melanocytes/drug effects , Melanocytes/metabolism , Middle Aged , Mitochondrial Membrane Transport Proteins/physiology , Putrescine/analogs & derivatives , Skin Pigmentation/drug effects
11.
Methods Mol Biol ; 2109: 55-65, 2020.
Article in English | MEDLINE | ID: mdl-31161578

ABSTRACT

The study of skin pigmentation requires determining the rate of melanin production in melanocytes and quantifying the rate of melanosome transfer to keratinocytes. Here, we describe a method to quantify melanosome transfer using immunofluorescence microscopy coupled with automated image analysis of in vitro human melanocytes and keratinocytes in co-culture. In this method, the number of melanin capped keratinocyte nuclei is quantified.


Subject(s)
Keratinocytes/cytology , Melanocytes/cytology , Melanosomes/transplantation , Cells, Cultured , Coculture Techniques , Humans , Keratinocytes/metabolism , Melanins/metabolism , Melanocytes/metabolism , Melanosomes/metabolism , Microscopy, Fluorescence , Radiographic Image Interpretation, Computer-Assisted
12.
J Invest Dermatol ; 138(12): 2653-2665, 2018 12.
Article in English | MEDLINE | ID: mdl-29906410

ABSTRACT

Wound healing is a dynamic process involving gene-expression changes that drive re-epithelialization. Here, we describe an essential role for polyamine regulator AMD1 in driving cell migration at the wound edge. The polyamines, putrescine, spermidine, and spermine are small cationic molecules that play essential roles in many cellular processes. We demonstrate that AMD1 is rapidly upregulated following wounding in human skin biopsies. Knockdown of AMD1 with small hairpin RNAs causes a delay in cell migration that is rescued by the addition of spermine. We further show that spermine can promote cell migration in keratinocytes and in human ex vivo wounds, where it significantly increases epithelial tongue migration. Knockdown of AMD1 prevents the upregulation of urokinase-type plasminogen activator/urokinase-type plasminogen activator receptor on wounding and results in a failure in actin cytoskeletal reorganization at the wound edge. We demonstrate that keratinocytes respond to wounding by modulating polyamine regulator AMD1 in order to regulate downstream gene expression and promote cell migration. This article highlights a previously unreported role for the regulation of polyamine levels and ratios in cellular behavior and fate.


Subject(s)
Adenosylmethionine Decarboxylase/metabolism , Cell Movement/genetics , Epidermis/physiology , Keratinocytes/physiology , Wound Healing , Wounds and Injuries/metabolism , Actin Cytoskeleton/metabolism , Adenosylmethionine Decarboxylase/genetics , Biopsy , Calcium Signaling , Cells, Cultured , Humans , RNA, Small Interfering/genetics , Re-Epithelialization/genetics , Spermine/metabolism , Up-Regulation , Wounds and Injuries/genetics
13.
Stem Cells ; 36(8): 1170-1178, 2018 08.
Article in English | MEDLINE | ID: mdl-29644784

ABSTRACT

Embryonic stem cells have the ability to self-renew or differentiate and these processes are under tight control. We previously reported that the polyamine regulator AMD1 is critical for embryonic stem cell self-renewal. The polyamines putrescine, spermidine, and spermine are essential organic cations that play a role in a wide array of cellular processes. Here, we explore the essential role of the polyamines in the promotion of self-renewal and identify a new stem cell regulator that acts downstream of the polyamines: MINDY1. MINDY1 protein levels are high in embryonic stem cells (ESCs) and are dependent on high polyamine levels. Overexpression of MINDY1 can promote ESC self-renewal in the absence of the usually essential cytokine Leukemia Inhibitory Factor (LIF). MINDY1 protein is prenylated and this modification is required for its ability to promote self-renewal. We go on to show that Mindy1 RNA is targeted for repression by mir-710 during Neural Precursor cell differentiation. Taken together, these data demonstrate that high polyamine levels are required for ESC self-renewal and that they function, in part, through promotion of high MINDY1 levels. Stem Cells 2018;36:1170-1178.


Subject(s)
Cell Self Renewal , Deubiquitinating Enzymes/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Polyamines/metabolism , Animals , Base Sequence , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Self Renewal/drug effects , Eflornithine/pharmacology , Embryonic Stem Cells/drug effects , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , HeLa Cells , Humans , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Protein Transport/drug effects
14.
Mol Plant ; 10(11): 1387-1399, 2017 11 06.
Article in English | MEDLINE | ID: mdl-28965832

ABSTRACT

5-Methylcytosine (m5C) is a well-characterized DNA modification, and is also predominantly reported in abundant non-coding RNAs in both prokaryotes and eukaryotes. However, the distribution and biological functions of m5C in plant mRNAs remain largely unknown. Here, we report transcriptome-wide profiling of RNA m5C in Arabidopsis thaliana by applying m5C RNA immunoprecipitation followed by a deep-sequencing approach (m5C-RIP-seq). LC-MS/MS and dot blot analyses reveal a dynamic pattern of m5C mRNA modification in various tissues and at different developmental stages. m5C-RIP-seq analysis identified 6045 m5C peaks in 4465 expressed genes in young seedlings. We found that m5C is enriched in coding sequences with two peaks located immediately after start codons and before stop codons, and is associated with mRNAs with low translation activity. We further demonstrated that an RNA (cytosine-5)-methyltransferase, tRNA-specific methyltransferase 4B (TRM4B), exhibits m5C RNA methyltransferase activity. Mutations in TRM4B display defects in root development and decreased m5C peaks. TRM4B affects the transcript levels of the genes involved in root development, which is positively correlated with their mRNA stability and m5C levels. Our results suggest that m5C in mRNA is a new epitranscriptome marker inArabidopsis, and that regulation of this modification is an integral part of gene regulatory networks underlying plant development.


Subject(s)
5-Methylcytosine/metabolism , Arabidopsis Proteins/metabolism , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Gene Expression Profiling , Gene Expression Regulation, Plant/genetics , Methylation , Plant Roots/metabolism , RNA, Messenger/genetics
15.
J Mol Cell Cardiol ; 112: 27-39, 2017 11.
Article in English | MEDLINE | ID: mdl-28865712

ABSTRACT

RATIONALE: Myocardial infarction (MI) triggers a dynamic microRNA response with the potential of yielding therapeutic targets. OBJECTIVE: We aimed to identify novel aberrantly expressed cardiac microRNAs post-MI with potential roles in adverse remodeling in a rat model, and to provide post-ischemic therapeutic inhibition of a candidate pathological microRNA in vivo. METHODS AND RESULTS: Following microRNA array profiling in rat hearts 2 and 14days post-MI, we identified a time-dependent up-regulation of miR-31 compared to sham-operated rats. A progressive increase of miR-31 (up to 91.4±11.3 fold) was detected in the infarcted myocardium by quantitative real-time PCR. Following target prediction analysis, reporter gene assays confirmed that miR-31 targets the 3´UTR of cardiac troponin-T (Tnnt2), E2F transcription factor 6 (E2f6), mineralocorticoid receptor (Nr3c2) and metalloproteinase inhibitor 4 (Timp4) mRNAs. In vitro, hypoxia and oxidative stress up-regulated miR-31 and suppressed target genes in cardiac cell cultures, whereas LNA-based oligonucleotide inhibition of miR-31 (miR-31i) reversed its repressive effect on target mRNAs. Therapeutic post-ischemic administration of miR-31i in rats silenced cardiac miR-31 and enhanced expression of target genes, while preserving cardiac structure and function at 2 and 4weeks post-MI. Left ventricular ejection fraction (EF) improved by 10% (from day 2 to 30 post-MI) in miR-31i-treated rats, whereas controls receiving scrambled LNA inhibitor or placebo incurred a 17% deterioration in EF. miR-31i decreased end-diastolic pressure and infarct size; attenuated interstitial fibrosis in the remote myocardium and enhanced cardiac output. CONCLUSION: miR-31 induction after MI is deleterious to cardiac function while its therapeutic inhibition in vivo ameliorates cardiac dysfunction and prevents the development of post-ischemic adverse remodeling.


Subject(s)
MicroRNAs/metabolism , Myocardial Ischemia/genetics , Ventricular Remodeling/genetics , Animals , Base Sequence , Cell Hypoxia/genetics , Cell Line , Gene Expression Profiling , Gene Silencing/drug effects , Male , Myocardial Ischemia/pathology , Myocardium/metabolism , Oligonucleotides/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/genetics , Rats , Up-Regulation/drug effects , Up-Regulation/genetics , Ventricular Remodeling/drug effects
16.
Sci Rep ; 6: 28112, 2016 06 27.
Article in English | MEDLINE | ID: mdl-27346849

ABSTRACT

The high proliferation rate of embryonic stem cells (ESCs) is thought to arise partly from very low expression of p21. However, how p21 is suppressed in ESCs has been unclear. We found that p53 binds to the p21 promoter in human ESCs (hESCs) as efficiently as in differentiated human mesenchymal stem cells, however it does not promote p21 transcription in hESCs. We observed an enrichment for both the repressive histone H3K27me3 and activating histone H3K4me3 chromatin marks at the p21 locus in hESCs, suggesting it is a suppressed, bivalent domain which overrides activation by p53. Reducing H3K27me3 methylation in hESCs rescued p21 expression, and ectopic expression of p21 in hESCs triggered their differentiation. Further, we uncovered a subset of bivalent promoters bound by p53 in hESCs that are similarly induced upon differentiation in a p53-dependent manner, whereas p53 promotes the transcription of other target genes which do not show an enrichment of H3K27me3 in ESCs. Our studies reveal a unique epigenetic strategy used by ESCs to poise undesired p53 target genes, thus balancing the maintenance of pluripotency in the undifferentiated state with a robust response to differentiation signals, while utilizing p53 activity to maintain genomic stability and homeostasis in ESCs.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/genetics , Histones/metabolism , Human Embryonic Stem Cells/metabolism , Tumor Suppressor Protein p53/metabolism , Base Sequence , Cell Differentiation , Cell Line , Epigenesis, Genetic , Fluorescent Antibody Technique, Indirect , Human Embryonic Stem Cells/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Methylation , Promoter Regions, Genetic/genetics , Protein Binding , Protein Stability , RNA Interference , RNA, Small Interfering/metabolism , Transcriptional Activation , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics
17.
RNA ; 22(6): 867-82, 2016 06.
Article in English | MEDLINE | ID: mdl-27090285

ABSTRACT

Recent footprinting studies have made the surprising observation that long noncoding RNAs (lncRNAs) physically interact with ribosomes. However, these findings remain controversial, and the overall proportion of cytoplasmic lncRNAs involved is unknown. Here we make a global, absolute estimate of the cytoplasmic and ribosome-associated population of stringently filtered lncRNAs in a human cell line using polysome profiling coupled to spike-in normalized microarray analysis. Fifty-four percent of expressed lncRNAs are detected in the cytoplasm. The majority of these (70%) have >50% of their cytoplasmic copies associated with polysomal fractions. These interactions are lost upon disruption of ribosomes by puromycin. Polysomal lncRNAs are distinguished by a number of 5' mRNA-like features, including capping and 5'UTR length. On the other hand, nonpolysomal "free cytoplasmic" lncRNAs have more conserved promoters and a wider range of expression across cell types. Exons of polysomal lncRNAs are depleted of endogenous retroviral insertions, suggesting a role for repetitive elements in lncRNA localization. Finally, we show that blocking of ribosomal elongation results in stabilization of many associated lncRNAs. Together these findings suggest that the ribosome is the default destination for the majority of cytoplasmic long noncoding RNAs and may play a role in their degradation.


Subject(s)
Cytoplasm/metabolism , RNA, Long Noncoding/metabolism , Ribosomes/metabolism , 5' Untranslated Regions , Binding Sites , HeLa Cells , Humans , Hydrolysis , In Situ Hybridization, Fluorescence , K562 Cells
18.
PLoS One ; 11(1): e0143235, 2016.
Article in English | MEDLINE | ID: mdl-26799392

ABSTRACT

The presence of multiple variants for many mRNAs is a major contributor to protein diversity. The processing of these variants is tightly controlled in a cell-type specific manner and has a significant impact on gene expression control. Here we investigate the differential translation rates of individual mRNA variants in embryonic stem cells (ESCs) and in ESC derived Neural Precursor Cells (NPCs) using polysome profiling coupled to RNA sequencing. We show that there are a significant number of detectable mRNA variants in ESCs and NPCs and that many of them show variant specific translation rates. This is correlated with differences in the UTRs of the variants with the 5'UTR playing a predominant role. We suggest that mRNA variants that contain alternate UTRs are under different post-transcriptional controls. This is likely due to the presence or absence of miRNA and protein binding sites that regulate translation rate. This highlights the importance of addressing translation rate when using mRNA levels as a read out of protein abundance. Additional analysis shows that many annotated non-coding mRNAs are present on the polysome fractions in ESCs and NPCs. We believe that the use of polysome fractionation coupled to RNA sequencing is a useful method for analysis of the translation state of many different RNAs in the cell.


Subject(s)
Embryonic Stem Cells/physiology , Protein Biosynthesis , RNA, Messenger/genetics , 3' Untranslated Regions , 5' Untranslated Regions , Animals , Cell Differentiation , Cells, Cultured , Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental , Insulin-Like Growth Factor II/genetics , Mice , Neural Stem Cells/physiology , Polyribosomes/genetics , Polyribosomes/metabolism , RNA Splicing , Ribonucleoproteins/genetics , Sequence Analysis, RNA
19.
Methods Mol Biol ; 1341: 143-55, 2016.
Article in English | MEDLINE | ID: mdl-26084596

ABSTRACT

Regulation of gene expression is essential to enable embryonic stem cells (ESCs) to either self-renew or to differentiate. Translational regulation of mRNA plays a major role in regulating gene expression and has been shown to be important for ESC differentiation. Sucrose gradients can be used to separate mRNAs based on the number of associated ribosomes and this can be used as a readout of the rate of translation. Following centrifugation through a sucrose gradient, mRNAs can be recovered, purified, and analyzed by quantitative real-time polymerase chain reaction (qRT-PCR) to determine their ribosomal load in different cell states. Here, we describe how to differentiate mouse ESCs to Neural Precursor Cells (NPCs) and analyze the rate of translation of individual mRNAs by qRT-PCR following polysome fractionation.


Subject(s)
Mouse Embryonic Stem Cells/metabolism , Neural Stem Cells/cytology , Protein Biosynthesis , RNA, Messenger/genetics , Animals , Cell Culture Techniques/methods , Cell Differentiation , Cell Fractionation/methods , Centrifugation, Density Gradient/methods , Mice , Mouse Embryonic Stem Cells/cytology , Neural Stem Cells/metabolism , Polyribosomes/genetics , RNA, Messenger/isolation & purification , Real-Time Polymerase Chain Reaction/methods
20.
Sci Rep ; 5: 9737, 2015 May 29.
Article in English | MEDLINE | ID: mdl-26024509

ABSTRACT

Oxidative stress (OS) is caused by an imbalance between pro- and anti-oxidant reactions leading to accumulation of reactive oxygen species within cells. We here investigate the effect of OS on the transcriptome of human fibroblasts. OS causes a rapid and transient global induction of transcription characterized by pausing of RNA polymerase II (PolII) in both directions, at specific promoters, within 30 minutes of the OS response. In contrast to protein-coding genes, which are commonly down-regulated, this novel divergent, PolII pausing-phenomenon leads to the generation of thousands of long noncoding RNAs (lncRNAs) with promoter-associated antisense lncRNAs transcripts (si-paancRNAs) representing the major group of stress-induced transcripts. OS causes transient dynamics of si-lncRNAs in nucleus and cytosol, leading to their accumulation at polysomes, in contrast to mRNAs, which get depleted from polysomes. We propose that si-lncRNAs represent a novel component of the transcriptional stress that is known to determine the outcome of immediate-early and later cellular stress responses and we provide insights on the fate of those novel mature lncRNA transcripts by showing that their association with polysomal complexes is significantly increased in OS.


Subject(s)
Genome, Human , Oxidative Stress , RNA, Messenger/genetics , RNA, Untranslated/genetics , Transcriptome , Binding Sites , Cell Line , Chromatin Immunoprecipitation , Computational Biology/methods , Fibroblasts/metabolism , Gene Expression Profiling , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Humans , Promoter Regions, Genetic , Protein Biosynthesis , RNA Polymerase II/metabolism , RNA Polymerase III/metabolism , RNA, Antisense/genetics , RNA, Long Noncoding/classification , RNA, Long Noncoding/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...