Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 51(10): 5193-5209, 2023 06 09.
Article in English | MEDLINE | ID: mdl-37070602

ABSTRACT

The long non-coding RNA EPR is expressed in epithelial tissues, binds to chromatin and controls distinct biological activities in mouse mammary gland cells. Because of its high expression in the intestine, in this study we have generated a colon-specific conditional targeted deletion (EPR cKO) to evaluate EPR in vivo functions in mice. EPR cKO mice display epithelium hyperproliferation, impaired mucus production and secretion, as well as inflammatory infiltration in the proximal portion of the large intestine. RNA sequencing analysis reveals a rearrangement of the colon crypt transcriptome with strong reduction of goblet cell-specific factors including those involved in the synthesis, assembly, transport and control of mucus proteins. Further, colon mucosa integrity and permeability are impaired in EPR cKO mice, and this results in higher susceptibility to dextran sodium sulfate (DSS)-induced colitis and tumor formation. Human EPR is down-regulated in human cancer cell lines as well as in human cancers, and overexpression of EPR in a colon cancer cell line results in enhanced expression of pro-apoptotic genes. Mechanistically, we show that EPR directly interacts with select genes involved in mucus metabolism whose expression is reduced in EPR cKO mice and that EPR deletion causes tridimensional chromatin organization changes.


Subject(s)
Cell Transformation, Neoplastic , Inflammation , Mucus , RNA, Long Noncoding , Animals , Humans , Mice , Cell Transformation, Neoplastic/immunology , Colon/metabolism , Disease Models, Animal , Inflammation/immunology , Intestinal Mucosa/metabolism , Mice, Inbred C57BL , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
2.
Nucleic Acids Res ; 50(13): 7608-7622, 2022 07 22.
Article in English | MEDLINE | ID: mdl-35748870

ABSTRACT

EPR is a long non-coding RNA (lncRNA) that controls cell proliferation in mammary gland cells by regulating gene transcription. Here, we report on Mettl7a1 as a direct target of EPR. We show that EPR induces Mettl7a1 transcription by rewiring three-dimensional chromatin interactions at the Mettl7a1 locus. Our data indicate that METTL7A1 contributes to EPR-dependent inhibition of TGF-ß signaling. METTL7A1 is absent in tumorigenic murine mammary gland cells and its human ortholog (METTL7A) is downregulated in breast cancers. Importantly, re-expression of METTL7A1 in 4T1 tumorigenic cells attenuates their transformation potential, with the putative methyltransferase activity of METTL7A1 being dispensable for its biological functions. We found that METTL7A1 localizes in the cytoplasm whereby it interacts with factors implicated in the early steps of mRNA translation, associates with ribosomes, and affects the levels of target proteins without altering mRNA abundance. Overall, our data indicates that METTL7A1-a transcriptional target of EPR-modulates translation of select transcripts.


Subject(s)
Breast Neoplasms , Methyltransferases/metabolism , RNA, Long Noncoding , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Proliferation , Chromatin/genetics , Female , Humans , Mice , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Ribosomes/metabolism
3.
Hematol Oncol ; 40(1): 40-47, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34679195

ABSTRACT

Long non-coding RNAs are emerging as essential regulators of gene expression, but their role in normal and neoplastic B cells is still largely uncharacterized. Here, we report on the expression pattern of the LINC00152 in normal B cells and Chronic Lymphocytic Leukemia B cell clones. Higher LINC00152 levels were consistently observed in memory B cell populations when compared to naïve B cells in the normal tissues analyzed [peripheral blood (PB), tonsils, and spleen]. In addition, independent stimulation via Immunoglobulins (IG), CD40, or Toll-like Receptor 9 (TLR9) upregulated LINC00152 in PB B cells. The expression of LINC00152 in a cohort of 107 early stage Binet A CLL patients was highly variable and did not correlate with known prognostic markers or clinical evolution. TLR9 stimulation, but not CD40 or IG challenge, was able to upregulate LINC00152 expression in CLL cells. In addition, LINC00152 silencing in CLL cell lines expressing LINC00152 failed to induce significant cell survival or apoptosis changes. These data suggest that, in normal B cells, the expression of LINC00152 is regulated by immunomodulatory signals, which are only partially effective in CLL cells. However, LINC00152 does not appear to contribute to CLL cell expansion and/or survival in a cohort of newly diagnosed CLL patients.


Subject(s)
Biomarkers, Tumor/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Palatine Tonsil/metabolism , RNA, Long Noncoding/metabolism , Spleen/metabolism , Biomarkers, Tumor/genetics , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Prognosis , Prospective Studies , RNA, Long Noncoding/genetics , Survival Rate
4.
Noncoding RNA ; 6(3)2020 Sep 17.
Article in English | MEDLINE | ID: mdl-32957640

ABSTRACT

Although mammals possess roughly the same number of protein-coding genes as worms, it is evident that the non-coding transcriptome content has become far broader and more sophisticated during evolution. Indeed, the vital regulatory importance of both short and long non-coding RNAs (lncRNAs) has been demonstrated during the last two decades. RNA binding proteins (RBPs) represent approximately 7.5% of all proteins and regulate the fate and function of a huge number of transcripts thus contributing to ensure cellular homeostasis. Transcriptomic and proteomic studies revealed that RBP-based complexes often include lncRNAs. This review will describe examples of how lncRNA-RBP networks can virtually control all the post-transcriptional events in the cell.

5.
Nucleic Acids Res ; 48(16): 9053-9066, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32756918

ABSTRACT

Long non-coding RNAs (lncRNAs) can affect multiple layers of gene expression to control crucial cellular functions. We have previously demonstrated that the lncRNA EPR, by controlling gene expression at different levels, affects cell proliferation and migration in cultured mammary gland cells and impairs breast tumor formation in an orthotopic transplant model in mice. Here, we used ChIRP-Seq to identify EPR binding sites on chromatin of NMuMG mammary gland cells overexpressing EPR and identified its trans binding sites in the genome. Then, with the purpose of relating EPR/chromatin interactions to the reshaping of the epitranscriptome landscape, we profiled histone activation marks at promoter/enhancer regions by ChIP-Seq. Finally, we integrated data derived from ChIRP-Seq, ChIP-Seq as well as RNA-Seq in a comprehensive analysis and we selected a group of bona fide direct transcriptional targets of EPR. Among them, we identified a subset of EPR targets whose expression is controlled by TGF-ß with one of them-Arrdc3-being able to modulate Epithelial to Mesenchymal Transition. This experimental framework allowed us to correlate lncRNA/chromatin interactions with the real outcome of gene expression and to start defining the gene network regulated by EPR as a component of the TGF-ß pathway.


Subject(s)
Arrestins/genetics , Breast Neoplasms/genetics , RNA, Long Noncoding/genetics , Transforming Growth Factor beta/genetics , Animals , Binding Sites/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Chromatin/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice , Transcriptome/genetics
6.
Nat Commun ; 10(1): 1969, 2019 04 29.
Article in English | MEDLINE | ID: mdl-31036808

ABSTRACT

Long noncoding RNAs (lncRNAs) are emerging as regulators of fundamental biological processes. Here we report on the characterization of an intergenic lncRNA expressed in epithelial tissues which we termed EPR (Epithelial cell Program Regulator). EPR is rapidly downregulated by TGF-ß and its sustained expression largely reshapes the transcriptome, favors the acquisition of epithelial traits, and reduces cell proliferation in cultured mammary gland cells as well as in an animal model of orthotopic transplantation. EPR generates a small peptide that localizes at epithelial cell junctions but the RNA molecule per se accounts for the vast majority of EPR-induced gene expression changes. Mechanistically, EPR interacts with chromatin and regulates Cdkn1a gene expression by affecting both its transcription and mRNA decay through its association with SMAD3 and the mRNA decay-promoting factor KHSRP, respectively. We propose that EPR enables epithelial cells to control proliferation by modulating waves of gene expression in response to TGF-ß.


Subject(s)
RNA Stability/genetics , RNA, Long Noncoding/genetics , Smad3 Protein/metabolism , Transcriptome/genetics , Transforming Growth Factor beta/pharmacology , Animals , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Humans , RNA, Long Noncoding/drug effects
8.
Biochim Biophys Acta Gene Regul Mech ; 1860(3): 291-298, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28088441

ABSTRACT

Resveratrol (RESV) is a natural polyphenolic compound endowed with anti-inflammatory, anti-proliferative, as well as pro-apoptotic activities that make it a potential anti-tumor compound. Here we show that RESV counteracts the TGF-ß-induced Epithelial to Mesenchymal Transition (EMT) phenotype in mammary gland cells and affects the alternative exon usage of pre-mRNAs that encode crucial factors in adhesion and migration -including CD44, ENAH, and FGFR2- in a panel of immortalized and transformed mammary gland cells. RESV causes a shift from the mesenchymal-specific forms of these factors to the respective epithelial forms and increases the expression of the RNA-binding proteins KHSRP and hnRNPA1. From a mechanistic point of view, we show that the combined silencing of KHSRP and hnRNPA1 prevents the RESV-dependent inclusion of the epithelial-type exons in the Cd44 pre-mRNA. Our findings support an unexpected regulatory mechanism where RESV limits EMT by controlling gene expression at post-transcriptional level.


Subject(s)
Alternative Splicing/drug effects , Epithelial-Mesenchymal Transition/drug effects , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Mammary Glands, Animal/metabolism , RNA-Binding Proteins/metabolism , Stilbenes/pharmacology , Animals , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Female , Heterogeneous Nuclear Ribonucleoprotein A1 , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Mice , Microfilament Proteins , RNA-Binding Proteins/genetics , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Resveratrol
9.
Cell Rep ; 16(4): 967-978, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27396342

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) confers several traits to cancer cells that are required for malignant progression. Here, we report that miR-27b-3p-mediated silencing of the single-strand RNA binding protein KHSRP is required for transforming growth factor ß (TGF-ß)-induced EMT in mammary gland cells. Sustained KHSRP expression limits TGF-ß-dependent induction of EMT factors and cell migration, whereas its knockdown in untreated cells mimics TGF-ß-induced EMT. Genome-wide sequencing analyses revealed that KHSRP controls (1) levels of mature miR-192-5p, a microRNA that targets a group of EMT factors, and (2) alternative splicing of a cohort of pre-mRNAs related to cell adhesion and motility including Cd44 and Fgfr2. KHSRP belongs to a ribonucleoprotein complex that includes hnRNPA1, and the two proteins cooperate in promoting epithelial-type exon usage of select pre-mRNAs. Thus, TGF-ß-induced KHSRP silencing is central in a pathway leading to gene-expression changes that contribute to the cellular changes linked to EMT.


Subject(s)
Alternative Splicing/genetics , Epithelial-Mesenchymal Transition/genetics , MicroRNAs/genetics , RNA Interference/physiology , RNA-Binding Proteins/genetics , Trans-Activators/genetics , Transforming Growth Factor beta/genetics , Animals , Cell Adhesion/genetics , Cell Line , Cell Line, Tumor , Cell Movement/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , HEK293 Cells , Humans , Hyaluronan Receptors/genetics , Mammary Glands, Animal , Mice , Receptor, Fibroblast Growth Factor, Type 2/genetics
10.
Wiley Interdiscip Rev RNA ; 7(2): 227-40, 2016.
Article in English | MEDLINE | ID: mdl-26708421

ABSTRACT

The single-stranded nucleic acid-binding protein KHSRP (KH-type splicing regulatory protein) modulates RNA life and gene expression at various levels. KHSRP controls important cellular functions as different as proliferation, differentiation, metabolism, and response to infectious agents. We summarize and discuss experimental evidence providing a potential link between changes in KHSRP expression/function and human diseases including neuromuscular disorders, obesity, type II diabetes, and cancer.


Subject(s)
Cell Differentiation , Disease Susceptibility , RNA-Binding Proteins/metabolism , Trans-Activators/metabolism , Animals , Gene Expression Regulation , Humans , Immunomodulation , Lipid Metabolism , Muscle Development , Muscles/physiology , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , Neurons/metabolism , RNA Processing, Post-Transcriptional , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Signal Transduction , Trans-Activators/chemistry , Trans-Activators/genetics , Virus Diseases/immunology , Virus Diseases/metabolism , Virus Diseases/virology
12.
Proc Natl Acad Sci U S A ; 111(47): E5023-8, 2014 Nov 25.
Article in English | MEDLINE | ID: mdl-25385579

ABSTRACT

Long noncoding RNAs (lncRNAs) interact with protein factors to regulate different layers of gene expression transcriptionally or posttranscriptionally. Here we report on the functional consequences of the unanticipated interaction of the RNA binding protein K homology-type splicing regulatory protein (KSRP) with the H19 lncRNA (H19). KSRP directly binds to H19 in the cytoplasm of undifferentiated multipotent mesenchymal C2C12 cells, and this interaction favors KSRP-mediated destabilization of labile transcripts such as myogenin. AKT activation induces KSRP dismissal from H19 and, as a consequence, myogenin mRNA is stabilized while KSRP is repurposed to promote maturation of myogenic microRNAs, thus favoring myogenic differentiation. Our data indicate that H19 operates as a molecular scaffold that facilitates effective association of KSRP with myogenin and other labile transcripts, and we propose that H19 works with KSRP to optimize an AKT-regulated posttranscriptional switch that controls myogenic differentiation.


Subject(s)
RNA, Long Noncoding/physiology , RNA, Messenger/metabolism , Animals , Cell Line , Humans , Protein Binding , RNA, Long Noncoding/metabolism , RNA-Binding Proteins/metabolism , Trans-Activators/metabolism
13.
Semin Cell Dev Biol ; 34: 2-8, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24845017

ABSTRACT

The single-strand-RNA binding protein KSRP is able to negatively regulate gene expression operating with at least two distinct and integrated postranscriptional mechanisms: (i) by promoting decay of unstable mRNAs and (ii) by favoring maturation from precursors of select microRNAs (miRNAs) including the prototypical tumor suppressor let-7. Studies performed in primary and cultured cells as well as in mice proved that the ability of KSRP to integrate different levels of gene expression is required for proper immune response, lipid metabolism, cell-fate decisions, tissue regeneration, and DNA damage response.


Subject(s)
RNA-Binding Proteins/physiology , Trans-Activators/physiology , Animals , Cell Differentiation , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation , Genes, MHC Class II , Humans , Lipid Metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism
14.
Mol Cell Biol ; 34(12): 2339-49, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24732799

ABSTRACT

White adipose tissue (WAT) releases fatty acids from stored triacylglycerol for an energy source. Here, we report that targeted deletion of KH-type splicing regulatory protein (KSRP), an RNA-binding protein that regulates gene expression at multiple levels, enhances lipolysis in epididymal WAT (eWAT) because of the upregulation of genes promoting lipolytic activity. Expression of microRNA 145 (miR-145) is decreased because of impaired primary miR-145 processing in Ksrp(-/-) eWAT. We show that miR-145 directly targets and represses Foxo1 and Cgi58, activators of lipolytic activity, and forced expression of miR-145 attenuates lipolysis. This study reveals a novel in vivo function of KSRP in controlling adipose lipolysis through posttranscriptional regulation of miR-145 expression.


Subject(s)
Adipose Tissue, White/metabolism , Lipolysis/genetics , MicroRNAs/metabolism , RNA-Binding Proteins/metabolism , Trans-Activators/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Adiposity , Animals , Cell Differentiation , Cell Size , Down-Regulation/genetics , Epididymis/metabolism , Fatty Acids/metabolism , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Gene Deletion , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Organ Size , Oxidation-Reduction , RNA Processing, Post-Transcriptional/genetics , Thermogenesis/genetics , Trans-Activators/deficiency , Triglycerides/metabolism
15.
Diabetes ; 63(9): 2949-61, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24722250

ABSTRACT

Brown adipose tissue oxidizes chemical energy for heat generation and energy expenditure. Promoting brown-like transformation in white adipose tissue (WAT) is a promising strategy for combating obesity. Here, we find that targeted deletion of KH-type splicing regulatory protein (KSRP), an RNA-binding protein that regulates gene expression at multiple levels, causes a reduction in body adiposity. The expression of brown fat-selective genes is increased in subcutaneous/inguinal WAT (iWAT) of Ksrp(-/-) mice because of the elevated expression of PR domain containing 16 and peroxisome proliferator-activated receptor gamma coactivator 1α, which are key regulators promoting the brown fat gene program. The expression of microRNA (miR)-150 in iWAT is decreased due to impaired primary miR-150 processing in the absence of KSRP. We show that miR-150 directly targets and represses Prdm16 and Ppargc1a, and that forced expression of miR-150 attenuates the elevated expression of brown fat genes caused by KSRP deletion. This study reveals the in vivo function of KSRP in controlling brown-like transformation of iWAT through post-transcriptional regulation of miR-150 expression.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , MicroRNAs/biosynthesis , Trans-Activators/deficiency , Adiposity/genetics , Animals , DNA-Binding Proteins/biosynthesis , Diet, High-Fat , Down-Regulation , Gene Expression Regulation , Male , Mice , MicroRNAs/genetics , Obesity/genetics , Obesity/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , RNA-Binding Proteins/physiology , Trans-Activators/physiology , Transcription Factors/biosynthesis , Up-Regulation
16.
Biochim Biophys Acta ; 1829(5): 469-79, 2013 May.
Article in English | MEDLINE | ID: mdl-23462617

ABSTRACT

Understanding the molecular mechanisms that control the balance between multipotency and differentiation is of great importance to elucidate the genesis of both developmental disorders and cell transformation events. To investigate the role of the RNA binding protein KSRP in controlling neural differentiation, we used the P19 embryonal carcinoma cell line that is able to differentiate into neuron-like cells under appropriate culture conditions. We have recently reported that KSRP controls the differentiative fate of multipotent mesenchymal cells owing to its ability to promote decay of unstable transcripts and to favor maturation of selected micro-RNAs (miRNAs) from precursors. Here we report that KSRP silencing in P19 cells favors neural differentiation increasing the expression of neuronal markers. Further, the expression of two master transcriptional regulators of neurogenesis, ASCL1 and JMJD3, was enhanced while the maturation of miR-200 family members from precursors was impaired in KSRP knockdown cells. These molecular changes can contribute to the reshaping of P19 cells transcriptome that follows KSRP silencing. Our data suggests that KSRP function is required to maintain P19 cells in a multipotent undifferentiated state and that its inactivation can orient cells towards neural differentiation.


Subject(s)
Gene Silencing , Neurogenesis/genetics , RNA-Binding Proteins/genetics , Trans-Activators/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line, Tumor , Jumonji Domain-Containing Histone Demethylases/metabolism , Mesenchymal Stem Cells/cytology , Mice , MicroRNAs/metabolism , RNA Precursors/metabolism , RNA Stability , RNA-Binding Proteins/metabolism , Teratocarcinoma , Trans-Activators/metabolism , Transcription, Genetic , Transcriptome
17.
Biochim Biophys Acta ; 1829(6-7): 689-94, 2013.
Article in English | MEDLINE | ID: mdl-23178464

ABSTRACT

KSRP is a single strand nucleic acid binding protein that controls gene expression at multiple levels. In this review we focus on the recent molecular, cellular, and structural insights into the mRNA decay promoting function of KSRP. We discuss also some aspects of KSRP-dependent microRNA maturation from precursors that are related to its mRNA destabilizing function. This article is part of a Special Issue entitled: RNA Decay mechanisms.


Subject(s)
MicroRNAs/genetics , RNA Stability/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Trans-Activators/genetics , Binding Sites , Cell Line , Gene Expression Regulation , Humans , RNA-Binding Proteins/metabolism
18.
Nat Struct Mol Biol ; 19(12): 1282-6, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23142982

ABSTRACT

Let-7 is an important tumor-suppressive microRNA (miRNA) that acts as an on-off switch for cellular differentiation and regulates the expression of a set of human oncogenes. Binding of the human KSRP protein to let-7 miRNA precursors positively regulates their processing to mature let-7, thereby contributing to control of cell proliferation, apoptosis and differentiation. Here we analyze the molecular basis for KSRP-let-7 precursor selectivity and show how the third KH domain of the protein recognizes a G-rich sequence in the pre-let-7 terminal loop and dominates the interaction. The structure of the KH3-RNA complex explains the protein recognition of this noncanonical KH target sequence, and we demonstrate that the specificity of this binding is crucial for the functional interaction between the protein and the miRNA precursor.


Subject(s)
MicroRNAs/biosynthesis , RNA-Binding Proteins/physiology , Trans-Activators/physiology , Humans , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular
19.
Cell Rep ; 2(5): 1159-68, 2012 Nov 29.
Article in English | MEDLINE | ID: mdl-23177623

ABSTRACT

MicroRNAs (miRNAs) are essential regulators of development, physiology, and evolution, and their biogenesis is strictly controlled at multiple levels. Regulatory proteins, such as KSRP, modulate rates and timing of enzymatic reactions responsible for maturation of select miRNAs from their primary transcripts in response to specific stimuli. Here, we show that KSRP silencing in mesenchymal C2C12 cells produces a change in the transcriptome largely overlapping that induced by bone morphogenetic protein 2 (BMP2) signaling activation. This induces osteoblastic differentiation while preventing myogenic differentiation. KSRP silencing- and BMP2-dependent myogenic miRNA (myomiR) maturation blockade is required for osteoblastic differentiation of C2C12 cells. Our results demonstrate that phosphorylated R-SMAD proteins, the transducers of BMP2 signal, associate with phosphorylated KSRP and block its interaction with primary myomiRs. This abrogates KSRP-dependent myomiR maturation, with SMAD4, SMAD5, and SMAD9 silencing being able to rescue KSRP function. Thus, SMAD-induced blockade of KSRP-dependent myomiR maturation is critical for orienting C2C12 cell differentiation toward osteoblastic lineage.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , MicroRNAs/metabolism , RNA-Binding Proteins/metabolism , Smad Proteins/metabolism , Trans-Activators/metabolism , Animals , Cell Differentiation , Cell Line , Cell Lineage , Mice , Osteoblasts/cytology , Osteoblasts/metabolism , Phosphorylation , RNA Interference , RNA, Small Interfering/metabolism , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , Signal Transduction , Smad4 Protein/metabolism , Smad5 Protein/metabolism , Smad6 Protein/metabolism , Trans-Activators/antagonists & inhibitors , Trans-Activators/genetics , Transcriptome
20.
PLoS Genet ; 8(7): e1002823, 2012.
Article in English | MEDLINE | ID: mdl-22844247

ABSTRACT

Gene silencing mediated by either microRNAs (miRNAs) or Adenylate/uridylate-rich elements Mediated mRNA Degradation (AMD) is a powerful way to post-transcriptionally modulate gene expression. We and others have reported that the RNA-binding protein KSRP favors the biogenesis of select miRNAs (including let-7 family) and activates AMD promoting the decay of inherently labile mRNAs. Different layers of interplay between miRNA- and AMD-mediated gene silencing have been proposed in cultured cells, but the relationship between the two pathways in living organisms is still elusive. We conditionally deleted Dicer in mouse pituitary from embryonic day (E) 9.5 through Cre-mediated recombination. In situ hybridization, immunohistochemistry, and quantitative reverse transcriptase-PCR revealed that Dicer is essential for pituitary morphogenesis and correct expression of hormones. Strikingly, αGSU (alpha glycoprotein subunit, common to three pituitary hormones) was absent in Dicer-deleted pituitaries. αGSU mRNA is unstable and its half-life increases during pituitary development. A transcriptome-wide analysis of microdissected E12.5 pituitaries revealed a significant increment of KSRP expression in conditional Dicer-deleted mice. We found that KSRP directly binds to αGSU mRNA, promoting its rapid decay; and, during pituitary development, αGSU expression displays an inverse temporal relationship to KSRP. Further, let-7b/c downregulated KSRP expression, promoting the degradation of its mRNA by directly binding to the 3'UTR. Therefore, we propose a model in which let-7b/c and KSRP operate within a negative feedback loop. Starting from E12.5, KSRP induces the maturation of let-7b/c that, in turn, post-transcriptionally downregulates the expression of KSRP itself. This event leads to stabilization of αGSU mRNA, which ultimately enhances the steady-state expression levels. We have identified a post-transcriptional regulatory network active during mouse pituitary development in which the expression of the hormone αGSU is increased by let7b/c through downregulation of KSRP. Our study unveils a functional crosstalk between miRNA- and AMD-dependent gene regulation during mammalian organogenesis events.


Subject(s)
MicroRNAs/genetics , Organogenesis/genetics , Pituitary Gland , RNA, Messenger , RNA-Binding Proteins/genetics , Trans-Activators/genetics , Animals , DEAD-box RNA Helicases/genetics , Embryonic Development/genetics , Feedback, Physiological , Gene Expression Regulation, Developmental , Glycoprotein Hormones, alpha Subunit/genetics , Glycoprotein Hormones, alpha Subunit/metabolism , HEK293 Cells , HeLa Cells , Humans , Mice , MicroRNAs/metabolism , NIH 3T3 Cells , Pituitary Gland/embryology , Pituitary Gland/growth & development , Pituitary Gland/metabolism , Pituitary Hormones/genetics , Pituitary Hormones/metabolism , RNA Stability/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Ribonuclease III/genetics , Trans-Activators/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...