Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Aging (Albany NY) ; 12(16): 15906-15917, 2020 08 29.
Article in English | MEDLINE | ID: mdl-32860669

ABSTRACT

Telomeres are transcribed into long non-coding RNAs known as Telomeric Repeat-Containing RNA (TERRA). They have been shown to be essential regulators of telomeres and to act as epigenomic modulators at extra-telomeric sites. However the role of TERRA during early embryonic development has never been investigated. Here, we show that TERRA is expressed in murine and bovine early development following a wave pattern. It starts at 4-cell stage, reaching a maximum at the 16-cell followed by a decline at the morula and blastocyst stages. Moreover, TERRA expression is not affected by increasing oocyte donor age whereas telomere length does. This indicates that TERRA expression is independent of the telomere length in early development. Our findings anticipate an essential role of TERRA in early stages of development and this might be useful in the future for a better understanding of age related female infertility.


Subject(s)
Cleavage Stage, Ovum/metabolism , DNA-Binding Proteins/metabolism , Maternal Age , Telomere Homeostasis , Telomere/metabolism , Transcription Factors/metabolism , Animals , Cattle , DNA-Binding Proteins/genetics , Embryo Culture Techniques , Female , Fertilization in Vitro , Gene Expression Regulation, Developmental , Male , Mice, Inbred C57BL , Oocyte Donation , Telomere/genetics , Time Factors , Transcription Factors/genetics
2.
Int J Cancer ; 146(2): 521-530, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31403184

ABSTRACT

It is critical to identify biomarkers and functional networks associated with aggressive thyroid cancer to anticipate disease progression and facilitate personalized patient management. We performed miRNome sequencing of 46 thyroid tumors enriched with advanced disease patients with a median follow-up of 96 months. MiRNome profiles correlated with tumor-specific histopathological and molecular features, such as stromal cell infiltration and tumor driver mutation. Differential expression analysis revealed a consistent hsa-miR-139-5p downexpression in primary carcinomas from patients with recurrent/metastatic disease compared to disease-free patients, sustained in paired local metastases and validated in publicly available thyroid cancer series. Exogenous expression of hsa-miR-139-5p significantly reduced migration and proliferation of anaplastic thyroid cancer cells. Proteomic analysis indicated RICTOR, SMAD2/3 and HNRNPF as putative hsa-miR-139-5p targets in our cell system. Abundance of HNRNPF mRNA, encoding an alternative splicing factor involved in cryptic exon inclusion/exclusion, inversely correlated with hsa-miR-139-5p expression in human tumors. RNA sequencing analysis revealed 174 splicing events differentially regulated upon HNRNPF repression in our cell system, affecting genes involved in RTK/RAS/MAPK and PI3K/AKT/MTOR signaling cascades among others. These results point at the hsa-miR-139-5p/HNRNPF axis as a novel regulatory mechanism associated with the modulation of major thyroid cancer signaling pathways and tumor virulence.


Subject(s)
Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Heterogeneous-Nuclear Ribonucleoprotein Group F-H/genetics , MicroRNAs/metabolism , Thyroid Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Alternative Splicing/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Disease-Free Survival , Female , Follow-Up Studies , Gene Expression Profiling , Heterogeneous-Nuclear Ribonucleoprotein Group F-H/metabolism , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis , Signal Transduction/genetics , Survival Rate , Thyroid Gland/pathology , Thyroid Neoplasms/mortality , Thyroid Neoplasms/pathology
3.
Elife ; 82019 08 20.
Article in English | MEDLINE | ID: mdl-31426913

ABSTRACT

The mechanisms that regulate pluripotency are still largely unknown. Here, we show that Telomere Repeat Binding Factor 1 (TRF1), a component of the shelterin complex, regulates the genome-wide binding of polycomb and polycomb H3K27me3 repressive marks to pluripotency genes, thereby exerting vast epigenetic changes that contribute to the maintenance of mouse ES cells in a naïve state. We further show that TRF1 mediates these effects by regulating TERRA, the lncRNAs transcribed from telomeres. We find that TERRAs are enriched at polycomb and stem cell genes in pluripotent cells and that TRF1 abrogation results in increased TERRA levels and in higher TERRA binding to those genes, coincidental with the induction of cell-fate programs and the loss of the naïve state. These results are consistent with a model in which TRF1-dependent changes in TERRA levels modulate polycomb recruitment to pluripotency and differentiation genes. These unprecedented findings explain why TRF1 is essential for the induction and maintenance of pluripotency.


Subject(s)
Epigenesis, Genetic , Induced Pluripotent Stem Cells/metabolism , Polycomb Repressive Complex 2/metabolism , RNA, Long Noncoding/metabolism , Telomeric Repeat Binding Protein 1/metabolism , Transcription, Genetic , Animals , Cell Differentiation , Cells, Cultured , Mice
4.
Stem Cell Reports ; 8(2): 460-475, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28162998

ABSTRACT

Reprogramming of differentiated cells into induced pluripotent stem cells has been recently achieved in vivo in mice. Telomeres are essential for chromosomal stability and determine organismal life span as well as cancer growth. Here, we study whether tissue dedifferentiation induced by in vivo reprogramming involves changes at telomeres. We find telomerase-dependent telomere elongation in the reprogrammed areas. Notably, we found highly upregulated expression of the TRF1 telomere protein in the reprogrammed areas, which was independent of telomere length. Moreover, TRF1 inhibition reduced in vivo reprogramming efficiency. Importantly, we extend the finding of TRF1 upregulation to pathological tissue dedifferentiation associated with neoplasias, in particular during pancreatic acinar-to-ductal metaplasia, a process that involves transdifferentiation of adult acinar cells into ductal-like cells due to K-Ras oncogene expression. These findings place telomeres as important players in cellular plasticity both during in vivo reprogramming and in pathological conditions associated with increased plasticity, such as cancer.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cellular Reprogramming/genetics , Telomere/genetics , Animals , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Cell Dedifferentiation/genetics , Cell Transformation, Neoplastic/metabolism , Chromatin Assembly and Disassembly/genetics , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/genetics , Gene Expression Regulation , Heterochromatin/genetics , Heterochromatin/metabolism , Mice , Mice, Transgenic , Protein Subunits/genetics , Stem Cells/cytology , Stem Cells/metabolism , Telomerase/metabolism , Telomere/metabolism , Telomere Homeostasis , Telomeric Repeat Binding Protein 1/genetics , Telomeric Repeat Binding Protein 1/metabolism , Cohesins
5.
Nat Commun ; 7: 12534, 2016 08 17.
Article in English | MEDLINE | ID: mdl-27531349

ABSTRACT

Telomeres are transcribed generating long non-coding RNAs known as TERRA. Deciphering the role of TERRA has been one of the unsolved issues of telomere biology in the past decade. This has been, in part, due to lack of knowledge on the TERRA loci, thus preventing functional genetic studies. Here, we describe that long non-coding RNAs with TERRA features are transcribed from the human 20q and Xp subtelomeres. Deletion of the 20q locus by using the CRISPR-Cas9 technology causes a dramatic decrease in TERRA levels, while deletion of the Xp locus does not result in decreased TERRA levels. Strikingly, 20q-TERRA ablation leads to dramatic loss of telomere sequences and the induction of a massive DNA damage response. These findings identify chromosome 20q as a main TERRA locus in human cells and represent the first demonstration in any organism of the essential role of TERRA in the maintenance of telomeres.


Subject(s)
RNA/metabolism , Telomere Homeostasis , Telomere/metabolism , Base Sequence , CRISPR-Cas Systems/genetics , Cell Line , Chromosomes, Human, Pair 20/genetics , Chromosomes, Human, X/genetics , Genetic Loci , Genotype , Humans , RNA, Long Noncoding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Deletion
6.
Nat Commun ; 5: 4723, 2014 Sep 03.
Article in English | MEDLINE | ID: mdl-25182072

ABSTRACT

Telomeric RNAs (TERRAs) are UUAGGG repeat-containing RNAs that are transcribed from the subtelomere towards the telomere. The precise genomic origin of TERRA has remained elusive. Using a whole-genome RNA-sequencing approach, we identify novel mouse transcripts arising mainly from the subtelomere of chromosome 18, and to a lesser extend chromosome 9, that resemble TERRA in several key aspects. Those transcripts contain UUAGGG-repeats and are heterogeneous in size, fluctuate in abundance in a TERRA-like manner during the cell cycle, are bound by TERRA RNA-binding proteins and are regulated in a manner similar to TERRA in response to stress and the induction of pluripotency. These transcripts are also found to associate with nearly all chromosome ends and downregulation of the transcripts that originate from chromosome 18 causes a reduction in TERRA abundance. Interestingly, downregulation of either chromosome 18 transcripts or TERRA results in increased number of telomere dysfunction-induced foci, suggesting a protective role at telomeres.


Subject(s)
Chromosomes, Mammalian/chemistry , Genome , RNA, Messenger/chemistry , RNA-Binding Proteins/metabolism , Telomere/chemistry , Transcription Factors/metabolism , Animals , Cell Cycle/genetics , Chromosomes, Mammalian/metabolism , Embryo, Mammalian , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression , Genes, Reporter , Genetic Loci , High-Throughput Nucleotide Sequencing , In Situ Hybridization, Fluorescence , Luciferases/genetics , Luciferases/metabolism , Mice , Primary Cell Culture , Protein Binding , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Repetitive Sequences, Nucleic Acid , Telomere/metabolism , Transcription Factors/genetics
7.
Nat Commun ; 5: 4190, 2014 Jun 27.
Article in English | MEDLINE | ID: mdl-24969639

ABSTRACT

HuR promotes myogenesis by stabilizing the MyoD, myogenin and p21 mRNAs during the fusion of muscle cells to form myotubes. Here we show that HuR, via a novel mRNA destabilizing activity, promotes the early steps of myogenesis by reducing the expression of the cell cycle promoter nucleophosmin (NPM). Depletion of HuR stabilizes the NPM mRNA, increases NPM protein levels and inhibits myogenesis, while its overexpression elicits the opposite effects. NPM mRNA destabilization involves the association of HuR with the decay factor KSRP as well as the ribonuclease PARN and the exosome. The C terminus of HuR mediates the formation of the HuR-KSRP complex and is sufficient for maintaining a low level of the NPM mRNA as well as promoting the commitment of muscle cells to myogenesis. We therefore propose a model whereby the downregulation of the NPM mRNA, mediated by HuR, KSRP and its associated ribonucleases, is required for proper myogenesis.


Subject(s)
ELAV Proteins/metabolism , Gene Expression Regulation, Developmental/genetics , Muscle Development/genetics , Muscle Fibers, Skeletal/metabolism , Nuclear Proteins/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Trans-Activators/metabolism , Animals , Humans , Mice , MyoD Protein/genetics , Myogenin/genetics , Nuclear Proteins/metabolism , Nucleophosmin , Promoter Regions, Genetic , RNA Stability
8.
Nat Commun ; 1: 33, 2010 Jun 29.
Article in English | MEDLINE | ID: mdl-20975687

ABSTRACT

Telomeres are transcribed from the telomeric C-rich strand, giving rise to UUAGGG repeat-containing telomeric transcripts or TERRA, which are novel structural components of telomeres. TERRA abundance is highly dependent on developmental status (including nuclear reprogramming), telomere length, cellular stresses, tumour stage and chromatin structure. However, the molecular mechanisms and factors controlling TERRA levels are still largely unknown. In this study, we identify a set of RNA-binding proteins, which endogenously bind and regulate TERRA in the context of primary mouse embryonic fibroblasts. The identification was carried out by biotin pull-down assays followed by LC-MALDI TOF/TOF mass spectrometry. Different members of the heterogeneous nuclear ribonucleoprotein family are among the ribonucleoprotein family that bind more abundantly to TERRA. Downregulation of TERRA-bound RBPs by small interfering RNA further shows that they can impact on TERRA abundance, their location and telomere lengthening. These findings anticipate an impact of TERRA-associated RBPs on telomere biology and telomeres diseases, such as cancer and aging.


Subject(s)
RNA-Binding Proteins/metabolism , Telomere/metabolism , Animals , Blotting, Northern , Blotting, Western , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Fluorescent Antibody Technique , Humans , Mice , RNA, Small Interfering/genetics , RNA-Binding Proteins/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Telomere/genetics
9.
Proc Natl Acad Sci U S A ; 106(46): 19393-8, 2009 Nov 17.
Article in English | MEDLINE | ID: mdl-19887628

ABSTRACT

Telomeres are heterochromatic structures at chromosome ends essential for chromosomal stability. Telomere shortening and the accumulation of dysfunctional telomeres are associated with organismal aging. Using telomerase-deficient TRF2-overexpressing mice (K5TRF2/Terc(-/-)) as a model for accelerated aging, we show that telomere shortening is paralleled by a gradual deregulation of the mammalian transcriptome leading to cumulative changes in a defined set of genes, including up-regulation of the mTOR and Akt survival pathways and down-regulation of cell cycle and DNA repair pathways. Increased DNA damage from dysfunctional telomeres leads to reduced deposition of H3K27me3 onto the inactive X chromosome (Xi), impaired association of the Xi with telomeric transcript accumulations (Tacs), and reactivation of an X chromosome-linked K5TRF2 transgene that is subjected to X-chromosome inactivation in female mice with sufficiently long telomeres. Exogenously induced DNA damage also disrupts Xi-Tacs, suggesting DNA damage at the origin of these alterations. Collectively, these findings suggest that critically short telomeres activate a persistent DNA damage response that alters gene expression programs in a nonstochastic manner toward cell cycle arrest and activation of survival pathways, as well as impacts the maintenance of epigenetic memory and nuclear organization, thereby contributing to organismal aging.


Subject(s)
Aging, Premature/genetics , DNA Damage/genetics , Skin/metabolism , Telomere/metabolism , X Chromosome Inactivation , Aging, Premature/metabolism , Aging, Premature/pathology , Animals , Cell Cycle/genetics , Female , Gene Expression Profiling , Keratin-15 , Keratin-5/genetics , Male , Mice , Mice, Transgenic , Skin/pathology , Telomerase/genetics , Telomerase/metabolism , Telomeric Repeat Binding Protein 2/genetics , Telomeric Repeat Binding Protein 2/metabolism , Transcription, Genetic
10.
Mol Cell Biol ; 29(16): 4341-51, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19528229

ABSTRACT

Activation of p38 mitogen-activated protein kinase (MAPK) plays an important role in the G(2)/M cell cycle arrest induced by DNA damage, but little is known about the role of this signaling pathway in the G(1)/S transition. Upregulation of the cyclin-dependent kinase inhibitor p21(Cip1) is thought to make a major contribution to the G(1)/S cell cycle arrest induced by gamma radiation. We show here that inhibition of p38 MAPK impairs p21(Cip1) accumulation and, as a result, the ability of cells to arrest in G(1) in response to gamma radiation. We found that p38 MAPK induces p21(Cip1) mRNA stabilization, without affecting its transcription or the stability of the protein. In particular, p38 MAPK phosphorylates the mRNA binding protein HuR on Thr118, which results in cytoplasmic accumulation of HuR and its enhanced binding to the p21(Cip1) mRNA. Our findings help to understand the emerging role of p38 MAPK in the cellular responses to DNA damage and reveal the existence of p53-independent networks that cooperate in modulating p21(Cip1) levels at the G(1)/S checkpoint.


Subject(s)
Antigens, Surface/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , G1 Phase/physiology , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , S Phase/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Antigens, Surface/genetics , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line/physiology , Cell Line/radiation effects , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA Damage , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , ELAV Proteins , ELAV-Like Protein 1 , Enzyme Activation , Gamma Rays , Humans , Mice , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Stability , RNA, Messenger/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA-Binding Proteins/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/genetics
11.
Nucleic Acids Res ; 37(8): 2658-71, 2009 May.
Article in English | MEDLINE | ID: mdl-19270063

ABSTRACT

The molecular basis underlying the aberrant DNA-methylation patterns in human cancer is largely unknown. Altered DNA methyltransferase (DNMT) activity is believed to contribute, as DNMT expression levels increase during tumorigenesis. Here, we present evidence that the expression of DNMT3b is post-transcriptionally regulated by HuR, an RNA-binding protein that stabilizes and/or modulates the translation of target mRNAs. The presence of a putative HuR-recognition motif in the DNMT3b 3'UTR prompted studies to investigate if this transcript associated with HuR. The interaction between HuR and DNMT3b mRNA was studied by immunoprecipitation of endogenous HuR ribonucleoprotein complexes followed by RT-qPCR detection of DNMT3b mRNA, and by in vitro pulldown of biotinylated DNMT3b RNAs followed by western blotting detection of HuR. These studies revealed that binding of HuR stabilized the DNMT3b mRNA and increased DNMT3b expression. Unexpectedly, cisplatin treatment triggered the dissociation of the [HuR-DNMT3b mRNA] complex, in turn promoting DNMT3b mRNA decay, decreasing DNMT3b abundance, and lowering the methylation of repeated sequences and global DNA methylation. In summary, our data identify DNMT3b mRNA as a novel HuR target, present evidence that HuR affects DNMT3b expression levels post-transcriptionally, and reveal the functional consequences of the HuR-regulated DNMT3b upon DNA methylation patterns.


Subject(s)
Antigens, Surface/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation , RNA Stability , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Antigens, Surface/analysis , Antineoplastic Agents/pharmacology , Base Sequence , Cell Line, Tumor , Cisplatin/pharmacology , DNA (Cytosine-5-)-Methyltransferases/metabolism , ELAV Proteins , ELAV-Like Protein 1 , Humans , Molecular Sequence Data , RNA-Binding Proteins/analysis , DNA Methyltransferase 3B
12.
Cancer Res ; 68(11): 4116-22, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18519670

ABSTRACT

An undifferentiated status and the epigenetic inactivation of tumor-suppressor genes are hallmarks of transformed cells. Promoter CpG island hypermethylation of differentiating genes, however, has rarely been reported. The Groucho homologue Transducin-like Enhancer of Split 1 (TLE1) is a multitasked transcriptional corepressor that acts through the acute myelogenous leukemia 1, Wnt, and Notch signaling pathways. We have found that TLE1 undergoes promoter CpG island hypermethylation-associated inactivation in hematologic malignancies, such as diffuse large B-cell lymphoma and AML. We also observed a mutual exclusivity of the epigenetic alteration of TLE1 and the cytogenetic alteration of AML1. TLE1 reintroduction in hypermethylated leukemia/lymphoma cells causes growth inhibition in colony assays and nude mice, whereas TLE1-short hairpin RNA depletion in unmethylated cells enhances tumor growth. We also show that these effects are mediated by TLE1 transcriptional repressor activity on its target genes, such as Cyclin D1, Colony-Stimulating Factor 1 receptor, and Hairy/Enhancer of Split 1. These data suggest that TLE1 epigenetic inactivation contributes to the development of hematologic malignancies by disrupting critical differentiation and growth-suppressing pathways.


Subject(s)
Epigenesis, Genetic , Hematologic Neoplasms/genetics , Repressor Proteins/genetics , Animals , Base Sequence , Cell Line, Tumor , Co-Repressor Proteins , CpG Islands , DNA Methylation , DNA Primers , Humans , Mice , Mice, Nude , Reverse Transcriptase Polymerase Chain Reaction
13.
Biochem J ; 409(1): 311-20, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17760565

ABSTRACT

Butyrate has antitumorigenic effects on colon cancer cells, inhibits cell growth and promotes differentiation and apoptosis. These effects depend on its intracellular concentration, which is regulated by its transport. We have analysed butyrate uptake kinetics in human colon adenocarcinoma cells sensitive to the apoptotic effects of butyrate (BCS-TC2, Caco-2 and HT-29), in butyrate-resistant cells (BCS-TC2.BR2) and in normal colonic cells (FHC). The properties of transport were analysed with structural analogues, specific inhibitors and different bicarbonate and sodium concentrations. Two carrier-mediated mechanisms were detected: a low-affinity/high-capacity (K(m)=109+/-16 mM in BCS-TC2 cells) anion exchanger and a high-affinity/low-capacity (K(m)=17.9+/-4.0 microM in BCS-TC2 cells) proton-monocarboxylate co-transporter that was energy-dependent and activated via PKCdelta (protein kinase Cdelta). All adenocarcinoma cells analysed express MCT (monocarboxylate transporter) 1, MCT4, ancillary protein CD147 and AE2 (anion exchanger 2). Silencing experiments show that MCT1, whose expression increases with butyrate treatment in butyrate-sensitive cells, plays a key role in high-affinity transport. Low-affinity uptake was mediated by a butyrate/bicarbonate antiporter along with a possible contribution of AE2 and MCT4. Butyrate treatment increased uptake in a time- and dose-dependent manner in butyrate-sensitive but not in butyrate-resistant cells. The two butyrate-uptake activities in human colon adenocarcinoma cells enable butyrate transport at different physiological conditions to maintain cell functionality. The high-affinity/low-capacity transport functions under low butyrate concentrations and may be relevant for the survival of carcinoma cells in tumour regions with low glucose and butyrate availability as well as for the normal physiology of colonocytes.


Subject(s)
Adenocarcinoma/metabolism , Butyrates/metabolism , Colonic Neoplasms/metabolism , Anion Transport Proteins/biosynthesis , Anions , Antiporters/biosynthesis , Basigin/biosynthesis , Biological Transport , Butyrates/pharmacokinetics , Cell Cycle Proteins/biosynthesis , Cell Line, Tumor , DNA Primers/chemistry , Glucose/metabolism , Humans , Kinetics , Oncogene Proteins/biosynthesis , Protein Kinase C/metabolism , RNA, Small Interfering/metabolism , SLC4A Proteins
14.
Cell Oncol ; 29(1): 1-17, 2007.
Article in English | MEDLINE | ID: mdl-17429137

ABSTRACT

The messenger RNA 3'-untranslated region (3'UTR) is emerging as critically important in regulating gene expression at posttranscriptional levels. The 3'UTR governs gene expression via orchestrated interactions between mRNA structural components (cis-elements) and specific trans-acting factors (RNA-binding proteins and non-coding RNAs). Alterations in any of these components can lead to disease. Here, we review the mutations in 3'UTR regulatory sequences as well as the aberrant levels, subcellular localization, and posttranslational modifications of trans-acting factors that can promote or enhance the malignant phenotype of cancer cells. A thorough understanding of these alterations and their impact upon 3'UTR-directed posttranscriptional gene regulation will uncover promising new targets for therapeutic intervention.


Subject(s)
3' Untranslated Regions/genetics , Gene Expression Regulation, Neoplastic , Neoplasms/genetics , RNA, Messenger/genetics , 3' Untranslated Regions/metabolism , Humans , Models, Biological , Mutation , Neoplasms/metabolism , Neoplasms/pathology , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism
15.
Carcinogenesis ; 28(9): 1877-84, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17449905

ABSTRACT

The Wnt-beta-catenin pathway is aberrantly activated in most colon cancers. DICKKOPF-1 (DKK-1) gene encodes an extracellular Wnt inhibitor that blocks the formation of signalling receptor complexes at the plasma membrane. We report that 1alpha,25-dihydroxyvitamin D3 [1,25(OH)2D3], the most active vitamin D metabolite, increases the level of DKK-1 RNA and protein in human SW480-ADH colon cancer cells. This effect is dose dependent, slow and depends on the presence of a transcription-competent nuclear vitamin D receptor (VDR). Accordingly, 1,25(OH)2D3 activates a 2300 bp fragment of the human DKK-1 gene promoter. Chromatin immunoprecipitation assays revealed that 1,25(OH)2D3 treatment induced a pattern of histone modifications which is compatible with transcriptionally active chromatin. DKK-1 is expressed at high level in colon cancer cell lines with a differentiated phenotype such as Caco-2 or HT-29. Exogenous expression of E-cadherin into SW480-ADH cells results in a strong adhesive phenotype and a 17-fold increase in DKK-1 RNA. In contrast, an E-cadherin blocking antibody inhibits 1,25(OH)2D3-induced differentiation of SW480-ADH cells and DKK-1 gene expression. Remarkably, in vivo treatment with the vitamin D analogue EB1089 induced DKK-1 protein expression in SW480-ADH cells xenografted in immunodeficient mice, and a correlation was observed in the expression of VDR and DKK-1 RNA in a series of 32 human colorectal tumours. These data indicate that 1,25(OH)2D3 activates the transcription of the DKK-1 gene, probably in an indirect way that is associated to the promotion of a differentiated phenotype. DKK-1 gene induction constitutes a novel mechanism of inhibition of Wnt signalling and antitumour action by 1,25(OH)2D3.


Subject(s)
Calcitriol/pharmacology , Cell Differentiation/physiology , Colonic Neoplasms/genetics , Intercellular Signaling Peptides and Proteins/genetics , Wnt Proteins/antagonists & inhibitors , Cell Differentiation/drug effects , Cell Line, Tumor , Chromatin/genetics , Chromatin/isolation & purification , DNA Primers , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunohistochemistry , Polymerase Chain Reaction , RNA, Neoplasm/genetics , Transcriptional Activation
16.
An. R. Acad. Farm ; 72(4): 563-581, oct. 2006. ilus, tab
Article in En | IBECS | ID: ibc-051852

ABSTRACT

La estabilidad del ARN mensajero está surgiendo como instrumento celular fundamental y efectivo para regular la expresión génica a nivel post-transcripcional. La estabilidad del ARNm se controla vía interacciones coordinadas entre componentes estructurales del ARNm (elementos cis) y factores trans específicos. Los determinantes de estabilidad de ARNm más conocidos y eficientes son los elementos ricos en adenina y uridina (ARE) que, a través de su unión con proteínas de unión a ARE (AUBPS), modulan la estabilidad de los transcritos y/o su traducción. Alteraciones en cualquiera de estos componentes puede dar lugar a enfermedades. Aquí revisamos las alteraciones genéticas en elementos regulatorios del 3’UTR, así como las aberraciones en los niveles, localización subcelular y modificaciones posttraslacionales de AUBPs que están asociadas a enfermedades humanas. Un conocimiento detallado de estas alteraciones y su impacto en la regulación de la estabilidad del ARNm revelará nuevas dianas para su aplicación terapéutica


mRNA stability is emerging as a fundamental and effective cellular tool to regulate gene expression at posttranscriptional levels. mRNA stability is controlled via orchestrated interactions between mRNA structural components (cis-elements) and specific trans-acting factors. The most widespread and efficient determinant of RNA stability are the adenylate and uridylate-rich elements (ARE) that, through binding of ARE-binding proteins (AUBPs), modulate the stability of transcripts and/or their translation. Alterations in any of these components can lead to disease. Here, we review the genetic alterations in 3’UTR regulatory sequences as well as the aberrant levels, subcellular localization, and posttranslational modifications of AUBPs that are linked to human diseases. A thorough understanding of these alterations and their impact on mRNA stability regulation will uncover promising new targets for therapeutic intervention


Subject(s)
Humans , Gene Expression Regulation , RNA Stability/genetics , RNA, Messenger/genetics , Alzheimer Disease/genetics , Inflammation/genetics , Thalassemia/genetics , Neoplasms/genetics , Transcription, Genetic , Uridine/genetics , Adenine , Protein Binding/genetics
17.
Dev Growth Differ ; 48(6): 381-90, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16872451

ABSTRACT

In our earlier attempt to identify genes involved in the maintenance of cellular pluripotency, we found that KH-domain protein Embryonal stem cell-specific gene 1 (Esg1) showed similar expression patterns to those of Oct3/4 (Pou5f1), whereas the forced repression of Oct3/4 in mouse embryonic stem cells immediately downregulated the expression of Esg1. Here we further confirm this overlap by in situ hybridization and immunohistochemical analyses. Both Esg1 transcript and protein exist in the egg and preimplantation embryos. At embryonic day 3.5, blastocyst stage, however, ESG1 protein was more abundant in the inner cell mass (ICM) than in trophectoderm (TE), whereas Esg1 transcript was detected in both the ICM and the TE, particularly in the polar trophectoderm. The presence of an RNA-binding KH-domain in ESG1 led us to search for and identify 902 target transcripts by microarray analysis of immunoprecipitated ESG1 complex. Interaction of 20 target mRNA with ESG1, including Cdc25a, Cdc42, Ezh2, Nfyc and Nr5a2, was further validated by reverse transcriptase-polymerase chain reaction of the immunoprecipitation material, supporting the notion that ESG1 is an RNA-binding protein which associates with specific target transcripts.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Blastocyst/metabolism , RNA-Binding Proteins/genetics , RNA/metabolism , Stem Cells/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/analysis , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blastocyst/chemistry , Blotting, Northern , Cell Proliferation , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental/genetics , Immunoblotting , Immunohistochemistry , In Situ Hybridization/methods , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Models, Biological , Morula/chemistry , Morula/cytology , Morula/metabolism , Octamer Transcription Factor-3/analysis , Octamer Transcription Factor-3/genetics , Oligonucleotide Array Sequence Analysis/methods , RNA/genetics , RNA-Binding Proteins/analysis , RNA-Binding Proteins/metabolism , Transcription Factors/analysis , Transcription Factors/genetics , Transcription Factors/metabolism
18.
Mol Cell Biol ; 25(21): 9520-31, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16227602

ABSTRACT

The RNA-binding protein TIA-1 (T-cell intracellular antigen 1) functions as a posttranscriptional regulator of gene expression and aggregates to form stress granules following cellular damage. TIA-1 was previously shown to bind mRNAs encoding tumor necrosis factor alpha (TNF-alpha) and cyclooxygenase 2 (COX-2), but TIA-1 target mRNAs have not been systematically identified. Here, immunoprecipitation (IP) of TIA-1-RNA complexes, followed by microarray-based identification and computational analysis of bound transcripts, was used to elucidate a common motif present among TIA-1 target mRNAs. The predicted TIA-1 motif was a U-rich, 30- to 37-nucleotide (nt)-long bipartite element forming loops of variable size and a bent stem. The TIA-1 motif was found in the TNF-alpha and COX-2 mRNAs and in 3,019 additional UniGene transcripts (approximately 3% of the UniGene database), localizing preferentially to the 3' untranslated region. The interactions between TIA-1 and target transcripts were validated by IP of endogenous mRNAs, followed by reverse transcription and PCR-mediated detection, and by pulldown of biotinylated RNAs, followed by Western blotting. Further studies using RNA interference revealed that TIA-1 repressed the translation of bound mRNAs. In summary, we report a signature motif present in mRNAs that associate with TIA-1 and provide support to the notion that TIA-1 represses the translation of target transcripts.


Subject(s)
RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Base Sequence , Cell Line, Tumor , Computational Biology , Cyclooxygenase 2/genetics , Humans , Immunoprecipitation , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , RNA-Binding Proteins/metabolism , Tumor Necrosis Factor-alpha/genetics
19.
J Biol Chem ; 280(24): 22819-26, 2005 Jun 17.
Article in English | MEDLINE | ID: mdl-15824116

ABSTRACT

In dividing cells, the RNA-binding protein HuR associates with and stabilizes labile mRNAs encoding proliferative proteins, events that are linked to the increased cytoplasmic presence of HuR. Here, assessment of HuR levels in various vascular pathologies (intimal hyperplasia, atherosclerosis and neointimal proliferation, sclerosis of arterialized saphenous venous graft, and fibromuscular dysplasia) revealed a distinct increase in HuR expression and cytoplasmic abundance within the intima and neointima layers. On the basis of these observations, we postulated a role for HuR in promoting the proliferation of vascular smooth muscle cells. To test this hypothesis directly, we investigated the expression, subcellular localization, and proliferative influence of HuR in human vascular smooth muscle cells (hVSMCs). Treatment of hVSMCs with platelet-derived growth factor increased HuR levels in the cytoplasm, thereby influencing the expression of metabolic, proliferative, and structural genes. Importantly, knockdown of HuR expression by using RNA interference caused a reduction of hVSMC proliferation, both basally and following platelet-derived growth factor treatment. We propose that HuR contributes to regulating hVSMC growth and homeostasis in pathologies associated with vascular smooth muscle proliferation.


Subject(s)
Antigens, Surface/chemistry , Gene Expression Regulation , Muscle, Smooth, Vascular/cytology , RNA-Binding Proteins/chemistry , RNA/chemistry , Biotinylation , Blotting, Western , Cell Proliferation , Cells, Cultured , Cytoplasm/metabolism , DNA, Complementary/metabolism , ELAV Proteins , ELAV-Like Protein 1 , Extracellular Matrix/metabolism , Humans , Immunohistochemistry , Microscopy, Fluorescence , Muscle, Smooth/metabolism , Oligonucleotide Array Sequence Analysis , Platelet-Derived Growth Factor/metabolism , Protein Binding , RNA Interference , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transfection
20.
RNA Biol ; 2(1): 11-3, 2005 Jan.
Article in English | MEDLINE | ID: mdl-17132932

ABSTRACT

The RNA-binding protein HuR regulates the stability and translation of target mRNAs. While no HuR mutations have been found in cancer, a link between HuR and malignant transformation has been suggested in cancers of the breast, colon, lung and ovary. We describe a paradigm consistent with a central role of HuR in oncogenesis.


Subject(s)
Antigens, Surface/metabolism , Cell Transformation, Neoplastic , RNA Processing, Post-Transcriptional , RNA-Binding Proteins/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Chromosomes, Human, Pair 19 , Colonic Neoplasms/metabolism , Cytoplasm/metabolism , ELAV Proteins , ELAV-Like Protein 1 , Humans , Mice , Mice, Nude , Protein Biosynthesis , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...