Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Front Mol Biosci ; 8: 696438, 2021.
Article in English | MEDLINE | ID: mdl-34395528

ABSTRACT

RNA polymerase (Pol) III transcribes small untranslated RNAs such as 5S ribosomal RNA, transfer RNAs, and U6 small nuclear RNA. Because of the functions of these RNAs, Pol III transcription is best known for its essential contribution to RNA maturation and translation. Surprisingly, it was discovered in the last decade that various inherited mutations in genes encoding nine distinct subunits of Pol III cause tissue-specific diseases rather than a general failure of all vital functions. Mutations in the POLR3A, POLR3C, POLR3E and POLR3F subunits are associated with susceptibility to varicella zoster virus-induced encephalitis and pneumonitis. In addition, an ever-increasing number of distinct mutations in the POLR3A, POLR3B, POLR1C and POLR3K subunits cause a spectrum of neurodegenerative diseases, which includes most notably hypomyelinating leukodystrophy. Furthermore, other rare diseases are also associated with mutations in genes encoding subunits of Pol III (POLR3H, POLR3GL) and the BRF1 component of the TFIIIB transcription initiation factor. Although the causal relationship between these mutations and disease development is widely accepted, the exact molecular mechanisms underlying disease pathogenesis remain enigmatic. Here, we review the current knowledge on the functional impact of specific mutations, possible Pol III-related disease-causing mechanisms, and animal models that may help to better understand the links between Pol III mutations and disease.

2.
Neuro Oncol ; 22(4): 550-562, 2020 04 15.
Article in English | MEDLINE | ID: mdl-31711240

ABSTRACT

BACKGROUND: Diffuse midline glioma (DMG) is a pediatric malignancy with poor prognosis. Most children die less than one year after diagnosis. Recently, mutations in histone H3 have been identified and are believed to be oncogenic drivers. Targeting this epigenetic abnormality using histone deacetylase (HDAC) inhibitors such as panobinostat (PS) is therefore a novel therapeutic option currently evaluated in clinical trials. METHODS: BH3 profiling revealed engagement in an irreversible apoptotic process of glioma cells exposed to PS confirmed by annexin-V/propidium iodide staining. Using proteomic analysis of 3 DMG cell lines, we identified 2 proteins deregulated after PS treatment. We investigated biological effects of their downregulation by silencing RNA but also combinatory effects with PS treatment in vitro and in vivo using a chick embryo DMG model. Electron microscopy was used to validate protein localization. RESULTS: Scaffolding proteins EBP50 and IRSp53 were upregulated by PS treatment. Reduction of these proteins in DMG cell lines leads to blockade of proliferation and migration, invasion, and an increase of apoptosis. EBP50 was found to be expressed in cytoplasm and nucleus in DMG cells, confirming known oncogenic locations of the protein. Treatment of glioma cells with PS together with genetic or chemical inhibition of EBP50 leads to more effective reduction of cell growth in vitro and in vivo. CONCLUSION: Our data reveal a specific relation between HDAC inhibitors and scaffolding protein deregulation which might have a potential for therapeutic intervention for cancer treatment.


Subject(s)
Glioma , Histone Deacetylases , Animals , Apoptosis , Cell Line, Tumor , Chick Embryo , Child , Glioma/drug therapy , Glioma/genetics , Histone Deacetylase Inhibitors/pharmacology , Histones , Humans , Panobinostat , Proteomics
3.
Oncotarget ; 8(25): 41211-41226, 2017 Jun 20.
Article in English | MEDLINE | ID: mdl-28476031

ABSTRACT

Glypican-3 (GPC3) is an oncogene, frequently upregulated in liver malignancies such as hepatocellular carcinoma (HCC) and hepatoblastoma and constitutes a potential molecular target for therapy in liver cancer. Using a functional screening system, we identified 10 new microRNAs controlling GPC3 expression in malignant liver cells, five of them e.g. miR-4510, miR-203a-3p, miR-548aa, miR-376b-3p and miR-548v reduce GPC3 expression. These 5 microRNAs were significantly downregulated in tumoral compared to non-tumoral liver and inhibited tumor cell proliferation. Interestingly, miR-4510 inversely correlated with GPC3 mRNA and protein in HCC samples. This microRNA also induced apoptosis of hepatoma cells and blocked tumor growth in vivo in the chick chorioallantoic membrane model. We further show that the tumor suppressive effect of miR-4510 is mediated through direct targeting of GPC3 mRNA and inactivation of Wnt/ß-catenin transcriptional activity and signaling pathway. Moreover, miR-4510 up-regulated the expression of several tumor suppressor genes while reducing the expression of other pro-oncogenes. In summary, we uncovered several new microRNAs targeting the oncogenic functions of GPC3. We provided strong molecular, cellular and in vivo evidences for the tumor suppressive activities of miR-4510 bringing to the fore the potential value of this microRNA in HCC therapy.


Subject(s)
Carcinoma, Hepatocellular/genetics , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Glypicans/genetics , Liver Neoplasms/genetics , MicroRNAs/genetics , Adolescent , Adult , Aged , Apoptosis/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Female , Glypicans/metabolism , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Middle Aged , Young Adult
4.
Hepatol Commun ; 1(2): 168-183, 2017 04.
Article in English | MEDLINE | ID: mdl-29404451

ABSTRACT

Hepatoblastoma (HBL) is the most common pediatric liver cancer. In this malignant neoplasm, beta-catenin protein accumulates and increases Wnt signaling due to recurrent activating mutations in the catenin-beta 1 (CTNNB1) gene. Therefore, beta-catenin is a key therapeutic target in HBL. However, controlling beta-catenin production with therapeutic molecules has been challenging. New biological studies could provide alternative therapeutic solutions for the treatment of HBL, especially for advanced tumors and metastatic disease. In this study, we identified microRNAs (miRNAs) that target beta-catenin and block HBL cell proliferation in vitro and tumor growth in vivo. Using our dual-fluorescence-FunREG system, we screened a library of 1,712 miRNA mimics and selected candidates inhibiting CTNNB1 expression through interaction with its untranslated regions. After validating the regulatory effect of nine miRNAs on beta-catenin in HBL cells, we measured their expression in patient samples. Let-7i-3p, miR-449b-3p, miR-624-5p, and miR-885-5p were decreased in tumors compared to normal livers. Moreover, they inhibited HBL cell growth and Wnt signaling activity in vitro partly through beta-catenin down-regulation. Additionally, miR-624-5p induced cell senescence in vitro, blocked experimental HBL growth in vivo, and directly targeted the beta-catenin 3'-untranslated region. Conclusion: Our results shed light on how beta-catenin-regulating miRNAs control HBL progression through Wnt signaling inactivation. In particular, miR-624-5p may constitute a promising candidate for miRNA replacement therapy for HBL patients. (Hepatology Communications 2017;1:168-183).

5.
Hepatology ; 57(1): 195-204, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22865282

ABSTRACT

UNLABELLED: Hepatocellular carcinoma (HCC) is the major primary liver cancer. Glypican-3 (GPC3), one of the most abnormally expressed genes in HCC, participates in liver carcinogenesis. Based on data showing that GPC3 expression is posttranscriptionally altered in HCC cells compared to primary hepatocytes, we investigated the implication of microRNAs (miRNAs) in GPC3 overexpression and HCC. To identify GPC3-regulating miRNAs, we developed a dual-fluorescence FunREG (functional, integrated, and quantitative method to measure posttranscriptional regulations) system that allowed us to screen a library of 876 individual miRNAs. Expression of candidate miRNAs and that of GPC3 messenger RNA (mRNA) was measured in 21 nontumoral liver and 112 HCC samples. We then characterized the phenotypic consequences of modulating expression of one candidate miRNA in HuH7 cells and deciphered the molecular mechanism by which this miRNA controls the posttranscriptional regulation of GPC3. We identified five miRNAs targeting GPC3 3'-untranslated region (UTR) and regulating its expression about the 876 tested. Whereas miR-96 and its paralog miR-1271 repressed GPC3 expression, miR-129-1-3p, miR-1291, and miR-1303 had an inducible effect. We report that miR-1271 expression is down-regulated in HCC tumor samples and inversely correlates with GPC3 mRNA expression in a particular subgroup of HCC. We also report that miR-1271 inhibits the growth of HCC cells in a GPC3-dependent manner and induces cell death. CONCLUSION: Using a functional screen, we found that miR-96, miR-129-1-3p, miR-1271, miR-1291, and miR-1303 differentially control GPC3 expression in HCC cells. In a subgroup of HCC, the up-regulation of GPC3 was associated with a concomitant down-regulation of its repressor miR-1271. Therefore, we propose that GPC3 overexpression and its associated oncogenic effects are linked to the down-regulation of miR-1271 in HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Gene Expression Regulation, Neoplastic , Glypicans/metabolism , Liver Neoplasms/metabolism , MicroRNAs/metabolism , 3' Untranslated Regions , Cell Line, Tumor , Hepatocytes/metabolism , Humans , RNA Processing, Post-Transcriptional
6.
Nucleic Acids Res ; 40(3): 1356-65, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22009679

ABSTRACT

Besides the fact that miR-96 and miR-182 belong to the miR-182/183 cluster, their seed region (UUGGCA, nucleotides 2-7) is identical suggesting potential common properties in mRNA target recognition and cellular functions. Here, we used the mRNA encoding Glypican-3, a heparan-sulfate proteoglycan, as a model target as its short 3' untranslated region is predicted to contain one miR-96/182 site, and assessed whether it is post-transcriptionally regulated by these two microRNAs. We found that miR-96 downregulated GPC3 expression by targeting its mRNA 3'-untranslated region and interacting with the predicted site. This downregulatory effect was due to an increased mRNA degradation and depended on Argonaute-2. Despite its seed similarity with miR-96, miR-182 was unable to regulate GPC3. This differential regulation was confirmed on two other targets, FOXO1 and FN1. By site-directed mutagenesis, we demonstrated that the miRNA nucleotide 8, immediately downstream the UUGGCA seed, plays a critical role in target recognition by miR-96 and miR-182. Our data suggest that because of a base difference at miRNA position 8, these two microRNAs control a completely different set of genes and therefore are functionally independent.


Subject(s)
Gene Expression Regulation , Glypicans/genetics , MicroRNAs/chemistry , 3' Untranslated Regions , Base Pairing , Base Sequence , Cell Line, Tumor , Cells, Cultured , Glypicans/metabolism , Humans , MicroRNAs/physiology , Molecular Sequence Data
7.
Biochem Soc Trans ; 38(6): 1608-14, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21118135

ABSTRACT

An increasing number of arguments, including altered microRNA expression, support the idea that post-transcriptional deregulation participates in gene disturbances found in diseased tissues. To evaluate this hypothesis, we developed a method which facilitates post-transcriptional investigations in a wide range of human cells and experimental conditions. This method, called FunREG (functional, integrated and quantitative method to measure post-transcriptional regulation), connects lentiviral transduction with a fluorescent reporter system and quantitative PCR. Using FunREG, we efficiently measured post-transcriptional regulation mediated either by selected RNA sequences or regulatory factors (microRNAs), and then evaluated the contribution of mRNA decay and translation efficiency in the observed regulation. We demonstrated the existence of gene-specific post-transcriptional deregulation in liver tumour cells, and also reported a molecular link between a transcript variant abrogating HDAC6 (histone deacetylase 6) regulation by miR-433 and a rare familial genetic disease. Because FunREG is sensitive, quantitative and easy to use, many applications can be envisioned in fundamental and pathophysiological research.


Subject(s)
Gene Expression Regulation , Polymerase Chain Reaction/methods , RNA Processing, Post-Transcriptional , Base Sequence , Genes, Reporter , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , RNA Stability/genetics , Transgenes
8.
Mol Cell Proteomics ; 8(8): 1777-88, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19411282

ABSTRACT

In the past 10 years, transcriptome and proteome analyses have provided valuable data on global gene expression and cell functional networks. However, when integrated,these analyses revealed partial correlations between mRNA expression levels and protein abundance thus suggesting that post-transcriptional regulations may be in part responsible for this discrepancy. In the present work, we report the development of a functional, integrated, and quantitative method to measure post-transcriptional regulations that we named FunREG. This method enables (i) quantitative measure of post-transcriptional regulations mediated by selected 3-untranslated regions and exogenous small interfering-RNA or micro-RNAs and (ii) comparison of these regulatory processes in physiologically relevant systems (e.g. cancer versus primary untransformed cells). We applied FunREG to the study of liver cancer, and we demonstrate for the first time the differential regulatory mechanisms controlling gene expression at a post-transcriptional level in normal and tumoral hepatic cells. As an example, translation efficiency mediated by heparin-binding epidermal growth factor 3-untranslated region was increased 3-fold in liver cancer cells compared with normal hepatocytes, whereas stability of an mRNA containing a portion of Cyclin D1 3-untranslated region was increased more than 2-fold in HepG2 cells compared with normal hepatocytes. Consequently we believe that the method presented herein may become an important tool in fundamental and medical research. This approach is convenient and easy to perform, accessible to any investigator, and should be adaptable to a large number of cell type, functional and chemical screens, as well as genome scale analyses. Finally FunREG may represent a helpful tool to reconcile transcriptome and proteome data.


Subject(s)
Molecular Biology/methods , RNA Processing, Post-Transcriptional , Transgenes/genetics , 3' Untranslated Regions/genetics , Cell Line, Tumor , Cells, Cultured , Flow Cytometry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Lentivirus/genetics , MicroRNAs/genetics , RNA, Small Interfering/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection
9.
Biochem J ; 400(2): 337-47, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-16834569

ABSTRACT

The ARE (AU-rich element) is a post-transcriptional element controlling both mRNA turnover and translation initiation by primarily inducing poly(A) tail shortening. The mechanisms by which the ARE-associated proteins induce deadenylation are still obscure. One possibility among others would be that an ARE-ARE-BP (ARE-binding protein) complex intervenes in the PABP [poly(A)-binding protein]-poly(A) tail association and facilitates poly(A) tail accessibility to deadenylases. Here, we show by several experimental approaches that AUF1 (AU-rich element RNA-binding protein 1)/hnRNP (heterogeneous nuclear ribonucleoprotein) D, an mRNA-destabilizing ARE-BP, can bind poly(A) sequence in vitro. First, endogenous AUF1 proteins from HeLa cells specifically bound poly(A), independently of PABP. Secondly, using polyadenylated RNA probes, we showed that (i) the four recombinant AUF1 isoforms bind poly(A) as efficiently as PABP, (ii) the AUF1 binding to poly(A) does not change when the polyadenylated probe contains the GM-CSF (granulocyte/macrophage-colony stimulating factor) ARE, suggesting that, in vitro, the AUF1-poly(A) association was independent of the ARE sequence itself. In vitro, the binding of AUF1 isoforms to poly(A) displayed oligomeric and co-operative properties and AUF1 efficiently displaced PABP from the poly(A). Finally, the AUF1 molar concentration in HeLa cytoplasm was only 2-fold lower than that of PABP, whereas in the nucleus, its molar concentration was similar to that of PABP. These in vitro results suggest that, in vivo, AUF1 could compete with PABP for the binding to poly(A). Altogether, our results may suggest a role for AUF1 in controlling PABP-poly(A) tail association.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein D/metabolism , Poly A/metabolism , Poly(A)-Binding Proteins/metabolism , Binding, Competitive , Cell Nucleus/metabolism , Cytoplasm/metabolism , HeLa Cells , Heterogeneous Nuclear Ribonucleoprotein D0 , Heterogeneous-Nuclear Ribonucleoprotein D/genetics , Humans , Kinetics , Poly(A)-Binding Proteins/genetics , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity
10.
Proteomics ; 4(2): 364-73, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14760706

ABSTRACT

Using a proteomic approach based on the two-dimensional (2-D) gel analysis of synthesized proteins, we investigated the involvement of the Snf1 kinase pathway in the regulation of gene expression during the diauxic shift in Saccharomyces cerevisiae. For this purpose, we used a mutant strain deleted for SNF4, the gene coding for the activator subunit of Snf1p. The levels of synthesis of 82 spots were found to be affected by the absence of Snf4p at the diauxic shift. Half of the proteins which exhibit a reduced synthesis in the mutant strain are proteins whose genes are controlled by the transcriptional activator Cat8p, a target of Snf1p. Proteins with an increased level of synthesis in the mutant strain were also observed. Among them are glycolytic enzymes whose synthesis is strongly reduced when wild-type cells enter the diauxic shift. This observation suggests that Snf1p exerts a negative control on the expression of glycolytic genes during the diauxic transition. The results obtained in this study were compiled with those previously obtained by similar proteomic approach with other regulatory factors involved in the diauxic shift. This compilation illustrates how 2-D gel electrophoresis can be used to elucidate the network of regulators participating to complex biological process.


Subject(s)
Carrier Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proteome/analysis , Saccharomyces cerevisiae Proteins/analysis , Saccharomyces cerevisiae/metabolism , Transcription Factors/metabolism , AMP-Activated Protein Kinases , Electrophoresis, Gel, Two-Dimensional , Gene Expression Regulation, Fungal/physiology , Mutation , Proteome/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction/physiology , Trans-Activators/metabolism
11.
J Biol Chem ; 278(46): 45391-6, 2003 Nov 14.
Article in English | MEDLINE | ID: mdl-12960168

ABSTRACT

In Saccharomyces cerevisiae the transition between the fermentative and the oxidative metabolism, called the diauxic shift, is associated with major changes in gene expression. In this study, we characterized a novel family of five genes whose expression is induced during the diauxic shift. These genes, FET3, FTR1, TIS11, SIT1, and FIT2, are involved in the iron uptake pathway. We showed that their induction at the diauxic shift is positively controlled by the Snf1/Snf4 kinase pathway. The transcriptional factor Aft1p, which is known to control their induction in response to iron limitation, is also required for their induction during the diauxic shift. The increase of the extracellular iron concentration does not affect this induction, indicating that glucose exhaustion by itself would be the signal. The possibility that the Snf1/Snf4 pathway was also involved in the induction of the same set of genes in response to iron starvation was considered. We demonstrate here that this is not the case. Thus, the two signals, glucose exhaustion and iron starvation, use two independent pathways to activate the same set of genes through the Aft1p transcriptional factor.


Subject(s)
Ceruloplasmin/metabolism , Iron/metabolism , Protein Serine-Threonine Kinases/physiology , Saccharomyces cerevisiae/metabolism , Blotting, Northern , Cation Transport Proteins/genetics , Ceruloplasmin/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Fungal , Glucose/metabolism , Glycoproteins/genetics , Immunoblotting , Membrane Transport Proteins/genetics , Nucleic Acid Hybridization , Open Reading Frames , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction , Time Factors , Transcription Factors/metabolism , Transcription, Genetic , Transcriptional Activation , Tristetraprolin
SELECTION OF CITATIONS
SEARCH DETAIL
...