39 results on '"Polycarpou-Schwarz M"'
Search Results
2. RNA surveillance by ADAR1 is required for vascular endothelial resilience
- Author
-
Tual-Chalot, S, primary, Ciliberti, G, additional, Polycarpou-Schwarz, M, additional, Amponsah-Offeh, M, additional, Turchinovich, A, additional, Dimmeler, S, additional, Gatsiou, A, additional, and Stellos, K, additional
- Published
- 2023
- Full Text
- View/download PDF
3. Endogenous suppression of double-stranded RNA-induced MDA5 autoinflammatory signaling is essential for the maintenance of vascular integrity in adult mice
- Author
-
Ciliberti, G., primary, Tual-Chalot, S., additional, Polycarpou-Schwarz, M., additional, Amponsah-Offeh, M., additional, Stellos, K., additional, Gatsiou, A., additional, Dimmeler, S., additional, and Bonini, F., additional
- Published
- 2023
- Full Text
- View/download PDF
4. Intronic ALU RNA editing couples mRNA stability with pre-mRNA processing of inflammatory gene expression in atherosclerosis
- Author
-
Sachse, M., primary, Polycarpou-Schwarz, M., additional, Bonini, F., additional, Ciliberti, G., additional, Georgiopoulos, G., additional, Stamatelopoulos, K., additional, Stellos, K., additional, and Gatsiou, A., additional
- Published
- 2023
- Full Text
- View/download PDF
5. M6A RNA methylation is integral for endothelial plasticity
- Author
-
Tual-Chalot, S., primary, Ciliberti, G., additional, Polycarpou-Schwarz, M., additional, Amponsah-Offeh, M., additional, Bonini, F., additional, Gatsiou, A., additional, and Stellos, K., additional
- Published
- 2023
- Full Text
- View/download PDF
6. RNA-binding protein HuR controls vascular endothelial cell inflammatory responses to tumor necrosis factor-A and is associated with atherosclerosis progression in humans
- Author
-
Sachse, M., primary, Georgiopoulos, G., additional, Tual-Chalot, S., additional, Sopova, K., additional, Polycarpou-Schwarz, M., additional, Amponsah-Offeh, M., additional, Ciliberti, G., additional, Bonini, F., additional, Mavraganis, G., additional, Bampatsias, D., additional, Delialis, D., additional, Gatsiou, A., additional, Stamatelopoulos, K., additional, and Stellos, K., additional
- Published
- 2023
- Full Text
- View/download PDF
7. Conserved loop I of U5 snRNA is dispensible for both catalytic steps of pre-mRNA splicing in HeLa nuclear extracts
- Author
-
Segault, V., Will, C., Polycarpou-Schwarz, M., Mattaj, I., Branlant, C., and Luehrmann, R.
- Published
- 1999
8. Genetic and physical interactions involving the yeast nuclear cap-binding complex.
- Author
-
Fortes, P, Kufel, J, Fornerod, M, Polycarpou-Schwarz, M, Lafontaine, Denis, Tollervey, D, Mattaj, I W, Fortes, P, Kufel, J, Fornerod, M, Polycarpou-Schwarz, M, Lafontaine, Denis, Tollervey, D, and Mattaj, I W
- Abstract
Yeast strains lacking the yeast nuclear cap-binding complex (yCBC) are viable, although impaired in growth. We have taken advantage of this observation to carry out a genetic screen for components that show synthetic lethality (SL) with a cbp20-Delta cbp80-Delta double mutation. One set of SL interactions was due to mutations that were complemented by components of U1 small nuclear RNP (snRNP) and the yeast splicing commitment complex. These interactions confirm the role of yCBC in commitment complex formation. Physical interaction of yCBC with the commitment complex components Mud10p and Mud2p, which may directly mediate yCBC function, was demonstrated. Unexpectedly, we identified multiple SL mutations that were complemented by Cbf5p and Nop58p. These are components of the two major classes of yeast small nucleolar RNPs, which function in the maturation of rRNA precursors. Mutants lacking yCBC were found to be defective in rRNA processing. Analysis of the yCBC deletion phenotype suggests that this is likely to be due to a defect in the splicing of a subset of ribosomal protein mRNA precursors., Journal Article, Research Support, Non-U.S. Gov't, info:eu-repo/semantics/published
- Published
- 1999
9. Involvement of the carboxyl terminus of vertebrate poly(A) polymerase in U1A autoregulation and in the coupling of splicing and polyadenylation.
- Author
-
Gunderson, S I, primary, Vagner, S, additional, Polycarpou-Schwarz, M, additional, and Mattaj, I W, additional
- Published
- 1997
- Full Text
- View/download PDF
10. A complex secondary structure in U1A pre-mRNA that binds two molecules of U1A protein is required for regulation of polyadenylation.
- Author
-
van Gelder, C.W., primary, Gunderson, S.I., additional, Jansen, E.J., additional, Boelens, W.C., additional, Polycarpou-Schwarz, M., additional, Mattaj, I.W., additional, and van Venrooij, W.J., additional
- Published
- 1993
- Full Text
- View/download PDF
11. Common and unique transcription factor requirements of human U1 and U6 snRNA genes.
- Author
-
Bernués, J., primary, Simmen, K.A., additional, Lewis, J.D., additional, Gunderson, S.I., additional, Polycarpou-Schwarz, M., additional, Moncollin, V., additional, Egly, J.M., additional, and Mattaj, I.W., additional
- Published
- 1993
- Full Text
- View/download PDF
12. Insulin and insulin-like growth factor I regulate a thyroid-specific nuclear protein that binds to the thyroglobulin promoter.
- Author
-
Santisteban, P, primary, Acebrón, A, additional, Polycarpou-Schwarz, M, additional, and Di Lauro, R, additional
- Published
- 1992
- Full Text
- View/download PDF
13. Cell-type-specific expression of the rat thyroperoxidase promoter indicates common mechanisms for thyroid-specific gene expression
- Author
-
Francis-Lang, H, primary, Price, M, additional, Polycarpou-Schwarz, M, additional, and Di Lauro, R, additional
- Published
- 1992
- Full Text
- View/download PDF
14. A complex secondary structure in U1A pre‐mRNA that binds two molecules of U1A protein is required for regulation of polyadenylation.
- Author
-
Gelder, C.W., Gunderson, S.I., Jansen, E.J., Boelens, W.C., Polycarpou‐Schwarz, M., Mattaj, I.W., and Venrooij, W.J.
- Abstract
The human U1A protein‐U1A pre‐mRNA complex and the relationship between its structure and function in inhibition of polyadenylation in vitro were investigated. Two molecules of U1A protein were shown to bind to a conserved region in the 3′ untranslated region of U1A pre‐mRNA. The secondary structure of this region was determined by a combination of theoretical prediction, phylogenetic sequence alignment, enzymatic structure probing and molecular genetics. The U1A binding sites form (part of) a complex secondary structure which is significantly different from the binding site of U1A protein on U1 snRNA. Studies with mutant pre‐mRNAs showed that the integrity of much of this structure is required for both high affinity binding to U1A protein and specific inhibition of polyadenylation in vitro. In particular, binding of a single molecule of U1A protein to U1A pre‐mRNA is not sufficient to produce efficient inhibition of polyadenylation.
- Published
- 1993
- Full Text
- View/download PDF
15. Drosophila SNF/D25 combines the functions of the two snRNP proteins U1A and U2B' that are encoded separately in human, potato, and yeast
- Author
-
Polycarpou-Schwarz M, Si, Gunderson, Kandels-Lewis S, Seraphin B, and Iain Mattaj
- Subjects
Base Sequence ,genetic processes ,fungi ,Molecular Sequence Data ,RNA-Binding Proteins ,Saccharomyces cerevisiae ,Ribonucleoprotein, U2 Small Nuclear ,Recombinant Proteins ,Ribonucleoprotein, U1 Small Nuclear ,Evolution, Molecular ,health occupations ,Animals ,Humans ,Drosophila ,Amino Acid Sequence ,Cloning, Molecular ,Sequence Alignment ,Research Article ,Solanum tuberosum - Abstract
The plant and vertebrate snRP proteins U1A and U2B' are structurally closely related, but bind to different U snRNAs. Two additional related snRNP proteins, the yeast U2B' protein and Drosophila SNF/D25 protein, are analyzed here. We show that the previously described yeast open reading frame YIB9w encodes yeast U2B' as judged by the fact that the protein encoded by YIB9w bindsto stem-loop IV of yeast U2 snRNA in vitro and is part of the U2 snRNP in vivo. In contrast to the human U2B' protein, specific binding of yeast U2B' to RNA in vitro can occur in the absence of an accessory U2A' protein. The Drosophila SNF-D25 protein, unlike all other U1A/U2B' proteins studied to date, is shown to be a component of both U1 and U2 snRNPs. In vitro, SNF/D25 binds to U1 snRNA on itsown and to U2 snRNA in the presence of either the human U2A' protein or of Drosophila nuclear extract. Thus, its RNA-binding properties are the sum of those exhibited by human or potato U1A and U2B' proteins. Implications for the role of SNF/D25 in alternative splicing, and for the evolution of the U1A/U2B' protein family, are discussed.
16. Insulin and insulin-like growth factor I regulate a thyroid-specific nuclear protein that binds to the thyroglobulin promoter
- Author
-
Pilar Santisteban, M Polycarpou-Schwarz, R Di Lauro, Alvaro Acebrón, Santisteban, P, Acebrón, A, Polycarpou Schwarz, M, and DI LAURO, Roberto
- Subjects
Chloramphenicol O-Acetyltransferase ,endocrine system ,Thyroid Nuclear Factor 1 ,medicine.medical_specialty ,Transcription, Genetic ,medicine.medical_treatment ,Molecular Sequence Data ,Thyroid Transcription Factor 1 ,Thyroid Gland ,Transfection ,Thyroglobulin ,Cell Line ,Insulin-like growth factor ,Endocrinology ,Insulin receptor substrate ,Internal medicine ,medicine ,Animals ,Insulin ,Insulin-Like Growth Factor I ,Promoter Regions, Genetic ,Molecular Biology ,Base Sequence ,biology ,GRB10 ,Nuclear Proteins ,General Medicine ,IRS2 ,Rats ,DNA-Binding Proteins ,Insulin receptor ,Organ Specificity ,biology.protein ,Protein Binding ,Transcription Factors - Abstract
The mechanism responsible for the stimulation of thyroglobulin (Tg) gene expression by insulin and insulin-like growth factor I (IGF-I) in rat thyroid FRTL-5 cells has been investigated. Both insulin and IGF-I stimulate transcription from the Tg promoter in a transient transfection assay demonstrating that the promoter used contains the DNA signals necessary for insulin and IGF-I regulation. Promoter mutations that interfere with the binding of thyroid transcription factor 1 (TTF-1), TTF-2, and the ubiquitous transcription factor abolish the insulin/IGF-I response, indicating that the three factors may be involved in the observed transcriptional control. Protein-DNA binding studies did not reveal any effect of insulin/IGF-I on the ubiquitous transcription factor and the TTF-1 binding capacity. Instead, TTF-2 is absent in nuclear extracts from cells depleted of serum and insulin. Addition of insulin or IGF-I restores the TTF-2 concentration to normal levels and requires ongoing protein synthesis. The insulin effect was maximal at 24 h and at a concentration of 1 microgram/ml. The same effect was observed with a 10-fold lower concentration of IGF-I. These results suggest that insulin (probably through the IGF-I receptor) and IGF-I modulate the levels of TTF-2, which results in an increased expression of the Tg gene.
- Published
- 1992
- Full Text
- View/download PDF
17. Cell-type-specific expression of the rat thyroperoxidase promoter indicates common mechanisms for thyroid-specific gene expression
- Author
-
M. Price, R Di Lauro, M Polycarpou-Schwarz, H Francis-Lang, Francis Lang, H, Price, M, Polycarpou Schwarz, M, and DI LAURO, Roberto
- Subjects
Thyroid Nuclear Factor 1 ,endocrine system ,Recombinant Fusion Proteins ,medicine.medical_treatment ,DNA Mutational Analysis ,Molecular Sequence Data ,Thyroid Transcription Factor 1 ,Thyroid Gland ,Iodide Peroxidase ,Methylation ,Cell Line ,Thyroid peroxidase ,Consensus Sequence ,Gene expression ,medicine ,Animals ,Deoxyribonuclease I ,Luciferases ,Promoter Regions, Genetic ,Transcription factor ,Gene ,Molecular Biology ,Regulation of gene expression ,Base Sequence ,biology ,Cell Biology ,Molecular biology ,Rats ,Gene Expression Regulation ,Peroxidases ,biology.protein ,Thyroglobulin ,Research Article ,Transcription Factors - Abstract
A 420-bp fragment from the 5' end of the rat thyroperoxidase (TPO) gene was fused to a luciferase reporter and shown to direct cell-type-specific expression when transfected into rat thyroid FRTL-5 cells. Analysis of this DNA fragment revealed four regions of the promoter which interact with DNA-binding proteins present in FRTL-5 cells. Mutation of the DNA sequence within any of these regions reduced TPO promoter activity. The trans-acting factors binding to these sequences were compared with thyroid transcription factor 1 (TTF-1) and TTF-2, previously identified as transcriptional activators of another thyroid-specific gene, the thyroglobulin (Tg) gene. Purified TTF-1 binds to three regions of TPO which are protected by FRTL-5 proteins. Two of the binding sites overlap with recognition sites for other DNA-binding proteins. One TTF-1 site can also bind a protein (UFB) present in the nuclei of both expressing and nonexpressing cells. TTF-1 binding to the proximal region overlaps with that for a novel protein present in FRTL-5 cells which can also recognize the promoter-proximal region of Tg. Using a combination of techniques, the factor binding to the fourth TPO promoter site was shown to be TTF-2. We conclude, therefore, that the FRTL-5-specific expression of two thyroid restricted genes, encoding TPO and Tg, relies on a combination of the same trans-acting factors present in thyroid cells.
- Published
- 1992
18. Effector T cell chemokine IP-10 predicts cardiac recovery and clinical outcomes post-myocardial infarction.
- Author
-
Sopova K, Tual-Chalot S, Mueller-Hennessen M, Vlachogiannis NI, Georgiopoulos G, Biener M, Sachse M, Turchinovich A, Polycarpou-Schwarz M, Spray L, Maneta E, Bennaceur K, Mohammad A, Richardson GD, Gatsiou A, Langer HF, Frey N, Stamatelopoulos K, Heineke J, Duerschmied D, Giannitsis E, Spyridopoulos I, and Stellos K
- Subjects
- Humans, Chemokine CXCL10, Heart, Retrospective Studies, Myocardial Infarction, ST Elevation Myocardial Infarction therapy
- Abstract
Background and Aims: Preclinical data suggest that activation of the adaptive immune system is critical for myocardial repair processes in acute myocardial infarction. The aim of the present study was to determine the clinical value of baseline effector T cell chemokine IP-10 blood levels in the acute phase of ST-segment elevation myocardial infarction (STEMI) for the prediction of the left ventricular function changes and cardiovascular outcomes after STEMI., Methods: Serum IP-10 levels were retrospectively quantified in two independent cohorts of STEMI patients undergoing primary percutaneous coronary intervention., Results: We report a biphasic response of the effector T cell trafficking chemokine IP-10 characterized by an initial increase of its serum levels in the acute phase of STEMI followed by a rapid reduction at 90min post reperfusion. Patients at the highest IP-10 tertile presented also with more CD4 effector memory T cells (CD4 T
EM cells), but not other T cell subtypes, in blood. In the Newcastle cohort (n=47), patients in the highest IP-10 tertile or CD4 TEM cells at admission exhibited an improved cardiac systolic function 12 weeks after STEMI compared to patients in the lowest IP-10 tertile. In the Heidelberg cohort (n=331), STEMI patients were followed for a median of 540 days for major adverse cardiovascular events (MACE). Patients presenting with higher serum IP-10 levels at admission had a lower risk for MACE after adjustment for traditional risk factors, CRP and high-sensitivity troponin-T levels (highest vs. rest quarters: HR [95% CI]=0.420 [0.218-0.808])., Conclusion: Increased serum levels of IP-10 in the acute phase of STEMI predict a better recovery in cardiac systolic function and less adverse events in patients after STEMI., Competing Interests: MM-H. reports consulting fees for Zoll CMS GmbH, research funding from Roche Diagnostics and BRAHMS Thermo Scientific. NF has received lecture fees from AstraZeneca, Boehringer Ingelheim, Bayer Vital and consulting fees for Boehringer Ingelheim, all of which not related to the subject in this manuscript. DD reports consulting and speaker fees from Bayer Healthcare, Boston Scientific, Daiichi Sankyo, AstraZeneca, and research grants from DFG, DZHK. EG has received honoraria for lectures from AstraZeneca, Daiichi Sankyo and Roche Diagnostics, consulting fees for BRAHMS Deutschland, Roche Diagnostics, AstraZeneca, Daiichi and Novo Nordisk, research funding from Daiichi and Roche Diagnostics. IS receives research grants from Kancera AB and Astra Zaneca. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Sopova, Tual-Chalot, Mueller-Hennessen, Vlachogiannis, Georgiopoulos, Biener, Sachse, Turchinovich, Polycarpou-Schwarz, Spray, Maneta, Bennaceur, Mohammad, Richardson, Gatsiou, Langer, Frey, Stamatelopoulos, Heineke, Duerschmied, Giannitsis, Spyridopoulos and Stellos.)- Published
- 2023
- Full Text
- View/download PDF
19. Iron regulatory protein (IRP)-mediated iron homeostasis is critical for neutrophil development and differentiation in the bone marrow.
- Author
-
Bonadonna M, Altamura S, Tybl E, Palais G, Qatato M, Polycarpou-Schwarz M, Schneider M, Kalk C, Rüdiger W, Ertl A, Anstee N, Bogeska R, Helm D, Milsom MD, and Galy B
- Subjects
- Animals, Hematopoiesis, Hemoglobins metabolism, Homeostasis, Iron metabolism, Iron Regulatory Protein 1 metabolism, Iron-Regulatory Proteins metabolism, Mammals metabolism, Mice, Oxygen metabolism, Bone Marrow metabolism, Neutrophils metabolism
- Abstract
Iron is mostly devoted to the hemoglobinization of erythrocytes for oxygen transport. However, emerging evidence points to a broader role for the metal in hematopoiesis, including the formation of the immune system. Iron availability in mammalian cells is controlled by iron-regulatory protein 1 (IRP1) and IRP2. We report that global disruption of both IRP1 and IRP2 in adult mice impairs neutrophil development and differentiation in the bone marrow, yielding immature neutrophils with abnormally high glycolytic and autophagic activity, resulting in neutropenia. IRPs promote neutrophil differentiation in a cell intrinsic manner by securing cellular iron supply together with transcriptional control of neutropoiesis to facilitate differentiation to fully mature neutrophils. Unlike neutrophils, monocyte count was not affected by IRP and iron deficiency, suggesting a lineage-specific effect of iron on myeloid output. This study unveils the previously unrecognized importance of IRPs and iron metabolism in the formation of a major branch of the innate immune system.
- Published
- 2022
- Full Text
- View/download PDF
20. IRE-dependent Regulation of Intestinal Dmt1 Prevails During Chronic Dietary Iron Deficiency but is Dispensable in Conditions of Acute Erythropoietic Stress.
- Author
-
Qatato M, Bonadonna M, Palais G, Ertl A, Schmidt G, Polycarpou-Schwarz M, Karim Z, and Galy B
- Published
- 2022
- Full Text
- View/download PDF
21. The S-phase-induced lncRNA SUNO1 promotes cell proliferation by controlling YAP1/Hippo signaling pathway.
- Author
-
Hao Q, Zong X, Sun Q, Lin YC, Song YJ, Hashemikhabir S, Hsu RY, Kamran M, Chaudhary R, Tripathi V, Singh DK, Chakraborty A, Li XL, Kim YJ, Orjalo AV, Polycarpou-Schwarz M, Moriarity BS, Jenkins LM, Johansson HE, Zhu YJ, Diederichs S, Bagchi A, Kim TH, Janga SC, Lal A, Prasanth SG, and Prasanth KV
- Subjects
- Co-Repressor Proteins metabolism, Cytoskeletal Proteins metabolism, DEAD-box RNA Helicases metabolism, HCT116 Cells, HeLa Cells, Humans, RNA, Long Noncoding metabolism, S Phase, Up-Regulation, Cell Proliferation genetics, Co-Repressor Proteins genetics, Cytoskeletal Proteins genetics, DEAD-box RNA Helicases genetics, Gene Expression Regulation, RNA, Long Noncoding genetics, Signal Transduction physiology
- Abstract
Cell cycle is a cellular process that is subject to stringent control. In contrast to the wealth of knowledge of proteins controlling the cell cycle, very little is known about the molecular role of lncRNAs (long noncoding RNAs) in cell-cycle progression. By performing genome-wide transcriptome analyses in cell-cycle-synchronized cells, we observed cell-cycle phase-specific induction of >2000 lncRNAs. Further, we demonstrate that an S-phase-upregulated lncRNA, SUNO1 , facilitates cell-cycle progression by promoting YAP1-mediated gene expression. SUNO1 facilitates the cell-cycle-specific transcription of WTIP , a positive regulator of YAP1, by promoting the co-activator, DDX5-mediated stabilization of RNA polymerase II on chromatin. Finally, elevated SUNO1 levels are associated with poor cancer prognosis and tumorigenicity, implying its pro-survival role. Thus, we demonstrate the role of a S-phase up-regulated lncRNA in cell-cycle progression via modulating the expression of genes controlling cell proliferation., Competing Interests: QH, XZ, QS, YL, YS, SH, RH, MK, RC, VT, DS, AC, XL, YK, MP, BM, LJ, YZ, SD, AB, TK, SJ, SP, KP No competing interests declared, AO Arturo V Orjalo is affiliated with LGC Biosearch Technologies and Genentech Inc, the author has no financial interests to declare, HJ Hans Johansson is affiliated with LGC Biosearch Technologies. The author has no financial interests to declare. AL Reviewing editor, eLife
- Published
- 2020
- Full Text
- View/download PDF
22. MIR100 host gene-encoded lncRNAs regulate cell cycle by modulating the interaction between HuR and its target mRNAs.
- Author
-
Sun Q, Tripathi V, Yoon JH, Singh DK, Hao Q, Min KW, Davila S, Zealy RW, Li XL, Polycarpou-Schwarz M, Lehrmann E, Zhang Y, Becker KG, Freier SM, Zhu Y, Diederichs S, Prasanth SG, Lal A, Gorospe M, and Prasanth KV
- Subjects
- Cell Differentiation genetics, Cell Division genetics, Cell Line, Tumor, Cell Proliferation genetics, Humans, RNA, Messenger genetics, RNA-Binding Proteins genetics, Cell Cycle genetics, ELAV-Like Protein 1 genetics, MicroRNAs genetics, RNA, Long Noncoding genetics
- Abstract
Long non-coding RNAs (lncRNAs) regulate vital biological processes, including cell proliferation, differentiation and development. A subclass of lncRNAs is synthesized from microRNA (miRNA) host genes (MIRHGs) due to pre-miRNA processing, and are categorized as miRNA-host gene lncRNAs (lnc-miRHGs). Presently, the cellular function of most lnc-miRHGs is not well understood. We demonstrate a miRNA-independent role for a nuclear-enriched lnc-miRHG in cell cycle progression. MIR100HG produces spliced and stable lncRNAs that display elevated levels during the G1 phase of the cell cycle. Depletion of MIR100HG-encoded lncRNAs in human cells results in aberrant cell cycle progression without altering the levels of miRNA encoded within MIR100HG. Notably, MIR100HG interacts with HuR/ELAVL1 as well as with several HuR-target mRNAs. Further, MIR100HG-depleted cells show reduced interaction between HuR and three of its target mRNAs, indicating that MIR100HG facilitates interaction between HuR and target mRNAs. Our studies have unearthed novel roles played by a MIRHG-encoded lncRNA in regulating RNA binding protein activity, thereby underscoring the importance of determining the function of several hundreds of lnc-miRHGs that are present in human genome.
- Published
- 2018
- Full Text
- View/download PDF
23. The Long Noncoding RNA Cancer Susceptibility 9 and RNA Binding Protein Heterogeneous Nuclear Ribonucleoprotein L Form a Complex and Coregulate Genes Linked to AKT Signaling.
- Author
-
Klingenberg M, Groß M, Goyal A, Polycarpou-Schwarz M, Miersch T, Ernst AS, Leupold J, Patil N, Warnken U, Allgayer H, Longerich T, Schirmacher P, Boutros M, and Diederichs S
- Subjects
- Animals, Apoptosis, Biomarkers, Tumor genetics, Carcinoma, Hepatocellular metabolism, Cell Line, Tumor, Chickens, Gene Expression Regulation, Neoplastic genetics, Humans, Liver metabolism, Liver pathology, Liver Neoplasms metabolism, RNA, Small Interfering, Signal Transduction, Carcinoma, Hepatocellular genetics, Heterogeneous-Nuclear Ribonucleoprotein L metabolism, Liver Neoplasms genetics, Proto-Oncogene Proteins c-akt metabolism, RNA, Long Noncoding metabolism
- Abstract
The identification of viability-associated long noncoding RNAs (lncRNAs) might be a promising rationale for new therapeutic approaches in liver cancer. Here, we applied an RNA interference screening approach in hepatocellular carcinoma (HCC) cell lines to find viability-associated lncRNAs. Among the multiple identified lncRNAs with a significant impact on HCC cell viability, we selected cancer susceptibility 9 (CASC9) due to the strength of its phenotype, expression, and up-regulation in HCC versus normal liver. CASC9 regulated viability across multiple HCC cell lines as shown by clustered regularly interspaced short palindromic repeats interference and single small interfering RNA (siRNA)-mediated and siRNA pool-mediated depletion of CASC9. Further, CASC9 depletion caused an increase in apoptosis and a decrease of proliferation. We identified the RNA binding protein heterogeneous nuclear ribonucleoprotein L (HNRNPL) as a CASC9 interacting protein by RNA affinity purification and validated it by native RNA immunoprecipitation. Knockdown of HNRNPL mimicked the loss-of-viability phenotype observed upon CASC9 depletion. Analysis of the proteome (stable isotope labeling with amino acids in cell culture) of CASC9-depleted and HNRNPL-depleted cells revealed a set of coregulated genes which implied a role of the CASC9:HNRNPL complex in AKT signaling and DNA damage sensing. CASC9 expression levels were elevated in patient-derived tumor samples compared to normal control tissue and had a significant association with overall survival of HCC patients. In a xenograft chicken chorioallantoic membrane model, we measured decreased tumor size after knockdown of CASC9. Conclusion: Taken together, we provide a comprehensive list of viability-associated lncRNAs in HCC; we identified the CASC9:HNRNPL complex as a clinically relevant viability-associated lncRNA/protein complex which affects AKT signaling and DNA damage sensing in HCC., (© 2018 by the American Association for the Study of Liver Diseases.)
- Published
- 2018
- Full Text
- View/download PDF
24. The cancer-associated microprotein CASIMO1 controls cell proliferation and interacts with squalene epoxidase modulating lipid droplet formation.
- Author
-
Polycarpou-Schwarz M, Groß M, Mestdagh P, Schott J, Grund SE, Hildenbrand C, Rom J, Aulmann S, Sinn HP, Vandesompele J, and Diederichs S
- Subjects
- Breast Neoplasms genetics, Breast Neoplasms metabolism, Breast Neoplasms pathology, Carcinogenesis genetics, Cell Cycle Checkpoints genetics, Cell Line, Tumor, Cell Movement genetics, Female, G1 Phase genetics, Gene Expression Regulation, Neoplastic genetics, Humans, MCF-7 Cells, Oncogenes genetics, Open Reading Frames genetics, RNA, Long Noncoding genetics, Resting Phase, Cell Cycle genetics, Cell Proliferation genetics, Lipid Droplets metabolism, Squalene Monooxygenase metabolism
- Abstract
Breast cancer is a leading cause of cancer-related death in women. Small open reading frame (sORF)-encoded proteins or microproteins constitute a new class of molecules often transcribed from presumed long non-coding RNA transcripts (lncRNAs). The translation of some of these sORFs has been confirmed, but their cellular function and importance remains largely unknown. Here, we report the identification and characterization of a novel microprotein of 10 kDa, which we named Cancer-Associated Small Integral Membrane Open reading frame 1 (CASIMO1). CASIMO1 RNA is overexpressed predominantly in hormone receptor-positive breast tumors. Its knockdown leads to decreased proliferation in multiple breast cancer cell lines. Its loss disturbs the organization of the actin cytoskeleton, leads to inhibition of cell motility, and causes a G
0 /G1 cell cycle arrest. The proliferation phenotype upon overexpression is observed only with CASIMO1 protein expression, but not with a non-translatable mutant attributing the effects to the sORF-derived protein rather than a lncRNA function. CASIMO1 microprotein interacts with squalene epoxidase (SQLE), a key enzyme in cholesterol synthesis and a known oncogene in breast cancer. Overexpression of CASIMO1 leads to SQLE protein accumulation without affecting its RNA levels and increased lipid droplet clustering, while knockdown of CASIMO1 decreased SQLE protein abundance and ERK phosphorylation downstream of SQLE. Importantly, SQLE knockdown mimicked the CASIMO1 knockdown phenotype and in turn SQLE overexpression fully rescued the effect of CASIMO1 knockdown. These findings establish CASIMO1 as the first functional microprotein that plays a role in carcinogenesis and is implicated in the cell lipid homeostasis.- Published
- 2018
- Full Text
- View/download PDF
25. Targeting LINC00673 expression triggers cellular senescence in lung cancer.
- Author
-
Roth A, Boulay K, Groß M, Polycarpou-Schwarz M, Mallette FA, Regnier M, Bida O, Ginsberg D, Warth A, Schnabel PA, Muley T, Meister M, Zabeck H, Hoffmann H, and Diederichs S
- Subjects
- Adenocarcinoma genetics, Cell Cycle Checkpoints genetics, Cell Line, Tumor, Cell Proliferation genetics, Gene Knockdown Techniques, Humans, Models, Biological, Mutation, RNA Interference, Signal Transduction, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Cellular Senescence genetics, Gene Expression Regulation, Neoplastic, Lung Neoplasms genetics, RNA, Long Noncoding genetics
- Abstract
Aberrant expression of noncoding RNAs plays a critical role during tumorigenesis. To uncover novel functions of long non-coding RNA (lncRNA) in lung adenocarcinoma, we used a microarray-based screen identifying LINC00673 with elevated expression in matched tumor versus normal tissue. We report that loss of LINC00673 is sufficient to trigger cellular senescence, a tumor suppressive mechanism associated with permanent cell cycle arrest, both in lung cancer and normal cells in a p53-dependent manner. LINC00673-depleted cells fail to efficiently transit from G1- to S-phase. Using a quantitative proteomics approach, we confirm the modulation of senescence-associated genes as a result of LINC00673 knockdown. In addition, we uncover that depletion of p53 in normal and tumor cells is sufficient to overcome LINC00673-mediated cell cycle arrest and cellular senescence. Furthermore, we report that overexpression of LINC00673 reduces p53 translation and contributes to the bypass of Ras-induced senescence. In summary, our findings highlight LINC00673 as a crucial regulator of proliferation and cellular senescence in lung cancer.
- Published
- 2018
- Full Text
- View/download PDF
26. A cautionary tale of sense-antisense gene pairs: independent regulation despite inverse correlation of expression.
- Author
-
Goyal A, Fiškin E, Gutschner T, Polycarpou-Schwarz M, Groß M, Neugebauer J, Gandhi M, Caudron-Herger M, Benes V, and Diederichs S
- Subjects
- Cell Line, DNA Damage, Humans, Nuclear Proteins biosynthesis, Nuclear Proteins genetics, Tumor Suppressor Protein p53 metabolism, Gene Expression Regulation, RNA, Antisense biosynthesis, RNA, Messenger biosynthesis
- Abstract
Long non-coding RNAs (lncRNAs) have been proven to play important roles in diverse cellular processes including the DNA damage response. Nearly 40% of annotated lncRNAs are transcribed in antisense direction to other genes and have often been implicated in their regulation via transcript- or transcription-dependent mechanisms. However, it remains unclear whether inverse correlation of gene expression would generally point toward a regulatory interaction between the genes. Here, we profiled lncRNA and mRNA expression in lung and liver cancer cells after exposure to DNA damage. Our analysis revealed two pairs of mRNA-lncRNA sense-antisense transcripts being inversely expressed upon DNA damage. The lncRNA NOP14-AS1 was strongly upregulated upon DNA damage, while the mRNA for NOP14 was downregulated, both in a p53-dependent manner. For another pair, the lncRNA LIPE-AS1 was downregulated, while its antisense mRNA CEACAM1 was upregulated. To test whether as expected the antisense genes would regulate each other resulting in this highly significant inverse correlation, we employed antisense oligonucleotides and RNAi to study transcript-dependent effects as well as dCas9-based transcriptional modulation by CRISPRi/CRISPRa for transcription-dependent effects. Surprisingly, despite the strong stimulus-dependent inverse correlation, our data indicate that neither transcript- nor transcription-dependent mechanisms explain the inverse regulation of NOP14-AS1:NOP14 or LIPE-AS1:CEACAM1 expression. Hence, sense-antisense pairs whose expression is strongly-positively or negatively-correlated can be nonetheless regulated independently. This highlights the requirement of individual experimental studies for each antisense pair and prohibits drawing conclusions on regulatory mechanisms from expression correlations., (© The Author(s) 2017. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2017
- Full Text
- View/download PDF
27. The long non-coding RNA LINC00152 is essential for cell cycle progression through mitosis in HeLa cells.
- Author
-
Nötzold L, Frank L, Gandhi M, Polycarpou-Schwarz M, Groß M, Gunkel M, Beil N, Erfle H, Harder N, Rohr K, Trendel J, Krijgsveld J, Longerich T, Schirmacher P, Boutros M, Erhardt S, and Diederichs S
- Subjects
- Alternative Splicing, Cell Cycle Checkpoints genetics, Cell Division genetics, Cell Proliferation, Gene Expression Profiling, Gene Expression Regulation, Gene Knockdown Techniques, HeLa Cells, Humans, Organ Specificity genetics, Proteomics methods, RNA Interference, RNA Transport, Time-Lapse Imaging, Cell Cycle genetics, Mitosis genetics, RNA, Long Noncoding genetics
- Abstract
In recent years, long non-coding RNA (lncRNA) research has identified essential roles of these transcripts in virtually all physiological cellular processes including tumorigenesis, but their functions and molecular mechanisms are poorly understood. In this study, we performed a high-throughput siRNA screen targeting 638 lncRNAs deregulated in cancer entities to analyse their impact on cell division by using time-lapse microscopy. We identified 26 lncRNAs affecting cell morphology and cell cycle including LINC00152. This transcript was ubiquitously expressed in many human cell lines and its RNA levels were significantly upregulated in lung, liver and breast cancer tissues. A comprehensive sequence analysis of LINC00152 revealed a highly similar paralog annotated as MIR4435-2HG and several splice variants of both transcripts. The shortest and most abundant isoform preferentially localized to the cytoplasm. Cells depleted of LINC00152 arrested in prometaphase of mitosis and showed reduced cell viability. In RNA affinity purification (RAP) studies, LINC00152 interacted with a network of proteins that were associated with M phase of the cell cycle. In summary, we provide new insights into the properties and biological function of LINC00152 suggesting that this transcript is crucial for cell cycle progression through mitosis and thus, could act as a non-coding oncogene.
- Published
- 2017
- Full Text
- View/download PDF
28. The lncRNA VELUCT strongly regulates viability of lung cancer cells despite its extremely low abundance.
- Author
-
Seiler J, Breinig M, Caudron-Herger M, Polycarpou-Schwarz M, Boutros M, and Diederichs S
- Subjects
- Cell Line, Tumor, Cell Proliferation, Cell Survival genetics, Chromatin metabolism, Gene Expression Regulation, Neoplastic, Gene Knockdown Techniques, Gene Silencing, Humans, RNA Stability genetics, RNA, Long Noncoding metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, RNA, Small Interfering metabolism, Lung Neoplasms genetics, Lung Neoplasms pathology, RNA, Long Noncoding genetics
- Abstract
Little is known about the function of most non-coding RNAs (ncRNAs). The majority of long ncRNAs (lncRNAs) is expressed at very low levels and it is a matter of intense debate whether these can be of functional relevance. Here, we identified lncRNAs regulating the viability of lung cancer cells in a high-throughput RNA interference screen. Based on our previous expression profiling, we designed an siRNA library targeting 638 lncRNAs upregulated in human cancer. In a functional siRNA screen analyzing the viability of lung cancer cells, the most prominent hit was a novel lncRNA which we called Viability Enhancing LUng Cancer Transcript (VELUCT). In silico analyses confirmed the non-coding properties of the transcript. Surprisingly, VELUCT was below the detection limit in total RNA from NCI-H460 cells by RT-qPCR as well as RNA-Seq, but was robustly detected in the chromatin-associated RNA fraction. It is an extremely low abundant lncRNA with an RNA copy number of less than one copy per cell. Blocking transcription with actinomycin D revealed that VELUCT RNA was highly unstable which may partially explain its low steady-state concentration. Despite its extremely low abundance, loss-of-function of VELUCT with three independent experimental approaches in three different lung cancer cell lines led to a significant reduction of cell viability: Next to four individual siRNAs, also two complex siPOOLs as well as two antisense oligonucleotides confirmed the strong and specific phenotype. In summary, the extremely low abundant lncRNA VELUCT is essential for regulation of cell viability in several lung cancer cell lines. Hence, VELUCT is the first example for a lncRNA that is expressed at a very low level, but has a strong loss-of-function phenotype. Thus, our study proves that at least individual low-abundant lncRNAs can play an important functional role., (© The Author(s) 2017. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2017
- Full Text
- View/download PDF
29. LIMT is a novel metastasis inhibiting lncRNA suppressed by EGF and downregulated in aggressive breast cancer.
- Author
-
Sas-Chen A, Aure MR, Leibovich L, Carvalho S, Enuka Y, Körner C, Polycarpou-Schwarz M, Lavi S, Nevo N, Kuznetsov Y, Yuan J, Azuaje F, Ulitsky I, Diederichs S, Wiemann S, Yakhini Z, Kristensen VN, Børresen-Dale AL, and Yarden Y
- Subjects
- Female, Humans, Breast Neoplasms pathology, Down-Regulation, Epidermal Growth Factor metabolism, Gene Expression Regulation, Neoplastic, RNA, Long Noncoding biosynthesis
- Abstract
Long noncoding RNAs (lncRNAs) are emerging as regulators of gene expression in pathogenesis, including cancer. Recently, lncRNAs have been implicated in progression of specific subtypes of breast cancer. One aggressive, basal-like subtype associates with increased EGFR signaling, while another, the HER2-enriched subtype, engages a kin of EGFR Based on the premise that EGFR-regulated lncRNAs might control the aggressiveness of basal-like tumors, we identified multiple EGFR-inducible lncRNAs in basal-like normal cells and overlaid them with the transcriptomes of over 3,000 breast cancer patients. This led to the identification of 11 prognostic lncRNAs. Functional analyses of this group uncovered LINC01089 (here renamed LncRNA Inhibiting Metastasis; LIMT), a highly conserved lncRNA, which is depleted in basal-like and in HER2-positive tumors, and the low expression of which predicts poor patient prognosis. Interestingly, EGF rapidly downregulates LIMT expression by enhancing histone deacetylation at the respective promoter. We also find that LIMT inhibits extracellular matrix invasion of mammary cells in vitro and tumor metastasis in vivo In conclusion, lncRNAs dynamically regulated by growth factors might act as novel drivers of cancer progression and serve as prognostic biomarkers., (© 2016 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2016
- Full Text
- View/download PDF
30. Rare Drosha splice variants are deficient in microRNA processing but do not affect general microRNA expression in cancer cells.
- Author
-
Grund SE, Polycarpou-Schwarz M, Luo C, Eichmüller SB, and Diederichs S
- Subjects
- Amino Acid Sequence, Cell Line, Cytoplasm metabolism, Gene Order, Humans, Molecular Sequence Data, Neoplasms metabolism, Protein Binding, Proteins metabolism, RNA-Binding Proteins, Ribonuclease III chemistry, Ribonuclease III metabolism, Sequence Alignment, Alternative Splicing, MicroRNAs metabolism, Neoplasms genetics, Ribonuclease III genetics
- Abstract
Drosha is a key enzyme in microRNA biogenesis, generating the precursor miRNA (pre-miRNA) by excising the stem-loop embedded in the primary transcripts (pri-miRNA). The specificity for the pri-miRNAs and determination of the cleavage site are provided by its binding partner DGCR8, which is necessary for efficient processing. The crucial Drosha domains for pri-miRNA cleavage are the middle part, the two enzymatic RNase III domains (RIIID), and the dsRNA binding domain (dsRBD) in the C-terminus. Here, we identify alternatively spliced transcripts in human melanoma and NT2 cell lines, encoding C-terminally truncated Drosha proteins lacking part of the RIIIDb and the entire dsRBD. Proteins generated from these alternative splice variants fail to bind to DGCR8 but still interact with Ewing sarcoma protein (EWS). In vitro as well as in vivo, the Drosha splice variants are deficient in pri-miRNA processing. However, the aberrant transcripts in melanoma cells do not consistently reduce mature miRNA levels compared with melanoma cell lines lacking those splice variants, possibly owing to their limited abundance. Our findings show that alternative processing-deficient Drosha splice variants exist in melanoma cells. In elevated amounts, these alternatively spliced transcripts could provide one potential mechanism accounting for the deregulation of miRNAs in cancer cells. On the basis of our results, the search for alternative inactive splice variants might be fruitful in different tumor entities to unravel the molecular basis of the previously observed decreased microRNA processing efficiency in cancer., (Copyright © 2012 Neoplasia Press, Inc.)
- Published
- 2012
- Full Text
- View/download PDF
31. Transient cyclical methylation of promoter DNA.
- Author
-
Kangaspeska S, Stride B, Métivier R, Polycarpou-Schwarz M, Ibberson D, Carmouche RP, Benes V, Gannon F, and Reid G
- Subjects
- Cell Line, Tumor, CpG Islands genetics, DNA genetics, Doxorubicin pharmacology, Estrogen Receptor alpha genetics, Estrogen Receptor alpha metabolism, Humans, RNA Polymerase II metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Time Factors, Transcription, Genetic drug effects, Trefoil Factor-1, Tumor Suppressor Proteins genetics, DNA metabolism, DNA Methylation drug effects, Gene Expression Regulation drug effects, Promoter Regions, Genetic genetics
- Abstract
Methylation of CpG dinucleotides is generally associated with epigenetic silencing of transcription and is maintained through cellular division. Multiple CpG sequences are rare in mammalian genomes, but frequently occur at the transcriptional start site of active genes, with most clusters of CpGs being hypomethylated. We reported previously that the proximal region of the trefoil factor 1 (TFF1, also known as pS2) and oestrogen receptor alpha (ERalpha) promoters could be partially methylated by treatment with deacetylase inhibitors, suggesting the possibility of dynamic changes in DNA methylation. Here we show that cyclical methylation and demethylation of CpG dinucleotides, with a periodicity of around 100 min, is characteristic for five selected promoters, including the oestrogen (E2)-responsive pS2 gene, in human cells. When the pS2 gene is actively transcribed, DNA methylation occurs after the cyclical occupancy of ERalpha and RNA polymerase II (polII). Moreover, we report conditions that provoke methylation cycling of the pS2 promoter in cell lines in which pS2 expression is quiescent and the proximal promoter is methylated. This coincides with a low-level re-expression of ERalpha and of pS2 transcripts.
- Published
- 2008
- Full Text
- View/download PDF
32. Thanatop: a novel 5-nitrofuran that is a highly active, cell-permeable inhibitor of topoisomerase II.
- Author
-
Polycarpou-Schwarz M, Müller K, Denger S, Riddell A, Lewis J, Gannon F, and Reid G
- Subjects
- Cell Cycle drug effects, Cell Line, Tumor, Cell Membrane Permeability, DNA Damage, Drug Screening Assays, Antitumor, Enzyme Inhibitors pharmacokinetics, Etoposide pharmacokinetics, Etoposide pharmacology, Humans, Nitrofurans pharmacokinetics, Enzyme Inhibitors pharmacology, Nitrofurans pharmacology, Topoisomerase II Inhibitors
- Abstract
A series of nitrofuran-based compounds were identified as inhibitors of estrogen signaling in a cell-based, high-throughput screen of a diverse library of small molecules. These highly related compounds were subsequently found to inhibit topoisomerase II in vitro at concentrations similar to that required for the inhibition of estrogen signaling in cells. The most potent nitrofuran discovered is approximately 10-fold more active than etoposide phosphate, a topoisomerase II inhibitor in clinical use. The nitrofurans also inhibit topoisomerase I activity, with approximately 20-fold less activity. Moreover, the nitrofurans, in contrast to etoposide, induce a profound cell cycle arrest in the G(0)-G(1) phase of the cell cycle, do not induce double-stranded DNA breaks, are not substrates for multidrug resistance protein-1 export from the cell, and are amenable to synthetic development. In addition, the nitrofurans synergize with etoposide phosphate in cell killing. Clonogenic assays done on a panel of human tumors maintained ex vivo in nude mice show that the most active compound identified in the screen is selective against tumors compared with normal hematopoietic stem cells. However, this compound had only moderate activity in a mouse xenograft model. This novel class of topoisomerase II inhibitor may provide additional chemotherapeutic strategies for the development of cytotoxic agents with proven clinical utility.
- Published
- 2007
- Full Text
- View/download PDF
33. Relationships and distinctions in iron-regulatory networks responding to interrelated signals.
- Author
-
Muckenthaler M, Richter A, Gunkel N, Riedel D, Polycarpou-Schwarz M, Hentze S, Falkenhahn M, Stremmel W, Ansorge W, and Hentze MW
- Subjects
- DNA, Complementary genetics, Deferoxamine pharmacology, Ferric Compounds pharmacology, HeLa Cells drug effects, HeLa Cells metabolism, Hemin pharmacology, Hemochromatosis genetics, Hemochromatosis metabolism, Hemochromatosis Protein, Histocompatibility Antigens Class I genetics, Histocompatibility Antigens Class I physiology, Humans, Hydrogen Peroxide pharmacology, Iron Chelating Agents pharmacology, Membrane Proteins genetics, Membrane Proteins physiology, Nitroprusside pharmacology, Oxidative Stress, Promoter Regions, Genetic drug effects, Protein Biosynthesis drug effects, Quaternary Ammonium Compounds pharmacology, Recombinant Fusion Proteins physiology, Transfection, Gene Expression Profiling, Gene Expression Regulation, Neoplastic drug effects, Iron metabolism, Oligonucleotide Array Sequence Analysis
- Abstract
Specialized cDNA-based microarrays (IronChips) were developed to investigate complex physiological gene-regulatory patterns in iron metabolism. Approximately 115 human cDNAs were strategically selected to represent genes involved either in iron metabolism or in interlinked pathways (eg, oxidative stress, nitric oxide [NO] metabolism, or copper metabolism), and were immobilized on glass slides. HeLa cells were treated with iron donors or iron chelators, or were subjected to oxidative stress (H(2)O(2)) or NO (sodium nitroprusside). In addition, we generated a stable transgenic HeLa cell line expressing the HFE gene under an inducible promoter. Gene-response patterns were recorded for all of these interrelated experimental stimuli, and analyzed for common and distinct responses that define signal-specific regulatory patterns. The resulting regulatory patterns reveal and define degrees of relationship between distinct signals. Remarkably, the gene responses elicited by the altered expression of the hemochromatosis protein HFE and by pharmacological iron chelation exhibit the highest degree of relatedness, both for iron-regulatory protein (IRP) and non-IRP target genes. This finding suggests that HFE expression directly affects the intracellular chelatable iron pool in the transgenic cell line. Furthermore, cells treated with the iron donors hemin or ferric ammonium citrate display response patterns that permit the identification of the iron-loaded state in both cases, and the discrimination between the sources of iron loading. These findings also demonstrate the broad utility of gene-expression profiling with the IronChip to study iron metabolism and related human diseases.
- Published
- 2003
- Full Text
- View/download PDF
34. Iron-dependent regulation of the divalent metal ion transporter.
- Author
-
Gunshin H, Allerson CR, Polycarpou-Schwarz M, Rofts A, Rogers JT, Kishi F, Hentze MW, Rouault TA, Andrews NC, and Hediger MA
- Subjects
- 3' Untranslated Regions, Biological Transport, Caco-2 Cells, Cation Transport Proteins metabolism, Cations, Divalent metabolism, Gene Expression Regulation, HeLa Cells, Humans, Iron Regulatory Protein 1, Iron Regulatory Protein 2, Iron-Regulatory Proteins, Iron-Sulfur Proteins metabolism, Nucleic Acid Conformation, Protein Binding, RNA Processing, Post-Transcriptional, RNA-Binding Proteins metabolism, Regulatory Sequences, Nucleic Acid, Cation Transport Proteins genetics, Iron metabolism, Iron-Binding Proteins
- Abstract
The first step in intestinal iron absorption is mediated by the H(+)-coupled Fe(2+) transporter called divalent cation transporter 1/divalent metal ion transporter 1 (DCT1/DMT1) (also known as natural resistance-associated macrophage protein 2). DCT1/DMT1 mRNA levels in the duodenum strongly increase in response to iron depletion. To study the mechanism of iron-dependent DCT1/DMT1 mRNA regulation, we investigated the endogenous expression of DCT1/DMT1 mRNA in various cell types. We found that only the iron responsive element (IRE)-containing form, which corresponds to one of two splice forms of DCT1/DMT1, is responsive to iron treatment and this responsiveness was cell type specific. We also examined the interaction of the putative 3'-UTR IRE with iron responsive binding proteins (IRP1 and IRP2), and found that IRP1 binds to the DCT1/DMT1-IRE with higher affinity compared to IRP2. This differential binding of IRP1 and IRP2 was also reported for the IREs of transferrin receptors, erythroid 5-aminolevulinate synthase and mitochondrial aconitase. We propose that regulation of DCT1/DMT1 mRNA by iron involves post-transcriptional regulation through the binding of IRP1 to the transporter's IRE, as well as other as yet unknown factors.
- Published
- 2001
- Full Text
- View/download PDF
35. Genetic and physical interactions involving the yeast nuclear cap-binding complex.
- Author
-
Fortes P, Kufel J, Fornerod M, Polycarpou-Schwarz M, Lafontaine D, Tollervey D, and Mattaj IW
- Subjects
- Gene Deletion, Genes, Lethal, Genetic Complementation Test, Nuclear Proteins genetics, Protein Binding, RNA Cap-Binding Proteins, RNA Precursors metabolism, RNA, Messenger biosynthesis, RNA, Ribosomal biosynthesis, RNA-Binding Proteins genetics, Nuclear Proteins metabolism, RNA Caps metabolism, RNA Splicing, RNA-Binding Proteins metabolism, Saccharomyces cerevisiae genetics
- Abstract
Yeast strains lacking the yeast nuclear cap-binding complex (yCBC) are viable, although impaired in growth. We have taken advantage of this observation to carry out a genetic screen for components that show synthetic lethality (SL) with a cbp20-Delta cbp80-Delta double mutation. One set of SL interactions was due to mutations that were complemented by components of U1 small nuclear RNP (snRNP) and the yeast splicing commitment complex. These interactions confirm the role of yCBC in commitment complex formation. Physical interaction of yCBC with the commitment complex components Mud10p and Mud2p, which may directly mediate yCBC function, was demonstrated. Unexpectedly, we identified multiple SL mutations that were complemented by Cbf5p and Nop58p. These are components of the two major classes of yeast small nucleolar RNPs, which function in the maturation of rRNA precursors. Mutants lacking yCBC were found to be defective in rRNA processing. Analysis of the yCBC deletion phenotype suggests that this is likely to be due to a defect in the splicing of a subset of ribosomal protein mRNA precursors.
- Published
- 1999
- Full Text
- View/download PDF
36. Conserved loop I of U5 small nuclear RNA is dispensable for both catalytic steps of pre-mRNA splicing in HeLa nuclear extracts.
- Author
-
Ségault V, Will CL, Polycarpou-Schwarz M, Mattaj IW, Branlant C, and Lührmann R
- Subjects
- Animals, Base Sequence, Cell Nucleus, Cell-Free System, HeLa Cells, Humans, Mutation, Nucleic Acid Conformation, RNA Precursors metabolism, RNA, Messenger metabolism, RNA, Small Nuclear chemistry, Xenopus, Conserved Sequence, RNA Splicing, RNA, Small Nuclear genetics, Ribonucleoprotein, U5 Small Nuclear metabolism
- Abstract
The function of conserved regions of the metazoan U5 snRNA was investigated by reconstituting U5 small nuclear ribonucleoprotein particles (snRNPs) from purified snRNP proteins and HeLa or Xenopus U5 snRNA mutants and testing their ability to restore splicing to U5-depleted nuclear extracts. Substitution of conserved nucleotides comprising internal loop 2 or deletion of internal loop 1 had no significant effect on the ability of reconstituted U5 snRNPs to complement splicing. However, deletion of internal loop 2 abolished U5 activity in splicing and spliceosome formation. Surprisingly, substitution of the invariant loop 1 nucleotides with a GAGA tetraloop had no effect on U5 activity. Furthermore, U5 snRNPs reconstituted from an RNA formed by annealing the 5' and 3' halves of the U5 snRNA, which lacked all loop 1 nucleotides, complemented both steps of splicing. Thus, in contrast to yeast, loop 1 of the human U5 snRNA is dispensable for both steps of splicing in HeLa nuclear extracts. This suggests that its function can be compensated for in vitro by other spliceosomal components: for example, by proteins associated with the U5 snRNP. Consistent with this idea, immunoprecipitation studies indicated that several functionally important U5 proteins associate stably with U5 snRNPs containing a GAGA loop 1 substitution.
- Published
- 1999
- Full Text
- View/download PDF
37. U1 snRNP inhibits pre-mRNA polyadenylation through a direct interaction between U1 70K and poly(A) polymerase.
- Author
-
Gunderson SI, Polycarpou-Schwarz M, and Mattaj IW
- Subjects
- Adenine metabolism, Amino Acid Sequence, Animals, Bacterial Proteins genetics, Cattle, Cell Nucleus chemistry, Cell Nucleus enzymology, Enzyme Activation physiology, HeLa Cells, Humans, Molecular Sequence Data, RNA Precursors genetics, RNA Splicing physiology, Ribonuclease H, Ribonucleoprotein, U1 Small Nuclear genetics, Saccharomyces cerevisiae chemistry, Saccharomyces cerevisiae enzymology, Bacterial Proteins metabolism, Escherichia coli Proteins, Gene Expression Regulation, Enzymologic, Polynucleotide Adenylyltransferase, RNA Precursors metabolism, Ribonucleoprotein, U1 Small Nuclear metabolism, Saccharomyces cerevisiae Proteins
- Abstract
It has previously been shown in vivo that bovine papillomavirus represses its late gene expression via a 5' splice site sequence located upstream of the late polyadenylation signal. Here, the mechanism of repression is determined by in vitro analysis. U1 snRNP binding to the 5' splice site results in inhibition of polyadenylation via a direct interaction with poly(A) polymerase (PAP). Although the inhibitory mechanism is similar to that used in U1A autoregulation, U1A within the U1 snRNP does not contribute to PAP inhibition. Instead the U1 70K protein, when bound to U1 snRNA, both interacts with and inhibits PAP. Conservation of the U1 70K inhibitory domains suggests that polyadenylation regulation via PAP inhibition may be more widespread than previously thought.
- Published
- 1998
- Full Text
- View/download PDF
38. Drosophila SNF/D25 combines the functions of the two snRNP proteins U1A and U2B' that are encoded separately in human, potato, and yeast.
- Author
-
Polycarpou-Schwarz M, Gunderson SI, Kandels-Lewis S, Seraphin B, and Mattaj IW
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Cloning, Molecular, Drosophila, Evolution, Molecular, Humans, Molecular Sequence Data, Recombinant Proteins genetics, Saccharomyces cerevisiae, Sequence Alignment, Solanum tuberosum, RNA-Binding Proteins genetics, Ribonucleoprotein, U1 Small Nuclear genetics, Ribonucleoprotein, U2 Small Nuclear genetics
- Abstract
The plant and vertebrate snRP proteins U1A and U2B' are structurally closely related, but bind to different U snRNAs. Two additional related snRNP proteins, the yeast U2B' protein and Drosophila SNF/D25 protein, are analyzed here. We show that the previously described yeast open reading frame YIB9w encodes yeast U2B' as judged by the fact that the protein encoded by YIB9w bindsto stem-loop IV of yeast U2 snRNA in vitro and is part of the U2 snRNP in vivo. In contrast to the human U2B' protein, specific binding of yeast U2B' to RNA in vitro can occur in the absence of an accessory U2A' protein. The Drosophila SNF-D25 protein, unlike all other U1A/U2B' proteins studied to date, is shown to be a component of both U1 and U2 snRNPs. In vitro, SNF/D25 binds to U1 snRNA on itsown and to U2 snRNA in the presence of either the human U2A' protein or of Drosophila nuclear extract. Thus, its RNA-binding properties are the sum of those exhibited by human or potato U1A and U2B' proteins. Implications for the role of SNF/D25 in alternative splicing, and for the evolution of the U1A/U2B' protein family, are discussed.
- Published
- 1996
39. Distinguishing specific from nonspecific complexes on southwestern blots by a rapid DMS protection assay.
- Author
-
Polycarpou-Schwarz M and Papavassiliou AG
- Subjects
- Animals, Base Sequence, Cell Line, Chemical Phenomena, Chemistry, Physical, DNA chemistry, DNA-Binding Proteins metabolism, Molecular Sequence Data, Rats, Blotting, Southern, DNA metabolism, Sulfuric Acid Esters
- Published
- 1993
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.