201
|
Nguyen LXT, Raval A, Garcia JS, Mitchell BS. Regulation of Ribosomal Gene Expression in Cancer. J Cell Physiol 2015; 230:1181-8. [DOI: 10.1002/jcp.24854] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 10/16/2014] [Indexed: 12/20/2022]
Affiliation(s)
- Le Xuan Truong Nguyen
- Departments of Medicine and Chemical and Systems Biology; Stanford Cancer Institute; Stanford University School of Medicine; Stanford California
| | - Aparna Raval
- Departments of Medicine and Chemical and Systems Biology; Stanford Cancer Institute; Stanford University School of Medicine; Stanford California
| | - Jacqueline S. Garcia
- Departments of Medicine and Chemical and Systems Biology; Stanford Cancer Institute; Stanford University School of Medicine; Stanford California
| | - Beverly S. Mitchell
- Departments of Medicine and Chemical and Systems Biology; Stanford Cancer Institute; Stanford University School of Medicine; Stanford California
| |
Collapse
|
202
|
Farley KI, Surovtseva Y, Merkel J, Baserga SJ. Determinants of mammalian nucleolar architecture. Chromosoma 2015; 124:323-31. [PMID: 25670395 DOI: 10.1007/s00412-015-0507-z] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 01/22/2015] [Accepted: 01/26/2015] [Indexed: 11/30/2022]
Abstract
The nucleolus is responsible for the production of ribosomes, essential machines which synthesize all proteins needed by the cell. The structure of human nucleoli is highly dynamic and is directly related to its functions in ribosome biogenesis. Despite the importance of this organelle, the intricate relationship between nucleolar structure and function remains largely unexplored. How do cells control nucleolar formation and function? What are the minimal requirements for making a functional nucleolus? Here we review what is currently known regarding mammalian nucleolar formation at nucleolar organizer regions (NORs), which can be studied by observing the dissolution and reformation of the nucleolus during each cell division. Additionally, the nucleolus can be examined by analyzing how alterations in nucleolar function manifest in differences in nucleolar architecture. Furthermore, changes in nucleolar structure and function are correlated with cancer, highlighting the importance of studying the determinants of nucleolar formation.
Collapse
Affiliation(s)
- Katherine I Farley
- Department of Molecular Biophysics & Biochemistry, Yale University School of Medicine, New Haven, CT, 06520, USA
| | | | | | | |
Collapse
|
203
|
|
204
|
Günther K, Paradowska-Dogan A, Bärmann B, Klein H, von Eichel-Streiber C, Hartley R, Weidner W, Behr R, Steger K. Expression of sperm-specific protamines impairs bacterial and eukaryotic cell proliferation. Histochem Cell Biol 2015; 143:599-609. [DOI: 10.1007/s00418-015-1309-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2015] [Indexed: 10/24/2022]
|
205
|
Farrell NP. Multi-platinum anti-cancer agents. Substitution-inert compounds for tumor selectivity and new targets. Chem Soc Rev 2015; 44:8773-85. [DOI: 10.1039/c5cs00201j] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Substitution-inert polynuclear platinum complexes are inherently dual-function anti-cancer agents combining extra and intra-cellular effects in one structural chemotype.
Collapse
Affiliation(s)
- N. P. Farrell
- Department of Chemistry
- Virginia Commonwealth University
- Richmond
- USA
| |
Collapse
|
206
|
Moreira-Ramos S, Rojas DA, Montes M, Urbina F, Miralles VJ, Maldonado E. Casein kinase 2 inhibits HomolD-directed transcription by Rrn7 in Schizosaccharomyces pombe. FEBS J 2014; 282:491-503. [PMID: 25410910 DOI: 10.1111/febs.13157] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Revised: 10/13/2014] [Accepted: 11/19/2014] [Indexed: 11/27/2022]
Abstract
In Schizosaccharomyces pombe, ribosomal protein gene (RPG) promoters contain a TATA analogue element called the HomolD box. The HomolD-binding protein Rrn7 forms a complex with the RNA polymerase II machinery. Despite the importance of ribosome biogenesis to cell survival, the mechanisms involved in the regulation of transcription of eukaryotic RPGs are unknown. In this study, we identified Rrn7 as a new substrate of the pleiotropic casein kinase 2 (CK2), which is a regulator of basal transcription. Recombinant Rrn7 from S. pombe, which is often used as a model organism for studying eukaryotic transcription, interacted with CK2 in vitro and in vivo. Furthermore, CK2-mediated phosphorylation of Rrn7 inhibited its HomolD-directed transcriptional activity and ability to bind to an oligonucleotide containing a HomolD box in vitro. Mutation of Rrn7 at Thr67 abolished these effects, indicating that this residue is a critical CK2 phosphorylation site. Finally, Rrn7 interacted with the regulatory subunit of CK2 in vivo, inhibition of CK2 in vivo potentiated ribosomal protein gene transcription, and chromatin immunoprecipitation analyses identified that the catalytic subunit of CK2 was associated with the rpk5 gene promoter in S. pombe. Taken together, these data suggest that CK2 inhibits ribosomal protein gene transcription in S. pombe via phosphorylation of Rrn7 at Thr67.
Collapse
|
207
|
Peterson EJ, Menon VR, Gatti L, Kipping R, Dewasinghe D, Perego P, Povirk LF, Farrell NP. Nucleolar targeting by platinum: p53-independent apoptosis follows rRNA inhibition, cell-cycle arrest, and DNA compaction. Mol Pharm 2014; 12:287-97. [PMID: 25407898 PMCID: PMC4334294 DOI: 10.1021/mp5006867] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
![]()
TriplatinNC
is a highly positively charged, substitution-inert
derivative of the phase II clinical anticancer drug, BBR3464. Such
substitution-inert complexes form a distinct subset of polynuclear
platinum complexes (PPCs) interacting with DNA and other biomolecules
through noncovalent interactions. Rapid cellular entry is facilitated
via interaction with cell surface glycosoaminoglycans and is a mechanism
unique to PPCs. Nanoscale secondary ion mass spectrometry (nanoSIMS)
showed rapid distribution within cytoplasmic and nucleolar compartments,
but not the nucleus. In this article, the downstream effects of nucleolar
localization are described. In human colon carcinoma cells, HCT116,
the production rate of 47S rRNA precursor transcripts was dramatically
reduced as an early event after drug treatment. Transcriptional inhibition
of rRNA was followed by a robust G1 arrest, and activation
of apoptotic proteins caspase-8, -9, and -3 and PARP-1 in a p53-independent
manner. Using cell synchronization and flow cytometry, it was determined
that cells treated while in G1 arrest immediately, but
cells treated in S or G2 successfully complete mitosis.
Twenty-four hours after treatment, the majority of cells finally arrest
in G1, but nearly one-third contained highly compacted
DNA; a distinct biological feature that cannot be associated with
mitosis, senescence, or apoptosis. This unique effect mirrored the
efficient condensation of tRNA and DNA in cell-free systems. The combination
of DNA compaction and apoptosis by TriplatinNC treatment conferred
striking activity in platinum-resistant and/or p53 mutant or null
cell lines. Taken together, our results support that the biological
activity of TriplatinNC reflects reduced metabolic deactivation (substitution-inert
compound not reactive to sulfur nucleophiles), high cellular accumulation,
and novel consequences of high-affinity noncovalent DNA binding, producing
a new profile and a further shift in the structure–activity
paradigms for antitumor complexes.
Collapse
Affiliation(s)
- Erica J Peterson
- Department of Chemistry and ‡Massey Cancer Center, Virginia Commonwealth University , Richmond, Virginia 23284, United States
| | | | | | | | | | | | | | | |
Collapse
|
208
|
Lopez-Camacho C, van Wijnen AJ, Lian JB, Stein JL, Stein GS. CBFβ and the leukemogenic fusion protein CBFβ-SMMHC associate with mitotic chromosomes to epigenetically regulate ribosomal genes. J Cell Biochem 2014; 115:2155-64. [PMID: 25079347 PMCID: PMC4199869 DOI: 10.1002/jcb.24892] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 07/25/2014] [Indexed: 01/01/2023]
Abstract
Mitotic bookmarking is an epigenetic control mechanism that sustains gene expression in progeny cells; it is often found in genes related to the maintenance of cellular phenotype and growth control. RUNX transcription factors regulate a broad spectrum of RNA Polymerase (Pol II) transcribed genes important for lineage commitment but also regulate RNA Polymerase I (Pol I) driven ribosomal gene expression, thus coordinating control of cellular identity and proliferation. In this study, using fluorescence microscopy and biochemical approaches we show that the principal RUNX co-factor, CBFβ, associates with nucleolar organizing regions (NORs) during mitosis to negatively regulate RUNX-dependent ribosomal gene expression. Of clinical relevance, we establish for the first time that the leukemogenic fusion protein CBFβ-SMMHC (smooth muscle myosin heavy chain) also associates with ribosomal genes in interphase chromatin and mitotic chromosomes to promote and epigenetically sustain regulation of ribosomal genes through RUNX factor interactions. Our results demonstrate that CBFβ contributes to the transcriptional regulation of ribosomal gene expression and provide further understanding of the epigenetic role of CBFβ-SMMHC in proliferation and maintenance of the leukemic phenotype.
Collapse
Affiliation(s)
- Cesar Lopez-Camacho
- Department of Biochemistry and Vermont Cancer Center, University of Vermont College of Medicine, Burlington, VT 05405 USA
| | - Andre J. van Wijnen
- Departments of Orthopedic Surgery and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Jane B. Lian
- Department of Biochemistry and Vermont Cancer Center, University of Vermont College of Medicine, Burlington, VT 05405 USA
| | - Janet L. Stein
- Department of Biochemistry and Vermont Cancer Center, University of Vermont College of Medicine, Burlington, VT 05405 USA
| | - Gary S. Stein
- Department of Biochemistry and Vermont Cancer Center, University of Vermont College of Medicine, Burlington, VT 05405 USA
| |
Collapse
|
209
|
Viktorovskaya OV, Schneider DA. Functional divergence of eukaryotic RNA polymerases: unique properties of RNA polymerase I suit its cellular role. Gene 2014; 556:19-26. [PMID: 25445273 DOI: 10.1016/j.gene.2014.10.035] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 10/16/2014] [Accepted: 10/17/2014] [Indexed: 02/08/2023]
Abstract
Eukaryotic cells express at least three unique nuclear RNA polymerases. The selective advantage provided by this enhanced complexity is a topic of fundamental interest in cell biology. It has long been known that the gene targets and transcription initiation pathways for RNA polymerases (Pols) I, II and III are distinct; however, recent genetic, biochemical and structural data suggest that even the core enzymes have evolved unique properties. Among the three eukaryotic RNA polymerases, Pol I is considered the most divergent. Transcription of the ribosomal DNA by Pol I is unmatched in its high rate of initiation, complex organization within the nucleolus and functional connection to ribosome assembly. Furthermore, ribosome synthesis is intimately linked to cell growth and proliferation. Thus, there is intense selective pressure on Pol I. This review describes key features of Pol I transcription, discusses catalytic activities of the enzyme and focuses on recent advances in understanding its unique role among eukaryotic RNA polymerases.
Collapse
Affiliation(s)
- Olga V Viktorovskaya
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, United States
| | - David A Schneider
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, United States.
| |
Collapse
|
210
|
Peltonen K, Colis L, Liu H, Jäämaa S, Zhang Z, Af Hällström T, Moore HM, Sirajuddin P, Laiho M. Small molecule BMH-compounds that inhibit RNA polymerase I and cause nucleolar stress. Mol Cancer Ther 2014; 13:2537-46. [PMID: 25277384 DOI: 10.1158/1535-7163.mct-14-0256] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Activation of the p53 pathway has been considered a therapeutic strategy to target cancers. We have previously identified several p53-activating small molecules in a cell-based screen. Two of the compounds activated p53 by causing DNA damage, but this modality was absent in the other four. We recently showed that one of these, BMH-21, inhibits RNA polymerase I (Pol I) transcription, causes the degradation of Pol I catalytic subunit RPA194, and has potent anticancer activity. We show here that three remaining compounds in this screen, BMH-9, BMH-22, and BMH-23, cause reorganization of nucleolar marker proteins consistent with segregation of the nucleolus, a hallmark of Pol I transcription stress. Further, the compounds destabilize RPA194 in a proteasome-dependent manner and inhibit nascent rRNA synthesis and expression of the 45S rRNA precursor. BMH-9- and BMH-22-mediated nucleolar stress was detected in ex vivo-cultured human prostate tissues indicating good tissue bioactivity. Testing of closely related analogues showed that their activities were chemically constrained. Viability screen for BMH-9, BMH-22, and BMH-23 in the NCI60 cancer cell lines showed potent anticancer activity across many tumor types. Finally, we show that the Pol I transcription stress by BMH-9, BMH-22, and BMH-23 is independent of p53 function. These results highlight the dominant impact of Pol I transcription stress on p53 pathway activation and bring forward chemically novel lead molecules for Pol I inhibition, and, potentially, cancer targeting.
Collapse
Affiliation(s)
- Karita Peltonen
- Center for Drug Research, University of Helsinki, Helsinki, Finland
| | - Laureen Colis
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hester Liu
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sari Jäämaa
- Center for Drug Research, University of Helsinki, Helsinki, Finland
| | - Zhewei Zhang
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Taija Af Hällström
- Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland
| | - Henna M Moore
- Center for Drug Research, University of Helsinki, Helsinki, Finland
| | - Paul Sirajuddin
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Marikki Laiho
- Center for Drug Research, University of Helsinki, Helsinki, Finland. Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland. Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
211
|
Randall E, Young V, Sierotzki H, Scalliet G, Birch PRJ, Cooke DEL, Csukai M, Whisson SC. Sequence diversity in the large subunit of RNA polymerase I contributes to Mefenoxam insensitivity in Phytophthora infestans. MOLECULAR PLANT PATHOLOGY 2014; 15:664-76. [PMID: 24521429 PMCID: PMC6638662 DOI: 10.1111/mpp.12124] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Phenylamide fungicides have been widely used for the control of oomycete-incited plant diseases for over 30 years. Insensitivity to this chemical class of fungicide was recorded early in its usage history, but the precise protein(s) conditioning insensitivity has proven difficult to determine. To determine the genetic basis of insensitivity and to inform strategies for the cloning of the gene(s) responsible, genetic crosses were established between Mefenoxam sensitive and intermediate insensitive isolates of Phytophthora infestans, the potato late blight pathogen. F1 progeny showed the expected semi-dominant phenotypes for Mefenoxam insensitivity and suggested the involvement of multiple loci, complicating the positional cloning of the gene(s) conditioning insensitivity to Mefenoxam. Instead, a candidate gene strategy was used, based on previous observations that the primary effect of phenylamide compounds is to inhibit ribosomal RNA synthesis. The subunits of RNA polymerase I (RNApolI) were sequenced from sensitive and insensitive isolates and F1 progeny. Single nucleotide polymorphisms (SNPs) specific to insensitive field isolates were identified in the gene encoding the large subunit of RNApolI. In a survey of field isolates, SNP T1145A (Y382F) showed an 86% association with Mefenoxam insensitivity. Isolates not showing this association belonged predominantly to one P. infestans genotype. The transfer of the 'insensitive' allele of RPA190 to a sensitive isolate yielded transgenic lines that were insensitive to Mefenoxam. These results demonstrate that sequence variation in RPA190 contributes to insensitivity to Mefenoxam in P. infestans.
Collapse
Affiliation(s)
- Eva Randall
- Cell and Molecular Sciences, James Hutton Institute, Invergowrie, Dundee, DD2 5DA, UK
| | | | | | | | | | | | | | | |
Collapse
|
212
|
Deschamps-Francoeur G, Garneau D, Dupuis-Sandoval F, Roy A, Frappier M, Catala M, Couture S, Barbe-Marcoux M, Abou-Elela S, Scott MS. Identification of discrete classes of small nucleolar RNA featuring different ends and RNA binding protein dependency. Nucleic Acids Res 2014; 42:10073-85. [PMID: 25074380 PMCID: PMC4150776 DOI: 10.1093/nar/gku664] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 07/08/2014] [Accepted: 07/09/2014] [Indexed: 12/13/2022] Open
Abstract
Small nucleolar RNAs (snoRNAs) are among the first discovered and most extensively studied group of small non-coding RNA. However, most studies focused on a small subset of snoRNAs that guide the modification of ribosomal RNA. In this study, we annotated the expression pattern of all box C/D snoRNAs in normal and cancer cell lines independent of their functions. The results indicate that C/D snoRNAs are expressed as two distinct forms differing in their ends with respect to boxes C and D and in their terminal stem length. Both forms are overexpressed in cancer cell lines but display a conserved end distribution. Surprisingly, the long forms are more dependent than the short forms on the expression of the core snoRNP protein NOP58, thought to be essential for C/D snoRNA production. In contrast, a subset of short forms are dependent on the splicing factor RBFOX2. Analysis of the potential secondary structure of both forms indicates that the k-turn motif required for binding of NOP58 is less stable in short forms which are thus less likely to mature into a canonical snoRNP. Taken together the data suggest that C/D snoRNAs are divided into at least two groups with distinct maturation and functional preferences.
Collapse
Affiliation(s)
- Gabrielle Deschamps-Francoeur
- Département de biochimie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec J1E 4K8, Canada
| | - Daniel Garneau
- Laboratoire de génomique fonctionnelle de l'Université de Sherbrooke, Québec J1E 4K8, Canada Département de microbiologie et d'infectiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec J1E 4K8, Canada
| | - Fabien Dupuis-Sandoval
- Département de biochimie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec J1E 4K8, Canada
| | - Audrey Roy
- Département de biochimie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec J1E 4K8, Canada
| | - Marie Frappier
- Département de biochimie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec J1E 4K8, Canada
| | - Mathieu Catala
- Laboratoire de génomique fonctionnelle de l'Université de Sherbrooke, Québec J1E 4K8, Canada Département de microbiologie et d'infectiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec J1E 4K8, Canada
| | - Sonia Couture
- Laboratoire de génomique fonctionnelle de l'Université de Sherbrooke, Québec J1E 4K8, Canada Département de microbiologie et d'infectiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec J1E 4K8, Canada
| | - Mélissa Barbe-Marcoux
- Département de biochimie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec J1E 4K8, Canada
| | - Sherif Abou-Elela
- Laboratoire de génomique fonctionnelle de l'Université de Sherbrooke, Québec J1E 4K8, Canada Département de microbiologie et d'infectiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec J1E 4K8, Canada
| | - Michelle S Scott
- Département de biochimie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec J1E 4K8, Canada
| |
Collapse
|
213
|
Diesch J, Hannan RD, Sanij E. Perturbations at the ribosomal genes loci are at the centre of cellular dysfunction and human disease. Cell Biosci 2014; 4:43. [PMID: 25949792 PMCID: PMC4422213 DOI: 10.1186/2045-3701-4-43] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Accepted: 07/27/2014] [Indexed: 01/05/2023] Open
Abstract
Ribosomal RNA (rRNA) gene (rDNA) transcription by RNA Polymerase I (Pol I) drives cell growth and underlies nucleolar structure and function, indirectly coordinating many fundamental cellular processes. The importance of keeping rDNA transcription under tight control is reflected by the fact that deranged Pol I transcription is a feature of cancer and other human disorders. In this review, we discuss multiple aspects of rDNA function including the relationship between Pol I transcription and proliferative capacity, the role of Pol I transcription in mediating nucleolar structure and integrity, and rDNA/nucleolar interactions with the genome and their influence on heterochromatin and global genome stability. Furthermore, we discuss how perturbations in the structure of the rDNA loci might contribute to human disease, in some cases independent of effects on ribosome biogenesis.
Collapse
Affiliation(s)
- Jeannine Diesch
- Growth Control Laboratory, Research Division, Peter MacCallum Cancer Centre, St. Andrews Place, East Melbourne, Victoria 3002, Australia ; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Ross D Hannan
- Growth Control Laboratory, Research Division, Peter MacCallum Cancer Centre, St. Andrews Place, East Melbourne, Victoria 3002, Australia ; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia ; Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia ; Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria 3010, Australia ; Division of Cancer Medicine, Peter MacCallum Cancer Centre, St. Andrews Place, East Melbourne, Victoria 3002, Australia ; School of Biomedical Sciences, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Elaine Sanij
- Growth Control Laboratory, Research Division, Peter MacCallum Cancer Centre, St. Andrews Place, East Melbourne, Victoria 3002, Australia ; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia ; Department of Pathology, University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
214
|
mTORC1 signaling controls multiple steps in ribosome biogenesis. Semin Cell Dev Biol 2014; 36:113-20. [PMID: 25148809 DOI: 10.1016/j.semcdb.2014.08.004] [Citation(s) in RCA: 188] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 08/07/2014] [Accepted: 08/11/2014] [Indexed: 02/06/2023]
Abstract
Ribosome biogenesis is critical for cells to generate the ribosomes they need for protein synthesis in order to survive, grow and proliferate. It is a complex process, involving the coordinated production of four different RNA species and about 80 proteins, as well as their assembly into functional ribosomal subunits. Given its high demand for amino acids and nucleotides, it is also a metabolically expensive process for the cell. The mammalian target of rapamycin complex 1 (mTORC1) is a protein kinases which is activated by nutrients, anabolic hormones and oncogenic signaling pathways. mTORC1 positively regulates several steps in ribosome biogenesis, including ribosomal RNA transcription, the synthesis of ribosomal proteins and other components required for ribosome assembly. mTORC1 can thus coordinate stimuli which promote ribosome production with the various steps involved in this process. Although important advances have been made in our understanding of mTORC1 signaling, major questions remain about the molecular mechanisms by which it regulates ribosome biogenesis.
Collapse
|
215
|
Knutson BA, Luo J, Ranish J, Hahn S. Architecture of the Saccharomyces cerevisiae RNA polymerase I Core Factor complex. Nat Struct Mol Biol 2014; 21:810-6. [PMID: 25132180 PMCID: PMC4219626 DOI: 10.1038/nsmb.2873] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 07/16/2014] [Indexed: 12/31/2022]
Abstract
Core Factor (CF) is a conserved RNA polymerase (Pol) I general transcription factor and is comprised of Rrn6, Rrn11, and the TFIIB-related subunit Rrn7. CF binds TBP, Pol I, and the regulatory factors Rrn3 and UAF. We used chemical crosslinking-mass spectrometry (CXMS) to determine the molecular architecture of CF and its interactions with TBP. The CF subunits assemble through an interconnected network of interactions between five structural domains that are conserved in orthologous subunits of the human Pol I factor SL1. The crosslinking-derived model was validated through a series of genetic and biochemical assays. Our combined results show the architecture of CF and the functions of the CF subunits in assembly of the complex. We extend these findings to model how CF assembles into the Pol I preinitiation complex, providing new insight into the roles of CF, TBP and Rrn3.
Collapse
Affiliation(s)
- Bruce A Knutson
- 1] Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA. [2]
| | - Jie Luo
- Institute for Systems Biology, Seattle, Washington, USA
| | | | - Steven Hahn
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| |
Collapse
|
216
|
Allen KD, Gourov AV, Harte C, Gao P, Lee C, Sylvain D, Splett JM, Oxberry WC, van de Nes PS, Troy-Regier MJ, Wolk J, Alarcon JM, Hernández AI. Nucleolar integrity is required for the maintenance of long-term synaptic plasticity. PLoS One 2014; 9:e104364. [PMID: 25089620 PMCID: PMC4121280 DOI: 10.1371/journal.pone.0104364] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 07/10/2014] [Indexed: 01/10/2023] Open
Abstract
Long-term memory (LTM) formation requires new protein synthesis and new gene expression. Based on our work in Aplysia, we hypothesized that the rRNA genes, stimulation-dependent targets of the enzyme Poly(ADP-ribose) polymerase-1 (PARP-1), are primary effectors of the activity-dependent changes in synaptic function that maintain synaptic plasticity and memory. Using electrophysiology, immunohistochemistry, pharmacology and molecular biology techniques, we show here, for the first time, that the maintenance of forskolin-induced late-phase long-term potentiation (L-LTP) in mouse hippocampal slices requires nucleolar integrity and the expression of new rRNAs. The activity-dependent upregulation of rRNA, as well as L-LTP expression, are poly(ADP-ribosyl)ation (PAR) dependent and accompanied by an increase in nuclear PARP-1 and Poly(ADP) ribose molecules (pADPr) after forskolin stimulation. The upregulation of PARP-1 and pADPr is regulated by Protein kinase A (PKA) and extracellular signal-regulated kinase (ERK)--two kinases strongly associated with long-term plasticity and learning and memory. Selective inhibition of RNA Polymerase I (Pol I), responsible for the synthesis of precursor rRNA, results in the segmentation of nucleoli, the exclusion of PARP-1 from functional nucleolar compartments and disrupted L-LTP maintenance. Taken as a whole, these results suggest that new rRNAs (28S, 18S, and 5.8S ribosomal components)--hence, new ribosomes and nucleoli integrity--are required for the maintenance of long-term synaptic plasticity. This provides a mechanistic link between stimulation-dependent gene expression and the new protein synthesis known to be required for memory consolidation.
Collapse
Affiliation(s)
- Kim D. Allen
- Department of Pathology, State University of New York, Downstate Medical Center, Brooklyn, New York, United States of America
- Department of Biology, School of Science, Health and Technology, City University of New York, Medgar Evers College, Brooklyn, New York, United States of America
| | - Andrei V. Gourov
- Department of Pathology, State University of New York, Downstate Medical Center, Brooklyn, New York, United States of America
| | - Christopher Harte
- Department of Pathology, State University of New York, Downstate Medical Center, Brooklyn, New York, United States of America
| | - Peng Gao
- Department of Pathology, State University of New York, Downstate Medical Center, Brooklyn, New York, United States of America
| | - Clarice Lee
- Department of Pathology, State University of New York, Downstate Medical Center, Brooklyn, New York, United States of America
| | - Darlene Sylvain
- Department of Pathology, State University of New York, Downstate Medical Center, Brooklyn, New York, United States of America
| | - Joshua M. Splett
- Department of Pathology, State University of New York, Downstate Medical Center, Brooklyn, New York, United States of America
| | - William C. Oxberry
- Department of Pathology, State University of New York, Downstate Medical Center, Brooklyn, New York, United States of America
- The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York, Downstate Medical Center, Brooklyn, New York, United States of America
| | - Paula S. van de Nes
- Departments of Physiology and Pharmacology, State University of New York, Downstate Medical Center, Brooklyn, New York, United States of America
| | - Matthew J. Troy-Regier
- Departments of Physiology and Pharmacology, State University of New York, Downstate Medical Center, Brooklyn, New York, United States of America
| | - Jason Wolk
- Department of Pathology, State University of New York, Downstate Medical Center, Brooklyn, New York, United States of America
| | - Juan M. Alarcon
- Department of Pathology, State University of New York, Downstate Medical Center, Brooklyn, New York, United States of America
- The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York, Downstate Medical Center, Brooklyn, New York, United States of America
- * E-mail: (JMA); (AIH)
| | - A. Iván Hernández
- Department of Pathology, State University of New York, Downstate Medical Center, Brooklyn, New York, United States of America
- The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York, Downstate Medical Center, Brooklyn, New York, United States of America
- * E-mail: (JMA); (AIH)
| |
Collapse
|
217
|
Abstract
Key Points
Akt/FLNA/TIF-90 signaling regulates rRNA synthesis in acute myelogenous leukemia cells. Direct targeting of Akt has potential therapeutic applications in acute myelogenous leukemia treatment.
Collapse
|
218
|
Rothblum K, Hu Q, Penrod Y, Rothblum LI. Selective inhibition of rDNA transcription by a small-molecule peptide that targets the interface between RNA polymerase I and Rrn3. Mol Cancer Res 2014; 12:1586-96. [PMID: 25033839 DOI: 10.1158/1541-7786.mcr-14-0229] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
UNLABELLED The interface between the polymerase I-associated factor Rrn3 and the 43-kDa subunit of RNA polymerase I is essential to the recruitment of Pol I to the preinitiation complex on the rDNA promoter. In silico analysis identified an evolutionarily conserved 22 amino acid peptide within rpa43 that is both necessary and sufficient to mediate the interaction between rpa43 and Rrn3. This peptide inhibited rDNA transcription in vitro, while a control peptide did not. To determine the effect of the peptide in cultured cells, the peptide was coupled to the HIV TAT peptide to facilitate transduction into cells. The wild-type peptide, but not control peptides, inhibited Pol I transcription and cell division. In addition, the peptide induced cell death, consistent with other observations that "nucleolar stress" results in the death of tumor cells. The 22mer is a small-molecule inhibitor of rDNA transcription that is specific for the interaction between Rrn3 and rpa43, as such it represents an original way to interfere with cell growth. IMPLICATIONS These results demonstrate a potentially novel pharmaceutical target for the therapeutic treatment of cancer cells.
Collapse
Affiliation(s)
- Katrina Rothblum
- Department of Cell Biology, University of Oklahoma College of Medicine, Oklahoma City, Oklahoma
| | - Qiyue Hu
- South Cove Community Health Care Center, Quincy, Massachusetts
| | - Yvonne Penrod
- Department of Cell Biology, University of Oklahoma College of Medicine, Oklahoma City, Oklahoma
| | - Lawrence I Rothblum
- Department of Cell Biology, University of Oklahoma College of Medicine, Oklahoma City, Oklahoma.
| |
Collapse
|
219
|
Su L, Zheng H, Li Z, Qiu J, Chen S, Liu J, Ou TM, Tan JH, Gu LQ, Huang ZS, Li D. Mechanistic studies on the anticancer activity of 2,4-disubstituted quinazoline derivative. Biochim Biophys Acta Gen Subj 2014; 1840:3123-30. [PMID: 25018006 DOI: 10.1016/j.bbagen.2014.07.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 06/23/2014] [Accepted: 07/02/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND Accelerated proliferation of solid tumor and hematologic cancer cells is related to accelerated transcription of ribosomal DNA by the RNA polymerase I to produce elevated level of ribosomal RNA. Therefore, down-regulation of RNA polymerase I transcription in cancer cells is an important anticancer therapeutic strategy. METHODS A variety of methods were used, including cloning, expression and purification of protein, electrophoretic mobility shift assay (EMSA), circular dichroic (CD) spectroscopy, CD-melting, isothermal titration calorimetry (ITC), chromatin immunoprecipitation (Ch-IP), RNA interference, RT-PCR, Western blot, and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) cell assay. RESULTS Our results showed that 2,4-disubstituted quinazoline derivative Sysu12d could down-regulate c-myc through stabilization of c-myc promoter G-quadruplex, resulting in down-regulation of nucleolin expression. Sysu12d could also disrupt nucleolin/G-quadruplex complex. Both of the above contributed to the down-regulation of ribosomal RNA synthesis, followed by activation of p53 and then cancer cell apoptosis. CONCLUSIONS These mechanistic studies set up the basis for further development of Sysu12d as a new type of lead compound for cancer treatment. GENERAL SIGNIFICANCE 2,4-Disubstituted quinazoline derivatives may have multi-functional effect for cancer treatment.
Collapse
Affiliation(s)
- Lijuan Su
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou University City, 132 Waihuan East Road, Guangzhou 510006, PR China
| | - Huaqin Zheng
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou University City, 132 Waihuan East Road, Guangzhou 510006, PR China
| | - Zeng Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou University City, 132 Waihuan East Road, Guangzhou 510006, PR China
| | - Jun Qiu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou University City, 132 Waihuan East Road, Guangzhou 510006, PR China
| | - Siqi Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou University City, 132 Waihuan East Road, Guangzhou 510006, PR China
| | - Jinggong Liu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou University City, 132 Waihuan East Road, Guangzhou 510006, PR China
| | - Tian-Miao Ou
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou University City, 132 Waihuan East Road, Guangzhou 510006, PR China
| | - Jia-Heng Tan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou University City, 132 Waihuan East Road, Guangzhou 510006, PR China
| | - Lian-Quan Gu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou University City, 132 Waihuan East Road, Guangzhou 510006, PR China
| | - Zhi-Shu Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou University City, 132 Waihuan East Road, Guangzhou 510006, PR China
| | - Ding Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou University City, 132 Waihuan East Road, Guangzhou 510006, PR China.
| |
Collapse
|
220
|
Rocchi L, Barbosa AJM, Onofrillo C, Del Rio A, Montanaro L. Inhibition of human dyskerin as a new approach to target ribosome biogenesis. PLoS One 2014; 9:e101971. [PMID: 25010840 PMCID: PMC4092089 DOI: 10.1371/journal.pone.0101971] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Accepted: 06/13/2014] [Indexed: 11/19/2022] Open
Abstract
The product of the DKC1 gene, dyskerin, is required for both ribosome biogenesis and telomerase complex stabilization. Targeting these cellular processes has been explored for the development of drugs to selectively or preferentially kill cancer cells. Presently, intense research is conducted involving the identification of new biological targets whose modulation may simultaneously interfere with multiple cellular functions that are known to be hyper-activated by neoplastic transformations. Here, we report, for the first time, the computational identification of small molecules able to inhibit dyskerin catalytic activity. Different insilico techniques were applied to select compounds and analyze the binding modes and the interaction patterns of ligands in the human dyskerin catalytic site. We also describe a newly developed and optimized fast real-time PCR assay that was used to detect dyskerin pseudouridylation activity invitro. The identification of new dyskerin inhibitors constitutes the first proof of principle that the pseudouridylation activity can be modulated by means of small molecule agents. Therefore, the presented results, obtained through the usage of computational tools and experimental validation, indicate an alternative therapeutic strategy to target ribosome biogenesis pathway.
Collapse
Affiliation(s)
- Laura Rocchi
- Department of Experimental, Diagnostic and Specialty Medicine, Alma Mater Studiorum, University of Bologna, Bologna, Italy
- Surgical Pathology Unit, University Hospital of Parma, Parma, Italy
| | - Arménio J. M. Barbosa
- Department of Experimental, Diagnostic and Specialty Medicine, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Carmine Onofrillo
- Department of Experimental, Diagnostic and Specialty Medicine, Alma Mater Studiorum, University of Bologna, Bologna, Italy
- “Giorgio Prodi” Interdepartmental Cancer Research Centre, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Alberto Del Rio
- Department of Experimental, Diagnostic and Specialty Medicine, Alma Mater Studiorum, University of Bologna, Bologna, Italy
- Institute for Organic Chemistry and Photoreactivity, National Research Council, Bologna, Italy
- * E-mail: (ADR); (LM)
| | - Lorenzo Montanaro
- Department of Experimental, Diagnostic and Specialty Medicine, Alma Mater Studiorum, University of Bologna, Bologna, Italy
- * E-mail: (ADR); (LM)
| |
Collapse
|
221
|
The nucleolus—guardian of cellular homeostasis and genome integrity. Chromosoma 2014; 122:487-97. [PMID: 24022641 DOI: 10.1007/s00412-013-0430-0] [Citation(s) in RCA: 148] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 08/05/2013] [Indexed: 01/25/2023]
Abstract
All organisms sense and respond to conditions that stress their homeostasis by downregulating the synthesis of rRNA and ribosome biogenesis, thus designating the nucleolus as the central hub in coordinating the cellular stress response. One of the most intriguing roles of the nucleolus, long regarded as a mere ribosome-producing factory, is its participation in monitoring cellular stress signals and transmitting them to the RNA polymerase I (Pol I) transcription machinery. As rRNA synthesis is a most energy-consuming process, switching off transcription of rRNA genes is an effective way of saving the energy required to maintain cellular homeostasis during acute stress. The Pol I transcription machinery is the key convergence point that collects and integrates a vast array of information from cellular signaling cascades to regulate ribosome production which, in turn, guides cell growth and proliferation. This review focuses on the mechanisms that link cell physiology to rDNA silencing, a prerequisite for nucleolar integrity and cell survival.
Collapse
|
222
|
Murano K, Okuwaki M, Momose F, Kumakura M, Ueshima S, Newbold RF, Nagata K. Reconstitution of human rRNA gene transcription in mouse cells by a complete SL1 complex. J Cell Sci 2014; 127:3309-19. [PMID: 24928901 DOI: 10.1242/jcs.146787] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
An important characteristic of the transcription of a ribosomal RNA gene (rDNA) mediated by DNA-dependent RNA polymerase (Pol) I is its stringent species specificity. SL1/TIF-IB is a key complex for species specificity, but its functional complex has not been reconstituted. Here, we established a novel and highly sensitive monitoring system for Pol I transcription to reconstitute the SL1 activity in which a transcript harboring a reporter gene synthesized by Pol I is amplified and converted into translatable mRNA by the influenza virus RNA-dependent RNA polymerase. Using this monitoring system, we reconstituted Pol I transcription from the human rDNA promoter in mouse cells by expressing four human TATA-binding protein (TBP)-associated factors (TAFIs) in the SL1 complex. The reconstituted SL1 also re-activated human rDNA transcription in mouse A9 cells carrying an inactive human chromosome 21 that contains the rDNA cluster. Chimeric SL1 complexes containing human and mouse TAFIs could be formed, but these complexes were inactive for human rDNA transcription. We conclude that four human TAFIs are necessary and sufficient to overcome the barrier of species specificity for human rDNA transcription in mouse cells.
Collapse
Affiliation(s)
- Kensaku Murano
- Department of Infection Biology, Faculty of Medicine and Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Mitsuru Okuwaki
- Department of Infection Biology, Faculty of Medicine and Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Fumitaka Momose
- Kitasato Institute for Life Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Michiko Kumakura
- Department of Infection Biology, Faculty of Medicine and Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Shuhei Ueshima
- Department of Infection Biology, Faculty of Medicine and Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Robert F Newbold
- Institute of Cancer Genetics and Pharmacogenomics, Division of Biosciences, School of Health Sciences and Social Care, Brunel University, Uxbridge, Middlesex UB8 3PH, UK
| | - Kyosuke Nagata
- Department of Infection Biology, Faculty of Medicine and Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
223
|
Sarshad AA, Corcoran M, Al-Muzzaini B, Borgonovo-Brandter L, Von Euler A, Lamont D, Visa N, Percipalle P. Glycogen synthase kinase (GSK) 3β phosphorylates and protects nuclear myosin 1c from proteasome-mediated degradation to activate rDNA transcription in early G1 cells. PLoS Genet 2014; 10:e1004390. [PMID: 24901984 PMCID: PMC4046919 DOI: 10.1371/journal.pgen.1004390] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 04/03/2014] [Indexed: 11/17/2022] Open
Abstract
Nuclear myosin 1c (NM1) mediates RNA polymerase I (pol I) transcription activation and cell cycle progression by facilitating PCAF-mediated H3K9 acetylation, but the molecular mechanism by which NM1 is regulated remains unclear. Here, we report that at early G1 the glycogen synthase kinase (GSK) 3β phosphorylates and stabilizes NM1, allowing for NM1 association with the chromatin. Genomic analysis by ChIP-Seq showed that this mechanism occurs on the rDNA as active GSK3β selectively occupies the gene. ChIP assays and transmission electron microscopy in GSK3β−/− mouse embryonic fibroblasts indicated that at G1 rRNA synthesis is suppressed due to decreased H3K9 acetylation leading to a chromatin state incompatible with transcription. We found that GSK3β directly phosphorylates the endogenous NM1 on a single serine residue (Ser-1020) located within the NM1 C-terminus. In G1 this phosphorylation event stabilizes NM1 and prevents NM1 polyubiquitination by the E3 ligase UBR5 and proteasome-mediated degradation. We conclude that GSK3β-mediated phosphorylation of NM1 is required for pol I transcription activation. Nuclear actin and myosin are essential regulators of gene expression. At the exit of mitosis, nuclear myosin 1c (NM1) mediates RNA polymerase I (pol I) transcription activation and cell cycle progression by modulating assembly of the chromatin remodeling complex WICH with the subunits WSTF and SNF2h and, crucially, facilitating H3K9 acetylation by the histone acetyl transferase PCAF. The molecular mechanism by which NM1 is regulated remains however unknown. Here, we conducted a genome-wide screen and demonstrate that GSK3β is selectively coupled to the rDNA transcription unit. In embryonic fibroblasts lacking GSK3β there is a significant drop in rRNA synthesis levels and the rDNA is devoid of actin, NM1 and SNF2h. Concomitantly with a transcriptional block we reveal decreased levels of histone H3 acetylation by the histone acetyl transferase PCAF. At G1, transcriptional repression in the GSK3β knockout mouse embryonic fibroblasts, leads to NM1 ubiquitination by the E3 ligase UBR5 and proteasome-mediated degradation. We conclude that GSK3β suppresses NM1 degradation through the ubiquitin-proteasome system, facilitates NM1 association with the rDNA chromatin and transcription activation at G1. We therefore propose a novel and fundamental role for GSK3β as essential regulator of rRNA synthesis and cell cycle progression.
Collapse
Affiliation(s)
- Aishe A Sarshad
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Martin Corcoran
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Bader Al-Muzzaini
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | | | - Anne Von Euler
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Douglas Lamont
- FingerPrints Proteomics Facility, College of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Neus Visa
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | | |
Collapse
|
224
|
Thomson E, Ferreira-Cerca S, Hurt E. Eukaryotic ribosome biogenesis at a glance. J Cell Sci 2014; 126:4815-21. [PMID: 24172536 DOI: 10.1242/jcs.111948] [Citation(s) in RCA: 206] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Ribosomes play a pivotal role in the molecular life of every cell. Moreover, synthesis of ribosomes is one of the most energetically demanding of all cellular processes. In eukaryotic cells, ribosome biogenesis requires the coordinated activity of all three RNA polymerases and the orchestrated work of many (>200) transiently associated ribosome assembly factors. The biogenesis of ribosomes is a tightly regulated activity and it is inextricably linked to other fundamental cellular processes, including growth and cell division. Furthermore, recent studies have demonstrated that defects in ribosome biogenesis are associated with several hereditary diseases. In this Cell Science at a Glance article and the accompanying poster, we summarise the current knowledge on eukaryotic ribosome biogenesis, with an emphasis on the yeast model system.
Collapse
Affiliation(s)
- Emma Thomson
- Biochemistry Center (BZH), University of Heidelberg, Im Neuenheimer Feld 328, 69120 Heidelberg, Germany
| | | | | |
Collapse
|
225
|
The nucleolar size is associated to the methylation status of ribosomal DNA in breast carcinomas. BMC Cancer 2014; 14:361. [PMID: 24884608 PMCID: PMC4062283 DOI: 10.1186/1471-2407-14-361] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Accepted: 04/30/2014] [Indexed: 01/21/2023] Open
Abstract
Background There is a body of evidence that shows a link between tumorigenesis and ribosome biogenesis. The precursor of mature 18S, 28S and 5.8S ribosomal RNAs is transcribed from the ribosomal DNA gene (rDNA), which exists as 300–400 copies in the human diploid genome. Approximately one half of these copies are epigenetically silenced, but the exact role of epigenetic regulation on ribosome biogenesis is not completely understood. In this study we analyzed the methylation profiles of the rDNA promoter and of the 5’ regions of 18S and 28S in breast cancer. Methods We analyzed rDNA methylation in 68 breast cancer tissues of which the normal counterpart was partially available (45/68 samples) using the MassARRAY EpiTYPER assay, a sensitive and quantitative method with single base resolution. Results We found that rDNA locus tended to be hypermethylated in tumor compared to matched normal breast tissues and that the DNA methylation level of several CpG units within the rDNA locus was associated to nuclear grade and to nucleolar size of tumor tissues. In addition we identified a subgroup of samples in which large nucleoli were associated with very limited or absent rDNA hypermethylation in tumor respect to matched normal tissue. Conclusions In conclusion, we suggest that rDNA is an important target of epigenetic regulation in breast tumors and that rDNA methylation level is associated to nucleolar size.
Collapse
|
226
|
Dichamp I, Séité P, Agius G, Barbarin A, Beby-Defaux A. Human papillomavirus 16 oncoprotein E7 stimulates UBF1-mediated rDNA gene transcription, inhibiting a p53-independent activity of p14ARF. PLoS One 2014; 9:e96136. [PMID: 24798431 PMCID: PMC4010441 DOI: 10.1371/journal.pone.0096136] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 04/03/2014] [Indexed: 11/18/2022] Open
Abstract
High-risk human papillomavirus oncoproteins E6 and E7 play a major role in HPV-related cancers. One of the main functions of E7 is the degradation of pRb, while E6 promotes the degradation of p53, inactivating the p14ARF-p53 pathway. pRb and p14ARF can repress ribosomal DNA (rDNA) transcription in part by targeting the Upstream Binding Factor 1 (UBF1), a key factor in the activation of RNA polymerase I machinery. We showed, through ectopic expression and siRNA silencing of p14ARF and/or E7, that E7 stimulates UBF1-mediated rDNA gene transcription, partly because of increased levels of phosphorylated UBF1, preventing the inhibitory function of p14ARF. Unexpectedly, activation of rDNA gene transcription was higher in cells co-expressing p14ARF and E7, compared to cells expressing E7 alone. We did not find a difference in P-UBF1 levels that could explain this data. However, p14ARF expression induced E7 to accumulate into the nucleolus, where rDNA transcription takes place, providing an opportunity for E7 to interact with nucleolar proteins involved in this process. GST-pull down and co-immunoprecipitation assays showed interactions between p14ARF, UBF1 and E7, although p14ARF and E7 are not able to directly interact. Co-expression of a pRb-binding-deficient mutant (E7C24G) and p14ARF resulted in EC24G nucleolar accumulation, but not in a significant higher activation of rDNA transcription, suggesting that the inactivation of pRb is involved in this phenomenon. Thus, p14ARF fails to prevent E7-mediated UBF1 phosphorylation, but could facilitate nucleolar pRb inactivation by targeting E7 to the nucleolus. While others have reported that p19ARF, the mouse homologue of p14ARF, inhibits some functions of E7, we showed that E7 inhibits a p53-independent function of p14ARF. These results point to a mutually functional interaction between p14ARF and E7 that might partly explain why the sustained p14ARF expression observed in most cervical pre-malignant lesions and malignancies may be ineffective.
Collapse
Affiliation(s)
- Isabelle Dichamp
- Unité de Virologie, Centre Hospitalier Universitaire de Poitiers, Faculté de Médecine et Pharmacie, Poitiers, France
| | - Paule Séité
- Equipe Emergente 2RCT «Récepteurs, Régulations, Cellules Tumorales», Université de Poitiers, Poitiers, France
| | - Gérard Agius
- Unité de Virologie, Centre Hospitalier Universitaire de Poitiers, Faculté de Médecine et Pharmacie, Poitiers, France
| | - Alice Barbarin
- Equipe Emergente 2RCT «Récepteurs, Régulations, Cellules Tumorales», Université de Poitiers, Poitiers, France
| | - Agnès Beby-Defaux
- Unité de Virologie, Centre Hospitalier Universitaire de Poitiers, Faculté de Médecine et Pharmacie, Poitiers, France
- Equipe Emergente 2RCT «Récepteurs, Régulations, Cellules Tumorales», Université de Poitiers, Poitiers, France
- * E-mail:
| |
Collapse
|
227
|
Burger K, Eick D. Functional ribosome biogenesis is a prerequisite for p53 destabilization: impact of chemotherapy on nucleolar functions and RNA metabolism. Biol Chem 2014; 394:1133-43. [PMID: 23640940 DOI: 10.1515/hsz-2013-0153] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 04/26/2013] [Indexed: 12/21/2022]
Abstract
The production and processing of ribosomal RNA is a complex and well-coordinated nucleolar process for ribosome biogenesis. Progress in understanding nucleolar structure and function has lead to the unexpected discovery of the nucleolus as a highly sensitive sensor of cellular stress and an important regulator of the tumor suppressor p53. Inhibition of ribosomal RNA metabolism has been shown to activate a signaling pathway for p53 induction. This review elucidates the potential of classical and recently developed chemotherapeutic drugs to stabilize p53 by inhibiting nucleolar functions.
Collapse
Affiliation(s)
- Kaspar Burger
- Department of Molecular Epigenetics, Helmholtz Zentrum München and Center for Integrated Protein Science Munich, Marchioninistrasse 25, 81377 Munich, Germany
| | | |
Collapse
|
228
|
Ponti D, Bellenchi GC, Puca R, Bastianelli D, Maroder M, Ragona G, Roussel P, Thiry M, Mercola D, Calogero A. The transcription factor EGR1 localizes to the nucleolus and is linked to suppression of ribosomal precursor synthesis. PLoS One 2014; 9:e96037. [PMID: 24787739 PMCID: PMC4006901 DOI: 10.1371/journal.pone.0096037] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 04/03/2014] [Indexed: 01/13/2023] Open
Abstract
EGR1 is an immediate early gene with a wide range of activities as transcription factor, spanning from regulation of cell growth to differentiation. Numerous studies show that EGR1 either promotes the proliferation of stimulated cells or suppresses the tumorigenic growth of transformed cells. Upon interaction with ARF, EGR1 is sumoylated and acquires the ability to bind to specific targets such as PTEN and in turn to regulate cell growth. ARF is mainly localized to the periphery of nucleolus where is able to negatively regulate ribosome biogenesis. Since EGR1 colocalizes with ARF under IGF-1 stimulation we asked the question of whether EGR1 also relocate to the nucleolus to interact with ARF. Here we show that EGR1 colocalizes with nucleolar markers such as fibrillarin and B23 in the presence of ARF. Western analysis of nucleolar extracts from HeLa cells was used to confirm the presence of EGR1 in the nucleolus mainly as the 100 kDa sumoylated form. We also show that the level of the ribosomal RNA precursor 47S is inversely correlated to the level of EGR1 transcripts. The EGR1 iseffective to regulate the synthesis of the 47S rRNA precursor. Then we demonstrated that EGR1 binds to the Upstream Binding Factor (UBF) leading us to hypothesize that the regulating activity of EGR1 is mediated by its interaction within the transcriptional complex of RNA polymerase I. These results confirm the presence of EGR1 in the nucleolus and point to a role for EGR1 in the control of nucleolar metabolism.
Collapse
Affiliation(s)
- Donatella Ponti
- Department of Medico-Surgical Sciences and Biotechnologies, University of Rome La Sapienza, Latina, Italy
| | | | - Rosa Puca
- Department of Medico-Surgical Sciences and Biotechnologies, University of Rome La Sapienza, Latina, Italy
| | - Daniela Bastianelli
- Department of Medico-Surgical Sciences and Biotechnologies, University of Rome La Sapienza, Latina, Italy
| | - Marella Maroder
- Department of Medico-Surgical Sciences and Biotechnologies, University of Rome La Sapienza, Latina, Italy
| | - Giuseppe Ragona
- Department of Medico-Surgical Sciences and Biotechnologies, University of Rome La Sapienza, Latina, Italy
| | - Pascal Roussel
- RNA Biology, FRE 3402 CNRS-Universitè Pierre et Marie Curie, Paris, France
| | - Marc Thiry
- Unit of Cell Biology, GIGA-Neuroscience, University of Liege, CHU SartTilman, Liege, Belgium
| | - Dan Mercola
- Department of Pathology and Laboratory Medicine, University of California, Irvine, California, United States of America
| | - Antonella Calogero
- Department of Medico-Surgical Sciences and Biotechnologies, University of Rome La Sapienza, Latina, Italy
- * E-mail:
| |
Collapse
|
229
|
Abstract
Long known as the center of ribosome synthesis, the nucleolus is connected to cell cycle regulation in more subtle ways. One is a surveillance system that reacts promptly when rRNA synthesis or processing is impaired, halting cell cycle progression. Conversely, the nucleolus also acts as a first-responder to growth-related stress signals. Here we review emerging concepts on how these "infraribosomal" links between the nucleolus and cell cycle progression operate in both forward and reverse gears. We offer perspectives on how new cancer therapeutic designs that target this infraribosomal mode of cell growth control may shape future clinical progress.
Collapse
Affiliation(s)
- Robert Y L Tsai
- Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, USA; and
| | - Thoru Pederson
- Program in Cell and Developmental Dynamics, Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
230
|
Wang Y, Huang JW, Castella M, Huntsman DG, Taniguchi T. p53 is positively regulated by miR-542-3p. Cancer Res 2014; 74:3218-27. [PMID: 24762395 DOI: 10.1158/0008-5472.can-13-1706] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The tumor suppressor p53 and miRNAs are linked through a complex network. Several miRNAs modulate p53 expression, while p53 regulates the transcription and/or biogenesis of several other miRNAs. Here, we report the development of a cell-based assay used with a library of human miRNA mimics in a high-throughput screen for miRNAs that modulate p53 expression. Overexpression of miRNA (miR)-542-3p in cancer cells elevated p53 expression, stimulated the expression of p53 targets, and inhibited cell proliferation. Mechanistically, miR-542-3p increased p53 protein stability by weakening interactions between p53 and its negative regulator MDM2. Furthermore, miR-542-3p suppressed ribosome biogenesis by downregulating a subset of ribosomal proteins such as RPS23, leading to upregulation of RPL11 and stabilization of p53. The 3'untranslated region in the RPS23 transcript contained a miR-542-3p-binding site, suggesting that RPS23 is a direct target of miR-542-3p. Our results define miR-542-3p as an important new positive regulator of p53 with potential applications in cancer treatment.
Collapse
Affiliation(s)
- Yemin Wang
- Authors' Affiliations: Divisions of Human Biology and Public Health Sciences, Howard Hughes Medical Institute, Fred Hutchinson Cancer Research Center; Molecular & Cellular Biology Program, University of Washington, Seattle, Washington; and Department of Pathology and Laboratory Medicine, Center for Translational and Applied Genomics, British Columbia Cancer Agency, University of British Columbia, Vancouver BC, CanadaAuthors' Affiliations: Divisions of Human Biology and Public Health Sciences, Howard Hughes Medical Institute, Fred Hutchinson Cancer Research Center; Molecular & Cellular Biology Program, University of Washington, Seattle, Washington; and Department of Pathology and Laboratory Medicine, Center for Translational and Applied Genomics, British Columbia Cancer Agency, University of British Columbia, Vancouver BC, Canada
| | - Jen-Wei Huang
- Authors' Affiliations: Divisions of Human Biology and Public Health Sciences, Howard Hughes Medical Institute, Fred Hutchinson Cancer Research Center; Molecular & Cellular Biology Program, University of Washington, Seattle, Washington; and Department of Pathology and Laboratory Medicine, Center for Translational and Applied Genomics, British Columbia Cancer Agency, University of British Columbia, Vancouver BC, CanadaAuthors' Affiliations: Divisions of Human Biology and Public Health Sciences, Howard Hughes Medical Institute, Fred Hutchinson Cancer Research Center; Molecular & Cellular Biology Program, University of Washington, Seattle, Washington; and Department of Pathology and Laboratory Medicine, Center for Translational and Applied Genomics, British Columbia Cancer Agency, University of British Columbia, Vancouver BC, Canada
| | - Maria Castella
- Authors' Affiliations: Divisions of Human Biology and Public Health Sciences, Howard Hughes Medical Institute, Fred Hutchinson Cancer Research Center; Molecular & Cellular Biology Program, University of Washington, Seattle, Washington; and Department of Pathology and Laboratory Medicine, Center for Translational and Applied Genomics, British Columbia Cancer Agency, University of British Columbia, Vancouver BC, Canada
| | - David George Huntsman
- Authors' Affiliations: Divisions of Human Biology and Public Health Sciences, Howard Hughes Medical Institute, Fred Hutchinson Cancer Research Center; Molecular & Cellular Biology Program, University of Washington, Seattle, Washington; and Department of Pathology and Laboratory Medicine, Center for Translational and Applied Genomics, British Columbia Cancer Agency, University of British Columbia, Vancouver BC, Canada
| | - Toshiyasu Taniguchi
- Authors' Affiliations: Divisions of Human Biology and Public Health Sciences, Howard Hughes Medical Institute, Fred Hutchinson Cancer Research Center; Molecular & Cellular Biology Program, University of Washington, Seattle, Washington; and Department of Pathology and Laboratory Medicine, Center for Translational and Applied Genomics, British Columbia Cancer Agency, University of British Columbia, Vancouver BC, Canada
| |
Collapse
|
231
|
Golomb L, Volarevic S, Oren M. p53 and ribosome biogenesis stress: the essentials. FEBS Lett 2014; 588:2571-9. [PMID: 24747423 DOI: 10.1016/j.febslet.2014.04.014] [Citation(s) in RCA: 166] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 04/04/2014] [Accepted: 04/04/2014] [Indexed: 12/18/2022]
Abstract
Cell proliferation and cell growth are two tightly linked processes, as the proliferation program cannot be executed without proper accumulation of cell mass, otherwise endangering the fate of the two daughter cells. It is therefore not surprising that ribosome biogenesis, a key element in cell growth, is regulated by many cell cycle regulators. This regulation is exerted transcriptionally and post-transcriptionally, in conjunction with numerous intrinsic and extrinsic signals. Those signals eventually converge at the nucleolus, the cellular compartment that is not only responsible for executing the ribosome biogenesis program, but also serves as a regulatory hub, responsible for integrating and transmitting multiple stress signals to the omnipotent cell fate gatekeeper, p53. In this review we discuss when, how and why p53 is activated upon ribosomal biogenesis stress, and how perturbation of this critical regulatory interplay may impact human disease.
Collapse
Affiliation(s)
- Lior Golomb
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Sinisa Volarevic
- Department of Molecular Medicine and Biotechnology, School of Medicine, University of Rijeka, Croatia
| | - Moshe Oren
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
232
|
Targeting RNA polymerase I to treat MYC-driven cancer. Oncogene 2014; 34:403-12. [PMID: 24608428 DOI: 10.1038/onc.2014.13] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 01/08/2014] [Accepted: 01/08/2014] [Indexed: 02/06/2023]
Abstract
The MYC oncoprotein and transcription factor is dysregulated in a majority of human cancers and is considered a major driver of the malignant phenotype. As such, developing drugs for effective inhibition of MYC in a manner selective to malignancies is a 'holy grail' of transcription factor-based cancer therapy. Recent advances in elucidating MYC biology in both normal cells and pathological settings were anticipated to bring inhibition of tumorigenic MYC function closer to the clinic. However, while the extensive array of cellular pathways that MYC impacts present numerous fulcrum points on which to leverage MYC's therapeutic potential, identifying the critical target(s) for MYC-specific cancer therapy has been difficult to achieve. Somewhat unexpectedly, MYC's fundamental role in regulating the 'housekeeping' process of ribosome biogenesis, one of the most ubiquitously required and conserved cell functions, may provide the Achilles' heel for therapeutically targeting MYC-driven tumors.
Collapse
|
233
|
Vlatković N, Boyd MT, Rubbi CP. Nucleolar control of p53: a cellular Achilles' heel and a target for cancer therapy. Cell Mol Life Sci 2014; 71:771-91. [PMID: 23685903 PMCID: PMC11113510 DOI: 10.1007/s00018-013-1361-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 04/07/2013] [Accepted: 04/30/2013] [Indexed: 02/07/2023]
Abstract
Nucleoli perform a crucial cell function, ribosome biogenesis, and of critical relevance to the subject of this review, they are also extremely sensitive to cellular stresses, which can cause loss of function and/or associated structural disruption. In recent years, we have learned that cells take advantage of this stress sensitivity of nucleoli, using them as stress sensors. One major protein regulated by this role of nucleoli is the tumor suppressor p53, which is activated in response to diverse cellular injuries in order to exert its onco-protective effects. Here we discuss a model of nucleolar regulation of p53, which proposes that key steps in the promotion of p53 degradation by the ubiquitin ligase MDM2 occur in nucleoli, thus providing an explanation for the observed link between nucleolar disruption and p53 stability. We review current evidence for this compartmentalization in p53 homeostasis and highlight current limitations of the model. Interestingly, a number of current chemotherapeutic agents capable of inducing a p53 response are likely to do so by targeting nucleolar functions and these compounds may serve to inform further improved therapeutic targeting of nucleoli.
Collapse
Affiliation(s)
- Nikolina Vlatković
- Cancer Research Centre, University of Liverpool, 200 London Rd, Liverpool, L3 9TA UK
| | - Mark T. Boyd
- Cancer Research Centre, University of Liverpool, 200 London Rd, Liverpool, L3 9TA UK
| | - Carlos P. Rubbi
- Cancer Research Centre, University of Liverpool, 200 London Rd, Liverpool, L3 9TA UK
| |
Collapse
|
234
|
Drygin D, O’Brien SE, Hannan RD, McArthur GA, Von Hoff DD. Targeting the nucleolus for cancer-specific activation of p53. Drug Discov Today 2014; 19:259-65. [DOI: 10.1016/j.drudis.2013.08.012] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 07/29/2013] [Accepted: 08/16/2013] [Indexed: 02/08/2023]
|
235
|
Villicaña C, Cruz G, Zurita M. The basal transcription machinery as a target for cancer therapy. Cancer Cell Int 2014; 14:18. [PMID: 24576043 PMCID: PMC3942515 DOI: 10.1186/1475-2867-14-18] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 02/21/2014] [Indexed: 01/11/2023] Open
Abstract
General transcription is required for the growth and survival of all living cells. However, tumor cells require extraordinary levels of transcription, including the transcription of ribosomal RNA genes by RNA polymerase I (RNPI) and mRNA by RNA polymerase II (RNPII). In fact, cancer cells have mutations that directly enhance transcription and are frequently required for cancer transformation. For example, the recent discovery that MYC enhances the transcription of the majority genes in the genome correlates with the fact that several transcription interfering drugs preferentially kill cancer cells. In recent years, advances in the mechanistic studies of the basal transcription machinery and the discovery of drugs that interfere with multiple components of transcription are being used to combat cancer. For example, drugs such as triptolide that targets the general transcription factors TFIIH and JQ1 to inhibit BRD4 are administered to target the high proliferative rate of cancer cells. Given the importance of finding new strategies to preferentially sensitize tumor cells, this review primarily focuses on several transcription inhibitory drugs to demonstrate that the basal transcription machinery constitutes a potential target for the design of novel cancer drugs. We highlight the drugs’ mechanisms for interfering with tumor cell survival, their importance in cancer treatment and the challenges of clinical application.
Collapse
Affiliation(s)
| | | | - Mario Zurita
- Departament of Developmental Genetics, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Mexico, Mexico.
| |
Collapse
|
236
|
Sheng J, Yu W, Gao X, Xu Z, Hu GF. Angiogenin stimulates ribosomal RNA transcription by epigenetic activation of the ribosomal DNA promoter. J Cell Physiol 2014; 229:521-9. [PMID: 24122807 DOI: 10.1002/jcp.24477] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 09/25/2013] [Indexed: 12/27/2022]
Abstract
Angiogenin (ANG) undergoes nuclear translocation and promotes ribosomal RNA (rRNA) transcription thereby enhancing cell growth and proliferation. However, the mode of action of ANG in stimulating rRNA transcription is unclear. Here, we show that ANG enhances the formation of RNA polymerase I (Pol I) pre-initiation complex at the ribosomal DNA (rDNA) promoter. ANG binds at the upstream control element (UCE) of the promoter and enhances promoter occupancy of RNA Pol I as well as the selectivity factor SL1 components TAFI 48 and TAFI 110. We also show that ANG increases the number of actively transcribing rDNA by epigenetic activation through promoter methylation and histone modification. ANG binds to histone H3, inhibits H3K9 methylation, and activates H3K4 methylation as well as H4 acetylation at the rDNA promoter. These data suggest that one of the mechanisms by which ANG stimulates rRNA transcription is through an epigenetic activation of rDNA promoter.
Collapse
Affiliation(s)
- Jinghao Sheng
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts; Institute of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | | | | | | | | |
Collapse
|
237
|
Paredes S, Villanova L, Chua KF. Molecular pathways: emerging roles of mammalian Sirtuin SIRT7 in cancer. Clin Cancer Res 2014; 20:1741-6. [PMID: 24536059 DOI: 10.1158/1078-0432.ccr-13-1547] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
SIRT7 belongs to the Sirtuin family of NAD-dependent enzymes, the members of which play diverse roles in aging, metabolism, and disease biology. Increased SIRT7 expression is observed in human cancers and growing evidence suggests important SIRT7 functions in fundamental cellular programs with an impact on oncogenic transformation and tumor biology. SIRT7 associates with chromatin, where it catalyzes selective deacetylation of lysine 18 on histone H3 (H3K18), an emerging epigenetic biomarker of aggressive tumors and poor clinical outcome in patients with cancer. Through H3K18 deacetylation at specific promoters, SIRT7 controls a tumor-suppressive gene expression program that stabilizes the transformed state of cancer cells. SIRT7 also orchestrates several molecular processes, including rRNA and tRNA synthesis, which ultimately promote the increased ribosome biogenesis necessary for tumor cell growth and proliferation. Remarkably, inactivation of SIRT7 can reverse the transformed phenotype of cancer cells and reduce their tumorigenicity in vivo. These findings place SIRT7 at the crossroads of chromatin signaling, metabolic, and tumor-regulatory pathways. Thus, SIRT7 is a promising pharmacologic target for epigenetic cancer therapy. The development of SIRT7 modulators may allow new therapeutic strategies that control tumor progression by reprogramming the chromatin landscape and biosynthetic machinery of cancer cells.
Collapse
Affiliation(s)
- Silvana Paredes
- Authors' Affiliations: Department of Medicine, Division of Endocrinology, Gerontology, and Metabolism, School of Medicine, Stanford University, Stanford; Geriatric Research, Education, and Clinical Center, VA Palo Alto Health Care System, Palo Alto, California; and Department of Experimental Medicine, Sapienza University, Rome, Italy
| | | | | |
Collapse
|
238
|
Liu R, Iadevaia V, Averous J, Taylor PM, Zhang Z, Proud CG. Impairing the production of ribosomal RNA activates mammalian target of rapamycin complex 1 signalling and downstream translation factors. Nucleic Acids Res 2014; 42:5083-96. [PMID: 24526220 PMCID: PMC4005692 DOI: 10.1093/nar/gku130] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Ribosome biogenesis is a key process for maintaining protein synthetic capacity in dividing or growing cells, and requires coordinated production of ribosomal proteins and ribosomal RNA (rRNA), including the processing of the latter. Signalling through mammalian target of rapamycin complex 1 (mTORC1) activates all these processes. Here, we show that, in human cells, impaired rRNA processing, caused by expressing an interfering mutant of BOP1 or by knocking down components of the PeBoW complex elicits activation of mTORC1 signalling. This leads to enhanced phosphorylation of its substrates S6K1 and 4E-BP1, and stimulation of proteins involved in translation initiation and elongation. In particular, we observe both inactivation and downregulation of the eukaryotic elongation factor 2 kinase, which normally inhibits translation elongation. The latter effect involves decreased expression of the eEF2K mRNA. The mRNAs for ribosomal proteins, whose translation is positively regulated by mTORC1 signalling, also remain associated with ribosomes. Therefore, our data demonstrate that disrupting rRNA production activates mTORC1 signalling to enhance the efficiency of the translational machinery, likely to help compensate for impaired ribosome production.
Collapse
Affiliation(s)
- Rui Liu
- Centre for Biological Sciences, University of Southampton, Highfield Campus, Southampton SO17 1BJ, UK, Centre de Recherche en Nutrition Humaine, INRA Clermont-Theix, Unité de Nutrition Humaine, 63122 Ceyrat, France and Division of Molecular Physiology, James Black Centre, College of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | | | | | | | | | | |
Collapse
|
239
|
Hannan RD, Drygin D, Pearson RB. Targeting RNA polymerase I transcription and the nucleolus for cancer therapy. Expert Opin Ther Targets 2014; 17:873-8. [PMID: 23862680 DOI: 10.1517/14728222.2013.818658] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The nucleoli are the site of the production of ribosomes, the protein synthetic apparatus of the cell. The presence of enlarged nucleoli, reflecting increased ribosomal gene transcription, has long been used by pathologists as an indicator of aggressive tumors. However, over the last 10 years a growing body of evidence has revealed that the nucleolus contains a dynamic cohort of over 4500 proteins, the majority of which have no function in ribosome production. The activity of some of these proteins is modulated by their regulated sequestration and release from the nucleolus. In particular, the nucleolus plays a central role in sensing cellular stress to modulate the abundance of the critical tumor suppressor protein p53. The finding that p53 activity is dysregulated in up to 50% of all human cancers highlights the importance of the nucleolar stress response in limiting malignant transformation. The development of drugs to selectively inhibit transcription of the ribosomal RNA genes in the nucleolus has paved the way for a new therapeutic approach to hijack nucleolar stress to selectively and non-genotoxically activate p53 in tumor cells. Here, we describe the potential application of this exciting new class of drugs for the treatment of human cancer.
Collapse
|
240
|
Léger K, Bär D, Savić N, Santoro R, Hottiger MO. ARTD2 activity is stimulated by RNA. Nucleic Acids Res 2014; 42:5072-82. [PMID: 24510188 PMCID: PMC4005644 DOI: 10.1093/nar/gku131] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
ADP-ribosyltransferases (ARTs) are important enzymes that regulate the genotoxic stress response and the maintenance of genome integrity. ARTD1 (PARP1) and ARTD2 (PARP2) are homologous proteins that modify themselves and target proteins by the addition of mono- and poly-ADP-ribose (PAR) moieties. Both enzymes have been described to be involved in the genotoxic stress response. Here, we characterize cellular PAR formation on hydrogen peroxide (H2O2) or N-methyl-N′-methyl-nitro-N-nitrosoguanidine (MNNG) stress, in combination with application of the RNA polymerase I inhibitor Actinomycin D (ActD), known to cause accumulation of short RNA polymerase I-dependent rRNA transcripts. Intriguingly, co-treatment with ActD substantially increased H2O2- or MNNG-induced PAR formation. In cells, this enhancement was predominantly mediated by ARTD2 and not ARTD1. In vitro experiments confirmed that ARTD2 is strongly activated by RNA and that the N-terminal SAP domain is important for the binding to RNA. Thus, our findings identify a new activator of ARTD2-dependent ADP-ribosylation, which has important implications for the future analysis of the biological role of ARTD2 in the nucleus.
Collapse
Affiliation(s)
- Karolin Léger
- Institute of Veterinary Biochemistry and Molecular Biology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland and Life Science Zurich Graduate School, University of Zurich, 8057 Zurich, Switzerland
| | | | | | | | | |
Collapse
|
241
|
Qiao X, Ding S, Liu F, Kucera GL, Bierbach U. Investigating the cellular fate of a DNA-targeted platinum-based anticancer agent by orthogonal double-click chemistry. J Biol Inorg Chem 2014; 19:415-26. [PMID: 24407462 DOI: 10.1007/s00775-013-1086-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Accepted: 12/21/2013] [Indexed: 12/11/2022]
Abstract
Confocal fluorescence microscopy was used to study a platinum-based anticancer agent in intact NCI-H460 lung cancer cells. Orthogonal copper-catalyzed azide-alkyne cycloaddition (click) reactions were used to simultaneously determine the cell-cycle-specific localization of the azide-functionalized platinum-acridine agent 1 and monitor its effects on nucleic acid metabolism. Copper-catalyzed postlabeling showed advantages over copper-free click chemistry using a dibenzocyclooctyne (DIBO)-modified reporter dye, which produced high background levels in microscopic images and failed to efficiently label platinum adducts in chromatin. Compound 1 was successfully labeled with the fluorophore DIBO to yield 1* (characterized by in-line high-performance liquid chromatography/electrospray mass spectrometry). 1 and 1* show a high degree of colocalization in the confocal images, but the ability of 1* to target the (compacted) chromatin was markedly reduced, most likely owing to the steric bulk introduced by the DIBO tag. Nuclear platinum levels correlated inversely with the ability of the cells to synthesize DNA and cause cell cycle arrest, as confirmed by bivariate flow cytometry analysis. In addition, a decrease in the level of cellular transcription, shrinkage of the nucleolar regions, and redistribution of RNA into the cytosol were observed. Postlabeling in conjunction with colocalization experiments is a useful tool for studying the cell killing mechanism of this type of DNA-targeted agent.
Collapse
Affiliation(s)
- Xin Qiao
- School of Pharmaceutical Sciences, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | | | | | | | | |
Collapse
|
242
|
Quin JE, Devlin JR, Cameron D, Hannan KM, Pearson RB, Hannan RD. Targeting the nucleolus for cancer intervention. Biochim Biophys Acta Mol Basis Dis 2014; 1842:802-16. [PMID: 24389329 DOI: 10.1016/j.bbadis.2013.12.009] [Citation(s) in RCA: 173] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 12/17/2013] [Indexed: 12/17/2022]
Abstract
The contribution of the nucleolus to cancer is well established with respect to its traditional role in facilitating ribosome biogenesis and proliferative capacity. More contemporary studies however, infer that nucleoli contribute a much broader role in malignant transformation. Specifically, extra-ribosomal functions of the nucleolus position it as a central integrator of cellular proliferation and stress signaling, and are emerging as important mechanisms for modulating how oncogenes and tumor suppressors operate in normal and malignant cells. The dependence of certain tumor cells to co-opt nucleolar processes to maintain their cancer phenotypes has now clearly been demonstrated by the application of small molecule inhibitors of RNA Polymerase I to block ribosomal DNA transcription and disrupt nucleolar function (Bywater et al., 2012 [1]). These drugs, which selectively kill tumor cells in vivo while sparing normal cells, have now progressed to clinical trials. It is likely that we have only just begun to scratch the surface of the potential of the nucleolus as a new target for cancer therapy, with "suppression of nucleolar stress" representing an emerging "hallmark" of cancer. This article is part of a Special Issue entitled: Role of the Nucleolus in Human Disease.
Collapse
Affiliation(s)
- Jaclyn E Quin
- Oncogenic Signalling and Growth Control Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Jennifer R Devlin
- Oncogenic Signalling and Growth Control Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Donald Cameron
- Oncogenic Signalling and Growth Control Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - Kate M Hannan
- Oncogenic Signalling and Growth Control Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Richard B Pearson
- Oncogenic Signalling and Growth Control Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, Victoria, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia; Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Ross D Hannan
- Oncogenic Signalling and Growth Control Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, Victoria, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia; Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia; Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia; School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland, Australia.
| |
Collapse
|
243
|
Hallgren J, Pietrzak M, Rempala G, Nelson PT, Hetman M. Neurodegeneration-associated instability of ribosomal DNA. Biochim Biophys Acta Mol Basis Dis 2014; 1842:860-8. [PMID: 24389328 DOI: 10.1016/j.bbadis.2013.12.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 12/17/2013] [Accepted: 12/20/2013] [Indexed: 12/12/2022]
Abstract
Homologous recombination (HR)-mediated instability of the repetitively organized ribosomal DNA (rDNA) has been proposed as a mediator of cell senescence in yeast triggering the DNA damage response. High individual variability in the content of human rDNA suggests that this genomic region remained relatively unstable throughout evolution. Therefore, quantitative real-time polymerase chain reaction was used to determine the genomic content of rDNA in post mortem samples of parietal cortex from 14 young and 9 elderly individuals with no diagnosis of a chronic neurodegenerative/neurological disease. In addition, rDNA content in that brain region was compared between 10 age-matched control individuals and 10 patients with dementia with Lewy bodies (DLB) which involves neurodegeneration of the cerebral cortex. Probing rRNA-coding regions of rDNA revealed no effects of aging on the rDNA content. Elevated rDNA content was observed in DLB. Conversely, in the DLB pathology-free cerebellum, lower genomic content of rDNA was present in the DLB group. In the parietal cortex, such a DLB-associated instability of rDNA was not accompanied by any major changes of cytosine-phosphate-guanine methylation of the rDNA promoter. As increased cerebro-cortical rDNA content was previously reported in Alzheimer's disease, neurodegeneration appears to be associated with instability of rDNA. The hypothetical origins and consequences of this phenomenon are discussed including possibilities that the DNA damage-induced recombination destabilizes rDNA and that differential content of rDNA affects heterochromatin formation, gene expression and/or DNA damage response. This article is part of a Special Issue entitled: Role of the Nucleolus in Human Disease.
Collapse
Affiliation(s)
- Justin Hallgren
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, KY 40292, USA; Department of Neurological Surgery, University of Louisville, Louisville, KY 40292, USA; Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY 40292, USA
| | - Maciej Pietrzak
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, KY 40292, USA; Department of Neurological Surgery, University of Louisville, Louisville, KY 40292, USA
| | - Grzegorz Rempala
- Division of Biostatistics, College of Public Health, Ohio State University, Columbus, OH 43210, USA
| | - Peter T Nelson
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536-0230, USA
| | - Michal Hetman
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, KY 40292, USA; Department of Neurological Surgery, University of Louisville, Louisville, KY 40292, USA; Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY 40292, USA.
| |
Collapse
|
244
|
Sarshad AA, Percipalle P. New Insight into Role of Myosin Motors for Activation of RNA Polymerases. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2014; 311:183-230. [DOI: 10.1016/b978-0-12-800179-0.00004-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
245
|
Akt activation enhances ribosomal RNA synthesis through casein kinase II and TIF-IA. Proc Natl Acad Sci U S A 2013; 110:20681-6. [PMID: 24297901 DOI: 10.1073/pnas.1313097110] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Transcription initiation factor I (TIF-IA) plays an essential role in regulating ribosomal RNA (rRNA) synthesis by tethering RNA polymerase I (Pol I) to the rDNA promoter. We have found that activated Akt enhances rRNA synthesis through the phosphorylation of casein kinase IIα (CK2α) on a threonine residue near its N terminus. CK2 in turn phosphorylates TIF-IA, thereby increasing rDNA transcription. Activated Akt also stabilizes TIF-IA, induces its translocation to the nucleolus, and enhances its interaction with Pol I. Treatment with AZD8055, an inhibitor of both Akt and mammalian target of rapamycin phosphorylation, but not with rapamycin, disrupts Akt-mediated TIF-IA stability, translocation, and activity. These data support a model in which activated Akt enhances rRNA synthesis both by preventing TIF-IA degradation and phosphorylating CK2α, which in turn phosphorylates TIF-IA. This model provides an explanation for the ability of activated Akt to promote cell proliferation and, potentially, transformation.
Collapse
|
246
|
Fernández-Tornero C, Moreno-Morcillo M, Rashid UJ, Taylor NMI, Ruiz FM, Gruene T, Legrand P, Steuerwald U, Müller CW. Crystal structure of the 14-subunit RNA polymerase I. Nature 2013; 502:644-9. [PMID: 24153184 DOI: 10.1038/nature12636] [Citation(s) in RCA: 157] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Accepted: 09/04/2013] [Indexed: 01/21/2023]
Abstract
Protein biosynthesis depends on the availability of ribosomes, which in turn relies on ribosomal RNA production. In eukaryotes, this process is carried out by RNA polymerase I (Pol I), a 14-subunit enzyme, the activity of which is a major determinant of cell growth. Here we present the crystal structure of Pol I from Saccharomyces cerevisiae at 3.0 Å resolution. The Pol I structure shows a compact core with a wide DNA-binding cleft and a tightly anchored stalk. An extended loop mimics the DNA backbone in the cleft and may be involved in regulating Pol I transcription. Subunit A12.2 extends from the A190 jaw to the active site and inserts a transcription elongation factor TFIIS-like zinc ribbon into the nucleotide triphosphate entry pore, providing insight into the role of A12.2 in RNA cleavage and Pol I insensitivity to α-amanitin. The A49-A34.5 heterodimer embraces subunit A135 through extended arms, thereby contacting and potentially regulating subunit A12.2.
Collapse
Affiliation(s)
- Carlos Fernández-Tornero
- 1] Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu 9, 28040 Madrid, Spain [2]
| | | | | | | | | | | | | | | | | |
Collapse
|
247
|
RNA polymerase I structure and transcription regulation. Nature 2013; 502:650-5. [DOI: 10.1038/nature12712] [Citation(s) in RCA: 164] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Accepted: 09/24/2013] [Indexed: 01/25/2023]
|
248
|
Polymerase I pathway inhibitor ameliorates experimental autoimmune encephalomyelitis. J Neuroimmunol 2013; 263:91-7. [DOI: 10.1016/j.jneuroim.2013.08.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 08/04/2013] [Accepted: 08/07/2013] [Indexed: 10/26/2022]
|
249
|
Waminal NE, Ryu KH, Choi SH, Kim HH. Randomly detected genetically modified (GM) maize (Zea mays L.) near a transport route revealed a fragile 45S rDNA phenotype. PLoS One 2013; 8:e74060. [PMID: 24040165 PMCID: PMC3767626 DOI: 10.1371/journal.pone.0074060] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 07/29/2013] [Indexed: 11/18/2022] Open
Abstract
Monitoring of genetically modified (GM) crops has been emphasized to prevent their potential effects on the environment and human health. Monitoring of the inadvertent dispersal of transgenic maize in several fields and transport routes in Korea was carried out by qualitative multiplex PCR, and molecular analyses were conducted to identify the events of the collected GM maize. Cytogenetic investigations through fluorescence in situ hybridization (FISH) of the GM maize were performed to check for possible changes in the 45S rDNA cluster because this cluster was reported to be sensitive to replication and transcription stress. Three GM maize kernels were collected from a transport route near Incheon port, Korea, and each was found to contain NK603, stacked MON863 x NK603, and stacked NK603 x MON810 inserts, respectively. Cytogenetic analysis of the GM maize containing the stacked NK603 x MON810 insert revealed two normal compact 5S rDNA signals, but the 45S rDNA showed a fragile phenotype, demonstrating a “beads-on-a-string” fragmentation pattern, which seems to be a consequence of genetic modification. Implications of the 45S rDNA cluster fragility in GM maize are also discussed.
Collapse
Affiliation(s)
- Nomar Espinosa Waminal
- Plant Biotechnology Institute, Department of Life Science, Sahmyook University, Seoul, Korea
- Department of Plant Science, Plant Genomics and Breeding Institute and Research Institute for Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul, Korea
| | - Ki Hyun Ryu
- Department of Horticulture, Biotechnology and Landscape Architecture, Seoul Women’s University, Seoul, Korea
| | - Sun-Hee Choi
- Department of Horticulture, Biotechnology and Landscape Architecture, Seoul Women’s University, Seoul, Korea
| | - Hyun Hee Kim
- Plant Biotechnology Institute, Department of Life Science, Sahmyook University, Seoul, Korea
- * E-mail:
| |
Collapse
|
250
|
Affiliation(s)
- Christine Galustian
- MRC Centre for Transplantation, Kings College London, Guys Hospital, Great Maze Pond, London, UK
| |
Collapse
|