Mechanisms of rDNA silencing and the Nucleolar Remodelling Complex (NoRC)

 

Peter C. McKeown

Genetics & Biotechnology Lab, Plant & AgriBiosciences Research Centre (PABC), School of Natural Sciences, National University of Ireland, Galway, Ireland
E-mail address: peter.mckeown@nuigalway.ie

*Address for correspondance: Peter C. McKeown, Aras de Brun, NUI Galway, University Road, Galway, Ireland

 

March 2014

 

Abstract

Protein synthesis in living cells requires functional ribosomes which are composed of ribosomal proteins and ribosomal RNA (rRNA) molecules. rRNA is transcribed from tandemly repeated ribosomal DNA (rDNA) which is organised into a nuclear compartment termed the nucleolus in S-phase cells. It is essential that rDNA transcription is properly regulated in order to meet the cell's requirements for ribosomes and hence protein synthesis without wasting metabolic energy. In the last twenty years many proteins involved in regulating this process have been identified, suggesting that most organisms contain multiple protein complexes that regulate rDNA packaging and transcription. Importantly, it has become clear that errors in the function of these proteins can permit aberrant cellular growth, including in several classes of cancer. In this review, I discuss the history of how protein complexes such as the Nucleolar Remodelling Complex (NoRC) were discovered, using examples from humans and from model research organisms from different biological groups. I will discuss recent discoveries of the critical roles of rDNA-binding complexes in nucleolar assembly, the widespread occurrence of regulatory non-coding RNAs which interact with these complexes, and the pathways which regulate rDNA transcription in response to cellular energy status. Finally, I will review the growing evidence that misregulation of rDNA transcription not only allows the growth of cancerous cells, but can trigger oncogenesis itself.

Introduction

All living cells have an essential requirement to transcribe rRNA genes (rDNA) to produce rRNA for use in ribosome synthesis. Ribosome production is necessary to support translation of mRNA and consumes a significant proportion of the energy available to a typical living cell. rDNA transcription, which is performed in eukaryotes by RNA polymerase I, must be correctly regulated in order to ensure that the cellular requirement for ribosomal subunits is met. Studies in many groups of organisms have revealed the existence of multiple interacting pathways which ensure this regulation by up- or down-regulate total rRNA synthesis via activation or silencing of rDNA, respectively. It has also become clear that dysregulation of these pathways is associated with many pathologies, and in mammals is associated with, and often prognostic for, the occurrence of cancer. Control of transcription at rDNA has also been used as a paradigm for understanding eukaryotic gene expression in general.

In this deep insight, key mechanisms that determine how rDNA transcription is controlled will be discussed. There is a particular focus on the pathways which act in mammalian cells, including those which have been implicated in tumourigenesis. However, reference is also made to some of the many insights into rDNA regulation revealed in other model biological systems, such as mutagenesis screens for loss of rDNA silencing in yeast, and some of the many epigenetic aspects of rDNA regulation discovered in hybrid plants of the Arabidopsis genus. As rRNA synthesis, the organisation of rDNA into nucleoli and the subsequent formation of ribosomes are all highly complex topics, this account will necessarily deal only with certain key discoveries, together with some highlights of the recent literature. The reader will be referred to many excellent reviews from the last few years for further details where appropriate.

In terms of organisation, this deep insight will begin with a general discussion of how RNA polymerase I acts to generate nascent rRNA molecules as precursors to ribosome biosynthesis, before turning to how this process is controlled. I will discuss the characteristics of rDNA regulation networks with particular biological or medical significance from different systems, before turning to aspects of how rDNA transcription has been linked to cancer.

a) the mammalian NoRC complex and other complexes which interact with or oppose its activity;
b) the links between rRDNA transcription and cellular energy status;
c) other multi-protein complexes in plants and yeast which shed light on other aspects of rRNA transcription control;
d) misregulation of rDNA regulation complexes and their links with cancer and other pathologies.

rDNA chromatin and the organisation of the nucleolus

Ribosomal RNA (rRNA) synthesis occurs in the nucleolus which assembles during the packaging and transcription of ribosomal DNA. In the nucleolus, tandemly repeated 45S ribosomal RNA genes (or rDNA) are transcribed to form 45S nascent pre-rRNA (Ballal et al., 1977). Each pre-rRNA is cleaved and processed to form the 18S, 5.8S and 25S rRNA molecules which are essential for the formation of ribosomes (Shaw and Jordan, 1995). The nucleolus is not bound by any membrane but self-assembles during the processes of rRNA transcription and subsequent processing (Mélèse and Xue, 1995). Nucleoli are stable enough to be extracted from culture cells (Andersen et al., 2002; McKeown et al., 2008) and studies in plants and animals have used mass spectrometry to identify proteins involved in the production of rRNA (Andersen et al., 2002; Pendle et al., 2005). Such studies have suggested that in many organisms nucleoli have also acquired additional cellular functions in stress response, splicing and small RNA biosynthesis (Pendle et al., 2005; Raška et al., 2006; Boisvert et al., 2007; Boulon et al., 2010), although these will not be considered further here.

In most eukaryotes, the rDNA locus consists of several hundred tandemly repeated rRNA genes, separated by linker regions. These are located at one or more large loci, which are also termed nucleolar organizing regions (NORs). The tandem repetition of rRNA genes is necessary as at least 20-40 rRNA genes are required to meet the production levels of rRNA in a typical dividing cell (Sollner-Webb et al., 1985). Furthermore, most eukaryotes contain many more rRNA genes than this (usually in the range of 50-500) and under most conditions, only a subset of the total are actively transcribed while the others are silenced by a dosage-control mechanism (Dammann et al., 1993; Russell and Zomerdijk, 2005). A consequence of this is that rRNA synthesis can be up- or down-regulated either by varying the number of active genes or by altering the transcription rate per gene, although the relative importance of these two pathways may vary between organisms (Dammann et al., 1993; French et al., 2003). Both processes are controlled by cellular signalling pathways of some complexity (Schmelzle and Hall, 2000; Stefanovsky et al., 2001; Grummt, 2003; Kim et al., 2003; Moss, 2004). The inactive rRNA genes are maintained in a transcriptionally silent, inactive state which requires the interaction of many cellular processes (McStay and Grummt, 2008). This silencing ensures that the cell's energy is not expended on unnecessary rRNA synthesis, that rRNA genes which have accumulated mutations or become pseudogenic are not transcribed, and that the activity of other RNA polymerases within the rDNA is prevented (Dammann et al., 1993). As we will see, preventing aberrant rRNA transcription may also be an important barrier to tumour formation in mammals, and failure to repress aberrant may trigger oncogenesis under some circumstances. Finally, classic studies demonstrated that the cells of hybrid organisms generally silence the entire rDNA inherited from one parental species and activate that from the other (Navashin, 1934). This phenomenon is now known as nucleolar dominance and has been used to illustrate many of the pathways that regulate rDNA activity.

A key issue for understanding rRNA transcription is therefore to understand the mechanistic basis of the switch between transcriptionally active and transcriptionally repressed rDNA. It was shown several decades ago that rDNA exists in two distinct conformations (that is, physical states within the nucleus), and that these can be stably inherited through mitosis (Conconi et al., 1989; Birch and Zomerdijk, 2008). The two forms of rDNA correspond to rRNA genes which are organised into different forms of chromatin, the DNA-protein superstructure into which DNA is 'packaged' following binding by histone octamers (Prior et al., 1983). The two forms of rDNA chromatin can be distinguished on the basis of their differential accessibility to the DNA-crosslinking drug, psoralen. One form has a highly compact psoralen-inaccessible organisation corresponding to densely-staining heterochromatin, and which is expected to be refractory to rRNA gene transcription. The other is a more dispersed, psoralen-accessible form which corresponds to the lightly-stained euchromatin and is expected to be permissive for transcription (Conconi et al., 1989; Dammann et al., 1993). This suggests a basic conceptual model in which the activation level of rRNA genes depends upon a switch between two different chromatin states, which either repress transcription by Pol I or induce it, respectively. This also corresponds with electron microscopy investigations which confirm that rDNA can be present in one of two forms within the cell, one of which is indeed densely compacted in the manner of heterochromatin, while the second is less dense in the manner of euchromatin.

Under the microscope, heterochromatin is visible within nucleoli as fibrillar centres or as 'knobs' arranged around the nucleolar periphery, while euchromatin may be present throughout the body of the nucleolus. Many lines of evidence support the supposition that the compacted heterochromatic rDNA is typically inactive, while that which more loosely organised is euchromatic and likely to be undergoing transcription (Raška et al., 2006). Understanding the features of these two chromatin states, and the protein complexes which induce transitions between them, is thus essential for understanding how rRNA transcription is controlled (Gerbi et al., 2003). Various reviews describe our current understanding of the importance of nucleolar organisation and its relationship with rDNA packaging, rRNA gene transcription and pre-rRNA processing in different taxa (Nierras et al., 1997; Raška et al., 2006; McStay and Grummt, 2008; Shaw and McKeown, 2011; Shaw and Brown, 2012).

Figure 1. Generalized model for the three chromatin states in which eukaryote rRNA genes may form. A) constitutively silent rDNA repeats organised into heterochromatin; B) rDNA repeats organised into euchromatin and actively transcribed by RNA Pol I (shown as a schematic core polymerase core and transcription factor; see text for details); C) 'poised' rDNA accessible to transcription machinery but not actively transcribed.

Early evidence on the nature of the molecular differences between active and inactive rRNA genes was suggested by the chemical manipulation of nucleolar dominance by aza-dC and trichostatin A, DNA methylation and histone deacetylation inhibitors, respectively (Reeder, 1985; Thompson and Flavell, 1988; Pikaard, 2000). As these pharmacological treatments altered the extent of nucleolar dominance, it was concluded that both DNA methylation and histone modification might differ between rDNA chromatin states in a manner related to the control of their transcription (Chen and Pikaard, 1997; Pikaard, 1999; McStay, 2006). Elucidating the details of what these modifications might be has been greatly accelerated by the development of the chromatin immunoprecipitation (ChIP) technique. For example, it has been shown that mammalian rRNA genes can be associated with either of two sets of covalent chromatin modifications, which correlate with their level of transcriptional activity (Santoro et al., 2002). DNA of silenced rRNA genes is highly methylated at cytosine residues, and is bound by histone octamers which are methylated at H3K9 (Figure 1A). Active rRNA genes are instead distinguished by DNA hypomethylation, and are bound to histone octamers incorporating H3K4 marks and widespread acetylation of many H3 and H4 lysine residues (Figure 1B). It has also been proposed that rDNA can also exist in a third, intermediate state, in which the chromatin of the rRNA genes is decondensed but they remain transcriptionally silent (Figure 1C). This is characterised by the simultaneous presence of histone modifications associated with euchromatin and heterochromatin on different sites and may correspond to a 'poised' euchromatic state (McKeown and Shaw, 2009; Xie et al., 2012).

Much research has therefore concentrated on determining what roles these chromatin modifications play at the molecular level, and to what extent they are causal for determining the activity of the rRNA genes with which they associate. Such studies have made use of many different biological systems and have exploited genetic, biochemical and cell biological techniques, including cell culture models for different human cancers and other diseases. These studies have demonstrated that while the proteins which control rDNA chromatin typically vary between different groups of eukaryotic organisms, the regulatory networks in which they act also have various key features in common. In the following section, the control of rDNA transcription in humans (and certain model systems) will be described.

rDNA silencing I - control of human RNA Pol I transcription by protein complexes

In eukaryote, 45S pre-rRNA is synthesised by a dedicated transcriptional system centred on the multimeric protein complex, RNA Polymerase I (hereafter RNA Pol I). RNA Pol I only catalyzed the transcription of rDNA, which is in turn not transcribed by any other polymerase system under normal conditions. For a general review of the biochemical structure of RNA Pol I, see (Vannini, 2013). In addition to the core subunits of RNA Pol I itself, its polymerase activity requires the action of several other proteins and protein complexes, many of which have regulatory potential. These RNA Pol I cofactors and transcription factors (TF) allow RNA Pol I to effect transcription at active rRNA genes and to determine the level of this.

In humans, the principal RNA Pol I TFs consist of the Upstream Binding Factor (UBF), the promoter selectivity factor (SL1, (Comai et al., 1992)) and the transcription termination factor (TTF-I, (Grummt, 2003)). These complexes remain associated with rDNA during the cell cycle, even during mitosis when rRNA gene transcription is silenced but are maintained in an inactivate state until transcription resumes in telophase (O'Mahony and Rothblum, 1991). Of these proteins, TTF-I is essential for activating rDNA transcription as its binding initiates an open chromatin conformation (Längst et al., 1997; Längst et al., 1998). This open chromatin structure is stabilised by the HMG-box protein, UBF, which binds ubiquitously across the entire rDNA locus (Roussel et al., 1993). The importance of UBF for supporting RNA Pol I-transcription is suggested by the fact that it largely supplants the histone octamer-based nucleosome as the basic subunit of chromatin at active rRNA genes (Zatsepina et al., 1993; O'Sullivan et al., 2002; Mais et al., 2005). UBF plays a particularly critical role at the rRNA gene promoter, where it serves as a scaffold for the binding of RNA Pol I transcription factors and other processing proteins (Mais et al., 2005; Prieto and McStay, 2007). As this facilitates RNA Pol I promoter escape (Panov et al., 2006), UBF therefore orchestrates the level of rRNA transcription at active rRNA genes (O'Mahony and Rothblum, 1991; O'Sullivan et al., 2002; Chen et al., 2004; Sanij et al., 2008). UBF activity is itself tightly regulated (Sanij and Hannan, 2009), including by post-translational modifications, which control how UBF reactivates rRNA transcription after it has temporarily ceased during mitosis (Voit et al., 1999; Meraner et al., 2006). Recent work has demonstrated that the control of RNA Pol I activity is both necessary and sufficient for the formation of the nucleolus in human cells (Grob et al., 2014).

RNA Pol I function also requires a complex termed FACT, (facilitates chromatin transcription), which can be co-precipitated with RNA Pol I (Birch et al., 2009). In contrast to UBF which regulates the initiation of transcription, FACT is specifically required for the efficient passage of polymerases through nucleosomes. In this way, FACT facilitates transcription by RNA Pol I, II and III and is thus essential for transcriptional elongation throughout the nucleus (its relationship with other RNA polymerases present in plants remains to be ascertained). FACT contains two core subunits, SSRP1 and Spt16. In accordance with the essential nature of FACT for transciption, cells in which either subunit is down-regulated display reduced transcription at the 3' regions of the rRNA genes (Birch et al., 2009).

Both initiation and elongation can be co-ordinately regulated independently of UBF and FACT by another large (2-3 MDa) complex, this time involved in maintaining an open chromatin structure throughout active rDNA repeats. This third key complex has been termed B-WICH and its core subunits include WSTF, SNF2h and myosin1 (Percipalle et al., 2006); the complex takes its name from the Williams syndrome transcription factor (WSTF). B-WICH was first reported to be required for efficient transcriptional activity of RNA Pol II. When its subunits are knocked down, the abundance of Pol III transcripts is also reduced, indicating that its role is not specific for control of rDNA activity (Cavellán et al., 2006; Percipalle et al., 2006). WSTF is also a component of other transcriptional complexes, leading to pleiotropic phenotypes when it is disrupted, and most of these also includes ncRNA molecules (Cavellán et al., 2006). The B-WICH complex appears to be required for the recruitment of some, but not all, of the histone acetyltransferases present at active rDNA (Vintermist et al., 2011) which presumably explains its mode of action.

The B-WICH complex physically interacts with the Cockayne's syndrome protein (CSB) protein, which also has multiple roles in controlling RNA Pol II transcription and DNA repair. CSB is itself required for RNA Pol I transcription, including performing ATP-dependent chromatin remodelling at active rDNA repeats (Lebedev et al., 2008). CSB can additionally act in an ATP-independent manner within another complex, termed CSB IP/150. This complex also includes XPG (a protein disrupted in certain cases of the human condition, Xeroderma Pigmentosum), RNA Pol I itself, and a transcription factor complex, TFIIH (Bradsher et al., 2002). TFIIH was originally described as a Pol II-specific TF, but has subsequently been shown to activate Pol I transcription in yeast and mouse as well (Iben et al., 2002). In human cells, CSB IP/150 performs this activation by recruiting a further histone acetyltransferase, PCAF, which leads to transcriptional initiation at rRNA genes which have already adopted an open, poised chromatin state (Shen et al., 2013). Curiously, CSB/TFIIH also recruits a histone methyltrasferase, G9a, which induces H3K9me2, which is generally considered a repressive chromatin mark (Yuan et al., 2007). The association of CSB with RNA Pol I within CSB IP/150 appears to be disrupted by mutations in the other components of the complex (Bradsher et al., 2002). This may be involved in the onset of Cockayne Syndrome, a recessive disorder associated with premature aging and neural degeneration.

As further evidence of the complexity of RNA Pol I transcriptional control, a further such complex has been reported from human cells rather recently (in 2012). This time, the complex was associated with the establishment of 'poised' or transcription-ready rRNA genes and was named 'nucleosome remodelling and deacetylation' complex (NuRD) (Xie et al., 2012). When cellular growth is attenuated and transcription at rDNA reduced, NuRD is enriched at rRNA gene promoters which are unmethylated, associated with RNA Pol I transcription factors, yet kept silent by the positioning of a key nucleosome. Notably, this complex is also tightly regulated with respect to the growth status of the mammalian cells in question, via pathways as yet unknown. The trend established by UBF and other complexes is that rDNA activity is regulated by the activity of multi-protein complexes, many with chromatin-modifying activities. These complexes, and doubtless more which remain to be discovered, are presumed to ensure RNA Pol I transcription occurs at an appropriate level via multiple, subtle interactions. This has been argued to be the main mode of rRNA synthesis in mammals under normal circumstances (Stefanovsky and Moss, 2006).

Control of rDNA transcription II - the role of silencing complexes

While UBF determines the activity of RNA Pol I at rRNA genes with a suitably open chromatin organisation, it is not responsible for determining the proportion of rRNA genes which are organised in this transcriptionally-permissive manner. In human cells, the principal determinant of the ratio of active and inactive rRNA genes is the multi-protein complex NoRC (nucleolar remodelling complex) which silences rRNA genes the transcriptional of which is not required. NoRC was discovered in the 1990s and was initially described as a complex of Snf2h/Smarca5 and the large (205 kDa) DNA-binding protein, Tip5 (transcription termination factor 1 (Ttf1)-interacting protein 5) - see Figure 2. At the sub-cellular level, NoRC was found to colocalise at the NORs. Since its discovery, it has been established that NoRC is essential both for blocking RNA Pol I transcription at these inactive rDNA loci, and moreover for catalyzing the assembly of rRNA genes into a heterochromatic conformation (Strohner et al., 2001). In other words, it regulates the level of rDNA transcription by decreasing the ratio of active:inactive rRNA genes, and is therefore complementary to the Pol I transcription factors which reduce or increase transcription at rRNA genes which are already active. NoRC therefore seems to play an antagonistic role to complexes such as B-WICH and CSB IP/150 (see above), although whether these complexes are capable of directly regulating NoRC is unclear. It has been suggested that complexes associated with euchromatic and heterochromatic rDNA might be recruited to rDNA at different stages during nuclear division, as active and inactive rRNA genes undergo replication at different timepoints (Yuan et al., 2007). The possibility of cross-talk occurring between these complexes merits further investigation.

Figure 2. Predicted model for the control of rDNA silencing by the Nucleolar Remodelling Complex (NoRC). NoRC consists of Tip5 and Snf2h. Tip5 is recruited to rRNA by transcription of a non-coding RNA termed pRNA (bold line). Binding of NoRC to rRNA genes leads to TTF-I dependent recruitment of chromatin remodelling enzymes (gray) and repression of rRNA transcription. Binding of NoRC to rDNA may require, or be stabilised by, Heterochromatin Protein 1 (HP1) and acetylated H4K16 residues (not shown).

The key role of NoRC in control of human rDNA transcription was first indicated by in vitro experiments showing that it was able to induce nucleosome sliding (of the sort required for rRNA gene silencing) along isolated DNA. This activity was dependent upon ATP, and the N-terminal tail of histone H4 (Strohner et al., 2001), indicating a direct affect upon nucleosome organisation. Based on its chromatin-remodelling capabilities and colocalisation with UBF in the nucleolus, NoRC was therefore recognised as a candidate regulator of rDNA transcription control. This hypothesis was proven shortly afterwards with the demonstration that association of NoRC with rDNA repeats caused rRNA genes to become heterochromatic and transcriptionally silent (Santoro et al., 2002). The biochemical nature of this heterochromatic DNA was indicated by the discovery that methylated rDNA genes could be immunoprecipitated in conjunction with the two components of NoRC (Tip5, Snf2). These proteins are additionally co-immunoprecipitated with hypoacetylated and hypermethylated histones, and with heterochromatin protein 1 (HP1). This indicated again that NoRC was a complex specifically associated with methylated, inactive rDNA, and that the rRNA genes in this rDNA was likely to be organised into heterochromatin. NoRC was therefore established as a key element of the heterochromatic fraction of rDNA previously identified in microscopy-based studies (Santoro et al., 2002).

The study of Santoro et al. furthermore determined that the role of NoRC in heterochromatic rDNA was likely to be causal rather than correlative. Over-expression of Tip5 caused a transfected rDNA reporter plasmid to become resistant to cleavage by the methylation-sensitive restriction enzyme HpaII, indicating that NoRC was able to induce DNA methylation in vivo (Santoro et al., 2002). As expected, treatment with the DNA methylation inhibitor 5-azacytidine reversed this effect (Santoro et al., 2002). NoRC is therefore responsible for inducing the methylation of rRNA gene promoters in mammalian cells. Importantly, the NoRC component Tip5 also acts as a binding site for the transcription termination factor TTF-I (Németh et al., 2004) which had previously been shown to be essential for repressive chromatin remodelling at the promoter of silenced rRNA genes (Längst et al., 1997; Längst et al., 1998). It is therefore believed that NoRC establishes and maintains the heterochromatic organisation of inactive rDNA repeats via recruitment of TTF-I and other enzymes which induce DNA methylation and the deposition of repressive histone modifications. These include H3K9 methylation and H4 hypoacetylation (Santoro et al., 2002) and may be mediated by Tip5 binding to H4K16ac via a bromodomain, which appears to stimulate recruitment of chromatin-remodelling enzymes such as HDAC1, DNMT1, DNMT3, and SNF2h to the rDNA (Zhou and Grummt, 2005), as shown in Figure 2. As an additional level of control, the binding of NoRC can be countered by an abundant nucleolar protein, nucleolin, which reduces the ability of TTF-I to bind to its target terminator region and thus reduces Tip5 recruitment (Cong et al., 2012).

Although initially described as a complex of proteins, it was subsequently discovered that NoRC also contains an essential RNA component. A non-coding RNA is produced from transcription of the rRNA gene promoter, termed pRNA, which binds to Tip5 and is required for targeting of the complex to the rDNA promoter. pRNA binding mediates DNMT3b association with the rRNA gene promoter (Bierhoff et al., 2011). pRNA-binding is dependent upon the acetylation of Tip5 by an interacting acetyltransferase, MOF (Males Absent on the First), which occurs only on lysine residue K633 and is essential for the gene-silencing activity of NoRC (Zhou et al., 2009). The pRNA also acts to allow the binding of poly(ADP-ribose)-polymerase-1 (PARP1) which is essential for NoRC to silence RNA Pol I and subsequent heterochromatinization of silent rDNA (Guetg et al., 2012).

rDNA silencing III - regulation of RNA Pol I silencing by cellular energy status

The manner in which the numerous complexes described above interact to control RNA Pol I activity remains a difficult issue, but the overall purpose of this complexity is clearly to allow rDNA activity to be attuned to the environment. One cellular condition to which rDNA appears to be particularly sensitive is the energetic status of the cell, which can (in mammals) regulate rRNA transcription by several different signalling pathways. This is unsurprising given the high demand that rRNA synthesis makes on the cell's ATP reserves, sometimes estimated as half of the cell's energetic output (Sollner-Webb et al., 1985; Grummt and Voit, 2010). Therefore, rDNA transcription must occur at a level suitable to meet the translational demand of the cell, while also taking account of the ATP available in the cell at that time.

The importance of this link, and its wide-ranging impacts on the healthy functioning of an organism, have been most widely established by studies of the budding yeast (Saccharomyces cerevisiae) protein, Silent information regulator 2 (Sir2). Sir2 is an NAD+-dependent protein deacetylase which is sensitive to cellular energy status (Fritze et al., 1997). Amongst other targets, Sir2 is able to deacetylate histones, removing H3-acetyl marks associated with open, transcriptionally active chromatin and potentially allowing the addition of H3-methyl marks, which are associated with transcriptionally inactive heterochromatin, Sir2 is known to be able to regulate rDNA transcription by RNA Pol I, and it has been proposed that this is linked to its histone/protein deacetylation capabilities (Fritze et al., 1997; Smith et al., 1998; Straight et al., 1999; Blander and Guarente, 2004; Machín et al., 2004; Ford et al., 2006). The ramifications of the role of Sir2 and related proteins (the so-called sirtuins) for human health is a topic of intense scientific debate, as several lines of evidence suggest that Sir2 is a key regulator of ageing and longevity in yeast, nematodes and metazoans. This may occur through mimicking the effects of caloric restriction. It has been claimed, for example, that increased cellular dosage of Sir2p in S. cerevisiae can expand yeast life span by suppressing genotoxic recombination between rDNA repeats of the sort discussed below (Lin et al., 2000). Here I will focus specifically on the role of Sir2 and related proteins in the direct control of transcription at the rDNA locus. For the details of the ongoing debate over the more controversial claims made for these proteins, the reader is referred to dedicated commentaries (Finkel et al., 2009; Lempiäinen and Shore, 2009; Imai and Guarente, 2010; Sebastián et al., 2012).

Figure 3. The regulation of rRNA gene transcription in response to cellular energy status. The eNOSC complex consists of NML (nucleomethylin), SuV (SUV39H1) and SIRT1 (sirtuin 1), and responds to signals indicating low cellular energy status by silencing rRNA genes and repressing ribosomal RNA production. At least one other pathway, involving mammalian Target Of Rapamycin (mTOR) operates in parallel. For details, and possible interaction of NMP with NMNAT1, see text.

Sirtuins share the common feature that they use NAD+ as a cofactor in the deacetylation of peptide targets, which may make them particularly suitable as sensors of cellular energy levels. The closest human homolog of ScSir2p is SIRTUIN 1 (SIRT1), which binds throughout rDNA repeats regardless of their transcriptional state, although the majority of it is distributed in other parts of the nucleoplasm (Michishita et al., 2005).The importance of the links between rDNA transcriptional activity and cellular metabolic status was underlined by the discovery that SIRT1 is part of a protein complex termed eNoSC within human cells (Murayama et al., 2008). This complex contains the H3K9me2-binding protein, Nucleomethylin (NML), SIRT1 and SUV39H1 (Figure 3). NML had previously been shown to be at least partially localised in the nucleolus (Andersen et al., 2005) and was identified by MS-ChIP to be bound to H3K9me2 at transcriptionally inactive rDNA (Murayama et al., 2008). In vivo over-expression of NML reduced the accumulation of nascent rRNA, an effect ablated by knock-down of SIRT1 (Murayama et al., 2008). The authors of this study proposed a model of 'co-ordinate binding' of NML and SIRT1 occurring specifically at silenced rRNA genes, with SIRT1 triggering H3K9 hypoacetylation and subsequent methylation at the same site. Methylation of hypoacetylated H3K9 was found to be at least partially due to the methyltransferase activity of SUV39H1, which also participates in the same complex. This suggests a model in which SUV39H1 and SIRT1 compete for modification of the same lysine residue. Many aspects of the regulation of SIRT1 within eNoSC remain unclear: two potentially significant points are that nucleomethylin (NML) interacts with a NAD+ synthesis enzyme called NMNAT1, which also contributes to the silencing of rDNA (Song et al., 2013) and physically associates with SIRT1 (see Figure 3); and that SIRT1 may be able to acetylate SUV39H1 in a manner that disrupts their binding. As a further complication, there is some evidence that NML - which has a methyltransferase-like domain - may act via methylation of some downstream target (Murayama et al., 2008). Although most commonly associated with core histones, many other proteins can be subject to methylation. eNoSC binding at rDNA is increased in HeLa cells under conditions that reduce available cellular energy (glucose starvation) and this has been found to protect such cells from energy deprivation-induced apoptosis. As the discovers of eNoSC point out, rDNA-silencing may be critical for this although interaction with other apoptosis-inducing pathways cannot be excluded (Murayama et al., 2008). eNoSC does not, however, appear to act on, or alter the methylation of, p53.

Other human sirtuins have different sub-cellular distributions, including the cytoplasm and mitochondria, while SIRT6 and SIRT7 are also nuclear. The only evidence for strong nucleolar enrichment was for SIRT7 (Michishita et al., 2005). SIRT7 is also of considerable interest for regulation of mammalian rDNA as it can also function as an activator of RNA Pol I (Ford et al., 2006). Although still relatively understudied, it has been shown that SIRT7 physically interacts with UBF and remains stably associated with both UBF and rDNA during mitosis (Grob et al., 2009; Tsai et al., 2012). After mitosis, SIRT7 is essential for the resumption of rRNA gene transcription during telophase, following a phosphorylation-induced conformation change performed by an unknown kinase (Grob et al., 2009). SIRT7 acts in vivo as an NAD+-dependent deacetylatase of the transcription-permissive histone modification H3K18Ac at gene promoters, thus reducing transcription (Barber et al., 2012). Misregulation of this activity has been linked with several aspects of tumour progression in human cells and in mice, and a significant increase in SIRT7 expression in breast cancer samples has been reported (Ashraf et al., 2006; Barber et al., 2012). No link between these correlations and the control of RNA Pol I has been reported, and the control of rDNA by SIRT7 has been suggested to be cell- or tissue-specific (Barber et al., 2012), although links between rDNA misregulation, SIRT7 and oncogenesis could easily remain to be discovered. Likewise, the requirement for NAD+, which varies in availability with a cell's energy status, may represent a significant link between to control of rRNA transcription in the same way that has been claimed for SIRT1.

In several eukayotic cells, another energy-sensing pathway based around signalling by Target of Rapamycin (TOR), which is a major component in the nutrient signalling machinery (Figure 3, right hand side), has been argued to control to the transcription of rRNA (Schmelzle and Hall, 2000; Kim et al., 2002). This pathway controls RNA Pol I transcription via chromatin remodelling at rDNA (Tsang et al., 2003), perhaps directly as TOR itself binds to rRNA gene promoters (Tsang et al., 2010). In yeast, TOR assists rDNA stability by increasing Sir2p association with rDNA (Ha and Huh, 2011) and also affects Rrn3p levels (Philippi et al., 2010). Whether mammalian TOR (mTOR) is able to interact with any of the sirtuins is not currently clear (Blagosklonny, 2010). Interestingly, the NuRD complex may also be regulated with regard to cellular energy status, as ATP is required to reverse the associated silencing (Xie et al., 2012). Again, the biological significance of this fact remains to be fully established.

The discovery of the presence of these multiple complexes poses several intriguing questions. One which remains to be resolved is the issue of how the different complexes which regulate rDNA function in human cell lines interact with one another. Another is why such a complex system is required to accomplish this regulation. The authors of Murayama et al., 2008 suggest an intriguing model in which NoRC is required for initiating silencing (following its binding to the transcriptional terminator element of rDNA via TTF-I - see Strohner et al., 2001) - but it is eNoSC, which appears to bind ubiquitously to rDNA throughout the nucleolus, that ensures chromatin silencing is propagated. This suggests coordinate regulation by different complexes might occur, or that different complexes might act to reinforce each other's activities in order to ensure that regulation of rRNA transcription and silencing are both robust and responsive.

Roles for histone modification in controlling RNA Pol I transcription

As noted above, the control of rDNA by NoRC is initiated when the complex binds to nucleosomes containing H4K16ac via a bromodomain in the Tip5 protein (Zhou and Grummt, 2005). This leads to the subsequent recruitment of multiple histone deacetylases and histone methyltransferases which induce a heterochromatic organisation at the locus and suggests a model in which different histone modifications direct various stages of euchromatin and heterochromatin formation at the rDNA locus. As testament to this, pharmacological inhibition of histone-modifying enzymes commonly leads to altered rRNA expression levels and nucleolar morphology, and changes to nucleolar dominance. The association between H4 and NoRC is not the only instance in which a major regulatory cascade requires binding to histones. H3/H4 dimer was identified as a core component of the major S. cerevisiae RNA Pol I TF, Upstream Activating Factor (UAF) (Keener et al., 1997), in addition to four non-histone protein components. Reduction of H3 synthesis inhibited rRNA transcription and was associated with reduced efficiency of multiple processes including initiation/elongation, and rRNA processing, although in some cases these were indirect effects (Nomura et al., 2004; Tongaonkar et al., 2005; Jones et al., 2007).

In mammalian cells, too, classic biochemical studies have shown that the H1 linker histone acts as a binding site for the abundant rDNA-associated protein, nucleolin (Erard et al., 1988) which is itself now known to have nucleosome chaperone activity (Angelov et al., 2006). Nucleolin has an affinity for unmethylated rRNA gene promoters and appears to ensure deposition of H3K4me3 and other histone modifications associated with a transcription-permissive state. On the other hand, nucleolin depletion causes H3K9me2 to accumulate (Cong et al., 2012). The B-WICH complex also triggers altered rDNA activity, this time by recruitment of histone acetyltransferases (Vintermist et al., 2011). Hence, suitably modified nucleosomes also have roles as downstream effectors of chromatin regulation at rDNA, acting to reinforce the open chromatin structure induced by nucleolin, and thereby helping to maintain rRNA transcription at active rDNA repeats. As other histone chaperones have also been shown to control the activity of rDNA in human cells (Kuzuhara and Horikoshi, 2004) and more recently in Arabidopsis (Li and Luan, 2010), histone chaperone activity may have more general roles in controlling the balance between transcribed and silenced rRNA genes.

Although covalent modifications at the H3K4 and H3K9 residues are well-known regulators of transcription and other cellular processes (Iizuka and Smith, 2003), additional histone modifications may also control rRNA transcription or other nucleolar functions, and it is possible that some are enriched or specific in the nucleolus (McKeown and Shaw, 2009). In mammals, for example, RNA Pol I transcription is encouraged via H3K56 acetylation (Chen et al., 2012), and as noted above H3K18 acetylation may be involved in regulation of rDNA by SIRT7. Active histone demethylation has also been implicated in rDNA control: JHDM1B is an evolutionarily conserved protein demethylase which regulates animal growth and may act as a tumour suppressor in mice (see Frescas et al., 2007 and references therein). JHDM1B binds to human rRNA genes in a stable manner reminiscent of UBF, and silences rRNA expression via demethylation of H3K4me3. This effect has an absolute requirement for its JmjC domain and appears to be a direct effect as H3K9me2 remains unaffected. Reduced expression of JHDM1B is associated with increased rRNA synthesis and accelerated cell growth, and has been reported to occur in brain tumour cells (Frescas et al., 2007).

Epigenetic control of RNA Pol I silencing and the importance of feedback

In the preceding sections, some of the protein complexes which regulate the silencing of rDNA in mammalian and yeast cells have been described. It has however been made clear that important details about how the different regulatory components interact with each other remain to be determined. An important conceptual contribution to this problem comes from research in plant hybrids, which has suggested that epigenetic regulators are able to establish 'self-reinforcing loops' at active and inactive rRNA genes. The details of how these loops are established, and their wider relevance, are discussed in the following section.

In hybrid organisms, it commonly occurs that chromatin remodelling of NORs inherited from different progenitors ensures that only the rDNA inherited from one parent is transcribed while the other remains silenced (so-called nucleolar dominance). The establishment of active or repressed states at the two sets of NORs in interspecific hybrids within the Arabidopsis genus involves differential DNA methylation reinforced by remodelling of the rRNA gene promoter by a complex involving the histone deacetylase HDA6, a histone deacetylase-like HDT protein, and an H3/H4 dimer (Lawrence et al., 2004; Earley et al., 2006). The same loop may also control differences in expression between different rRNA genes within the same NOR. hda6 mutants show aberrant accumulation of rDNA-encoded small RNAs, leading to the suggestion that siRNA-triggered pathways might also be involved in this chromatin remodelling loop as in animals. Furthermore, two mutants defective in siRNA biogenesis (dcl3, rdr2) disrupt the normal patterns of nucleolar dominance when crossed in an inter-specific manner (Preuss et al., 2008). The promoters and intergenic spacers of the silenced rRNA genes derived from A. thaliana were associated with the production of 24nt-siRNAs from both DNA strands, which were lost if DCL3 activity was ablated. In agreement with this, nucleolar dominance in Arabidopsis requires Pol IV and Pol V (Pontes 2006); exactly how these siRNA direct DRM2-mediated methylation, or even if they are causative for silencing at all, remains unclear. The model proposed by Preuss et al., 2008 was that transcription (possibly from promoter-like sequences in the intergenic spacers) directs 24-nt siRNA production from one of the rRNA alleles (Figure 4).

Figure 4. The regulation of rRNA gene transcription in the model plant, Arabidopsis thaliana. As in human cells, recruitment of rRNA silencing machinery is effected by production of non coding RNA from the rDNA repeats (e.g. 24 nt-siRNAs; short black lines); these lead to recruitment of DNA methyltransferases and methyl-binding domain proteins (MDB6, MDB10) and histone deacetylases (HDA6) which maintain silencing of rRNA genes by chromatin remodelling. Compared with rRNA gene regulation in humans, there is no NoRC, and no role has been reported for the plant ortholog of HP1.

The demonstration that noncoding RNA (ncRNA) is involved in rDNA silencing in Arabidopsis marks a point in common between animals and plants, as a long ncRNA plays a similar role in mouse (Mayer et al., 2006; Mayer et al., 2008). On the other hand, no role has been reported for shorter RNA in the silencing of inactive rDNA in mammals (as far as is known), nor for long ncRNA in plants. A further point regarding comparisons between different eukaryote taxa is that there is no obvious ortholog or even functional analog of Tip5 in Arabidopsis, suggesting that there is no direct equivalent of NoRC in plants. Physical association with the siRNA/epigenetic machinery might be possible however - supported by colocalisation within Cajal bodies which may lie within the nucleolus (Pontes et al., 2006) and could also therefore allow physical association with rDNA. Curiously, antisense transcription of human rDNA can also occur, this time acting to promote H4K20me3, a repressive chromatin mark (Bierhoff et al., 2011). It can be concluded that effects of non-coding RNA (whether siRNAs or lncRNAs) on RNA Pol I transcription are likely to be widespread, although the details of how they act are likely to vary between species, and interact with other regulators in complex ways.

Nucleolar dominance also depends on an Arabidopsis methylcytosine binding domain protein, MBD6, being localised to silenced rRNA by the de novo DNA methyltransferase DRM2 (Preuss et al., 2008). This pathway also includes other effectors of RNAi (DCL3, RDR2) and leads to methylation of the sequence, and subsequent binding of MBD6 (Figure 4). This change is coincident with association with heterochromatic histone marks and DNA condensation, which are presumed to ensure that silencing spreads across the entire NOR and is maintained in a robust manner. This study also demonstrated that mutants and/or RNAi knock-downs of genes encoding many other DNA methyltransferases, RDR proteins and DCL proteins did not affect nucleolar dominance (Preuss et al., 2008). Therefore, epigenetic regulation of rDNA activity is under the control of a particular subset of siRNA and chromatin-modifying pathways, at least in the A. thaliana X A. arenosa system being studied. The precise role of MBD6 was not clear at this time - when knocked down, it caused siRNA levels to increase slightly, suggesting that it might be responsible for negative feedback within this loop of the system.

Presumably, the complex pathways which control rDNA silencing in Arabidopsis are necessary to ensure that its chromatin state - and hence the epigenetic regulation of rRNA transcription - is stable both temporally and spatially. Its self-reinforcing nature means that it can act across very large genomic regions (many megabases in length), and be transmitted with fidelity through nuclear divisions, regardless of cell differentiation. It should however be added that the actual purpose of nucleolar dominance remains unclear, although a likely hypothesis is that it prevents the production of hybrid ribosomes which might function with reduced efficiency. This leads to the important conclusion that nucleolar dominance represents a modification of endogenous rRNA control, albeit in a hybrid background, with many regulatory features preserved. However, nucleolar dominance does differ in other ways (e.g. the differing requirements for de novo DNA methylation (Preuss et al., 2008; Earley et al., 2010)), perhaps because it has evolved to effect a stable silencing effect, rather than allowing environmental response.

An interesting recent report identified that the rRNA genes of Arabidopsis thaliana are not as uniform as previously thought. Rather, they include at least four variants forms, which show distinct features in their regulation (Pontvianne et al., 2010). The possibility of rRNA gene variants being functionally distinct (either under wild-type conditions, or following tumourigenesis) would be an interesting question to address. In Arabidopsis, it has been shown that different H3K9 and K27 histone methyltransferases are responsible for control of expression of these rRNA gene variants in A. thaliana and in nucleolar dominance between different rDNA from different parents in A. thaliana X A. arenosa hybrids (Pontvianne et al., 2012). This again suggests that silencing of rDNA during nucleolar dominance is in some respects a guide to rDNA control in non-hybrid organisms, and in other respects is different. Curiously, mutation of certain histone methyltransferases also leads to preferential replication of certainly variants (Pontvianne et al., 2012), implicating the covalent modification of histones in preventing replication slippage as well .

Silencing of aberrant RNA Pol II transcription in nucleoli

A further role for the rDNA silencing pathways discussed above came from Saccharomyces cerevisiae and concerned the mechanisms which ensure that sequences at the rDNA locus are not promiscuously transcribed by RNA Pol II. RNA Pol I is the most specific of the RNA polymerases found in eukaryotes, being solely responsible for transcription of rDNA. rDNA transcription is in turn specific for RNA Pol I, and is not transcribed by RNA Pol II machinery (Grummt, 2003). It has been known for some years that this is because of chromatin-related pathways which act to prevent Pol II transcription from occurring and hence ensure polymerase fidelity. Accordingly, if an rRNA gene is cloned into a different genomic location, then transcription by Pol II can instead occur.

In S. cerevisiae, repression of RNA Pol II transcription within the rDNA repeats requires the HDAC Sir2p (Smith and Boeke, 1997; Smith et al., 1998). The repressive activity of Sir2p requires the structural chromatin protein, condensin which causes rDNA to adopt the correct chromatin organisation and is essential for ensuring that sufficient Sir2p levels is retained at the rDNA locus (Smith et al., 1998; Machín et al., 2004). ScSir2 is a component of another rDNA-silencing complex which has been termed RENT (regulator of nucleolar silencing and telophase exit (Straight et al., 1999)) which is anchored to rDNA by the component protein Net1 (the name derives from the essential role of RENT in coordinating mitotic exit and the resumption of rRNA transcription (Cockell and Gasser, 1999)). Yeast mutant screens have suggested that Set1p is also essential for blocking RNA Pol II transcription in the rDNA locus, this time acting via deposition of H3K4me3 which in this instance is associated with transcriptional silencing (Briggs et al., 2001). This activity is independent of Sir2, and hence of the RENT complex (Bryk et al., 2002), and presumably acts to reinforce the repression of RNA Pol II activity. The role of histone modifications in preventing RNA Pol II transcription at rDNA in mammals is not clear, although there is evidence that rDNA methylation is required (Gagnon-Kugler et al., 2009) which indicates that correct chromatin organisation is again essential.

The repression of Pol II-mediated transcription from the intergenic spacers of rDNA is another important function of HDA6 in Arabidopsis thaliana, and an hda6 allele was found to produce Pol II-transcribed RNA from cryptic promoters throughout the rDNA. This in turn leads to siRNA production, which normally causes induction of heterochromatin but does not appear to do so in this case (Earley et al., 2010). Instead, the rDNA in question was found to be associated with histone modifications permissive for transcription such as H3K9ac, H3K14ac, H4K16ac. This indicates that siRNA production does not necessarily lead to the induction of repressive, heterochromatin-forming loops (Earley et al., 2010). This lead to the authors proposing a model in which HDA6 directly regulates Pol I and Pol II transcription, completely blocking the latter within the rDNA. In the most simple scenario, it could achieve this via deacetylation of histones. In this model, DNA methylation acts in a reinforcing role, perhaps by ensuring that the HDA6 protein remains concentrated at rDNA.

Before concluding this section, it is interesting to note that correct chromatin organisation of the rDNA repeats may be required not only for regulating their transcriptional activity, but also for their stability (Peng and Karpen, 2006). This report demonstrated that, in Drosophila melanogaster, chromatin-regulation pathways (including an RNAi pathway involving Dicer-2 and Su(var)3-9, and those responsible for dimethylation of H3K9) are necessary to prevent the occurrence of inter-repeat recombination. This ligase-mediated recombination, to which tandemly repeated DNA sequences are always prone, has the potential to liberate genotoxic circular DNA molecules, which were termed extrachromosomal circular repeat DNA (eccDNA). This may explain why so many pathways have evolved to prevent unregulated Pol II to occur at rDNA repeats. How chromatin actually prevents inter-repeat recombination between repeated DNA sequences is not clear, although in human cells an analogous process is known to involve the TTF-I complex (Guetg et al., 2010). Increased copy-number of certain rRNA gene variants is observed in histone methyltransferase mutants in Arabidopsis, which may suggest that correct histone modification also contributes to rDNA stability in plants (Pontvianne et al., 2012). In mammals, too, recent work has suggested that NoRC, or at least one of its component proteins, is important for the overall chromatin organisation of the rDNA repeats. The authors indicate that over-expression of Tip5 causes general changes to DNaseI accessibility (Zillner et al., 2013), a standard measure of chromatin compaction. This need not, however, be a direct effect. Given that a similar effect is mimicked by serum starvation, it is possible that there the eNoSC complex may also influence this process, although the direct physical association of Tip5 with the nuclear matrix reported here (Zillner et al., 2013) could be an additional way of amplifying local chromatin remodelling into an rDNA-wide effect. As an interesting aside, TTF-I may also have more general functions in maintaining heterochromatin stability throughout eukaryotic nuclei. Its ability to bind and stabilise heterochromatin repeats is for example essential for genomic integrity at centromeric repeats (Guetg et al., 2010) and suggests that its role at rDNA has evolved to exploit its chromatin remodelling capabilities.

Disruption of rDNA silencing and cancer prognosis

Nucleoli have long been observed to exhibit altered morphologies in different classes of cancer cell (Derenzini et al., 2000; Boisvert et al., 2007; Montanaro et al., 2008). In fact, such changes have been used as diagnostic and prognostic indicators since the early twentieth century (Shiue et al., 2010). The first report of such a link appears to have been by Pianese in 1896 (recorded in Donati et al., 2012) and was well-established by the later part of the twentieth century (Gani, 1976). This led to the hypothesis that there might be a correlation between tumourigenesis and the levels of rRNA/ribosome synthesis within the cell. At the molecular level, it has been noted that at least seven proto-oncogenes and/or tumour suppressors can alter overall levels of translation (Ruggero and Pandolfi, 2003). Furthermore, multiple ribosomal proteins are also found to be over-expressed in many different tumour types. Correlations between elevated rRNA levels and malignant transformation have also been observed (Ruggero and Pandolfi, 2003; White, 2008). This correlation has been shown to be consistent in a study of six different tumour types, and intriguingly tends to become stronger with advancing cancer stage (Williamson et al., 2006). rRNA has also been found to be over-expressed in many prostate cancer cell lines (Uemura et al., 2012).

Such studies provide convincing evidence that the changes to nucleolar morphology observed in many cancers are likely to be due to increased rRNA synthesis within these cells. This also seems to be correlated with enhanced translational capacity at the ribosome. However, most commentators traditionally considered this to be a simple consequence of the altered metabolic state of tumour cells following the resumption of proliferative growth (Donati et al., 2012). In other words, the activation level of the rDNA and associated nucleolar changes are analogous to the effects observed in non-cancerous cells when they are actively growing (e.g. in S-phase of the cell cycle). As cancer cells typically display elevated metabolic rates and hence have greater translational requirements compared with non-cancer cells, this could potentially explain the correlation between altered nucleolar morphology/rDNA transcription levels in cancer. Altered nucleolar organisation and levels of ribosome components have therefore been regarded as useful prognostic markers, but have not been considered from the perspective of cancer causation.

Cancer causation and RNA Pol I misregulation

As described above, RNA Pol I activity is associated with rapid cell growth and division during cancer, leading to changes to nucleolar morphology of potential prognostic use. However, some researchers have argued that there may also be a causal relationship between misregulation of rDNA and the occurrence of cancer (Ruggero and Pandolfi, 2003; White, 2008; Montanaro et al., 2012). In support of this, a recent study by Bywater et al. has demonstrated that aberrant hyperactivation of Pol I is causally required for malignancy in several tumours (see Bywater et al., 2012; associated commentary (Hannan et al., 2012)) and many oncogenic and tumour-prevention pathways upregulate RNA Pol I transcription at rDNA. One immediate consequent of this is that RNA Pol I has emerged as a potential chemotherapy target. One drug which may have important therapeutic potential in this regard is quarfloxin, a specific RNA Pol I-inhibitor which has been investigated for use in the treatment of neuroendocrine carcinoma (Drygin et al., 2009). Quarfloxin appears to confirm the relationship between altered rRNA transcription and prognosis of certain cancer types and has also been proposed for use in clinical trials for treatment of lymphoma and leukemia.

If RNA Pol I can be causative for cancer progression, this also suggests that there should exist cellular mechanisms which regulate rRNA transcriptionas a means of tumour prevention. rRNA synthesis is generally limited by tumour supressors such as p53, ARF, pRB and PTEN via a combination of direct and indirect mechanisms (Budde and Grummt, 1999; Zhai and Comai, 2000; White, 2008; Wang et al., 2013). For example, in cell lines derived from gastric cancers which had lost expression of the tumour suppressor ZNF545, restoration of its expression suppressed cell proliferation and induced apoptosis due to inhibition of rRNA transcription. Importantly, this silencing was linked to restoration of the normal heterochromatin marks at the promoters, including HP1β binding and H3K4 hypomethylation (Wang et al., 2013). Direct regulators of RNA Pol I transcription activity act by various mechanisms, including promoter remodelling, PIC formation, and elevated elongation rates. Such regulators include known oncogenes such as AML1-ETO and the tumor suppressors p53, pRb, and p14ARF (Hannan et al., 2012). Various lines of evidence indicate that these pathways often target RNA Pol I regulators/TFs such as UBF or RRN3. For example, the Retinoblastoma protein is required for suppression of UBF (Cavanaugh et al., 1995), possibly by preventing SL-1 binding (Hannan et al., 2000). Studies such as these indicate that rDNA misregulation during tumourigenesis, either by increased transcription by RNA Pol I at active rRNA genes, re-activation of previously silenced rRNA genes, or both.

The human TF complex, c-Myc, is of particular interest in the study of eukaryote transcription as it is able to regulate RNA Pol I, II and III (Gomez-Roman et al., 2006). It has therefore been proposed to coordinate rDNA transcription with the biosynthesis of other ribosome components (reviewed (Lempiäinen and Shore, 2009)) i.e. the ribosomal proteins (translated from mRNA transcribed by RNA Pol II) and the 5S rRNA (transcribed by RNA Pol III). The gene encoding c-Myc is considered a proto-oncogene, which may be due to far-reaching effects of c-Myc on transcription. In principle, c-Myc could be able to single-handedly increase the concentration of ribosomes within a cell and so drive growth and tumourigenesis (Arabi et al., 2005; Grandori et al., 2005; van Riggelen et al., 2010). For example, rRNA upregulation in prostate cancer lines closely correlates with levels c-Myc levels. c-Myc may be particularly important for aberrant activity of the Pol I complex during tumourigenesis (Uemura et al., 2012) although it should be noted that probably induces many pleiotropic effects on other cellular processes as well. This may make it difficult to describe the precise nature of the link and is one reason why c-Myc has not been successfully targeted by chemotherapy. Identification of some of its functional partners may help to resolve these complications (Chan et al., 2011). One possible part of this network is the kinase ERK, which is able to stabilise c-Myc by phosphorylation and can also activate both UBF and RRN3 in the same way, providing simultaneous routes to rDNA activation (Stefanovsky et al., 2001). As a final point, it has been reported SIRT7, a potential rRNA gene regulator associated with tumour progression (see above) may also regulate the expression of ribosomal proteins (Barber et al., 2012). It is possible that certain other rDNA regulators, including sirtuins, might also have be able to alter cellular translation in a concerted way reminiscent of c-Myc.

Another route to aberrant rRNA transcription (this time independent of c-Myc and ERK) is mediated by the growth factor receptor ErbB2, which acts as a TF for RNA Pol I. ErbB2 is upregulated in many cancers and is associated with increased metastasis and other aggressive traits. It appears to act by increasing the binding affinity of the polymerase to rDNA at transcription sites within the nucleolus, identified by visualisation of BrUTP incorporation sites (Li et al., 2011). Co-immunoprecipitation experiments indicated that ErbB2 binds to RNA Pol I as part of a complex with β-actin, and this binding leads to increased transcription levels in vivo (Li et al., 2011). Over-expression of ErbB2 lead to increased protein synthesis, indicating that RNA Pol I up-regulation can indeed remove the limiting factors on total cellular translation. rRNA gene silencing can also act to limit tumourigenesis via the tumour suppressor ARF, which triggers TTF-I nucleolar exit and thus reduced rRNA transcription. ARF and one of its target proteins, MDM2 (which has E3 ubiquitin ligase activity) compete for binding to TTF-I at overlapping sites. When levels of ARF are artificially ablated, MDM2 accumulates, binds to TTF-I at increased levels and catalyzes its ubiquitinylation. This reduces the concentration of TTF-I in the cell via by targeting it for proteosomic degradation (Lessard et al., 2012).

To understand the cellular networks that are proposed to link RNA Pol I regulation to cancer, it is also necessary to consider in more detail why misregulation might be associated with tumour progression, beyond the observation that cancer cells have high metabolic requirements. One fact which may be critical is the discovery that the key oncogene p53, which as noted above may regulate RNA Pol I activity, might also be partially controlled by it. It has been reported that the relative levels of ribosomal RNA (transcribed by RNA Pol I) and the rate of ribosomal protein synthesis can lead to changes in p53 levels in mammalian cells (Donati et al., 2011a). The likely complexity of any such interactions is demonstrated by research from the same group suggesting that Pol I can also control cellular proliferation via a p53-independent pathway. This pathway may instead involve E2F-1 (Donati et al., 2011b).

Sensing of reduced cellular energy status in the nucleolus by eNoSC has been proposed to play a key role in p53 accumulation, eventually leading to apoptosis or cell cycle arrest (Kumazawa et al., 2011). This is argued to occur via reduced rRNA synthesis which leads to release of Myb-binding protein 1a (MYBBP1A) from the nucleolus. This protein catalyzes acetylation of p53, which is a key element in causing it to accumulate and function (Kumazawa et al., 2011). Conversely, failure to down-regulate RNA Pol I could therefore block p53 hypomethylation as MYBBP1A would remain associated with the nucleolus even under pro-apoptopic conditions. This mechanism provides an attractive explanation of how p53-mediated cell cycle arrest could rely upon correct transcription of rRNA genes, and a route towards therapeutic intervention (Donati et al., 2012). It has previously been argued that hypoxia-induced acidification of human culture cells can promote the binding of rDNA to von Hippel-Lindau tumor suppressor protein (VHL) which in turn reduces the RNA Pol I-activating capacity of UBF (Mekhail et al., 2006). This would indicate that hypoxia can directly reduce ribosome production via pH-sensitive protein-protein interactions as well as via adverse affects on cellular energy status and is an additional pathway of relevance for disrupted response of the RNA Pol I machinery during tumourigenesis.

The sirtuin HDACs have also been proposed to play roles in cancer. HsSIRT1, for example, has been implicated in different forms of human cancer although determining precisely what role it is playing is not always clear (Deng, 2009). It has the potential to play a key part in cancer progression due to its interactions with p53 and other tumour supressors. The regulation of SIRT1 is correspondingly complex, as reviewed elsewhere (Liu et al., 2009), so its activities - and their significance to rDNA silencing - will only be briefly summarised here. SIRT1 is able to deacetylate p53 and physically interacts in vivo with several other proteins which regulate this deacetylation ability. Its expression is in turn controlled by p53 via two binding sites in its promoter, which repress its expression; SIRT1 can also repress its own expression via an autoregulatory loop as part of a complex with HIC1 (Chen et al., 2005). A particularly intriguing possibility is that SIRT1 might contribute to cancer development through a positive feedback loop on c-MYC expression. It has been proposed that this could perpetuate aberrant upregulation of c-MYC and suppression of apoptosis during colorectal cancer, for example (Menssen et al., 2012). Given its many interactions, and the fact that SIRT1 is present in in different domains of the nucleus, the issue of how these different roles are integrated remains an open question. Nor is it clear what role its control of rRNA transcription might play in its other cellular activities. For a discussion of sirtuin inhibitors as candidates for use in chemotherapy see (Liu et al., 2009).

rDNA silencing and further links to disease

A degree of care is needed as even when genes or pathways have been implicated in misregulating RNA Pol I, it is not necessarily the case that this is related to their oncogenic potential. It is likely that the significance of altered rDNA silencing may vary between cancer types and stages, and may be causative in some instances but a downstream consequence of malignant transformation in others. Such considerations should be borne in mind when evaluating the therapeutic potential of altering rRNA gene expression in any given case. Many chemotherapy agents affect RNA Pol I transcription, although of their nature most of these affect many other cellular processes as well so how critical their effects on the rDNA are remains unclear (Drygin et al., 2010).

Given what has been said previously concerning the potential genotoxic nature of recombination at repeated DNA, it is finally possible that mediators of rDNA silencing - such as those which control its organisation at the chromatin level - may also act as oncogenes under some circumstances. For example, the risk of rDNA which has deviated from their chromatin organisation producing DNA minicircles by inter-repeat recombination has been argued to have potentially carcinogenic effects (Peng and Karpen, 2007). The NML-interacting protein NMNAT1 may also have roles in preventing DNA breakage, and is correlated with increased DNA damage in lung cancer (Song et al., 2013), although whether this is linked to its role in down-regulating rRNA transcription is uncertain. NoRC also plays a role in preventing potentially carcinogenic chromosome-breakage events during nuclear division by maintaining cellular heterochromatin (Postepska-Igielska et al., 2013). However, this has been argued to be principally due to its roles in stabilising heterochromatin at the centromeres, telomeres and associated chromosomal regions, rather than at the rDNA locus (Postepska-Igielska et al., 2013). On the other hand, general effects on genome stability may involve its association with rDNA as well as other tandemly repeated regions.

Although they will not be considered in detail here, it should be noted that misregulation of rDNA transcription has also been observed various other human pathologies, and may be causal in at least some cases. These disorders often occur due to disruption of the epigenetic pathways which regulate RNA Pol I activity and include various hypertrophies and atrophies (Hannan et al., 2012). Such diseases may share common pathways with cancer in that they are typically associated with altered cellular growth rates. Defects in components of the RNA Pol I machinery itself are also causative for a range of rare congenital pathologies collectively termed ribosomopathies (Narla and Ebert, 2010).

Summary and conclusion

Correct regulation of rDNA transcription is a key part of the integration between energy status and translational capacity. Altered rDNA transcription (and associated changes to nucleolar morphology) have been correlated with the occurrence of cancer. Research over the last twenty years has succeeded in identifying some of the key protein complexes and chromatin-remodelling pathways which control this regulation in humans and in model systems. The transcription of rDNA by RNA Pol I can be silenced by the activity of NoRC and other complexes which form the rRNA genes into a repressive chromatin organisation. At active local, the exact level of transcription depends upon complexes with TF-activity, and especially UBF. The interactions between the various complexes which can activate or silence rRNA gene transcription are still only poorly understood, but it is clear that they represent the components of a sophisticated regulatory network. One key function is to link rRNA production to a cell's metabolic requirements and to the its energy reserves. Perturbation of these regulatory networks is therefore detrimental for the cell's function and has been linked to various pathologies. Elucidation of how misregulation of rDNA transcription of this kind contributes to the initiation and maintenance of cancer cell growth will be the focus of future research initiatives. Within this area, a particular question that will need to be addressed is the extent to which normalizing rRNA gene expression is able to restrict the growth of human cell lines and, if so, the nature of the molecular mechanisms which regulate this.

Bibliography

Chromosome alterations caused by hybridization and their bearing upon certain general genetic problems.
Navashin M.
Cytologia. 1934; 5: 169-203.
 
The nucleoli of cultured human lymphocytes. I. Nucleolar morphology in relation to transformation and the DNA cycle.
Gani R.
Exp Cell Res. 1976 Feb;97(2):249-58.
PMID 1248517
 
Fidelity of synthesis of preribosomal RNA in isolated nucleoli and nucleolar chromatin.
Ballal NR, Choi YC, Mouche R, Busche H.
Proc Natl Acad Sci U S A. 1977 Jun;74(6):2446-50.
PMID 196290
 
Reversible changes in nucleosome structure and histone H3 accessibility in transcriptionally active and inactive states of rDNA chromatin.
Prior CP, Cantor CR, Johnson EM, Littau VC, Allfrey VG.
Cell. 1983 Oct;34(3):1033-42.
PMID 6313204
 
Mechanisms of nucleolar dominance in animals and plants.
Reeder RH.
J Cell Biol. 1985 Nov;101(5 Pt 1):2013-6. (REVIEW)
PMID 3932369
 
Transcription of cloned eukaryotic ribosomal RNA genes.
Sollner-Webb B, Tower J.
Annu Rev Biochem. 1986;55:801-30. (REVIEW)
PMID 3527052
 
A major nucleolar protein, nucleolin, induces chromatin decondensation by binding to histone H1.
Erard MS, Belenguer P, Caizergues-Ferrer M, Pantaloni A, Amalric F.
Eur J Biochem. 1988 Aug 15;175(3):525-30.
PMID 3409881
 
DNase I sensitivity of ribosomal RNA genes in chromatin and nucleolar dominance in wheat.
Thompson WF, Flavell RB.
J Mol Biol. 1988 Dec 5;204(3):535-48.
PMID 3225845
 
Two different chromatin structures coexist in ribosomal RNA genes throughout the cell cycle.
Conconi A, Widmer RM, Koller T, Sogo JM.
Cell. 1989 Jun 2;57(5):753-61.
PMID 2720786
 
Identification of two forms of the RNA polymerase I transcription factor UBF.
O'Mahony DJ, Rothblum LI.
Proc Natl Acad Sci U S A. 1991 Apr 15;88(8):3180-4.
PMID 2014238
 
The TATA-binding protein and associated factors are integral components of the RNA polymerase I transcription factor, SL1.
Comai L, Tanese N, Tjian R.
Cell. 1992 Mar 6;68(5):965-76.
PMID 1547496
 
Chromatin structures and transcription of rDNA in yeast Saccharomyces cerevisiae.
Dammann R, Lucchini R, Koller T, Sogo JM.
Nucleic Acids Res. 1993 May 25;21(10):2331-8.
PMID 8506130
 
Localization of the RNA polymerase I transcription factor hUBF during the cell cycle.
Roussel P, Andre C, Masson C, Geraud G, Hernandez-Verdun D.
J Cell Sci. 1993 Feb;104 ( Pt 2):327-37.
PMID 8505363
 
The RNA polymerase I-specific transcription initiation factor UBF is associated with transcriptionally active and inactive ribosomal genes.
Zatsepina OV, Voit R, Grummt I, Spring H, Semenov MV, Trendelenburg MF.
Chromosoma. 1993 Nov;102(9):599-611.
PMID 8306821
 
Activity of RNA polymerase I transcription factor UBF blocked by Rb gene product.
Cavanaugh AH, Hempel WM, Taylor LJ, Rogalsky V, Todorov G, Rothblum LI.
Nature. 1995 Mar 9;374(6518):177-80.
PMID 7877691
 
The nucleolus: an organelle formed by the act of building a ribosome.
Melese T, Xue Z.
Curr Opin Cell Biol. 1995 Jun;7(3):319-24. (REVIEW)
PMID 7662360
 
The nucleolus.
Shaw PJ, Jordan EG.
Annu Rev Cell Dev Biol. 1995;11:93-121. (REVIEW)
PMID 8689574
 
Epigenetic silencing of RNA polymerase I transcription: a role for DNA methylation and histone modification in nucleolar dominance.
Chen ZJ, Pikaard CS.
Genes Dev. 1997 Aug 15;11(16):2124-36.
PMID 9284051
 
Direct evidence for SIR2 modulation of chromatin structure in yeast rDNA.
Fritze CE, Verschueren K, Strich R, Easton Esposito R.
EMBO J. 1997 Nov 3;16(21):6495-509.
PMID 9351831
 
Histones H3 and H4 are components of upstream activation factor required for the high-level transcription of yeast rDNA by RNA polymerase I.
Keener J, Dodd JA, Lalo D, Nomura M.
Proc Natl Acad Sci U S A. 1997 Dec 9;94(25):13458-62.
PMID 9391047
 
RNA polymerase I transcription on nucleosomal templates: the transcription termination factor TTF-I induces chromatin remodeling and relieves transcriptional repression.
Langst G, Blank TA, Becker PB, Grummt I.
EMBO J. 1997 Feb 17;16(4):760-8.
PMID 9049305
 
Does Saccharomyces need an organized nucleolus?
Nierras CR, Liebman SW, Warner JR.
Chromosoma. 1997 Jun;105(7-8):444-51.
PMID 9211972
 
An unusual form of transcriptional silencing in yeast ribosomal DNA.
Smith JS, Boeke JD.
Genes Dev. 1997 Jan 15;11(2):241-54.
PMID 9009206
 
TTF-I determines the chromatin architecture of the active rDNA promoter.
Langst G, Becker PB, Grummt I.
EMBO J. 1998 Jun 1;17(11):3135-45.
PMID 9606195
 
Distribution of a limited Sir2 protein pool regulates the strength of yeast rDNA silencing and is modulated by Sir4p.
Smith JS, Brachmann CB, Pillus L, Boeke JD.
Genetics. 1998 Jul;149(3):1205-19.
PMID 9649515
 
p53 represses ribosomal gene transcription.
Budde A, Grummt I.
Oncogene. 1999 Jan 28;18(4):1119-24.
PMID 10023689
 
The nucleolus: nucleolar space for RENT.
Cockell MM, Gasser SM.
Curr Biol. 1999 Jul 29-Aug 12;9(15):R575-6. (REVIEW)
PMID 10469557
 
Nucleolar dominance and silencing of transcription.
Pikaard CS.
Trends Plant Sci. 1999 Dec;4(12):478-483.
PMID 10562732
 
Net1, a Sir2-associated nucleolar protein required for rDNA silencing and nucleolar integrity.
Straight AF, Shou W, Dowd GJ, Turck CW, Deshaies RJ, Johnson AD, Moazed D.
Cell. 1999 Apr 16;97(2):245-56.
PMID 10219245
 
Phosphorylation by G1-specific cdk-cyclin complexes activates the nucleolar transcription factor UBF.
Voit R, Hoffmann M, Grummt I.
EMBO J. 1999 Apr 1;18(7):1891-9.
PMID 10202152
 
Nucleolar size indicates the rapidity of cell proliferation in cancer tissues.
Derenzini M, Trere D, Pession A, Govoni M, Sirri V, Chieco P.
J Pathol. 2000 Jun;191(2):181-6.
PMID 10861579
 
Rb and p130 regulate RNA polymerase I transcription: Rb disrupts the interaction between UBF and SL-1.
Hannan KM, Hannan RD, Smith SD, Jefferson LS, Lun M, Rothblum LI.
Oncogene. 2000 Oct 12;19(43):4988-99.
PMID 11042686
 
Requirement of NAD and SIR2 for life-span extension by calorie restriction in Saccharomyces cerevisiae.
Lin SJ, Defossez PA, Guarente L.
Science. 2000 Sep 22;289(5487):2126-8.
PMID 11000115
 
The epigenetics of nucleolar dominance.
Pikaard CS.
Trends Genet. 2000 Nov;16(11):495-500. (REVIEW)
PMID 11074291
 
TOR, a central controller of cell growth.
Schmelzle T, Hall MN.
Cell. 2000 Oct 13;103(2):253-62. (REVIEW)
PMID 11057898
 
Repression of RNA polymerase I transcription by the tumor suppressor p53.
Zhai W, Comai L.
Mol Cell Biol. 2000 Aug;20(16):5930-8.
PMID 10913176
 
Histone H3 lysine 4 methylation is mediated by Set1 and required for cell growth and rDNA silencing in Saccharomyces cerevisiae.
Briggs SD, Bryk M, Strahl BD, Cheung WL, Davie JK, Dent SY, Winston F, Allis CD.
Genes Dev. 2001 Dec 15;15(24):3286-95.
PMID 11751634
 
An immediate response of ribosomal transcription to growth factor stimulation in mammals is mediated by ERK phosphorylation of UBF.
Stefanovsky VY, Pelletier G, Hannan R, Gagnon-Kugler T, Rothblum LI, Moss T.
Mol Cell. 2001 Nov;8(5):1063-73.
PMID 11741541
 
NoRC--a novel member of mammalian ISWI-containing chromatin remodeling machines.
Strohner R, Nemeth A, Jansa P, Hofmann-Rohrer U, Santoro R, Langst G, Grummt I.
EMBO J. 2001 Sep 3;20(17):4892-900.
PMID 11532953
 
Directed proteomic analysis of the human nucleolus.
Andersen JS, Lyon CE, Fox AH, Leung AK, Lam YW, Steen H, Mann M, Lamond AI.
Curr Biol. 2002 Jan 8;12(1):1-11.
PMID 11790298
 
CSB is a component of RNA pol I transcription.
Bradsher J, Auriol J, Proietti de Santis L, Iben S, Vonesch JL, Grummt I, Egly JM.
Mol Cell. 2002 Oct;10(4):819-29.
PMID 12419226
 
Evidence that Set1, a factor required for methylation of histone H3, regulates rDNA silencing in S. cerevisiae by a Sir2-independent mechanism.
Bryk M, Briggs SD, Strahl BD, Curcio MJ, Allis CD, Winston F.
Curr Biol. 2002 Jan 22;12(2):165-70.
PMID 11818070
 
TFIIH plays an essential role in RNA polymerase I transcription.
Iben S, Tschochner H, Bier M, Hoogstraten D, Hozak P, Egly JM, Grummt I.
Cell. 2002 May 3;109(3):297-306.
PMID 12015980
 
mTOR interacts with raptor to form a nutrient-sensitive complex that signals to the cell growth machinery.
Kim DH, Sarbassov DD, Ali SM, King JE, Latek RR, Erdjument-Bromage H, Tempst P, Sabatini DM.
Cell. 2002 Jul 26;110(2):163-75.
PMID 12150925
 
UBF binding in vivo is not restricted to regulatory sequences within the vertebrate ribosomal DNA repeat.
O'Sullivan AC, Sullivan GJ, McStay B.
Mol Cell Biol. 2002 Jan;22(2):657-68.
PMID 11756560
 
The nucleolar remodeling complex NoRC mediates heterochromatin formation and silencing of ribosomal gene transcription.
Santoro R, Li J, Grummt I.
Nat Genet. 2002 Nov;32(3):393-6. Epub 2002 Oct 7.
PMID 12368916
 
In exponentially growing Saccharomyces cerevisiae cells, rRNA synthesis is determined by the summed RNA polymerase I loading rate rather than by the number of active genes.
French SL, Osheim YN, Cioci F, Nomura M, Beyer AL.
Mol Cell Biol. 2003 Mar;23(5):1558-68.
PMID 12588976
 
The nucleolus: a site of ribonucleoprotein maturation.
Gerbi SA, Borovjagin AV, Lange TS.
Curr Opin Cell Biol. 2003 Jun;15(3):318-25. (REVIEW)
PMID 12787774
 
Life on a planet of its own: regulation of RNA polymerase I transcription in the nucleolus.
Grummt I.
Genes Dev. 2003 Jul 15;17(14):1691-702. (REVIEW)
PMID 12865296
 
Functional consequences of histone modifications.
Iizuka M, Smith MM.
Curr Opin Genet Dev. 2003 Apr;13(2):154-60. (REVIEW)
PMID 12672492
 
GbetaL, a positive regulator of the rapamycin-sensitive pathway required for the nutrient-sensitive interaction between raptor and mTOR.
Kim DH, Sarbassov DD, Ali SM, Latek RR, Guntur KV, Erdjument-Bromage H, Tempst P, Sabatini DM.
Mol Cell. 2003 Apr;11(4):895-904.
PMID 12718876
 
Does the ribosome translate cancer?
Ruggero D, Pandolfi PP.
Nat Rev Cancer. 2003 Mar;3(3):179-92. (REVIEW)
PMID 12612653
 
Chromatin-mediated regulation of nucleolar structure and RNA Pol I localization by TOR.
Tsang CK, Bertram PG, Ai W, Drenan R, Zheng XF.
EMBO J. 2003 Nov 17;22(22):6045-56.
PMID 14609951
 
The Sir2 family of protein deacetylases.
Blander G, Guarente L.
Annu Rev Biochem. 2004;73:417-35. (REVIEW)
PMID 15189148
 
Upstream binding factor association induces large-scale chromatin decondensation.
Chen D, Belmont AS, Huang S.
Proc Natl Acad Sci U S A. 2004 Oct 19;101(42):15106-11. Epub 2004 Oct 11.
PMID 15477594
 
A nuclear FK506-binding protein is a histone chaperone regulating rDNA silencing.
Kuzuhara T, Horikoshi M.
Nat Struct Mol Biol. 2004 Mar;11(3):275-83. Epub 2004 Feb 8.
PMID 14981505
 
A concerted DNA methylation/histone methylation switch regulates rRNA gene dosage control and nucleolar dominance.
Lawrence RJ, Earley K, Pontes O, Silva M, Chen ZJ, Neves N, Viegas W, Pikaard CS.
Mol Cell. 2004 Feb 27;13(4):599-609.
PMID 14992728
 
Condensin regulates rDNA silencing by modulating nucleolar Sir2p.
Machin F, Paschos K, Jarmuz A, Torres-Rosell J, Pade C, Aragon L.
Curr Biol. 2004 Jan 20;14(2):125-30.
PMID 14738734
 
At the crossroads of growth control; making ribosomal RNA.
Moss T.
Curr Opin Genet Dev. 2004 Apr;14(2):210-7. (REVIEW)
PMID 15196469
 
The chromatin remodeling complex NoRC and TTF-I cooperate in the regulation of the mammalian rRNA genes in vivo.
Nemeth A, Strohner R, Grummt I, Langst G.
Nucleic Acids Res. 2004 Aug 3;32(14):4091-9. Print 2004.
PMID 15292447
 
Transcription of rDNA in the yeast Saccharomyces cerevisiae.
Nomura M, Nogi Y, Oakes M.
Molecular Biology Intelligence Unit. 2004; The Nucleolus, 128-153.
 
Nucleolar proteome dynamics.
Andersen JS, Lam YW, Leung AK, Ong SE, Lyon CE, Lamond AI, Mann M.
Nature. 2005 Jan 6;433(7021):77-83.
PMID 15635413
 
c-Myc associates with ribosomal DNA and activates RNA polymerase I transcription.
Arabi A, Wu S, Ridderstrale K, Bierhoff H, Shiue C, Fatyol K, Fahlen S, Hydbring P, Soderberg O, Grummt I, Larsson LG, Wright AP.
Nat Cell Biol. 2005 Mar;7(3):303-10. Epub 2005 Feb 20.
PMID 15723053
 
Tumor suppressor HIC1 directly regulates SIRT1 to modulate p53-dependent DNA-damage responses.
Chen WY, Wang DH, Yen RC, Luo J, Gu W, Baylin SB.
Cell. 2005 Nov 4;123(3):437-48.
PMID 16269335
 
c-Myc binds to human ribosomal DNA and stimulates transcription of rRNA genes by RNA polymerase I.
Grandori C, Gomez-Roman N, Felton-Edkins ZA, Ngouenet C, Galloway DA, Eisenman RN, White RJ.
Nat Cell Biol. 2005 Mar;7(3):311-8.
PMID 15723054
 
UBF-binding site arrays form pseudo-NORs and sequester the RNA polymerase I transcription machinery.
Mais C, Wright JE, Prieto JL, Raggett SL, McStay B.
Genes Dev. 2005 Jan 1;19(1):50-64. Epub 2004 Dec 14.
PMID 15598984
 
Evolutionarily conserved and nonconserved cellular localizations and functions of human SIRT proteins.
Michishita E, Park JY, Burneskis JM, Barrett JC, Horikawa I.
Mol Biol Cell. 2005 Oct;16(10):4623-35. Epub 2005 Aug 3.
PMID 16079181
 
Proteomic analysis of the Arabidopsis nucleolus suggests novel nucleolar functions.
Pendle AF, Clark GP, Boon R, Lewandowska D, Lam YW, Andersen J, Mann M, Lamond AI, Brown JW, Shaw PJ.
Mol Biol Cell. 2005 Jan;16(1):260-9. Epub 2004 Oct 20.
PMID 15496452
 
RNA-polymerase-I-directed rDNA transcription, life and works.
Russell J, Zomerdijk JC.
Trends Biochem Sci. 2005 Feb;30(2):87-96. (REVIEW)
PMID 15691654
 
Histones are required for transcription of yeast rRNA genes by RNA polymerase I.
Tongaonkar P, French SL, Oakes ML, Vu L, Schneider DA, Beyer AL, Nomura M.
Proc Natl Acad Sci U S A. 2005 Jul 19;102(29):10129-34. Epub 2005 Jul 7.
PMID 16002464
 
The PHD finger/bromodomain of NoRC interacts with acetylated histone H4K16 and is sufficient for rDNA silencing.
Zhou Y, Grummt I.
Curr Biol. 2005 Aug 9;15(15):1434-8.
PMID 16085498
 
Nucleolin is a histone chaperone with FACT-like activity and assists remodeling of nucleosomes.
Angelov D, Bondarenko VA, Almagro S, Menoni H, Mongelard F, Hans F, Mietton F, Studitsky VM, Hamiche A, Dimitrov S, Bouvet P.
EMBO J. 2006 Apr 19;25(8):1669-79. Epub 2006 Apr 6.
PMID 16601700
 
Altered sirtuin expression is associated with node-positive breast cancer.
Ashraf N, Zino S, Macintyre A, Kingsmore D, Payne AP, George WD, Shiels PG.
Br J Cancer. 2006 Oct 23;95(8):1056-61. Epub 2006 Sep 26.
PMID 17003781
 
The WSTF-SNF2h chromatin remodeling complex interacts with several nuclear proteins in transcription.
Cavellan E, Asp P, Percipalle P, Farrants AK.
J Biol Chem. 2006 Jun 16;281(24):16264-71. Epub 2006 Apr 9.
PMID 16603771
 
Erasure of histone acetylation by Arabidopsis HDA6 mediates large-scale gene silencing in nucleolar dominance.
Earley K, Lawrence RJ, Pontes O, Reuther R, Enciso AJ, Silva M, Neves N, Gross M, Viegas W, Pikaard CS.
Genes Dev. 2006 May 15;20(10):1283-93. Epub 2006 Apr 28.
PMID 16648464
 
Mammalian Sir2 homolog SIRT7 is an activator of RNA polymerase I transcription.
Ford E, Voit R, Liszt G, Magin C, Grummt I, Guarente L.
Genes Dev. 2006 May 1;20(9):1075-80. Epub 2006 Apr 17.
PMID 16618798
 
Activation by c-Myc of transcription by RNA polymerases I, II and III.
Gomez-Roman N, Felton-Edkins ZA, Kenneth NS, Goodfellow SJ, Athineos D, Zhang J, Ramsbottom BA, Innes F, Kantidakis T, Kerr ER, Brodie J, Grandori C, White RJ.
Biochem Soc Symp. 2006;(73):141-54. (REVIEW)
PMID 16626295
 
Intergenic transcripts regulate the epigenetic state of rRNA genes.
Mayer C, Schmitz KM, Li J, Grummt I, Santoro R.
Mol Cell. 2006 May 5;22(3):351-61.
PMID 16678107
 
Nucleolar dominance: a model for rRNA gene silencing.
McStay B.
Genes Dev. 2006 May 15;20(10):1207-14. (REVIEW)
PMID 16702398
 
Restriction of rRNA synthesis by VHL maintains energy equilibrium under hypoxia.
Mekhail K, Rivero-Lopez L, Khacho M, Lee S.
Cell Cycle. 2006 Oct;5(20):2401-13. Epub 2006 Oct 16.
PMID 17102617
 
Acetylation of UBF changes during the cell cycle and regulates the interaction of UBF with RNA polymerase I.
Meraner J, Lechner M, Loidl A, Goralik-Schramel M, Voit R, Grummt I, Loidl P.
Nucleic Acids Res. 2006 Mar 31;34(6):1798-806. Print 2006.
PMID 16582105
 
UBF activates RNA polymerase I transcription by stimulating promoter escape.
Panov KI, Friedrich JK, Russell J, Zomerdijk JC.
EMBO J. 2006 Jul 26;25(14):3310-22. Epub 2006 Jul 6.
PMID 16858408
 
The chromatin remodelling complex WSTF-SNF2h interacts with nuclear myosin 1 and has a role in RNA polymerase I transcription.
Percipalle P, Fomproix N, Cavellan E, Voit R, Reimer G, Kruger T, Thyberg J, Scheer U, Grummt I, Farrants AK.
EMBO Rep. 2006 May;7(5):525-30. Epub 2006 Mar 3.
PMID 16514417
 
The Arabidopsis chromatin-modifying nuclear siRNA pathway involves a nucleolar RNA processing center.
Pontes O, Li CF, Costa Nunes P, Haag J, Ream T, Vitins A, Jacobsen SE, Pikaard CS.
Cell. 2006 Jul 14;126(1):79-92.
PMID 16839878
 
Structure and function of the nucleolus in the spotlight.
Raska I, Shaw PJ, Cmarko D.
Curr Opin Cell Biol. 2006 Jun;18(3):325-34. Epub 2006 May 9. (REVIEW)
PMID 16687244
 
Regulation of rRNA synthesis in human and mouse cells is not determined by changes in active gene count.
Stefanovsky V, Moss T.
Cell Cycle. 2006 Apr;5(7):735-9. Epub 2006 Apr 1.
PMID 16582637
 
Nascent pre-rRNA overexpression correlates with an adverse prognosis in alveolar rhabdomyosarcoma.
Williamson D, Lu YJ, Fang C, Pritchard-Jones K, Shipley J.
Genes Chromosomes Cancer. 2006 Sep;45(9):839-45.
PMID 16770781
 
The multifunctional nucleolus.
Boisvert FM, van Koningsbruggen S, Navascues J, Lamond AI.
Nat Rev Mol Cell Biol. 2007 Jul;8(7):574-85. (REVIEW)
PMID 17519961
 
JHDM1B/FBXL10 is a nucleolar protein that represses transcription of ribosomal RNA genes.
Frescas D, Guardavaccaro D, Bassermann F, Koyama-Nasu R, Pagano M.
Nature. 2007 Nov 8;450(7167):309-13.
PMID 17994099
 
RNA polymerase I in yeast transcribes dynamic nucleosomal rDNA.
Jones HS, Kawauchi J, Braglia P, Alen CM, Kent NA, Proudfoot NJ.
Nat Struct Mol Biol. 2007 Feb;14(2):123-30. Epub 2007 Jan 28.
PMID 17259992
 
H3K9 methylation and RNA interference regulate nucleolar organization and repeated DNA stability.
Peng JC, Karpen GH.
Nat Cell Biol. 2007 Jan;9(1):25-35. Epub 2006 Dec 10.
PMID 17159999
 
Recruitment of factors linking transcription and processing of pre-rRNA to NOR chromatin is UBF-dependent and occurs independent of transcription in human cells.
Prieto JL, McStay B.
Genes Dev. 2007 Aug 15;21(16):2041-54.
PMID 17699751
 
Activation of RNA polymerase I transcription by cockayne syndrome group B protein and histone methyltransferase G9a.
Yuan X, Feng W, Imhof A, Grummt I, Zhou Y.
Mol Cell. 2007 Aug 17;27(4):585-95.
PMID 17707230
 
Structure and function of ribosomal RNA gene chromatin.
Birch JL, Zomerdijk JC.
Biochem Soc Trans. 2008 Aug;36(Pt 4):619-24. doi: 10.1042/BST0360619. (REVIEW)
PMID 18631128
 
Truncated Cockayne syndrome B protein represses elongation by RNA polymerase I.
Lebedev A, Scharffetter-Kochanek K, Iben S.
J Mol Biol. 2008 Oct 3;382(2):266-74. doi: 10.1016/j.jmb.2008.07.018. Epub 2008 Jul 16.
PMID 18656484
 
The structure of NoRC-associated RNA is crucial for targeting the chromatin remodelling complex NoRC to the nucleolus.
Mayer C, Neubert M, Grummt I.
EMBO Rep. 2008 Aug;9(8):774-80. doi: 10.1038/embor.2008.109. Epub 2008 Jul 4.
PMID 18600236
 
Preparation of Arabidopsis nuclei and nucleoli.
McKeown P, Pendle AF, Shaw PJ.
Methods Mol Biol. 2008;463:67-75. doi: 10.1007/978-1-59745-406-3_5.
PMID 18951161
 
The epigenetics of rRNA genes: from molecular to chromosome biology.
McStay B, Grummt I.
Annu Rev Cell Dev Biol. 2008;24:131-57. doi: 10.1146/annurev.cellbio.24.110707.175259. (REVIEW)
PMID 18616426
 
Nucleolus, ribosomes, and cancer.
Montanaro L, Trere D, Derenzini M.
Am J Pathol. 2008 Aug;173(2):301-10. doi: 10.2353/ajpath.2008.070752. Epub 2008 Jun 26. (REVIEW)
PMID 18583314
 
Epigenetic control of rDNA loci in response to intracellular energy status.
Murayama A, Ohmori K, Fujimura A, Minami H, Yasuzawa-Tanaka K, Kuroda T, Oie S, Daitoku H, Okuwaki M, Nagata K, Fukamizu A, Kimura K, Shimizu T, Yanagisawa J.
Cell. 2008 May 16;133(4):627-39. doi: 10.1016/j.cell.2008.03.030.
PMID 18485871
 
Multimegabase silencing in nucleolar dominance involves siRNA-directed DNA methylation and specific methylcytosine-binding proteins.
Preuss SB, Costa-Nunes P, Tucker S, Pontes O, Lawrence RJ, Mosher R, Kasschau KD, Carrington JC, Baulcombe DC, Viegas W, Pikaard CS.
Mol Cell. 2008 Dec 5;32(5):673-84. doi: 10.1016/j.molcel.2008.11.009.
PMID 19061642
 
UBF levels determine the number of active ribosomal RNA genes in mammals.
Sanij E, Poortinga G, Sharkey K, Hung S, Holloway TP, Quin J, Robb E, Wong LH, Thomas WG, Stefanovsky V, Moss T, Rothblum L, Hannan KM, McArthur GA, Pearson RB, Hannan RD.
J Cell Biol. 2008 Dec 29;183(7):1259-74. doi: 10.1083/jcb.200805146. Epub 2008 Dec 22.
PMID 19103806
 
RNA polymerases I and III, non-coding RNAs and cancer.
White RJ.
Trends Genet. 2008 Dec;24(12):622-9. doi: 10.1016/j.tig.2008.10.003. Epub 2008 Nov 6. (REVIEW)
PMID 18980784
 
FACT facilitates chromatin transcription by RNA polymerases I and III.
Birch JL, Tan BC, Panov KI, Panova TB, Andersen JS, Owen-Hughes TA, Russell J, Lee SC, Zomerdijk JC.
EMBO J. 2009 Apr 8;28(7):854-65. doi: 10.1038/emboj.2009.33. Epub 2009 Feb 12.
PMID 19214185
 
SIRT1, is it a tumor promoter or tumor suppressor?
Deng CX.
Int J Biol Sci. 2009;5(2):147-52. Epub 2009 Jan 21. (REVIEW)
PMID 19173036
 
Anticancer activity of CX-3543: a direct inhibitor of rRNA biogenesis.
Drygin D, Siddiqui-Jain A, O'Brien S, Schwaebe M, Lin A, Bliesath J, Ho CB, Proffitt C, Trent K, Whitten JP, Lim JK, Von Hoff D, Anderes K, Rice WG.
Cancer Res. 2009 Oct 1;69(19):7653-61. doi: 10.1158/0008-5472.CAN-09-1304. Epub 2009 Sep 8.
PMID 19738048
 
Recent progress in the biology and physiology of sirtuins.
Finkel T, Deng CX, Mostoslavsky R.
Nature. 2009 Jul 30;460(7255):587-91. doi: 10.1038/nature08197. (REVIEW)
PMID 19641587
 
Loss of human ribosomal gene CpG methylation enhances cryptic RNA polymerase II transcription and disrupts ribosomal RNA processing.
Gagnon-Kugler T, Langlois F, Stefanovsky V, Lessard F, Moss T.
Mol Cell. 2009 Aug 28;35(4):414-25. doi: 10.1016/j.molcel.2009.07.008.
PMID 19716787
 
Involvement of SIRT7 in resumption of rDNA transcription at the exit from mitosis.
Grob A, Roussel P, Wright JE, McStay B, Hernandez-Verdun D, Sirri V.
J Cell Sci. 2009 Feb 15;122(Pt 4):489-98. doi: 10.1242/jcs.042382. Epub 2009 Jan 27.
PMID 19174463
 
Growth control and ribosome biogenesis.
Lempiainen H, Shore D.
Curr Opin Cell Biol. 2009 Dec;21(6):855-63. doi: 10.1016/j.ceb.2009.09.002. Epub 2009 Sep 30. (REVIEW)
PMID 19796927
 
The critical role of the class III histone deacetylase SIRT1 in cancer.
Liu T, Liu PY, Marshall GM.
Cancer Res. 2009 Mar 1;69(5):1702-5. doi: 10.1158/0008-5472.CAN-08-3365. Epub 2009 Feb 24. (REVIEW)
PMID 19244112
 
Chromatin: linking structure and function in the nucleolus.
McKeown PC, Shaw PJ.
Chromosoma. 2009 Feb;118(1):11-23. doi: 10.1007/s00412-008-0184-2. Epub 2008 Oct 17. (REVIEW)
PMID 18925405
 
The role of UBF in regulating the structure and dynamics of transcriptionally active rDNA chromatin.
Sanij E, Hannan RD.
Epigenetics. 2009 Aug 16;4(6):374-82. Epub 2009 Aug 6. (REVIEW)
PMID 19717978
 
Reversible acetylation of the chromatin remodelling complex NoRC is required for non-coding RNA-dependent silencing.
Zhou Y, Schmitz KM, Mayer C, Yuan X, Akhtar A, Grummt I.
Nat Cell Biol. 2009 Aug;11(8):1010-6. doi: 10.1038/ncb1914. Epub 2009 Jul 5.
PMID 19578370
 
Noncoding transcripts in sense and antisense orientation regulate the epigenetic state of ribosomal RNA genes.
Bierhoff H, Schmitz K, Maass F, Ye J, Grummt I.
Cold Spring Harb Symp Quant Biol. 2010;75:357-64. doi: 10.1101/sqb.2010.75.060. Epub 2011 Apr 18.
PMID 21502405
 
Linking calorie restriction to longevity through sirtuins and autophagy: any role for TOR.
Blagosklonny MV.
Cell Death Dis. 2010;1:e12. doi: 10.1038/cddis.2009.17.
PMID 21364614
 
The nucleolus under stress.
Boulon S, Westman BJ, Hutten S, Boisvert FM, Lamond AI.
Mol Cell. 2010 Oct 22;40(2):216-27. doi: 10.1016/j.molcel.2010.09.024. (REVIEW)
PMID 20965417
 
The RNA polymerase I transcription machinery: an emerging target for the treatment of cancer.
Drygin D, Rice WG, Grummt I.
Annu Rev Pharmacol Toxicol. 2010;50:131-56. doi: 10.1146/annurev.pharmtox.010909.105844. (REVIEW)
PMID 20055700
 
Mechanisms of HDA6-mediated rRNA gene silencing: suppression of intergenic Pol II transcription and differential effects on maintenance versus siRNA-directed cytosine methylation.
Earley KW, Pontvianne F, Wierzbicki AT, Blevins T, Tucker S, Costa-Nunes P, Pontes O, Pikaard CS.
Genes Dev. 2010 Jun 1;24(11):1119-32. doi: 10.1101/gad.1914110.
PMID 20516197
 
Linking rDNA transcription to the cellular energy supply.
Grummt I, Voit R.
Cell Cycle. 2010 Jan 15;9(2):225-6. Epub 2010 Jan 12.
PMID 20023389
 
The NoRC complex mediates the heterochromatin formation and stability of silent rRNA genes and centromeric repeats.
Guetg C, Lienemann P, Sirri V, Grummt I, Hernandez-Verdun D, Hottiger MO, Fussenegger M, Santoro R.
EMBO J. 2010 Jul 7;29(13):2135-46. doi: 10.1038/emboj.2010.17. Epub 2010 Feb 18.
PMID 20168299
 
Ten years of NAD-dependent SIR2 family deacetylases: implications for metabolic diseases.
Imai S, Guarente L.
Trends Pharmacol Sci. 2010 May;31(5):212-20. doi: 10.1016/j.tips.2010.02.003. Epub 2010 Mar 11. (REVIEW)
PMID 20226541
 
AtFKBP53 is a histone chaperone required for repression of ribosomal RNA gene expression in Arabidopsis.
Li H, Luan S.
Cell Res. 2010 Mar;20(3):357-66. doi: 10.1038/cr.2010.22. Epub 2010 Feb 9.
PMID 20142844
 
Ribosomopathies: human disorders of ribosome dysfunction.
Narla A, Ebert BL.
Blood. 2010 Apr 22;115(16):3196-205. doi: 10.1182/blood-2009-10-178129. Epub 2010 Mar 1. (REVIEW)
PMID 20194897
 
TOR-dependent reduction in the expression level of Rrn3p lowers the activity of the yeast RNA Pol I machinery, but does not account for the strong inhibition of rRNA production.
Philippi A, Steinbauer R, Reiter A, Fath S, Leger-Silvestre I, Milkereit P, Griesenbeck J, Tschochner H.
Nucleic Acids Res. 2010 Sep;38(16):5315-26. doi: 10.1093/nar/gkq264. Epub 2010 Apr 25.
PMID 20421203
 
Nucleolin is required for DNA methylation state and the expression of rRNA gene variants in Arabidopsis thaliana.
Pontvianne F, Abou-Ellail M, Douet J, Comella P, Matia I, Chandrasekhara C, Debures A, Blevins T, Cooke R, Medina FJ, Tourmente S, Pikaard CS, Saez-Vasquez J.
PLoS Genet. 2010 Nov 24;6(11):e1001225. doi: 10.1371/journal.pgen.1001225.
PMID 21124873
 
Nucleolar organization, growth control and cancer.
Shiue CN, Arabi A, Wright AP.
Epigenetics. 2010 Apr 1;5(3):200-5. Epub 2010 Apr 1.
PMID 20305389
 
mTOR binds to the promoters of RNA polymerase I- and III-transcribed genes.
Tsang CK, Liu H, Zheng XF.
Cell Cycle. 2010 Mar 1;9(5):953-7. Epub 2010 Mar 7.
PMID 20038818
 
MYC as a regulator of ribosome biogenesis and protein synthesis.
van Riggelen J, Yetil A, Felsher DW.
Nat Rev Cancer. 2010 Apr;10(4):301-9. doi: 10.1038/nrc2819. (REVIEW)
PMID 20332779
 
AKT promotes rRNA synthesis and cooperates with c-MYC to stimulate ribosome biogenesis in cancer.
Chan JC, Hannan KM, Riddell K, Ng PY, Peck A, Lee RS, Hung S, Astle MV, Bywater M, Wall M, Poortinga G, Jastrzebski K, Sheppard KE, Hemmings BA, Hall MN, Johnstone RW, McArthur GA, Hannan RD, Pearson RB.
Sci Signal. 2011 Aug 30;4(188):ra56. doi: 10.1126/scisignal.2001754.
PMID 21878679
 
The balance between rRNA and ribosomal protein synthesis up- and downregulates the tumour suppressor p53 in mammalian cells.
Donati G, Bertoni S, Brighenti E, Vici M, Trere D, Volarevic S, Montanaro L, Derenzini M.
Oncogene. 2011a Jul 21;30(29):3274-88. doi: 10.1038/onc.2011.48. Epub 2011 Mar 14.
PMID 21399665
 
Selective inhibition of rRNA transcription downregulates E2F-1: a new p53-independent mechanism linking cell growth to cell proliferation.
Donati G, Brighenti E, Vici M, Mazzini G, Trere D, Montanaro L, Derenzini M.
J Cell Sci. 2011b Sep 1;124(Pt 17):3017-28. doi: 10.1242/jcs.086074.
PMID 21878508
 
Rapamycin increases rDNA stability by enhancing association of Sir2 with rDNA in Saccharomyces cerevisiae.
Ha CW, Huh WK.
Nucleic Acids Res. 2011 Mar;39(4):1336-50. doi: 10.1093/nar/gkq895. Epub 2010 Oct 14.
PMID 20947565
 
Novel nucleolar pathway connecting intracellular energy status with p53 activation.
Kumazawa T, Nishimura K, Kuroda T, Ono W, Yamaguchi C, Katagiri N, Tsuchiya M, Masumoto H, Nakajima Y, Murayama A, Kimura K, Yanagisawa J.
J Biol Chem. 2011 Jun 10;286(23):20861-9. doi: 10.1074/jbc.M110.209916. Epub 2011 Apr 6.
PMID 21471221
 
Nuclear ErbB2 enhances translation and cell growth by activating transcription of ribosomal RNA genes.
Li LY, Chen H, Hsieh YH, Wang YN, Chu HJ, Chen YH, Chen HY, Chien PJ, Ma HT, Tsai HC, Lai CC, Sher YP, Lien HC, Tsai CH, Hung MC.
Cancer Res. 2011 Jun 15;71(12):4269-79. doi: 10.1158/0008-5472.CAN-10-3504. Epub 2011 May 9.
PMID 21555369
 
The structure of rDNA chromatin.
Shaw PJ, McKeown PC.
In The Nucleolus. Protein Reviews Volume 15, 2011, pp 43-55.
 
The chromatin remodelling complex B-WICH changes the chromatin structure and recruits histone acetyl-transferases to active rRNA genes.
Vintermist A, Bohm S, Sadeghifar F, Louvet E, Mansen A, Percipalle P, Ostlund Farrants AK.
PLoS One. 2011 Apr 29;6(4):e19184. doi: 10.1371/journal.pone.0019184.
PMID 21559432
 
SIRT7 links H3K18 deacetylation to maintenance of oncogenic transformation.
Barber MF, Michishita-Kioi E, Xi Y, Tasselli L, Kioi M, Moqtaderi Z, Tennen RI, Paredes S, Young NL, Chen K, Struhl K, Garcia BA, Gozani O, Li W, Chua KF.
Nature. 2012 Jul 5;487(7405):114-8. doi: 10.1038/nature11043.
PMID 22722849
 
Inhibition of RNA polymerase I as a therapeutic strategy to promote cancer-specific activation of p53.
Bywater MJ, Poortinga G, Sanij E, Hein N, Peck A, Cullinane C, Wall M, Cluse L, Drygin D, Anderes K, Huser N, Proffitt C, Bliesath J, Haddach M, Schwaebe MK, Ryckman DM, Rice WG, Schmitt C, Lowe SW, Johnstone RW, Pearson RB, McArthur GA, Hannan RD.
Cancer Cell. 2012 Jul 10;22(1):51-65. doi: 10.1016/j.ccr.2012.05.019.
PMID 22789538
 
The histone H3 lysine 56 acetylation pathway is regulated by target of rapamycin (TOR) signaling and functions directly in ribosomal RNA biogenesis.
Chen H, Fan M, Pfeffer LM, Laribee RN.
Nucleic Acids Res. 2012 Aug;40(14):6534-46. doi: 10.1093/nar/gks345. Epub 2012 May 2.
PMID 22553361
 
Interaction of nucleolin with ribosomal RNA genes and its role in RNA polymerase I transcription.
Cong R, Das S, Ugrinova I, Kumar S, Mongelard F, Wong J, Bouvet P.
Nucleic Acids Res. 2012 Oct;40(19):9441-54. doi: 10.1093/nar/gks720. Epub 2012 Aug 2.
PMID 22859736
 
Ribosome biogenesis and control of cell proliferation: p53 is not alone.
Donati G, Montanaro L, Derenzini M.
Cancer Res. 2012 Apr 1;72(7):1602-7. doi: 10.1158/0008-5472.CAN-11-3992. Epub 2012 Jan 26.
PMID 22282659
 
Inheritance of silent rDNA chromatin is mediated by PARP1 via noncoding RNA.
Guetg C, Scheifele F, Rosenthal F, Hottiger MO, Santoro R.
Mol Cell. 2012 Mar 30;45(6):790-800. doi: 10.1016/j.molcel.2012.01.024. Epub 2012 Mar 8.
PMID 22405650
 
The cellular abundance of the essential transcription termination factor TTF-I regulates ribosome biogenesis and is determined by MDM2 ubiquitinylation.
Lessard F, Stefanovsky V, Tremblay MG, Moss T.
Nucleic Acids Res. 2012 Jul;40(12):5357-67. doi: 10.1093/nar/gks198. Epub 2012 Mar 1.
PMID 22383580
 
The c-MYC oncoprotein, the NAMPT enzyme, the SIRT1-inhibitor DBC1, and the SIRT1 deacetylase form a positive feedback loop.
Menssen A, Hydbring P, Kapelle K, Vervoorts J, Diebold J, Luscher B, Larsson LG, Hermeking H.
Proc Natl Acad Sci U S A. 2012 Jan 24;109(4):E187-96. doi: 10.1073/pnas.1105304109. Epub 2011 Dec 21.
PMID 22190494
 
Changes in ribosome biogenesis may induce cancer by down-regulating the cell tumor suppressor potential.
Montanaro L, Trere D, Derenzini M.
Biochim Biophys Acta. 2012 Jan;1825(1):101-10. doi: 10.1016/j.bbcan.2011.10.006. Epub 2011 Nov 3. (REVIEW)
PMID 22079382
 
Histone methyltransferases regulating rRNA gene dose and dosage control in Arabidopsis.
Pontvianne F, Blevins T, Chandrasekhara C, Feng W, Stroud H, Jacobsen SE, Michaels SD, Pikaard CS.
Genes Dev. 2012 May 1;26(9):945-57. doi: 10.1101/gad.182865.111.
PMID 22549957
 
From sirtuin biology to human diseases: an update.
Sebastian C, Satterstrom FK, Haigis MC, Mostoslavsky R.
J Biol Chem. 2012 Dec 14;287(51):42444-52. doi: 10.1074/jbc.R112.402768. Epub 2012 Oct 18. (REVIEW)
PMID 23086954
 
Nucleoli: composition, function, and dynamics.
Shaw P, Brown J.
Plant Physiol. 2012 Jan;158(1):44-51. doi: 10.1104/pp.111.188052. Epub 2011 Nov 14. (REVIEW)
PMID 22082506
 
Functional proteomics establishes the interaction of SIRT7 with chromatin remodeling complexes and expands its role in regulation of RNA polymerase I transcription.
Tsai YC, Greco TM, Boonmee A, Miteva Y, Cristea IM.
Mol Cell Proteomics. 2012 May;11(5):60-76. doi: 10.1074/mcp.A111.015156.
PMID 22586326
 
Overexpression of ribosomal RNA in prostate cancer is common but not linked to rDNA promoter hypomethylation.
Uemura M, Zheng Q, Koh CM, Nelson WG, Yegnasubramanian S, De Marzo AM.
Oncogene. 2012 Mar 8;31(10):1254-63. doi: 10.1038/onc.2011.319. Epub 2011 Aug 8.
PMID 21822302
 
The chromatin remodeling complex NuRD establishes the poised state of rRNA genes characterized by bivalent histone modifications and altered nucleosome positions.
Xie W, Ling T, Zhou Y, Feng W, Zhu Q, Stunnenberg HG, Grummt I, Tao W.
Proc Natl Acad Sci U S A. 2012 May 22;109(21):8161-6. doi: 10.1073/pnas.1201262109. Epub 2012 May 8.
PMID 22570494
 
Dysregulation of RNA polymerase I transcription during disease.
Hannan KM, Sanij E, Rothblum LI, Hannan RD, Pearson RB.
Biochim Biophys Acta. 2013 Mar-Apr;1829(3-4):342-60. doi: 10.1016/j.bbagrm.2012.10.014. Epub 2012 Nov 12. (REVIEW)
PMID 23153826
 
The chromatin remodelling complex NoRC safeguards genome stability by heterochromatin formation at telomeres and centromeres.
Postepska-Igielska A1, Krunic D, Schmitt N, Greulich-Bode KM, Boukamp P, Grummt I.
EMBO Rep. 2013 Aug;14(8):704-10. doi: 10.1038/embor.2013.87. Epub 2013 Jun 25.
PMID 23797874
 
The chromatin remodeling factor CSB recruits histone acetyltransferase PCAF to rRNA gene promoters in active state for transcription initiation.
Shen M, Zhou T, Xie W, Ling T, Zhu Q, Zong L, Lyu G, Gao Q, Zhang F, Tao W.
PLoS One. 2013 May 7;8(5):e62668. doi: 10.1371/journal.pone.0062668. Print 2013.
PMID 23667505
 
A structural perspective on RNA polymerase I and RNA polymerase III transcription machineries.
Vannini A.
Biochim Biophys Acta. 2013 Mar-Apr;1829(3-4):258-64. doi: 10.1016/j.bbagrm.2012.09.009. Epub 2012 Sep 29. (REVIEW)
PMID 23031840
 
Zinc-finger protein 545 is a novel tumour suppressor that acts by inhibiting ribosomal RNA transcription in gastric cancer.
Wang S, Cheng Y, Du W, Lu L, Zhou L, Wang H, Kang W, Li X, Tao Q, Sung JJ, Yu J.
Gut. 2013 Jun;62(6):833-41. doi: 10.1136/gutjnl-2011-301776. Epub 2012 May 12.
PMID 22580414
 
Large-scale organization of ribosomal DNA chromatin is regulated by Tip5.
Zillner K, Filarsky M, Rachow K, Weinberger M, Langst G, Nemeth A.
Nucleic Acids Res. 2013 May 1;41(10):5251-62. doi: 10.1093/nar/gkt218. Epub 2013 Apr 10.
PMID 23580549
 
Construction of synthetic nucleoli in human cells reveals how a major functional nuclear domain is formed and propagated through cell division.
Grob A, Colleran C, McStay B.
Genes Dev. 2014 Feb 1;28(3):220-30. doi: 10.1101/gad.234591.113. Epub 2014 Jan 21.
PMID 24449107
 
Written2014-03Peter C McKeown
& Biotechnology Lab, Plant & AgriBiosciences Research Centre (PABC), School of Natural Sciences, National University of Ireland, Galway, Ireland

Citation

This paper should be referenced as such :
PC McKeown
Mechanisms of rDNA silencing, the Nucleolar Remodelling Complex (NoRC)
Atlas Genet Cytogenet Oncol Haematol. 2014;18(10):763-783.
Free journal version : [ pdf ]   [ DOI ]
On line version : http://AtlasGeneticsOncology.org/Deep/NoRCID20134.htm